1
|
Zheng Q, Wang X. Alzheimer's disease: insights into pathology, molecular mechanisms, and therapy. Protein Cell 2025; 16:83-120. [PMID: 38733347 PMCID: PMC11786724 DOI: 10.1093/procel/pwae026] [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: 03/04/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024] Open
Abstract
Alzheimer's disease (AD), the leading cause of dementia, is characterized by the accumulation of amyloid plaques and neurofibrillary tangles in the brain. This condition casts a significant shadow on global health due to its complex and multifactorial nature. In addition to genetic predispositions, the development of AD is influenced by a myriad of risk factors, including aging, systemic inflammation, chronic health conditions, lifestyle, and environmental exposures. Recent advancements in understanding the complex pathophysiology of AD are paving the way for enhanced diagnostic techniques, improved risk assessment, and potentially effective prevention strategies. These discoveries are crucial in the quest to unravel the complexities of AD, offering a beacon of hope for improved management and treatment options for the millions affected by this debilitating disease.
Collapse
Affiliation(s)
- Qiuyang Zheng
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Xin Wang
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
| |
Collapse
|
2
|
Aivalioti E, Georgiopoulos G, Tual-Chalot S, Bampatsias D, Delialis D, Sopova K, Drakos SG, Stellos K, Stamatelopoulos K. Amyloid-beta metabolism in age-related neurocardiovascular diseases. Eur Heart J 2025; 46:250-272. [PMID: 39527015 PMCID: PMC11735085 DOI: 10.1093/eurheartj/ehae655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/13/2024] [Accepted: 09/15/2024] [Indexed: 11/16/2024] Open
Abstract
Epidemiological evidence suggests the presence of common risk factors for the development and prognosis of both cardio- and cerebrovascular diseases, including stroke, Alzheimer's disease, vascular dementia, heart, and peripheral vascular diseases. Accumulation of harmful blood signals may induce organotypic endothelial dysfunction affecting blood-brain barrier function and vascular health in age-related diseases. Genetic-, age-, lifestyle- or cardiovascular therapy-associated imbalance of amyloid-beta (Aβ) peptide metabolism in the brain and periphery may be the missing link between age-related neurocardiovascular diseases. Genetic polymorphisms of genes related to Aβ metabolism, lifestyle modifications, drugs used in clinical practice, and Aβ-specific treatments may modulate Aβ levels, affecting brain, vascular, and cardiac diseases. This narrative review elaborates on the effects of interventions on Aβ metabolism in the brain, cerebrospinal fluid, blood, and peripheral heart or vascular tissues. Implications for clinical applicability, gaps in knowledge, and future perspectives of Aβ as the link among age-related neurocardiovascular diseases are also discussed.
Collapse
Affiliation(s)
- Evmorfia Aivalioti
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, PO Box 11528, 80 Vas. Sofias Str., Athens, Greece
| | - Georgios Georgiopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, PO Box 11528, 80 Vas. Sofias Str., Athens, Greece
- School of Biomedical Engineering and Imaging Sciences, King’s College, London, UK
- Department of Physiology, School of Medicine, University of Patras, Patra, Greece
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Centre for Life, Newcastle Upon Tyne, NE1 3BZ, UK
| | - Dimitrios Bampatsias
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, PO Box 11528, 80 Vas. Sofias Str., Athens, Greece
- Division of Cardiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Dimitrios Delialis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, PO Box 11528, 80 Vas. Sofias Str., Athens, Greece
| | - Kateryna Sopova
- Department of Cardiovascular Research, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13–17, D-68167 Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Mannheim, Germany
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Stavros G Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT, USA
- Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Centre for Life, Newcastle Upon Tyne, NE1 3BZ, UK
- Department of Cardiovascular Research, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13–17, D-68167 Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Mannheim, Germany
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, PO Box 11528, 80 Vas. Sofias Str., Athens, Greece
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Centre for Life, Newcastle Upon Tyne, NE1 3BZ, UK
| |
Collapse
|
3
|
Liu Y, Li T, Xiong J. Alzheimer's disease and diabetes-associated cognitive dysfunction: the microglia link? Metab Brain Dis 2025; 40:85. [PMID: 39754611 DOI: 10.1007/s11011-024-01516-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025]
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of amyloid-β (Aβ) plaques and the aggregation of tau protein, resulting in intense memory loss and dementia. Diabetes-associated cognitive dysfunction (DACD) is a complication of diabetes mellitus, which is associated with decreased cognitive function and impaired memory. A growing body of literature emphasize the involvement of microglia in AD and DACD. Although AD and DACD share some common features related to symptomatology and pathophysiology, the characteristics and heterogeneity of microglia remain largely unknown in these two diseases. In this study, multiple bioinformatics analyses were performed to analyze the frequency, altered genes, cell-cell communication, and subtypes of microglia in AD and DACD mouse models based on two publicly single-nucleus RNA sequencing (snRNA-Seq) datasets. The results revealed that the frequency of microglia was increased in both AD and DACD mouse models when compared with control mice. After analyzing the differentially expressed genes of microglia from the two mouse models, only six common upregulated genes were found. The CellChat analysis revealed the complex cell-cell communication network (microglia clusters with other cell types) in 5XFAD vs. control mice and db/db vs. control mice. The microglia subtypes and their transcription factor activity profile in 5XFAD mice were different from that in db/db mice. In summary, this study provided some insights into the alterations of microglia in 5XFAD and db/db mice, which might open up potential avenues for the microglial-targeted therapy in AD and DACD.
