1
|
Qiu NZ, Hou HM, Guo TY, Lv YL, Zhou Y, Zhang FF, Zhang F, Wang XD, Chen W, Gao YF, Chen MH, Zhang XH, Zhang HT, Wang H. Phosphodiesterase 8 (PDE8): Distribution and Cellular Expression and Association with Alzheimer's Disease. Neurochem Res 2024; 49:1993-2004. [PMID: 38782837 DOI: 10.1007/s11064-024-04156-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/19/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
Phosphodiesterase 8 (PDE8), as a member of PDE superfamily, specifically promotes the hydrolysis and degradation of intracellular cyclic adenosine monophosphate (cAMP), which may be associated with pathogenesis of Alzheimer's disease (AD). However, little is currently known about potential role in the central nervous system (CNS). Here we investigated the distribution and expression of PDE8 in brain of mouse, which we believe can provide evidence for studying the role of PDE8 in CNS and the relationship between PDE8 and AD. Here, C57BL/6J mice were used to observe the distribution patterns of two subtypes of PDE8, PDE8A and PDE8B, in different sexes in vivo by western blot (WB). Meanwhile, C57BL/6J mice were also used to demonstrate the distribution pattern of PDE8 in selected brain regions and localization in neural cells by WB and multiplex immunofluorescence staining. Furthermore, the triple transgenic (3×Tg-AD) mice and wild type (WT) mice of different ages were used to investigate the changes of PDE8 expression in the hippocampus and cerebral cortex during the progression of AD. PDE8 was found to be widely expressed in multiple tissues and organs including heart, kidney, stomach, brain, and liver, spleen, intestines, and uterus, with differences in expression levels between the two subtypes of PDE8A and PDE8B, as well as two sexes. Meanwhile, PDE8 was widely distributed in the brain, especially in areas closely related to cognitive function such as cerebellum, striatum, amygdala, cerebral cortex, and hippocampus, without differences between sexes. Furthermore, PDE8A was found to be expressed in neuronal cells, microglia and astrocytes, while PDE8B is only expressed in neuronal cells and microglia. PDE8A expression in the hippocampus of both female and male 3×Tg-AD mice was gradually increased with ages and PDE8B expression was upregulated only in cerebral cortex of female 3×Tg-AD mice with ages. However, the expression of PDE8A and PDE8B was apparently increased in both cerebral cortex and hippocampus in both female and male 10-month-old 3×Tg-AD mice compared WT mice. These results suggest that PDE8 may be associated with the progression of AD and is a potential target for its prevention and treatment in the future.
Collapse
Affiliation(s)
- Nian-Zhuang Qiu
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, China
| | - Hui-Mei Hou
- Development Planning and Discipline Construction Department, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, China
| | - Tian-Yang Guo
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, China
| | - Yu-Li Lv
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, China
| | - Yao Zhou
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, China
| | - Fang-Fang Zhang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, China
| | - Feng Zhang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, China
| | - Xiao-Dan Wang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, China
| | - Wei Chen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, China
| | - Yong-Feng Gao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, China
| | - Mei-Hua Chen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, China
| | - Xue-Hui Zhang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, China.
| | - Han-Ting Zhang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, China.
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, 266073, Shandong, China.
| | - Hao Wang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, China.
| |
Collapse
|
2
|
Gong L, Liu D, Zhang B, Yu S, Xi C. Sex-Specific Entorhinal Cortex Functional Connectivity in Cognitively Normal Older Adults with Amyloid-β Pathology. Mol Neurobiol 2024:10.1007/s12035-024-04243-z. [PMID: 38867110 DOI: 10.1007/s12035-024-04243-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 05/06/2024] [Indexed: 06/14/2024]
Abstract
Sex and apolipoprotein E (APOE) genotype have been shown to influence the risk and progression of Alzheimer's disease (AD). However, the impact of these factors on the functional connectivity of the entorhinal cortex (ERC) in clinically unpaired older adults (CUOA) with amyloid-β (Aβ +) pathology remains unclear. A total of 1022 cognitively normal older adults with Aβ + (603 females and 586 APOE ε4 +) from the Anti-Amyloid Treatment in Asymptomatic Alzheimer's (A4) study were included in this study. The 2 × 2 (gender, 2 APOE genotypes) analysis of covariance was performed to compare the demographic information, cognitive performance, and volumetric MRI data among these groups. Voxel-wise comparisons of bilateral ERC functional connectivity (FC) were conducted, and partial correlation analyses were used to explore the associations between cognitive performance and ERC-FC strength. We found that the APOE genotype influenced ERC functional connectivity mainly in the sensorimotor network (SMN). Males exhibited higher ERC-FC in the salience network (SN), while females displayed higher ERC-FC in the default mode network (DMN), executive control network (ECN), and reward network. The interplay of sex and APOE genotype on ERC-FC was observed in the SMN and cerebellar lobe. The ERC-FC was associated with executive function and memory performance in individuals with CUOA-Aβ + . Our findings provide evidence of sex-specific ERC functional connectivity compensation mechanism in cognitively normal older adults with Aβ + pathology. This study may contribute to a better understanding of the mechanisms underlying the early stages of AD and may help develop personalized interventions in preclinical AD.
