1
|
Morrison L, Dyer AH, Dolphin H, Batten I, Reddy C, Widdowson M, Woods CP, Gibney J, Bourke NM, Kennelly SP. Circulating Interleukin-17A is associated with executive function in middle aged adults with and without type 2 diabetes. Brain Behav Immun Health 2024; 41:100862. [PMID: 39350951 PMCID: PMC11440310 DOI: 10.1016/j.bbih.2024.100862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/19/2024] [Accepted: 09/14/2024] [Indexed: 10/04/2024] Open
Abstract
Midlife cardiovascular risk factors such as Type 2 Diabetes (T2DM) and obesity are associated with the later development of cognitive impairment and dementia. Systemic inflammation is postulated as a crucial mechanism, yet there are few studies examining this at the earliest stages prior to overt cognitive impairment. To assess this, we recruited a cohort of middle-aged cognitively-unimpaired individuals with and without uncomplicated T2DM. Comprehensive neuropsychological assessment was performed at baseline and at 4-year follow-up. Ten serum chemokines and cytokines (Eotaxin, MCP-1, MIP-1β, CXCL10, IL-6, IL-10, IL12p70, IL-17A, IFN-γ and TNF-α) were measured at both baseline and follow-up using high-sensitivity assays. Overall, 136 participants were recruited including 90 with uncomplicated midlife T2DM (age 52.6 ± 8.3; 47% female) and 46 without (age 52.9 ± 8.03; 61% female). Cognitive trajectories were stable over time and did not differ with T2DM. Yet on cross-sectional analyses at both baseline and follow-up, greater circulating IL-17A was consistently associated with poorer performance on tests of executive function/attention (β: 0.21; -0.40, -0.02, p = 0.03 at baseline; β: 0.26; -0.46, -0.05, p = 0.02 at follow-up). Associations persisted on covariate adjustment and did not differ by T2DM status. In summary, we provide evidence that greater circulating IL-17A levels were associated with poorer executive function in midlife, independent of T2DM. Long-term follow-up of this and other cohorts will further elucidate the earliest stages in the relationship between systemic inflammation and cognitive decline to provide further mechanistic insights and potentially identify those at greatest risk for later cognitive decline.
Collapse
Affiliation(s)
- Laura Morrison
- Tallaght Institute for Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Ireland
| | - Adam H Dyer
- Tallaght Institute for Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Ireland
| | - Helena Dolphin
- Tallaght Institute for Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Ireland
| | - Isabella Batten
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Ireland
| | - Conor Reddy
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland
| | - Matthew Widdowson
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, Ireland
| | - Conor P Woods
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, Ireland
| | - James Gibney
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, Ireland
| | - Nollaig M Bourke
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Ireland
| | - Sean P Kennelly
- Tallaght Institute for Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Ireland
| |
Collapse
|
2
|
Geloso MC, Zupo L, Corvino V. Crosstalk between peripheral inflammation and brain: Focus on the responses of microglia and astrocytes to peripheral challenge. Neurochem Int 2024; 180:105872. [PMID: 39362496 DOI: 10.1016/j.neuint.2024.105872] [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: 08/09/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
A growing body of evidence supports the link between peripheral inflammation and impairment of neurologic functions, including mood and cognitive abilities. The pathogenic event connecting peripheral inflammation and brain dysfunction is represented by neuroinflammation, a pathogenic phenomenon that provides an important contribution to neurodegeneration and cognitive decline also in Alzheimer's, Parkinson's, Huntington's diseases, as well as in Multiple Sclerosis. It is driven by resident brain immune cells, microglia and astrocytes, that acquire an activated phenotype in response to proinflammatory molecules moving from the periphery to the brain parenchyma. Although a huge progress has been made in clarifying cellular and molecular mechanisms bridging peripheral and central inflammation, a clear picture has not been achieved so far. Therefore, experimental models are of crucial relevance to clarify knowledge gaps in this regard. Many findings demonstrate that systemic inflammation induced by pathogen-associated molecular patterns, such as lipopolysaccharide (LPS), is able to trigger neuroinflammation. Therefore, LPS-administration is widely considered a useful tool to study this phenomenon. On this basis, the present review will focus on in vivo studies based on acute and subacute effects of systemic administration of LPS, with special attention on the state of art of microglia and astrocyte response to peripheral challenge.
Collapse
Affiliation(s)
- Maria Concetta Geloso
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Gemelli Science and Technology Park (GSTeP)-Organoids Research Core Facility, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy.
| | - Luca Zupo
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Valentina Corvino
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| |
Collapse
|
3
|
Rodriguez Moore G, Melo-Escobar I, Stegner D, Bracko O. One immune cell to bind them all: platelet contribution to neurodegenerative disease. Mol Neurodegener 2024; 19:65. [PMID: 39334369 PMCID: PMC11438031 DOI: 10.1186/s13024-024-00754-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Alzheimer's disease (AD) and related dementias (ADRD) collectively affect a significant portion of the aging population worldwide. The pathological progression of AD involves not only the classical hallmarks of amyloid beta (Aβ) plaque buildup and neurofibrillary tangle development but also the effects of vasculature and chronic inflammatory processes. Recently, platelets have emerged as central players in systemic and neuroinflammation. Studies have shown that patients with altered platelet receptor expression exhibit accelerated cognitive decline independent of traditional risk factors. Additionally, platelets from AD patients exhibit heightened unstimulated activation compared to control groups. Platelet granules contain crucial AD-related proteins like tau and amyloid precursor protein (APP). Dysregulation of platelet exocytosis contributes to disease phenotypes characterized by increased bleeding, stroke, and cognitive decline risk. Recent studies have indicated that these effects are not associated with the quantity of platelets present in circulation. This underscores the hypothesis that disruptions in platelet-mediated inflammation and healing processes may play a crucial role in the development of ADRD. A thorough look at platelets, encompassing their receptors, secreted molecules, and diverse roles in inflammatory interactions with other cells in the circulatory system in AD and ADRD, holds promising prospects for disease management and intervention. This review discusses the pivotal roles of platelets in ADRD.
Collapse
Affiliation(s)
| | - Isabel Melo-Escobar
- Department of Biology, University of Miami, Coral Gables, FL, 33146, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - David Stegner
- Institute for Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians University of Würzburg, Würzburg, Germany
| | - Oliver Bracko
- Department of Biology, University of Miami, Coral Gables, FL, 33146, USA.