Collapse
Affiliation(s)
- Yaqiong Liu
- Regenerative Medicine Institute (REMEDI), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Tao Li
- Regenerative Medicine Institute (REMEDI), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Juliang Xiong
- Department of Pharmacy, the Second Affiliated Hospital of Shaoyang University, Shaoyang, Hunan, China.
| |
Collapse
|
4
|
Zhao B, Zang P, Quan M, Wang Q, Guo D, Jia J, Wang W. The Effect of APOE ε4 on Alzheimer's Disease Fluid Biomarkers: A Cross-Sectional Study Based on the COAST. CNS Neurosci Ther 2025; 31:e70202. [PMID: 39749650 PMCID: PMC11696244 DOI: 10.1111/cns.70202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/10/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
AIMS To analyze the effect of APOE ε4 on fluid biomarkers and the correlations between blood molecules and CSF biomarkers in AD patients. METHODS This study enrolled 575 AD patients, 131 patients with non-AD dementia, and 112 cognitively normal (CN) participants, and AD patients were divided into APOE ε4 carriers and non-carriers. Cerebrospinal fluid (CSF) biomarkers and blood-derived biomolecules were compared between AD and CN groups, between non-AD dementia and CN groups, as well as within APOE ε4 subgroups of AD patients. Utilizing Spearman's correlation analysis and quantile regression analysis, the relationships between blood-derived biomolecules and CSF biomarkers were analyzed in APOE ε4 carriers and non-carriers. RESULTS The levels of CSF biomarkers and blood molecules exhibited significant differences between the AD and CN groups, including Aβ42, t-tau, p-tau 181, high-density lipoprotein, low-density lipoprotein (LDL), and uric acid. In AD patients, APOE ε4 carriers had increased levels of CSF t-tau, p-tau 181, and plasma LDL. In the correlation and regression analyses, the negative relationships between plasma TG and t-tau, between plasma TG and p-tau 181 levels, as well as the positive relationship between serum IgA and CSF Aβ42, were observed significantly in APOE ε4+ AD groups, but not in APOE ε4- AD group. CONCLUSION APOE ε4 is associated with accelerated progression of AD pathology. The blood-derived biomolecules correlated with CSF biomarkers in APOE ε4 carriers are related to neuroinflammation and lipid metabolism, which may indicate the role of APOE ε4 in AD pathophysiology and offer insights for diagnostic and therapeutic strategies for AD. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT03653156.