Collapse
Affiliation(s)
- Liang Gong
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, 610017, Sichuan, China
| | - Duan Liu
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, 610017, Sichuan, China
| | - Bei Zhang
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, 610017, Sichuan, China
| | - Siyi Yu
- Department of Acupuncture & Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China.
| | - Chunhua Xi
- Department of Neurology, The Third Affiliated Hospital of Anhui Medical University, Huaihe Road 390, Heifei, 230061, Anhui, China.
| |
Collapse
|
3
|
Chen YH, Wang ZB, Liu XP, Xu JP, Mao ZQ. Sex differences in the relationship between depression and Alzheimer's disease-mechanisms, genetics, and therapeutic opportunities. Front Aging Neurosci 2024; 16:1301854. [PMID: 38903903 PMCID: PMC11188317 DOI: 10.3389/fnagi.2024.1301854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 04/25/2024] [Indexed: 06/22/2024] Open
Abstract
Depression and Alzheimer's disease (AD) are prevalent neuropsychiatric disorders with intriguing epidemiological overlaps. Their interrelation has recently garnered widespread attention. Empirical evidence indicates that depressive disorders significantly contribute to AD risk, and approximately a quarter of AD patients have comorbid major depressive disorder, which underscores the bidirectional link between AD and depression. A growing body of evidence substantiates pervasive sex differences in both AD and depression: both conditions exhibit a higher incidence among women than among men. However, the available literature on this topic is somewhat fragmented, with no comprehensive review that delineates sex disparities in the depression-AD correlation. In this review, we bridge these gaps by summarizing recent progress in understanding sex-based differences in mechanisms, genetics, and therapeutic prospects for depression and AD. Additionally, we outline key challenges in the field, holding potential for improving treatment precision and efficacy tailored to male and female patients' distinct needs.
Collapse
Affiliation(s)
- Yu-Han Chen
- The First Clinical Medical School, Hebei North University, Zhangjiakou, China
| | - Zhi-Bo Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Xi-Peng Liu
- Department of Neurosurgery, The First Affiliated Hospital of Hebei North, Zhangjiakou, China
| | - Jun-Peng Xu
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhi-Qi Mao
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
4
|
de Vries LE, Jongejan A, Monteiro Fortes J, Balesar R, Rozemuller AJM, Moerland PD, Huitinga I, Swaab DF, Verhaagen J. Gene-expression profiling of individuals resilient to Alzheimer's disease reveals higher expression of genes related to metallothionein and mitochondrial processes and no changes in the unfolded protein response. Acta Neuropathol Commun 2024; 12:68. [PMID: 38664739 PMCID: PMC11046840 DOI: 10.1186/s40478-024-01760-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/10/2024] [Indexed: 04/28/2024] Open
Abstract
Some individuals show a discrepancy between cognition and the amount of neuropathological changes characteristic for Alzheimer's disease (AD). This phenomenon has been referred to as 'resilience'. The molecular and cellular underpinnings of resilience remain poorly understood. To obtain an unbiased understanding of the molecular changes underlying resilience, we investigated global changes in gene expression in the superior frontal gyrus of a cohort of cognitively and pathologically well-defined AD patients, resilient individuals and age-matched controls (n = 11-12 per group). 897 genes were significantly altered between AD and control, 1121 between resilient and control and 6 between resilient and AD. Gene set enrichment analysis (GSEA) revealed that the expression of metallothionein (MT) and of genes related to mitochondrial processes was higher in the resilient donors. Weighted gene co-expression network analysis (WGCNA) identified gene modules related to the unfolded protein response, mitochondrial processes and synaptic signaling to be differentially associated with resilience or dementia. As changes in MT, mitochondria, heat shock proteins and the unfolded protein response (UPR) were the most pronounced changes in the GSEA and/or WGCNA, immunohistochemistry was used to further validate these processes. MT was significantly increased in astrocytes in resilient individuals. A higher proportion of the mitochondrial gene MT-CO1 was detected outside the cell body versus inside the cell body in the resilient compared to the control group and there were higher levels of heat shock protein 70 (HSP70) and X-box-binding protein 1 spliced (XBP1s), two proteins related to heat shock proteins and the UPR, in the AD donors. Finally, we show evidence for putative sex-specific alterations in resilience, including gene expression differences related to autophagy in females compared to males. Taken together, these results show possible mechanisms involving MTs, mitochondrial processes and the UPR by which individuals might maintain cognition despite the presence of AD pathology.