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
4
|
Yao L, Li MY, Wang KC, Liu YZ, Zheng HZ, Zhong Z, Ma SQ, Yang HM, Sun MM, He M, Huang HP, Wang HF. Abnormal resting-state functional connectivity of hippocampal subregions in type 2 diabetes mellitus-associated cognitive decline. Front Psychiatry 2024; 15:1360623. [PMID: 39376966 PMCID: PMC11456530 DOI: 10.3389/fpsyt.2024.1360623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 09/03/2024] [Indexed: 10/09/2024] Open
Abstract
Objective Type 2 diabetes mellitus (T2DM) over time predisposes to inflammatory responses and abnormalities in functional brain networks that damage learning, memory, or executive function. The hippocampus is a key region often reporting connectivity abnormalities in memory disorders. Here, we investigated peripheral inflammatory responses and resting-state functional connectivity (RSFC) changes characterized of hippocampal subregions in type 2 diabetes-associated cognitive decline (T2DACD). Methods The study included 16 patients with T2DM, 16 patients with T2DACD and 25 healthy controls (HCs). Subjects were assessed for cognitive performance, tested for the expression of inflammatory factors IL-6, IL-10 and TNF-α in peripheral serum, underwent resting-state functional magnetic resonance imaging scans, and analyzed for RSFC using the hippocampal subregions as seeds. We also calculated the correlation between cognitive performance and RSFC of hippocampal subregion, and analyzed the significantly altered RSFC values of T2DACD for Receiver Operating Characteristic (ROC) analysis. Results T2DACD patients showed a decline in their ability to complete cognitive assessment scales and experimental paradigms, and T2DM did not show abnormal cognitive performance. IL-6 expression was increased in peripheral serum in both T2DACD and T2DM. Compared with HCs, T2DACD showed abnormalities RSFC of the left anterior hippocampus with left precentral gyrus and left angular gyrus. T2DM showed abnormalities RSFC of the left middle hippocampus with right medial frontal gyrus, right anterior and middle hippocampus with left precuneus, left anterior hippocampus with right precuneus and right posterior middle temporal gyrus. Compared with T2DM, T2DACD showed abnormalities RSFC of the left posterior hippocampus and right middle hippocampus with left precuneus. In addition, RSFC in the left posterior hippocampus with left precuneus of T2DACD was positively correlated with Flanker conflict response time (r=0.766, P=0.001). In the ROC analysis, the significantly altered RSFC values of T2DACD achieved significant performance. Conclusions T2DACD showed a significant decrease in attentional inhibition and working memory, peripheral pro-inflammatory response increased, and abnormalities RSFC of the hippocampal subregions with default mode network and sensory-motor network. T2DM did not show a significant cognitive decline, but peripheral pro-inflammatory response increased and abnormalities RSFC of the hippocampus subregions occurred in the brain. In addition, the left precuneus may be a key brain region in the conversion of T2DM to T2DACD. The results of this study may provide a basis for the preliminary diagnosis of T2DACD.
Collapse
Affiliation(s)
- Lin Yao
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Meng-Yuan Li
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Kang-Cheng Wang
- College of Psychology, Shandong Normal University, Jinan, Shandong, China
| | - Yan-Ze Liu
- Acupuncture and Tuina Center, The Third Affiliated Clinical Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hai-Zhu Zheng
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zhen Zhong
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Shi-Qi Ma
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hong-Mei Yang
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Meng-Meng Sun
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Min He
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hai-Peng Huang
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hong-Feng Wang
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
5
|
Yutani R, Venketaraman V, Sheren N. Treatment of Acute and Long-COVID, Diabetes, Myocardial Infarction, and Alzheimer's Disease: The Potential Role of a Novel Nano-Compound-The Transdermal Glutathione-Cyclodextrin Complex. Antioxidants (Basel) 2024; 13:1106. [PMID: 39334765 PMCID: PMC11429141 DOI: 10.3390/antiox13091106] [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: 06/23/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Oxidative stress (OS) occurs from excessive reactive oxygen species or a deficiency of antioxidants-primarily endogenous glutathione (GSH). There are many illnesses, from acute and post-COVID-19, diabetes, myocardial infarction to Alzheimer's disease, that are associated with OS. These dissimilar illnesses are, in order, viral infections, metabolic disorders, ischemic events, and neurodegenerative disorders. Evidence is presented that in many illnesses, (1) OS is an early initiator and significant promotor of their progressive pathophysiologic processes, (2) early reduction of OS may prevent later serious and irreversible complications, (3) GSH deficiency is associated with OS, (4) GSH can likely reduce OS and restore adaptive physiology, (5) effective administration of GSH can be accomplished with a novel nano-product, the GSH/cyclodextrin (GC) complex. OS is an overlooked pathological process of many illnesses. Significantly, with the GSH/cyclodextrin (GC) complex, therapeutic administration of GSH is now available to reduce OS. Finally, rigorous prospective studies are needed to confirm the efficacy of this therapeutic approach.
Collapse
Affiliation(s)
- Ray Yutani
- Department of Family Medicine, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Vishwanath Venketaraman
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Nisar Sheren
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
6
|
Donner L, Krüger I, Pfeiler S, Gerdes N, Schaller M, Kelm M, Elvers M. Reduced platelet activation and thrombus formation in male transgenic model mice of Alzheimer's disease suggests early sex-specific differences in platelet pathophysiology. Mol Cell Neurosci 2024; 130:103952. [PMID: 39002827 DOI: 10.1016/j.mcn.2024.103952] [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: 03/25/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and characterized by extracellular amyloid-β (Aβ) plaques, intracellular neurofibrillary tau tangles and neurodegeneration. Over 80 % of AD patients also exhibit cerebral amyloid angiopathy (CAA). CAA is a cerebrovascular disease caused by deposition of Aβ in the walls of cerebral blood vessels leading to vessel damage and impairment of normal blood flow. To date, different studies suggest that platelet function, including activation, adhesion and aggregation, is altered in AD due to vascular Aβ deposition. For example, the transgenic AD model mice APP23 mice that exhibit CAA and parenchymal Aβ plaques, show pre-activated platelets in the blood circulation and increased platelet integrin activation leading to a pro-thrombotic phenotype in these mice late stages of AD. However, it is still an open question whether or not platelets exhibit changes in their activation profile before they are exposed to vascular Aβ deposits. Therefore, the present study examined platelets from middle-aged transgenic APP23 mice at the age of 8-10 months. At this age, APP23 mice show amyloid plaques in the brain parenchyma but not in the vasculature. Our analyses show that these APP23 mice have unaltered platelet numbers and size, and unaltered surface expression of glycoproteins. However, the number of dense granules in transgenic platelets was increased while the release was unaltered. Male, but not female APP23 mice, exhibited reduced platelet activation after stimulation of the thrombin receptor PAR4 and decreased thrombus stability on collagen under flow conditions ex vivo compared to control mice. In an arterial thrombosis model in vivo, male APP23 mice showed attenuated occlusion of the injured artery compared to controls. These findings provide clear evidence for early changes in platelet activation and thrombus formation in male mice before development of overt CAA. Furthermore, reduced platelet activation and thrombus formation suggest sex-specific differences in platelet physiology in AD that has to be considered in future studies of platelets and their role in AD.