Collapse
Affiliation(s)
- Bote Zhao
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- National Center for Neurological Disorders and National Clinical Research Center for Geriatric DiseasesBeijingChina
- Center of Alzheimer's Disease, Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersBeijingChina
| | - Peixi Zang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Department of NeurologyGansu Provincial HospitalLanzhou CityGansu ProvinceChina
| | - Meina Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- National Center for Neurological Disorders and National Clinical Research Center for Geriatric DiseasesBeijingChina
- Center of Alzheimer's Disease, Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersBeijingChina
| | - Qianqian Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- National Center for Neurological Disorders and National Clinical Research Center for Geriatric DiseasesBeijingChina
- Center of Alzheimer's Disease, Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersBeijingChina
| | - Dongmei Guo
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- National Center for Neurological Disorders and National Clinical Research Center for Geriatric DiseasesBeijingChina
- Center of Alzheimer's Disease, Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersBeijingChina
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- National Center for Neurological Disorders and National Clinical Research Center for Geriatric DiseasesBeijingChina
- Center of Alzheimer's Disease, Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersBeijingChina
| | - Wei Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- National Center for Neurological Disorders and National Clinical Research Center for Geriatric DiseasesBeijingChina
- Center of Alzheimer's Disease, Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersBeijingChina
| |
Collapse
|
5
|
Xu F, Albadry M, Döding A, Chen X, Dirsch O, Schulze-Späte U, Dahmen U. The effects of saturated and unsaturated fatty acids on MASLD: a Mendelian randomization analysis and in vivo experiment. Eur J Nutr 2024; 64:52. [PMID: 39718605 DOI: 10.1007/s00394-024-03560-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024]
Abstract
BACKGROUND Excessive intake of fatty acids is a key factor contributing to metabolic dysfunction-associated steatotic liver disease (MASLD). However, the effects of saturated fatty acids (SFA) and unsaturated fatty acids (UFA) on the development of MASLD are uncertain. Therefore, we conducted two-sample Mendelian randomization studies and animal experiments to explore the effects of SFA, monounsaturated fatty acids (MUFA) and polyunsaturated fatty acids (PUFA) on the risk of developing MASLD. METHODS The genetic summary data of exposures and outcome were retrieved from genome-wide association studies (GWASs) and used for five Mendelian randomization methods. A comprehensive sensitivity analysis was performed to verify the robustness of the results. Mice were subjected to different diets followed by assessment of severity of steatosis based on a histological score and determination of hepatic triglyceride levels to investigate the relationships between SFA, MUFA, PUFA and MASLD. RESULTS The Mendelian randomization results showed that MUFA (odds ratio: 1.441, 95% confidence interval: 1.078-1.927, P = 0.014) was causally associated with the incidence of MASLD. SFA and PUFA were not causally associated with the incidence of MASLD. Sensitivity analysis did not identify any significant bias in the results. The animal experiment results showed that a MUFA-enriched diet significantly contributed to the development of hepatic steatosis (P < 0.001). CONCLUSION SFA and PUFA did not have a significant causal effect on MASLD, but MUFA intake is a risk factor for MASLD. A MUFA-enriched diet increased the incidence of macrovesicular steatosis and the hepatic triglyceride levels. Therefore, replacing MUFA intake with a moderate intake of PUFA might help reduce the risk of MASLD.
Collapse
Affiliation(s)
- Fengming Xu
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747, Jena, Germany
- Else Kröner Graduate School for Medical Students "JSAM", Jena University Hospital, 07747, Jena, Germany
- Department of Infectious Diseases, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Mohamed Albadry
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747, Jena, Germany
- Department of Pathology, Faculty of Veterinary Medicine, Menoufia University, Shebin El Kom , 6131567, Egypt
| | - Annika Döding
- Section of Geriodontics, Department of Conservative Dentistry and Periodontics, Jena University Hospital, 07743, Jena, Germany
| | - Xinpei Chen
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747, Jena, Germany
| | - Olaf Dirsch
- Institute for Pathology, BG Klinikum Berlin, 12683, Berlin, Germany
| | - Ulrike Schulze-Späte
- Section of Geriodontics, Department of Conservative Dentistry and Periodontics, Jena University Hospital, 07743, Jena, Germany
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747, Jena, Germany.