Collapse
Affiliation(s)
- Luuk E de Vries
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands.
| | - Aldo Jongejan
- Amsterdam UMC Location University of Amsterdam, Epidemiology and Data Science, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Public Health, Methodology, Amsterdam, The Netherlands
- Amsterdam Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands
| | - Jennifer Monteiro Fortes
- Department of Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | - Rawien Balesar
- Department of Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | - Annemieke J M Rozemuller
- Department of Pathology, Amsterdam Neuroscience, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | - Perry D Moerland
- Amsterdam UMC Location University of Amsterdam, Epidemiology and Data Science, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Public Health, Methodology, Amsterdam, The Netherlands
- Amsterdam Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands
| | - Inge Huitinga
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Dick F Swaab
- Department of Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | - Joost Verhaagen
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands.
- Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
5
|
de Vries LE, Huitinga I, Kessels HW, Swaab DF, Verhaagen J. The concept of resilience to Alzheimer's Disease: current definitions and cellular and molecular mechanisms. Mol Neurodegener 2024; 19:33. [PMID: 38589893 PMCID: PMC11003087 DOI: 10.1186/s13024-024-00719-7] [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: 08/23/2023] [Accepted: 03/20/2024] [Indexed: 04/10/2024] Open
Abstract
Some individuals are able to maintain their cognitive abilities despite the presence of significant Alzheimer's Disease (AD) neuropathological changes. This discrepancy between cognition and pathology has been labeled as resilience and has evolved into a widely debated concept. External factors such as cognitive stimulation are associated with resilience to AD, but the exact cellular and molecular underpinnings are not completely understood. In this review, we discuss the current definitions used in the field, highlight the translational approaches used to investigate resilience to AD and summarize the underlying cellular and molecular substrates of resilience that have been derived from human and animal studies, which have received more and more attention in the last few years. From these studies the picture emerges that resilient individuals are different from AD patients in terms of specific pathological species and their cellular reaction to AD pathology, which possibly helps to maintain cognition up to a certain tipping point. Studying these rare resilient individuals can be of great importance as it could pave the way to novel therapeutic avenues for AD.
Collapse
Affiliation(s)
- Luuk E de Vries
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands.
| | - Inge Huitinga
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands
| | - Helmut W Kessels
- Swammerdam Institute for Life Sciences, Amsterdam Neuroscience, University of Amsterdam, 1098 XH, Amsterdam, the Netherlands
| | - Dick F Swaab
- Department of Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, Netherlands
| | - Joost Verhaagen
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
6
|
Ali M, Garcia P, Lunkes LP, Sciortino A, Thomas M, Heurtaux T, Grzyb K, Halder R, Coowar D, Skupin A, Buée L, Blum D, Buttini M, Glaab E. Single cell transcriptome analysis of the THY-Tau22 mouse model of Alzheimer's disease reveals sex-dependent dysregulations. Cell Death Discov 2024; 10:119. [PMID: 38453894 PMCID: PMC10920792 DOI: 10.1038/s41420-024-01885-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/09/2024] Open
Abstract
Alzheimer's disease (AD) progression and pathology show pronounced sex differences, but the factors driving these remain poorly understood. To gain insights into early AD-associated molecular changes and their sex dependency for tau pathology in the cortex, we performed single-cell RNA-seq in the THY-Tau22 AD mouse model. By examining cell type-specific and cell type-agnostic AD-related gene activity changes and their sex-dimorphism for individual genes, pathways and cellular sub-networks, we identified both statistically significant alterations and interpreted the upstream mechanisms controlling them. Our results confirm several significant sex-dependent alterations in gene activity in the THY-Tau22 model mice compared to controls, with more pronounced alterations in females. Both changes shared across multiple cell types and cell type-specific changes were observed. The differential genes showed significant over-representation of known AD-relevant processes, such as pathways associated with neuronal differentiation, programmed cell death and inflammatory responses. Regulatory network analysis of these genes revealed upstream regulators that modulate many of the downstream targets with sex-dependent changes. Most key regulators have been previously implicated in AD, such as Egr1, Klf4, Chchd2, complement system genes, and myelin-associated glycoproteins. Comparing with similar data from the Tg2576 AD mouse model and human AD patients, we identified multiple genes with consistent, cell type-specific and sex-dependent alterations across all three datasets. These shared changes were particularly evident in the expression of myelin-associated genes such as Mbp and Plp1 in oligodendrocytes. In summary, we observed significant cell type-specific transcriptomic changes in the THY-Tau22 mouse model, with a strong over-representation of known AD-associated genes and processes. These include both sex-neutral and sex-specific patterns, characterized by consistent shifts in upstream master regulators and downstream target genes. Collectively, these findings provide insights into mechanisms influencing sex-specific susceptibility to AD and reveal key regulatory proteins that could be targeted for developing treatments addressing sex-dependent AD pathology.