Collapse
Affiliation(s)
- Lili Donner
- Department of Vascular- and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine University, Düsseldorf, Germany
| | - Irena Krüger
- Department of Vascular- and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine University, Düsseldorf, Germany
| | - Susanne Pfeiler
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Norbert Gerdes
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital, Heinrich-Heine University, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | | | - Malte Kelm
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital, Heinrich-Heine University, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Margitta Elvers
- Department of Vascular- and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine University, Düsseldorf, Germany.
| |
Collapse
|
7
|
Nefodova A, Rudyk M, Dovhyi R, Dovbynchuk T, Dzubenko N, Tolstanova G, Skivka L. Systemic inflammation in Aβ 1-40-induced Alzheimer's disease model: New translational opportunities. Brain Res 2024; 1837:148960. [PMID: 38679313 DOI: 10.1016/j.brainres.2024.148960] [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: 01/09/2024] [Revised: 03/21/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
Alzheimer disease (AD) is the most frequent cause of dementia, and the most common neurodegenerative disease, which is characterized by memory impairment, neuronal death, and synaptic loss in the hippocampus. Sporadic late-onset AD, which accounts for over 95 % of disease cases, is a multifactorial pathology with complex etiology and pathogenesis. Nowadays, neuroinflammation is considered the third most important component of AD pathogenesis in addition to amyloid peptide generation and deposition. Neuroinflammation is associated with the impairment of blood-brain barrier and leakage of inflammatory mediators into the periphery with developing systemic inflammatory responses. Systemic inflammation is currently considered one of the therapeutic targets for AD treatment, that necessitates in-depth study of this phenomenon in appropriate non-transgenic animal models. This study was aimed to explore systemic inflammatory manifestations in rats with Aβ1-40-induced AD. The impairment of spatial memory and cognitive flexibility in Aβ1-40-lesioned rats was accompanied by pronounced systemic inflammation, which was confirmed by commonly accepted biomarkers: increased hematological indices of systemic inflammation (NLR, dNLR, LMR, PLR and SII), signs of anemia of inflammation or chronic diseases, and pro-inflammatory polarized activation of circulating phagocytes. In addition, markers of systemic inflammation strongly correlated with disorders of remote cognitive flexibility in Aβ1-40-lesioned rats. These results indicate, that Aβ1-40-induced AD model permits the investigation of specific element of the disease - systemic inflammation in addition to well reproduced neuroinflammation and impairment of spatial memory and cognitive flexibility. It increases translational value of this well-known model.
Collapse
Affiliation(s)
- Anastasiia Nefodova
- Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 2, Hlushkov Avenue, Kyiv 03022, Ukraine
| | - Mariia Rudyk
- Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 2, Hlushkov Avenue, Kyiv 03022, Ukraine.
| | - Roman Dovhyi
- Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 2, Hlushkov Avenue, Kyiv 03022, Ukraine
| | - Taisa Dovbynchuk
- Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 2, Hlushkov Avenue, Kyiv 03022, Ukraine
| | - Nataliia Dzubenko
- Educational and Scientific Institute of High Technologies, Taras Shevchenko University of Kyiv, 4g, Hlushkov Avenue, Kyiv 03022, Ukraine
| | - Ganna Tolstanova
- Educational and Scientific Institute of High Technologies, Taras Shevchenko University of Kyiv, 4g, Hlushkov Avenue, Kyiv 03022, Ukraine
| | - Larysa Skivka
- Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 2, Hlushkov Avenue, Kyiv 03022, Ukraine
| |
Collapse
|
8
|
Casanova R, Walker KA, Justice JN, Anderson A, Duggan MR, Cordon J, Barnard RT, Lu L, Hsu FC, Sedaghat S, Prizment A, Kritchevsky SB, Wagenknecht LE, Hughes TM. Associations of plasma proteomics and age-related outcomes with brain age in a diverse cohort. GeroScience 2024; 46:3861-3873. [PMID: 38438772 PMCID: PMC11226584 DOI: 10.1007/s11357-024-01112-4] [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: 12/07/2023] [Accepted: 02/26/2024] [Indexed: 03/06/2024] Open
Abstract
Machine learning models are increasingly being used to estimate "brain age" from neuroimaging data. The gap between chronological age and the estimated brain age gap (BAG) is potentially a measure of accelerated and resilient brain aging. Brain age calculated in this fashion has been shown to be associated with mortality, measures of physical function, health, and disease. Here, we estimate the BAG using a voxel-based elastic net regression approach, and then, we investigate its associations with mortality, cognitive status, and measures of health and disease in participants from Atherosclerosis Risk in Communities (ARIC) study who had a brain MRI at visit 5 of the study. Finally, we used the SOMAscan assay containing 4877 proteins to examine the proteomic associations with the MRI-defined BAG. Among N = 1849 participants (age, 76.4 (SD 5.6)), we found that increased values of BAG were strongly associated with increased mortality and increased severity of the cognitive status. Strong associations with mortality persisted when the analyses were performed in cognitively normal participants. In addition, it was strongly associated with BMI, diabetes, measures of physical function, hypertension, prevalent heart disease, and stroke. Finally, we found 33 proteins associated with BAG after a correction for multiple comparisons. The top proteins with positive associations to brain age were growth/differentiation factor 15 (GDF-15), Sushi, von Willebrand factor type A, EGF, and pentraxin domain-containing protein 1 (SEVP 1), matrilysin (MMP7), ADAMTS-like protein 2 (ADAMTS), and heat shock 70 kDa protein 1B (HSPA1B) while EGF-receptor (EGFR), mast/stem-cell-growth-factor-receptor (KIT), coagulation-factor-VII, and cGMP-dependent-protein-kinase-1 (PRKG1) were negatively associated to brain age. Several of these proteins were previously associated with dementia in ARIC. These results suggest that circulating proteins implicated in biological aging, cellular senescence, angiogenesis, and coagulation are associated with a neuroimaging measure of brain aging.