| |
Collapse
|
6
|
Moore A, Ritchie MD. Is the Relationship Between Cardiovascular Disease and Alzheimer's Disease Genetic? A Scoping Review. Genes (Basel) 2024; 15:1509. [PMID: 39766777 PMCID: PMC11675426 DOI: 10.3390/genes15121509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Cardiovascular disease (CVD) and Alzheimer's disease (AD) are two diseases highly prevalent in the aging population and often co-occur. The exact relationship between the two diseases is uncertain, though epidemiological studies have demonstrated that CVDs appear to increase the risk of AD and vice versa. This scoping review aims to examine the current identified overlapping genetics between CVDs and AD at the individual gene level and at the shared pathway level. METHODS Following PRISMA-ScR guidelines for a scoping review, we searched the PubMed and Scopus databases from 1990 to October 2024 for articles that involved (1) CVDs, (2) AD, and (3) used statistical methods to parse genetic relationships. RESULTS Our search yielded 2918 articles, of which 274 articles passed screening and were organized into two main sections: (1) evidence of shared genetic risk; and (2) shared mechanisms. The genes APOE, PSEN1, and PSEN2 reportedly have wide effects across the AD and CVD spectrum, affecting both cardiac and brain tissues. Mechanistically, changes in three main pathways (lipid metabolism, blood pressure regulation, and the breakdown of the blood-brain barrier (BBB)) contribute to subclinical and etiological changes that promote both AD and CVD progression. However, genetic studies continue to be limited by the availability of longitudinal data and lack of cohorts that are representative of diverse populations. CONCLUSIONS Highly penetrant familial genes simultaneously increase the risk of CVDs and AD. However, in most cases, sets of dysregulated genes within larger-scale mechanisms, like changes in lipid metabolism, blood pressure regulation, and BBB breakdown, increase the risk of both AD and CVDs and contribute to disease progression.
Collapse
Affiliation(s)
- Anni Moore
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Marylyn D. Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Division of Informatics, Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
7
|
Niemeyer CS, Merle L, Bubak AN, Baxter BD, Gentile Polese A, Colon-Reyes K, Vang S, Hassell JE, Bruce KD, Nagel MA, Restrepo D. Olfactory and trigeminal routes of HSV-1 CNS infection with regional microglial heterogeneity. J Virol 2024; 98:e0096824. [PMID: 39475273 PMCID: PMC11575344 DOI: 10.1128/jvi.00968-24] [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: 06/03/2024] [Accepted: 09/29/2024] [Indexed: 11/06/2024] Open
Abstract
Herpes simplex virus type 1 (HSV-1) primarily targets the oral and nasal epithelia before establishing latency in the trigeminal ganglion (TG) and other peripheral ganglia. HSV-1 can also infect and become latent in the central nervous system (CNS) independent of latency in the TGs. Recent studies suggest entry to the CNS via two distinct routes: the TG-brainstem connection and olfactory nerve; however, to date, there is no characterization of brain regions targeted during HSV-1 primary infection. Furthermore, the immune response by microglia may also contribute to the heterogeneity between different brain regions. However, the response to HSV-1 by microglia has not been characterized in a region-specific manner. This study investigated the time course of HSV-1 spread within the olfactory epithelium (OE) and CNS following intranasal inoculation and the corresponding macrophage/microglial response in a C57BL/6 mouse model. We found an apical to basal spread of HSV-1 within the OE and underlying tissue accompanied by an inflammatory response of macrophages. OE infection was followed by infection of a small subset of brain regions targeted by the TG in the brainstem and other cranial nerve nuclei, including the vagus and hypoglossal nerve. Furthermore, other brain regions were positive for HSV-1 antigens, such as the locus coeruleus (LC), raphe nucleus (RaN), and hypothalamus while sparing the hippocampus and cortex. Within each brain region, microglia activation also varied widely. These findings provide critical insights into the region-specific dissemination of HSV-1 within the CNS, elucidating potential mechanisms linking viral infection to neurological and neurodegenerative diseases.IMPORTANCEThis study shows how herpes simplex virus type 1 (HSV-1) spreads within the brain after infecting the nasal passages. Our data reveal the distinct pattern of HSV-1 through the brain during a non-encephalitic infection. Furthermore, microglial activation was also temporally and spatially specific, with some regions of the brain having sustained microglial activation even in the absence of viral antigens. Previous reports have identified specific brain regions found to be positive for HSV-1 infection; however, to date, there has not been a concise investigation of the anatomical spread of HSV-1 and the brain regions consistently vulnerable to viral entry and spread. Understanding these region-specific differences in infection and immune response is crucial because it links HSV-1 infection to potential triggers for neurological and neurodegenerative diseases.