Collapse
Affiliation(s)
- Muhammad Ali
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Pierre Garcia
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Laetitia P Lunkes
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Alessia Sciortino
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Melanie Thomas
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Tony Heurtaux
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, 8 avenue du Swing, L-4367, Belvaux, Luxembourg
- Luxembourg Center of Neuropathology, L-3555, Dudelange, Luxembourg
| | - Kamil Grzyb
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Rashi Halder
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Djalil Coowar
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Alex Skupin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Luc Buée
- University of Lille, Inserm, CHU Lille, UMR-S1172 Lille Neuroscience & Cognition (LilNCog), Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - David Blum
- University of Lille, Inserm, CHU Lille, UMR-S1172 Lille Neuroscience & Cognition (LilNCog), Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Manuel Buttini
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
7
|
Lynch MA. A case for seeking sex-specific treatments in Alzheimer's disease. Front Aging Neurosci 2024; 16:1346621. [PMID: 38414633 PMCID: PMC10897030 DOI: 10.3389/fnagi.2024.1346621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/15/2024] [Indexed: 02/29/2024] Open
Abstract
There is no satisfactory explanation for the sex-related differences in the incidence of many diseases and this is also true of Alzheimer's disease (AD), where females have a higher lifetime risk of developing the disease and make up about two thirds of the AD patient population. The importance of understanding the cause(s) that account for this disproportionate distribution cannot be overestimated, and is likely to be a significant factor in the search for therapeutic strategies that will combat the disease and, furthermore, potentially point to a sex-targeted approach to treatment. This review considers the literature in the context of what is known about the impact of sex on processes targeted by drugs that are in clinical trial for AD, and existing knowledge on differing responses of males and females to these drugs. Current knowledge strongly supports the view that trials should make assessing sex-related difference in responses a priority with a focus on exploring the sex-stratified treatments.
Collapse
|
8
|
da Silva SP, de Castro CCM, Rabelo LN, Engelberth RC, Fernández-Calvo B, Fiuza FP. Neuropathological and sociodemographic factors associated with the cortical amyloid load in aging and Alzheimer's disease. GeroScience 2024; 46:621-643. [PMID: 37870702 PMCID: PMC10828279 DOI: 10.1007/s11357-023-00982-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/10/2023] [Indexed: 10/24/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia and is characterized by a progressive decline in cognitive abilities. A pathological hallmark of AD is a region-specific accumulation of the amyloid-beta protein (Aβ). Here, we explored the association between regional Aβ deposition, sociodemographic, and local biochemical factors. We quantified the Aβ burden in postmortem cortical samples from parietal (PCx) and temporal (TCx) regions of 27 cognitively unimpaired (CU) and 15 AD donors, aged 78-100 + years. Histological images of Aβ immunohistochemistry and local concentrations of pathological and inflammatory proteins were obtained at the "Aging, Dementia and TBI Study" open database. We used the area fraction fractionator stereological methodology to quantify the Aβ burden in the gray and white matter within each cortical region. We found higher Aβ burdens in the TCx of AD octogenarians compared to CU ones. We also found higher Aβ loads in the PCx of AD nonagenarians than in AD octogenarians. Moreover, AD women exhibited increased Aβ deposition compared to CU women. Interestingly, we observed a negative correlation between education years and Aβ burden in the white matter of both cortices in CU samples. In AD brains, the Aβ40, Aβ42, and pTau181 isoforms of Aβ and Tau proteins were positively correlated with the Aβ burden. Additionally, in the TCx of AD donors, the proinflammatory cytokine TNFα showed a positive correlation with the Aβ load. These novel findings contribute to understanding the interplay between sociodemographic characteristics, local inflammatory signaling, and the development of AD-related pathology in the cerebral cortex.