Collapse
Affiliation(s)
- Ramon Casanova
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC, USA.
| | | | - Jamie N Justice
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Andrea Anderson
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC, USA
| | | | | | - Ryan T Barnard
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC, USA
| | - Lingyi Lu
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC, USA
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC, USA
| | - Sanaz Sedaghat
- School of Public Health, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Anna Prizment
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Stephen B Kritchevsky
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Lynne E Wagenknecht
- Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Timothy M Hughes
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
9
|
Liu Z, Liu M, Xiong Y, Wang Y, Bu X. Crosstalk between bone and brain in Alzheimer's disease: Mechanisms, applications, and perspectives. Alzheimers Dement 2024; 20:5720-5739. [PMID: 38824621 PMCID: PMC11350061 DOI: 10.1002/alz.13864] [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: 02/05/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 06/04/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that involves multiple systems in the body. Numerous recent studies have revealed bidirectional crosstalk between the brain and bone, but the interaction between bone and brain in AD remains unclear. In this review, we summarize human studies of the association between bone and brain and provide an overview of their interactions and the underlying mechanisms in AD. We review the effects of AD on bone from the aspects of AD pathogenic proteins, AD risk genes, neurohormones, neuropeptides, neurotransmitters, brain-derived extracellular vesicles (EVs), and the autonomic nervous system. Correspondingly, we elucidate the underlying mechanisms of the involvement of bone in the pathogenesis of AD, including bone-derived hormones, bone marrow-derived cells, bone-derived EVs, and inflammation. On the basis of the crosstalk between bone and the brain, we propose potential strategies for the management of AD with the hope of offering novel perspectives on its prevention and treatment. HIGHLIGHTS: The pathogenesis of AD, along with its consequent changes in the brain, may involve disturbing bone homeostasis. Degenerative bone disorders may influence the progression of AD through a series of pathophysiological mechanisms. Therefore, relevant bone intervention strategies may be beneficial for the comprehensive management of AD.
Collapse
Affiliation(s)
- Zhuo‐Ting Liu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease (Third Military Medical University)ChongqingChina
| | - Ming‐Han Liu
- Department of OrthopaedicsXinqiao Hospital, Third Military Medical UniversityChongqingChina
| | - Yan Xiong
- Department of OrthopaedicsDaping Hospital, Third Military Medical UniversityChongqingChina
| | - Yan‐Jiang Wang
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease (Third Military Medical University)ChongqingChina
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
| | - Xian‐Le Bu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease (Third Military Medical University)ChongqingChina
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
| |
Collapse
|
10
|
Yu S, Chen X, Yang T, Cheng J, Liu E, Jiang L, Song M, Shu H, Ma Y. Revealing the mechanisms of blood-brain barrier in chronic neurodegenerative disease: an opportunity for therapeutic intervention. Rev Neurosci 2024; 0:revneuro-2024-0040. [PMID: 38967133 DOI: 10.1515/revneuro-2024-0040] [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: 03/16/2024] [Accepted: 05/30/2024] [Indexed: 07/06/2024]
Abstract
The brain microenvironment is tightly regulated, and the blood-brain barrier (BBB) plays a pivotal role in maintaining the homeostasis of the central nervous system. It effectively safeguards brain tissue from harmful substances in peripheral blood. However, both acute pathological factors and age-related biodegradation have the potential to compromise the integrity of the BBB and are associated with chronic neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD), as well as Epilepsy (EP). This association arises due to infiltration of peripheral foreign bodies including microorganisms, immune-inflammatory mediators, and plasma proteins into the central nervous system when the BBB is compromised. Nevertheless, these partial and generalized understandings do not prompt a shift from passive to active treatment approaches. Therefore, it is imperative to acquire a comprehensive and in-depth understanding of the intricate molecular mechanisms underlying vascular disease alterations associated with the onset and progression of chronic neurodegenerative disorders, as well as the subsequent homeostatic changes triggered by BBB impairment. The present article aims to systematically summarize and review recent scientific work with a specific focus on elucidating the fundamental mechanisms underlying BBB damage in AD, PD, and EP as well as their consequential impact on disease progression. These findings not only offer guidance for optimizing the physiological function of the BBB, but also provide valuable insights for developing intervention strategies aimed at early restoration of BBB structural integrity, thereby laying a solid foundation for designing drug delivery strategies centered around the BBB.
Collapse
Affiliation(s)
- Sixun Yu
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan Province, China
| | - Xin Chen
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
| | - Tao Yang
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
| | - Jingmin Cheng
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
| | - Enyu Liu
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
| | - Lingli Jiang
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
| | - Min Song
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
| | - Haifeng Shu
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan Province, China
| | - Yuan Ma
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| |
Collapse
|
11
|
Wang N, Cai L, Pei X, Lin Z, Huang L, Liang C, Wei M, Shao L, Guo T, Huang F, Luo H, Zheng H, Chen XF, Leng L, Zhang YW, Wang X, Zhang J, Guo K, Wang Z, Zhang H, Zhao Y, Xu H. Microglial apolipoprotein E particles contribute to neuronal senescence and synaptotoxicity. iScience 2024; 27:110006. [PMID: 38868202 PMCID: PMC11167441 DOI: 10.1016/j.isci.2024.110006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/13/2023] [Accepted: 05/14/2024] [Indexed: 06/14/2024] Open
Abstract
Apolipoprotein E (apoE) plays a crucial role in the pathogenesis of Alzheimer's disease (AD). Microglia exhibit a substantial upregulation of apoE in AD-associated circumstances, despite astrocytes being the primary source of apoE expression and secretion in the brain. Although the role of astrocytic apoE in the brain has been extensively investigated, it remains unclear that whether and how apoE particles generated from astrocytes and microglia differ in biological characteristic and function. Here, we demonstrate the differences in size between apoE particles generated from microglia and astrocytes. Microglial apoE particles impair neurite growth and synapses, and promote neuronal senescence, whereas depletion of GPNMB (glycoprotein non-metastatic melanoma protein B) in microglial apoE particles mitigated these deleterious effects. In addition, human APOE4-expressing microglia are more neurotoxic than APOE3-bearing microglia. For the first time, these results offer concrete evidence that apoE particles produced by microglia are involved in neuronal senescence and toxicity.