Collapse
Affiliation(s)
- Christy S Niemeyer
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laetitia Merle
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Andrew N Bubak
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - B Dnate' Baxter
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Arianna Gentile Polese
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Katherine Colon-Reyes
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sandy Vang
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - James E Hassell
- Department of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kimberley D Bruce
- Department of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Maria A Nagel
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Diego Restrepo
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
8
|
Liu S, Li X, Fan P, Gu Y, Yang A, Wang W, Zhou L, Chen H, Zheng F, Lin J, Xu Z, Zhao Q. The potential role of transcription factor sterol regulatory element binding proteins (SREBPs) in Alzheimer's disease. Biomed Pharmacother 2024; 180:117575. [PMID: 39442239 DOI: 10.1016/j.biopha.2024.117575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
Sterol regulatory element binding proteins (SREBPs) are a series of cholesterol-related transcription factors. Their role in regulating brain cholesterol biosynthesis, amyloid accumulation, and tau tangles formation has been intensively studied in protein-protein interaction analysis based on genes in clinical databases. SREBPs play an important role in maintaining cholesterol homeostasis in the brain. There are three subtypes of SREBPs, SREBP-1a stimulates the expression of genes related to cholesterol and fatty acid synthesis, SREBP-1c stimulates adipogenesis, and SREBP-2 stimulates cholesterol synthase and LDL receptors. SREBP-2 is activated in response to cholesterol depletion and stimulates a compensatory upregulation of cholesterol uptake and synthesis. Previous studies have shown that inhibition of SREBP-2 reduces cholesterol and amyloid accumulation, and new research suggests that SREBPs play a multifaceted role in Alzheimer's disease. Here, we highlight the importance of SREBPs in AD, in terms of multiple pathways regulating cholesterol in the brain, and primarily demonstrate the potential of SREBP-2 inhibitors. There was a trend towards a significant increase in the expression levels of different SREBP isoforms in AD patients compared to healthy controls. Therefore, there is a close link between SREBPs and AD, and this review analyses the potential role of SREBPs in the treatment of AD. In addition, we systematically reviewed the research progress of SREBPs in AD, and this review will provide more innovative insights into the pathogenesis and treatment of AD and new strategies for drug development in AD.
Collapse
Affiliation(s)
- Siyuan Liu
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Xinzhu Li
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Panpan Fan
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Yujia Gu
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Aizhu Yang
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Weiyi Wang
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Lijun Zhou
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Huanhua Chen
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Fangyuan Zheng
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Junjie Lin
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Zihua Xu
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Qingchun Zhao
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| |
Collapse
|
9
|
Dunacka J, Świątek G, Wrona D. High Behavioral Reactivity to Novelty as a Susceptibility Factor for Memory and Anxiety Disorders in Streptozotocin-Induced Neuroinflammation as a Rat Model of Alzheimer's Disease. Int J Mol Sci 2024; 25:11562. [PMID: 39519114 PMCID: PMC11546707 DOI: 10.3390/ijms252111562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/18/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Individual differences in responsiveness to environmental factors, including stress reactivity and anxiety levels, which differ between high (HR) and low (LR) responders to novelty, might be risk factors for development of memory and anxiety disorders in sporadic Alzheimer's disease (sAD). In the present study, we investigated whether behavioral characteristics of the HR and LR rats, influence the progression of sAD (neuroinflammation, β-amyloid peptide, behavioral activity related to memory (Morris water maze) and anxiety (elevated plus maze, white and illuminated open field test) in streptozotocin (STZ)-induced neuroinflammation as a model of early pathophysiological alterations in sAD. Early (45 days) in disease progression, there was a more severe impairment of reference memory and higher levels of anxiety in HRs compared with LRs. Behavioral depression in HRs was associated with higher expression of β-amyloid deposits, particularly in the NAcS, and activation of microglia (CD68+ cells) in the hypothalamus, as opposed to less inflammation in the hippocampus, particularly in CA1, compared with LRs in late (90 days) sAD progression. Our findings suggest that rats with higher behavioral activity and increased responsivity to stressors show more rapid progression of disease and anxiety disorders compared with low responders to novelty in the STZ-induced sAD model.