Collapse
Affiliation(s)
- Sayonara P da Silva
- Graduate Program in Neuroengineering, Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, RN, 59280-000, Brazil
| | - Carla C M de Castro
- Graduate Program in Neuroengineering, Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, RN, 59280-000, Brazil
| | - Lívia N Rabelo
- Graduate Program in Neuroengineering, Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, RN, 59280-000, Brazil
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande Do Norte, Natal, Brazil
| | - Rovena C Engelberth
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande Do Norte, Natal, Brazil
| | - Bernardino Fernández-Calvo
- Department of Psychology, University of Córdoba, Córdoba, Spain
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- Department of Psychology, Federal University of Paraíba, João Pessoa, Brazil
| | - Felipe P Fiuza
- Graduate Program in Neuroengineering, Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, RN, 59280-000, Brazil.
| |
Collapse
|
9
|
Hou Y, Caldwell JZK, Lathia JD, Leverenz JB, Pieper AA, Cummings J, Cheng F. Microglial immunometabolism endophenotypes contribute to sex difference in Alzheimer's disease. Alzheimers Dement 2024; 20:1334-1349. [PMID: 37985399 PMCID: PMC10916937 DOI: 10.1002/alz.13546] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/20/2023] [Accepted: 10/05/2023] [Indexed: 11/22/2023]
Abstract
INTRODUCTION The molecular mechanisms that contribute to sex differences, in particular female predominance, in Alzheimer's disease (AD) prevalence, symptomology, and pathology, are incompletely understood. METHODS To address this problem, we investigated cellular metabolism and immune responses ("immunometabolism endophenotype") across AD individuals as a function of sex with diverse clinical diagnosis of cognitive status at death (cogdx), Braak staging, and Consortium to Establish a Registry for AD (CERAD) scores using human cortex metabolomics and transcriptomics data from the Religious Orders Study / Memory and Aging Project (ROSMAP) cohort. RESULTS We identified sex-specific metabolites, immune and metabolic genes, and pathways associated with the AD diagnosis and progression. We identified female-specific elevation in glycerophosphorylcholine and N-acetylglutamate, which are AD inflammatory metabolites involved in interleukin (IL)-17 signaling, C-type lectin receptor, interferon signaling, and Toll-like receptor pathways. We pinpointed distinct microglia-specific immunometabolism endophenotypes (i.e., lipid- and amino acid-specific IL-10 and IL-17 signaling pathways) between female and male AD subjects. In addition, female AD subjects showed evidence of diminished excitatory neuron and microglia communications via glutamate-mediated immunometabolism. DISCUSSION Our results point to new understanding of the molecular basis for female predominance in AD, and warrant future independent validations with ethnically diverse patient cohorts to establish a likely causal relationship of microglial immunometabolism in the sex differences in AD. HIGHLIGHTS Sex-specific immune metabolites, gene networks and pathways, are associated with Alzheimer's disease pathogenesis and disease progression. Female AD subjects exhibit microglial immunometabolism endophenotypes characterized by decreased glutamate metabolism and elevated interleukin-10 pathway activity. Female AD subjects showed a shift in glutamate-mediated cell-cell communications between excitatory neurons to microglia and astrocyte.
Collapse
Affiliation(s)
- Yuan Hou
- Genomic Medicine InstituteCleveland ClinicLerner Research InstituteClevelandOhioUSA
| | - Jessica Z. K. Caldwell
- Department of Molecular MedicineCleveland Clinic Lerner College of MedicineCase Western Reserve UniversityClevelandOhioUSA
- Lou Ruvo Center for Brain HealthCleveland ClinicNeurological InstituteLas VegasNevadaUSA
| | - Justin D. Lathia
- Department of Molecular MedicineCleveland Clinic Lerner College of MedicineCase Western Reserve UniversityClevelandOhioUSA
- Department of Cardiovascular & Metabolic ScienceCleveland ClinicLerner Research InstituteClevelandOhioUSA
| | - James B. Leverenz
- Lou Ruvo Center for Brain HealthCleveland ClinicNeurological InstituteClevelandOhioUSA
| | - Andrew A. Pieper
- Brain Health Medicines CenterHarrington Discovery InstituteUniversity Hospitals Cleveland Medical CenterClevelandOhioUSA
- Department of PsychiatryCase Western Reserve UniversityClevelandOhioUSA
- Geriatric PsychiatryGRECCLouis Stokes Cleveland VA Medical CenterClevelandOhioUSA
- Institute for Transformative Molecular MedicineSchool of MedicineCase Western Reserve UniversityClevelandOhioUSA
- Department of NeurosciencesSchool of MedicineCase Western Reserve UniversityClevelandOhioUSA
- Department of PathologySchool of MedicineCase Western Reserve UniversityClevelandOhioUSA
| | - Jeffrey Cummings
- Chambers‐Grundy Center for Transformative NeuroscienceDepartment of Brain HealthSchool of Integrated Health SciencesUniversity of Nevada Las VegasLas VegasNevadaUSA