Collapse
Affiliation(s)
- Na Wang
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Lujian Cai
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Xinyu Pei
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Zhihao Lin
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Lihong Huang
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen 361102, China
| | - Chensi Liang
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Min Wei
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Lin Shao
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Tiantian Guo
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Fang Huang
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing 400016, China
| | - Hong Luo
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Honghua Zheng
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Xiao-fen Chen
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Lige Leng
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Yun-wu Zhang
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Xin Wang
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen 361102, China
| | - Jie Zhang
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Kai Guo
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing 400016, China
| | - Zhanxiang Wang
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Hongsheng Zhang
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing 400016, China
| | - Yingjun Zhao
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Huaxi Xu
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
12
|
Kivimäki M, Walker KA. Severe infections as a gateway to dementia. NATURE AGING 2024; 4:752-754. [PMID: 38783151 DOI: 10.1038/s43587-024-00643-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Affiliation(s)
- Mika Kivimäki
- Brain Sciences, University College London, London, UK.
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| |
Collapse
|
13
|
Lorenzini L, Zanella L, Sannia M, Baldassarro VA, Moretti M, Cescatti M, Quadalti C, Baldi S, Bartolucci G, Di Gloria L, Ramazzotti M, Clavenzani P, Costanzini A, De Giorgio R, Amedei A, Calzà L, Giardino L. Experimental colitis in young Tg2576 mice accelerates the onset of an Alzheimer's-like clinical phenotype. Alzheimers Res Ther 2024; 16:116. [PMID: 38773640 PMCID: PMC11110243 DOI: 10.1186/s13195-024-01471-2] [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: 02/14/2024] [Accepted: 05/01/2024] [Indexed: 05/24/2024]
Abstract
Systemic inflammation and neuroinflammation affect the natural course of the sporadic form of Alzheimer's disease (AD), as supported by epidemiological and preclinical data, and several epidemiological studies indicate a higher prevalence of AD in patients with inflammatory bowel disease. In this study, we explored whether colitis induced by dextran sulfate sodium (DSS) in young, presymptomatic/preplaque mice worsens and/or anticipates age-dependent cognitive impairment in Tg2576, a widely used mouse model of AD. We demonstrated that DSS colitis induced in young Tg2576 mice anticipates the onset age of learning and memory deficit in the Morris water maze test. To explore potential mechanisms behind the acceleration of cognitive decline in Tg2576 mice by DSS colitis, we focused on gut microbiota, systemic inflammation and neuroinflammation markers. We observed a Firmicutes/Bacteroidetes ratio change in Tg2576 DSS animals comparable to that of elderly Tg2576 mice, suggesting accelerated microbiota aging in Tg2576 DSS mice, a change not observed in C57BL6 DSS mice. We also observed substantial differences between Tg2576 and WT mice in several inflammation and neuroinflammation-related parameters as early as 3 months of age, well before plaque deposition, a picture which evolved rapidly (between 3 and 5.5 months of age) in contrast to Tg2576 and WT littermates not treated with DSS. In detail, following induction of DSS colitis, WT and Tg2576 mice exhibited contrasting features in the expression level of inflammation-evoked astrocyte-associated genes in the hippocampus. No changes in microglial features occurred in the hippocampus between the experimental groups, whereas a reduced glial fibrillary acidic protein immunoreactivity was observed in Tg2576 vs. WT mice. This finding may reflect an atrophic, "loss-of-function" profile, further exacerbated by DSS where a decreased of GFAP mRNA expression level was detected. In conclusion, we suggest that as-yet unidentified peripheral mediators evoked by DSS colitis and involving the gut-brain axis emphasize an astrocyte "loss-of-function" profile present in young Tg2576 mice, leading to impaired synaptic morphological and functional integrity as a very early sign of AD.
Collapse
Affiliation(s)
- Luca Lorenzini
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, Bologna, Italy
| | - Lorenzo Zanella
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, Bologna, Italy
| | | | | | - Marzia Moretti
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, Bologna, Italy
| | | | - Corinne Quadalti
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Tolara di Sopra 41/E, Bologna, 40064, Ozzano Emilia, Italy
| | - Simone Baldi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Gianluca Bartolucci
- Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, Florence, Italy
| | - Leandro Di Gloria
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Matteo Ramazzotti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Paolo Clavenzani
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, Bologna, Italy
| | - Anna Costanzini
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Roberto De Giorgio
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Laura Calzà
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Tolara di Sopra 41/E, Bologna, 40064, Ozzano Emilia, Italy.
| | - Luciana Giardino
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, Bologna, Italy
| |
Collapse
|
14
|
Scholz R, Brösamle D, Yuan X, Beyer M, Neher JJ. Epigenetic control of microglial immune responses. Immunol Rev 2024; 323:209-226. [PMID: 38491845 DOI: 10.1111/imr.13317] [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: 12/18/2023] [Accepted: 03/02/2024] [Indexed: 03/18/2024]
Abstract
Microglia, the major population of brain-resident macrophages, are now recognized as a heterogeneous population comprising several cell subtypes with different (so far mostly supposed) functions in health and disease. A number of studies have performed molecular characterization of these different microglial activation states over the last years making use of "omics" technologies, that is transcriptomics, proteomics and, less frequently, epigenomics profiling. These approaches offer the possibility to identify disease mechanisms, discover novel diagnostic biomarkers, and develop new therapeutic strategies. Here, we focus on epigenetic profiling as a means to understand microglial immune responses beyond what other omics methods can offer, that is, revealing past and present molecular responses, gene regulatory networks and potential future response trajectories, and defining cell subtype-specific disease relevance through mapping non-coding genetic variants. We review the current knowledge in the field regarding epigenetic regulation of microglial identity and function, provide an exemplary analysis that demonstrates the advantages of performing joint transcriptomic and epigenomic profiling of single microglial cells and discuss how comprehensive epigenetic analyses may enhance our understanding of microglial pathophysiology.
Collapse
Affiliation(s)
- Rebekka Scholz
- Immunogenomics & Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Desirée Brösamle
- Biomedical Center (BMC), Biochemistry, Faculty of Medicine, LMU Munich, Munich, Germany
- Neuroimmunology and Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Xidi Yuan
- Biomedical Center (BMC), Biochemistry, Faculty of Medicine, LMU Munich, Munich, Germany
- Neuroimmunology and Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Marc Beyer
- Immunogenomics & Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and University of Bonn and West German Genome Center, Bonn, Germany
| | - Jonas J Neher
- Biomedical Center (BMC), Biochemistry, Faculty of Medicine, LMU Munich, Munich, Germany
- Neuroimmunology and Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
15
|
Dark HE, Duggan MR, Walker KA. Plasma biomarkers for Alzheimer's and related dementias: A review and outlook for clinical neuropsychology. Arch Clin Neuropsychol 2024; 39:313-324. [PMID: 38520383 PMCID: PMC11484593 DOI: 10.1093/arclin/acae019] [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: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 03/25/2024] Open
Abstract
Recent technological advances have improved the sensitivity and specificity of blood-based biomarkers for Alzheimer's disease and related dementias. Accurate quantification of amyloid-ß peptide, phosphorylated tau (pTau) isoforms, as well as markers of neurodegeneration (neurofilament light chain [NfL]) and neuro-immune activation (glial fibrillary acidic protein [GFAP] and chitinase-3-like protein 1 [YKL-40]) in blood has allowed researchers to characterize neurobiological processes at scale in a cost-effective and minimally invasive manner. Although currently used primarily for research purposes, these blood-based biomarkers have the potential to be highly impactful in the clinical setting - aiding in diagnosis, predicting disease risk, and monitoring disease progression. Whereas plasma NfL has shown promise as a non-specific marker of neuronal injury, plasma pTau181, pTau217, pTau231, and GFAP have demonstrated desirable levels of sensitivity and specificity for identification of individuals with Alzheimer's disease pathology and Alzheimer's dementia. In this forward looking review, we (i) provide an overview of the most commonly used blood-based biomarkers for Alzheimer's disease and related dementias, (ii) discuss how comorbid medical conditions, demographic, and genetic factors can inform the interpretation of these biomarkers, (iii) describe ongoing efforts to move blood-based biomarkers into the clinic, and (iv) highlight the central role that clinical neuropsychologists may play in contextualizing and communicating blood-based biomarker results for patients.
Collapse
Affiliation(s)
- Heather E Dark
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Michael R Duggan
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| |
Collapse
|
16
|
Umoh IO, dos Reis HJ, de Oliveira ACP. Molecular Mechanisms Linking Osteoarthritis and Alzheimer's Disease: Shared Pathways, Mechanisms and Breakthrough Prospects. Int J Mol Sci 2024; 25:3044. [PMID: 38474288 PMCID: PMC10931612 DOI: 10.3390/ijms25053044] [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: 12/31/2023] [Revised: 02/04/2024] [Accepted: 02/09/2024] [Indexed: 03/14/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease mostly affecting the elderly population. It is characterized by cognitive decline that occurs due to impaired neurotransmission and neuronal death. Even though deposition of amyloid beta (Aβ) peptides and aggregation of hyperphosphorylated TAU have been established as major pathological hallmarks of the disease, other factors such as the interaction of genetic and environmental factors are believed to contribute to the development and progression of AD. In general, patients initially present mild forgetfulness and difficulty in forming new memories. As it progresses, there are significant impairments in problem solving, social interaction, speech and overall cognitive function of the affected individual. Osteoarthritis (OA) is the most recurrent form of arthritis and widely acknowledged as a whole-joint disease, distinguished by progressive degeneration and erosion of joint cartilage accompanying synovitis and subchondral bone changes that can prompt peripheral inflammatory responses. Also predominantly affecting the elderly, OA frequently embroils weight-bearing joints such as the knees, spine and hips leading to pains, stiffness and diminished joint mobility, which in turn significantly impacts the patient's standard of life. Both infirmities can co-occur in older adults as a result of independent factors, as multiple health conditions are common in old age. Additionally, risk factors such as genetics, lifestyle changes, age and chronic inflammation may contribute to both conditions in some individuals. Besides localized peripheral low-grade inflammation, it is notable that low-grade systemic inflammation prompted by OA can play a role in AD pathogenesis. Studies have explored relationships between systemic inflammatory-associated diseases like obesity, hypertension, dyslipidemia, diabetes mellitus and AD. Given that AD is the most common form of dementia and shares similar risk factors with OA-both being age-related and low-grade inflammatory-associated diseases, OA may indeed serve as a risk factor for AD. This work aims to review literature on molecular mechanisms linking OA and AD pathologies, and explore potential connections between these conditions alongside future prospects and innovative treatments.
Collapse
Affiliation(s)
| | - Helton Jose dos Reis
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Federal University of Minas Gerais, Av. Antonio Carlos 6627, Belo Horizonte 31270-901, MG, Brazil;
| | - Antonio Carlos Pinheiro de Oliveira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Federal University of Minas Gerais, Av. Antonio Carlos 6627, Belo Horizonte 31270-901, MG, Brazil;
| |
Collapse
|
17
|
Ko YK, Kim E, Lee EJ, Nam SJ, Kim Y, Kim S, Choi SY, Kim HY, Choi Y. Enrichment of infection-associated bacteria in the low biomass brain bacteriota of Alzheimer's disease patients. PLoS One 2024; 19:e0296307. [PMID: 38335187 PMCID: PMC10857729 DOI: 10.1371/journal.pone.0296307] [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: 09/14/2023] [Accepted: 12/08/2023] [Indexed: 02/12/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease accompanied by neuroimmune inflammation in the frontal cortex and hippocampus. Recently, the presence of bacteria in AD-affected brains has been documented, prompting speculation about their potential role in AD-associated neuroinflammation. However, the characterization of bacteriota in human brains affected by AD remains inconclusive. This study aimed to investigate potential associations between specific bacteria and AD pathology by examining brain tissues from AD-associated neurodegenerative regions (frontal cortex and hippocampus) and the non-AD-associated hypothalamus. Employing 16S rRNA gene sequencing, 30 postmortem brain tissue samples from four individuals with normal brain histology (N) and four AD patients were analyzed, along with three blank controls. A remarkably low biomass characterized the brain bacteriota, with their overall structures delineated primarily by brain regions rather than the presence of AD. While most analyzed parameters exhibited no significant distinction in the brain bacteriota between the N and AD groups, the unique detection of Cloacibacterium normanense in the AD-associated neurodegenerative regions stood out. Additionally, infection-associated bacteria, as opposed to periodontal pathogens, were notably enriched in AD brains. This study's findings provide valuable insights into potential link between bacterial infection and neuroinflammation in AD.