| | - Feixiong Cheng
- Genomic Medicine InstituteCleveland ClinicLerner Research InstituteClevelandOhioUSA
- Department of Molecular MedicineCleveland Clinic Lerner College of MedicineCase Western Reserve UniversityClevelandOhioUSA
| |
Collapse
|
10
|
Perluigi M, Di Domenico F, Butterfield DA. Oxidative damage in neurodegeneration: roles in the pathogenesis and progression of Alzheimer disease. Physiol Rev 2024; 104:103-197. [PMID: 37843394 PMCID: PMC11281823 DOI: 10.1152/physrev.00030.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/30/2023] [Accepted: 05/24/2023] [Indexed: 10/17/2023] Open
Abstract
Alzheimer disease (AD) is associated with multiple etiologies and pathological mechanisms, among which oxidative stress (OS) appears as a major determinant. Intriguingly, OS arises in various pathways regulating brain functions, and it seems to link different hypotheses and mechanisms of AD neuropathology with high fidelity. The brain is particularly vulnerable to oxidative damage, mainly because of its unique lipid composition, resulting in an amplified cascade of redox reactions that target several cellular components/functions ultimately leading to neurodegeneration. The present review highlights the "OS hypothesis of AD," including amyloid beta-peptide-associated mechanisms, the role of lipid and protein oxidation unraveled by redox proteomics, and the antioxidant strategies that have been investigated to modulate the progression of AD. Collected studies from our groups and others have contributed to unraveling the close relationships between perturbation of redox homeostasis in the brain and AD neuropathology by elucidating redox-regulated events potentially involved in both the pathogenesis and progression of AD. However, the complexity of AD pathological mechanisms requires an in-depth understanding of several major intracellular pathways affecting redox homeostasis and relevant for brain functions. This understanding is crucial to developing pharmacological strategies targeting OS-mediated toxicity that may potentially contribute to slow AD progression as well as improve the quality of life of persons with this severe dementing disorder.
Collapse
Affiliation(s)
- Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
11
|
Chang K, Ling JP, Redding-Ochoa J, An Y, Li L, Dean SA, Blanchard TG, Pylyukh T, Barrett A, Irwin KE, Moghekar A, Resnick SM, Wong PC, Troncoso JC. Loss of TDP-43 splicing repression occurs early in the aging population and is associated with Alzheimer's disease neuropathologic changes and cognitive decline. Acta Neuropathol 2023; 147:4. [PMID: 38133681 DOI: 10.1007/s00401-023-02653-2] [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: 08/25/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 12/23/2023]
Abstract
LATE-NC, the neuropathologic changes of limbic-predominant age-related TAR DNA-binding protein 43 kDa (TDP-43) encephalopathy are frequently associated with Alzheimer's disease (AD) and cognitive impairment in older adults. The association of TDP-43 proteinopathy with AD neuropathologic changes (ADNC) and its impact on specific cognitive domains are not fully understood and whether loss of TDP-43 function occurs early in the aging brain remains unknown. Here, using a large set of autopsies from the Baltimore Longitudinal Study of Aging (BLSA) and another younger cohort, we were able to study brains from subjects 21-109 years of age. Examination of these brains show that loss of TDP-43 splicing repression, as judged by TDP-43 nuclear clearance and expression of a cryptic exon in HDGFL2, first occurs during the 6th decade, preceding by a decade the appearance of TDP-43+ neuronal cytoplasmic inclusions (NCIs). We corroborated this observation using a monoclonal antibody to demonstrate a cryptic exon-encoded neoepitope within HDGFL2 in neurons exhibiting nuclear clearance of TDP-43. TDP-43 nuclear clearance is associated with increased burden of tau pathology. Age at death, female sex, high CERAD neuritic plaque score, and high Braak neurofibrillary stage significantly increase the odds of LATE-NC. Faster rates of cognitive decline on verbal memory (California Verbal Learning Test immediate recall), visuospatial ability (Card Rotations Test), mental status (MMSE) and semantic fluency (Category Fluency Test) were associated with LATE-NC. Notably, the effects of LATE-NC on verbal memory and visuospatial ability are independent of ADNC. However, the effects of TDP-43 nuclear clearance in absence of NCI on the longitudinal trajectories and levels of cognitive measures are not significant. These results establish that loss of TDP-43 splicing repression is an early event occurring in the aging population during the development of TDP-43 proteinopathy and is associated with increased tau pathology. Furthermore, LATE-NC correlates with high levels of ADNC but also has an impact on specific memory and visuospatial functions in aging that is independent of AD.