Collapse
Affiliation(s)
- Yeon Kyeong Ko
- Department of Immunology and Molecular Microbiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Eunbi Kim
- Department of Immunology and Molecular Microbiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Eun-Jae Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Soo Jeong Nam
- Department of Pathology, Asan Medical Center, Seoul, Republic of Korea
| | - Yeshin Kim
- Department of Neurology, Kangwon National University Hospital, Kangwon National University College of Medicine, Chuncheon, Republic of Korea
| | - Seongheon Kim
- Department of Neurology, Kangwon National University Hospital, Kangwon National University College of Medicine, Chuncheon, Republic of Korea
| | - Se-Young Choi
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Hyun Young Kim
- Department of Immunology and Molecular Microbiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Youngnim Choi
- Department of Immunology and Molecular Microbiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
18
|
Gao H, Fang B, Sun Z, Du X, Guo H, Zhao L, Zhang M. Effect of Human Milk Oligosaccharides on Learning and Memory in Mice with Alzheimer's Disease. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:1067-1081. [PMID: 38112024 DOI: 10.1021/acs.jafc.3c05949] [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: 12/20/2023]
Abstract
Alzheimer's disease (AD) is distinguished by cognitive dysfunction and neuroinflammation in the brain. 2'-Fucosyllactose (2'-FL) is a major human milk oligosaccharide (HMO) that is abundantly present in breast milk and has been demonstrated to exhibit immunomodulatory effects. However, the role of 2'-FL and HMO in gut microbiota modulation in relation to AD remains insufficiently investigated. This study aimed to elucidate the preventive effect of the 2'-FL and HMO impact of AD and the relevant mechanism involved. Here, the behavioral results showed that 2'-FL and HMO intervention decreased the expression of Tau phosphorylation and amyloid-β (Aβ), inhibited neuroinflammation, and restored cognitive impairment in AD mice. The metagenomic analysis proved that 2'-FL and HMO intervention restored the dysbiosis of the gut microbiota in AD. Notably, 2'-FL and HMO intervention significantly enhanced the relative abundance of Clostridium and Akkermansia. The metabolomics results showed that 2'-FL and HMO enhanced the oleoyl-l-carnitine metabolism as potential drivers. More importantly, the levels of oleoyl-l-carnitine were positively correlated with the abundances of Clostridium and Akkermansia. These results indicated that 2'-FL and HMO had therapeutic potential to prevent AD-induced cognitive impairment, which is of great significance for the treatment of AD.
Collapse
Affiliation(s)
- Haina Gao
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Bing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Zhe Sun
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Xiaoyu Du
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Huiyuan Guo
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Liang Zhao
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Beijing Laboratory of Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Ming Zhang
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
19
|
Chen K, Gupta R, Martín‐Ávila A, Cui M, Xie Z, Yang G. Anesthesia-induced hippocampal-cortical hyperactivity and tau hyperphosphorylation impair remote memory retrieval in Alzheimer's disease. Alzheimers Dement 2024; 20:494-510. [PMID: 37695022 PMCID: PMC10843666 DOI: 10.1002/alz.13464] [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: 03/23/2023] [Revised: 07/25/2023] [Accepted: 08/16/2023] [Indexed: 09/12/2023]
Abstract
INTRODUCTION Anesthesia often exacerbates memory recall difficulties in individuals with Alzheimer's disease (AD), but the underlying mechanisms remain unclear. METHODS We used in vivo Ca2+ imaging, viral-based circuit tracing, and chemogenetic approaches to investigate anesthesia-induced remote memory impairment in mouse models of presymptomatic AD. RESULTS Our study identified pyramidal neuron hyperactivity in the anterior cingulate cortex (ACC) as a significant contributor to anesthesia-induced remote memory impairment. This ACC hyperactivation arises from the disinhibition of local inhibitory circuits and increased excitatory inputs from the hippocampal CA1 region. Inhibiting hyperactivity in the CA1-ACC circuit improved memory recall after anesthesia. Moreover, anesthesia led to increased tau phosphorylation in the hippocampus, and inhibiting this hyperphosphorylation prevented ACC hyperactivity and subsequent memory impairment. DISCUSSION Hippocampal-cortical hyperactivity plays a role in anesthesia-induced remote memory impairment. Targeting tau hyperphosphorylation shows promise as a therapeutic strategy to mitigate anesthesia-induced neural network dysfunction and retrograde amnesia in AD.
Collapse
Affiliation(s)
- Kai Chen
- Department of AnesthesiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Riya Gupta
- Barnard College of Columbia UniversityNew YorkNew YorkUSA
| | | | - Meng Cui
- Department of BiologyPurdue UniversityWest LafayetteIndianaUSA
| | - Zhongcong Xie
- Geriatric Anesthesia Research UnitDepartment of AnesthesiaCritical Care and Pain MedicineMassachusetts General Hospital and Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Guang Yang
- Department of AnesthesiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| |
Collapse
|
20
|
Ayyubova G, Kodali M, Upadhya R, Madhu LN, Attaluri S, Somayaji Y, Shuai B, Rao S, Shankar G, Shetty AK. Extracellular vesicles from hiPSC-NSCs can prevent peripheral inflammation-induced cognitive dysfunction with inflammasome inhibition and improved neurogenesis in the hippocampus. J Neuroinflammation 2023; 20:297. [PMID: 38087314 PMCID: PMC10717852 DOI: 10.1186/s12974-023-02971-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Extracellular vesicles (EVs) released by human induced pluripotent stem cell-derived neural stem cells (hiPSC-NSCs) are enriched with miRNAs and proteins capable of mediating robust antiinflammatory activity. The lack of tumorigenic and immunogenic properties and ability to permeate the entire brain to incorporate into microglia following intranasal (IN) administrations makes them an attractive biologic for curtailing chronic neuroinflammation in neurodegenerative disorders. We tested the hypothesis that IN administrations of hiPSC-NSC-EVs can alleviate chronic neuroinflammation and cognitive impairments induced by the peripheral lipopolysaccharide (LPS) challenge. Adult male, C57BL/6J mice received intraperitoneal injections of LPS (0.75 mg/kg) for seven consecutive days. Then, the mice received either vehicle (VEH) or hiPSC-NSC-EVs (~ 10 × 109 EVs/administration, thrice over 6 days). A month later, mice in all groups were investigated for cognitive function with behavioral tests and euthanized for histological and biochemical studies. Mice receiving VEH after LPS displayed deficits in associative recognition memory, temporal pattern processing, and pattern separation. Such impairments were associated with an increased incidence of activated microglia presenting NOD-, LRR-, and pyrin domain containing 3 (NLRP3) inflammasomes, elevated levels of NLRP3 inflammasome mediators and end products, and decreased neurogenesis in the hippocampus. In contrast, the various cognitive measures in mice receiving hiPSC-NSC-EVs after LPS were closer to naive mice. Significantly, these mice displayed diminished microglial activation, NLRP3 inflammasomes, proinflammatory cytokines, and a level of neurogenesis matching age-matched naïve controls. Thus, IN administrations of hiPSC-NSC-EVs are an efficacious approach to reducing chronic neuroinflammation-induced cognitive impairments.
Collapse
Affiliation(s)
- Gunel Ayyubova
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Leelavathi N Madhu
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Yogish Somayaji
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Shama Rao
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Goutham Shankar
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA.
| |
Collapse
|
21
|
Munteanu C, Iordan DA, Hoteteu M, Popescu C, Postoiu R, Onu I, Onose G. Mechanistic Intimate Insights into the Role of Hydrogen Sulfide in Alzheimer's Disease: A Recent Systematic Review. Int J Mol Sci 2023; 24:15481. [PMID: 37895161 PMCID: PMC10607039 DOI: 10.3390/ijms242015481] [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: 09/22/2023] [Revised: 10/15/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023] Open
Abstract
In the rapidly evolving field of Alzheimer's Disease (AD) research, the intricate role of Hydrogen Sulfide (H2S) has garnered critical attention for its diverse involvement in both pathological substrates and prospective therapeutic paradigms. While conventional pathophysiological models of AD have primarily emphasized the significance of amyloid-beta (Aβ) deposition and tau protein hyperphosphorylation, this targeted systematic review meticulously aggregates and rigorously appraises seminal contributions from the past year elucidating the complex mechanisms of H2S in AD pathogenesis. Current scholarly literature accentuates H2S's dual role, delineating its regulatory functions in critical cellular processes-such as neurotransmission, inflammation, and oxidative stress homeostasis-while concurrently highlighting its disruptive impact on quintessential AD biomarkers. Moreover, this review illuminates the nuanced mechanistic intimate interactions of H2S in cerebrovascular and cardiovascular pathology associated with AD, thereby exploring avant-garde therapeutic modalities, including sulfurous mineral water inhalations and mud therapy. By emphasizing the potential for therapeutic modulation of H2S via both donors and inhibitors, this review accentuates the imperative for future research endeavors to deepen our understanding, thereby potentially advancing novel diagnostic and therapeutic strategies in AD.
Collapse
Affiliation(s)
- Constantin Munteanu
- Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iași, Romania;
- Teaching Emergency Hospital “Bagdasar-Arseni” (TEHBA), 041915 Bucharest, Romania; (M.H.); (R.P.); (G.O.)
| | - Daniel Andrei Iordan
- Department of Individual Sports and Kinetotherapy, Faculty of Physical Education and Sport, ‘Dunarea de Jos’ University of Galati, 800008 Galati, Romania;
| | - Mihail Hoteteu
- Teaching Emergency Hospital “Bagdasar-Arseni” (TEHBA), 041915 Bucharest, Romania; (M.H.); (R.P.); (G.O.)
| | - Cristina Popescu
- Teaching Emergency Hospital “Bagdasar-Arseni” (TEHBA), 041915 Bucharest, Romania; (M.H.); (R.P.); (G.O.)
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila” (UMPCD), 020022 Bucharest, Romania
| | - Ruxandra Postoiu
- Teaching Emergency Hospital “Bagdasar-Arseni” (TEHBA), 041915 Bucharest, Romania; (M.H.); (R.P.); (G.O.)
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila” (UMPCD), 020022 Bucharest, Romania
| | - Ilie Onu
- Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iași, Romania;
| | - Gelu Onose
- Teaching Emergency Hospital “Bagdasar-Arseni” (TEHBA), 041915 Bucharest, Romania; (M.H.); (R.P.); (G.O.)
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila” (UMPCD), 020022 Bucharest, Romania
| |
Collapse
|
22
|
Zheng J, Du X, Yang L, Fu H. Causal relationships between delirium and Alzheimer's disease: a bidirectional two-sample Mendelian randomization study. Eur J Med Res 2023; 28:271. [PMID: 37550780 PMCID: PMC10405368 DOI: 10.1186/s40001-023-01245-w] [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: 06/19/2023] [Accepted: 07/26/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND Previous observational studies have reported that delirium has an association with an increased risk of Alzheimer's disease (AD), and that patients with AD have a higher risk of developing delirium. However, due to the limitations of observational study, it is challenging to confirm whether delirium has a causal effect on AD or reverse causation exists. METHODS A bidirectional two-sample Mendelian randomization (MR) was performed to investigate the relationship between delirium and AD. Summary statistics from genome-wide association studies of delirium and AD phenotypes were utilized. Inverse-variance weighted (IVW) method was used as the main analysis approach, and additional analyses were performed using MR Egger, weighted median, simple mode and weighted mode to ensure result accuracy. Heterogeneity and horizontal pleiotropy were assessed using Cochran's Q statistics and MR Egger intercept, separately. RESULTS The MR analyses showed that genetically predicted delirium was not associated with AD (IVW: odds ratio [OR] 0.98, 95% CI 0.91-1.05, P = 0.544; MR Egger: OR 0.98, 95% CI 0.83-1.15, P = 0.780; weighted median: OR 0.96, 95% CI 0.88-1.05, P = 0.323; simple mode: OR 0.91, 95% CI 0.80-1.04, P = 0.212; weighted mode: OR 0.93, 95% CI 0.83-1.05, P = 0.277). However, in the reverse direction, AD was associated with delirium (IVW: OR 1.32, 95% CI 1.13-1.54, P = 3.91E-04; MR Egger: OR 1.42, 95% CI 1.02-1.98, P = 5.60E-02; Weighted median: OR 1.39, 95% CI 1.18-1.63, P = 8.22E-05; Simple mode: OR 1.41, 95% CI 1.10-1.80, P = 1.41E-02; Weighted mode: OR 1.39, 95% CI 1.16-1.67, P = 3.23E-03). CONCLUSION Based on the results of our MR study, there is no bidirectional causality between delirium and AD, delirium is not associated with an increased risk of AD, while genetically predicted AD is a potential causal risk factor for delirium.
Collapse
Affiliation(s)
- Jiang Zheng
- Department of Anesthesiology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Xiaohui Du
- Department of Anesthesiology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Liu Yang
- Department of Neurology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Hong Fu
- Department of Anesthesiology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China.
| |
Collapse
|