Collapse
Affiliation(s)
- Koping Chang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department and Graduate Institute of Pathology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, 100225, Taiwan
| | - Jonathan P Ling
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Javier Redding-Ochoa
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Ling Li
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Office of the Chief Medical Examiner, State of Maryland, Baltimore, MD, 21223, USA
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Stephanie A Dean
- Office of the Chief Medical Examiner, State of Maryland, Baltimore, MD, 21223, USA
| | - Thomas G Blanchard
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Tatiana Pylyukh
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Alexander Barrett
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Katherine E Irwin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Philip C Wong
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Juan C Troncoso
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
12
|
Zhang Y, Lu J, Wang M, Zuo C, Jiang J. Influence of Gender on Tau Precipitation in Alzheimer's Disease According to ATN Research Framework. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:565-575. [PMID: 38223687 PMCID: PMC10781910 DOI: 10.1007/s43657-022-00076-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 08/26/2022] [Accepted: 09/01/2022] [Indexed: 01/16/2024]
Abstract
Tau proteins accumulation and their spreading pattern were affected by gender in cognitive impairment patients, especially in the progression of Alzheimer's disease (AD). However, it was unclear whether the gender effects for tau deposition influenced by amyloid deposition. The aim of this study was to investigate gender differences for tau depositions in Aβ positive (A+) subjects. In this study, tau and amyloid positron emission tomography images, structural magnetic resonance imaging images, and demographic information were collected from 179 subjects in Alzheimer's Disease Neuroimaging Initiative (ADNI) database and 63 subjects from Huashan Hospital. Subjects were classified as T+ or T- according to the presence or absence of tau (T) biomarkers. We used two-sample t test and one-way analysis of variance test to analyze the effect of gender with adjusting for age, years of education, and Minimum Mental State Examination. In the ADNI cohort, we found differences in Tau deposition in fusiform gyrus, inferior temporal gyrus, middle temporal gyrus and parahippocampal gyrus between the female T+ (FT+) and male T+ (MT+) groups (p < 0.05). Tau deposition did not differ significantly between female T- (FT-) and male T- (MT-) subjects (p > 0.05). In the Huashan Hospital cohort, there was no difference in Tau deposition between FT+ and MT+ (p > 0.05). The results show that tau depositions significantly increased in females in above brain regions. Our findings suggest that tau deposition is influenced by gender in the A+ subjects. This result has important clinical implications for the development of gender-guided early interventions for patients with both Tau and Amyloid depositions. Supplementary Information The online version contains supplementary material available at 10.1007/s43657-022-00076-9.
Collapse
Affiliation(s)
- Ying Zhang
- School of Communication and Information Engineering, Shanghai University, Shanghai, 200444 China
| | - Jiaying Lu
- PET Center and National Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, 201206 China
| | - Min Wang
- School of Communication and Information Engineering, Shanghai University, Shanghai, 200444 China
| | - Chuantao Zuo
- PET Center and National Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, 201206 China
| | - Jiehui Jiang
- Institute of Biomedical Engineering, School of Life Science, Shanghai University, Shanghai, 200444 China
| |
Collapse
|
13
|
Chen XL, Fortes JM, Hu YT, van Iersel J, He KN, van Heerikhuize J, Balesar R, Swaab D, Bao AM. Sexually dimorphic age-related molecular differences in the entorhinal cortex of cognitively intact elderly: Relation to early Alzheimer's changes. Alzheimers Dement 2023; 19:3848-3857. [PMID: 36960685 DOI: 10.1002/alz.13037] [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: 10/18/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 03/25/2023]
Abstract
INTRODUCTION Women are more vulnerable to Alzheimer's disease (AD) than men. The entorhinal cortex (EC) is one of the earliest structures affected in AD. We identified in cognitively intact elderly different molecular changes in the EC in relation to age. METHODS Changes in 12 characteristic molecules in relation to age were determined by quantitative immunohistochemistry or in situ hybridization in the EC. They were arbitrarily grouped into sex steroid-related molecules, markers of neuronal activity, neurotransmitter-related molecules, and cholinergic activity-related molecules. RESULTS The changes in molecules indicated increasing local estrogenic and neuronal activity accompanied by a higher and faster hyperphosphorylated tau accumulation in women's EC in relation to age, versus a mainly stable local estrogenic/androgenic and neuronal activity in men's EC. DISCUSSION EC employs a different neurobiological strategy in women and men to maintain cognitive function, which seems to be accompanied by an earlier start of AD in women. HIGHLIGHTS Local estrogen system is activated with age only in women's entorhinal cortex (EC). EC neuronal activity increased with age only in elderly women with intact cognition. Men and women have different molecular strategies to retain cognition with aging. P-tau accumulation in the EC was higher and faster in cognitively intact elderly women.
Collapse
Affiliation(s)
- Xin-Lu Chen
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Jennifer Monteiro Fortes
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Yu-Ting Hu
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Juliet van Iersel
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Kang-Ning He
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Joop van Heerikhuize
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Rawien Balesar
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Dick Swaab
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Ai-Min Bao
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
14
|
Bayram E, Coughlin DG, Litvan I. Sex Differences for Clinical Correlates of Alzheimer's Pathology in People with Lewy Body Pathology. Mov Disord 2022; 37:1505-1515. [PMID: 35531707 PMCID: PMC9308759 DOI: 10.1002/mds.29044] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Lewy body (LB) dementias have limited clinical diagnostic accuracy because of frequent copathologies contributing to clinical heterogeneity. Although sex differences in clinical prevalence and frequency of pure LB pathology were shown, differences for clinicopathological correlations are less known. OBJECTIVE The aim of this study was to determine sex differences for clinical associations of Alzheimer's disease (AD) copathology in those with LB pathology. METHODS Data were from National Alzheimer's Coordinating Center for 223 women and 468 men with limbic or neocortical LB, separated into two groups as those with high likelihood and low/intermediate likelihood for LB clinical phenotype based on pathology. Clinical associations of sex and interaction of sex and pathology for the clinical phenotype were analyzed. RESULTS More severe AD copathology was associated with worse cognitive decline and lower likelihood of LB disease clinical phenotype. Women with more severe AD copathology and tau had worse cognitive decline and higher likelihood of AD clinical phenotype than men. Men with more severe AD copathology had lower likelihood of LB clinical phenotype than women. Interaction of sex and pathology was more pronounced in those aged between 70 and 80 years. CONCLUSIONS AD copathology reduces the likelihood of LB clinical phenotype for both women and men; however, men may be at higher risk for LB disease underdiagnosis and women at higher risk for dementia. The use of both LB and AD biomarkers, even when LB or AD pathology is not clinically expected, is necessary for the accurate clinical diagnosis of both LB diseases and AD. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Ece Bayram
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego
| | - David G. Coughlin
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego
| | - Irene Litvan
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego
| |
Collapse
|
15
|
Mast N, Petrov AM, Prendergast E, Bederman I, Pikuleva IA. Brain Acetyl-CoA Production and Phosphorylation of Cytoskeletal Proteins Are Targets of CYP46A1 Activity Modulation and Altered Sterol Flux. Neurotherapeutics 2021; 18:2040-2060. [PMID: 34235635 PMCID: PMC8609074 DOI: 10.1007/s13311-021-01079-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2021] [Indexed: 02/04/2023] Open
Abstract
Cholesterol and 24-hydroxycholesterol are the most abundant brain sterols and represent the substrate and product, respectively, of cytochrome P450 46A1 (CYP46A1), a CNS-specific enzyme. CYP46A1 controls cholesterol elimination and turnover in the brain, the two processes that determine the rate of brain sterol flux through the plasma membranes and thereby the properties of these membranes. Brain sterol flux is decreased in Cyp46a1-/- mice compared to wild-type mice and increased in 5XFAD mice (a model of Alzheimer's disease) when they are treated with a small dose of efavirenz, a CYP46A1 activator. Herein, we first assessed the brain proteome (synaptosomal fractions) and phospho-proteome (synaptosomal fractions and brain homogenates) of efavirenz-treated and control 5XFAD mice. Then, based on the pattern of protein abundance change, we conducted acetyl-CoA measurements (brain homogenates and mitochondria) and metabolic profiling (brain homogenates). The phospho-proteomics datasets were used for comparative analyses with the datasets obtained by us previously on mice with the same changes (efavirenz-treated and control 5XFAD mice from a different treatment paradigm) or with changes in the opposite direction (Cyp46a1-/- vs wild-type mice) in brain sterol flux. We found that CYP46A1 activity or the rate of brain sterol flux affects acetyl-CoA-related metabolic pathways as well as phosphorylation of cytoskeletal and other proteins. Knowledge of the key roles of acetyl-CoA and cytoskeletal phosphorylation in cell biology expands our understanding of the significance of CYP46A1-mediated cholesterol 24-hydroxylation in the brain and provides an additional explanation for why CYP46A1 activity modulations are beneficial in mouse models of different brain diseases.
Collapse
Affiliation(s)
- Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Alexey M Petrov
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center, Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, 420111, Kazan, Russia
- Institute of Neuroscience, Kazan State Medial University, 49 Butlerova Street, 420012, Kazan, Russia
| | - Erin Prendergast
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Ilya Bederman
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|