Collapse
Affiliation(s)
| | | | - Danuta Wrona
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, 59 Wita Stwosza Str., 80-308 Gdansk, Poland; (J.D.); (G.Ś.)
| |
Collapse
|
10
|
Barrantes FJ. Cognitive synaptopathy: synaptic and dendritic spine dysfunction in age-related cognitive disorders. Front Aging Neurosci 2024; 16:1476909. [PMID: 39420927 PMCID: PMC11484076 DOI: 10.3389/fnagi.2024.1476909] [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: 08/06/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Cognitive impairment is a leading component of several neurodegenerative and neurodevelopmental diseases, profoundly impacting on the individual, the family, and society at large. Cognitive pathologies are driven by a multiplicity of factors, from genetic mutations and genetic risk factors, neurotransmitter-associated dysfunction, abnormal connectomics at the level of local neuronal circuits and broader brain networks, to environmental influences able to modulate some of the endogenous factors. Otherwise healthy older adults can be expected to experience some degree of mild cognitive impairment, some of which fall into the category of subjective cognitive deficits in clinical practice, while many neurodevelopmental and neurodegenerative diseases course with more profound alterations of cognition, particularly within the spectrum of the dementias. Our knowledge of the underlying neuropathological mechanisms at the root of this ample palette of clinical entities is far from complete. This review looks at current knowledge on synaptic modifications in the context of cognitive function along healthy ageing and cognitive dysfunction in disease, providing insight into differential diagnostic elements in the wide range of synapse alterations, from those associated with the mild cognitive changes of physiological senescence to the more profound abnormalities occurring at advanced clinical stages of dementia. I propose the term "cognitive synaptopathy" to encompass the wide spectrum of synaptic pathologies associated with higher brain function disorders.
Collapse
Affiliation(s)
- Francisco J. Barrantes
- Laboratory of Molecular Neurobiology, Biomedical Research Institute, Pontifical Catholic University of Argentina (UCA), Argentine Scientific and Technological Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
11
|
Wang J, Cao G, Liu Y, Chen S, Li H, Zheng B. Mendelian randomization study on simvastatin and gastric cancer: exploring the therapeutic potential of statins in oncology. Transl Cancer Res 2024; 13:4671-4677. [PMID: 39430821 PMCID: PMC11483371 DOI: 10.21037/tcr-24-576] [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: 04/07/2024] [Accepted: 08/01/2024] [Indexed: 10/22/2024]
Abstract
Background Gastric cancer ranks as the fifth most prevalent cancer and the third leading cause of cancer-related mortality worldwide, Statins, renowned for their cholesterol-lowering effects, have garnered interest for their potential roles in cancer prevention and treatment due to their pleiotropic effects, such as anti-proliferative, pro-apoptotic, and anti-inflammatory properties. This study aims to investigate the therapeutic potential of simvastatin, a widely prescribed statin, in the context of gastric cancer using Mendelian randomization (MR) to explore a possible causal relationship between simvastatin use and gastric cancer risk. Methods We conducted a two-sample MR analysis utilizing summary statistics from genome-wide association studies (GWAS). Data from the Integrative Epidemiology Unit (IEU) Open GWAS project included 462,933 participants and 9,851,867 single nucleotide polymorphisms (SNPs) for simvastatin, and 476,116 participants with 24,188,662 SNPs for gastric cancer. Instrumental variables screening criteria were stringent, resulting in 41 valid SNPs as instrumental variables. The MR analysis was performed using the inverse variance weighting (IVW), supplemented by MR-Egger, weighted median estimator (WME), weighted mode, and simple mode approaches. Heterogeneity and pleiotropy were assessed using IVW, MR-Egger tests, and the MR-PRESSO method. Results The IVW and WME analyses indicated a significant protective effect of simvastatin against gastric cancer [IVW: odds ratio (OR) =0.1459, 95% confidence interval (CI): -3.502 to -0.346, P=0.01; WME: OR =0.0347, 95% CI: -3.521 to 0.1610, P=0.03]. There was no significant difference between the results of the two MR analyses before and after the removal of outliers (P=0.76), and the Egger-intercept for horizontal pleiotropy testing was not significant (P=0.38). Leave-one-out sensitivity analysis supported the robustness of our findings. Conclusions This MR study provides evidence for a potential protective effect of simvastatin against gastric cancer, suggesting its consideration as an adjunct to traditional cancer therapies.
Collapse
Affiliation(s)
- Jiazhong Wang
- Department of Thyroid Surgery, Second Affiliated Hospital, Xi’an Jiaotong University School of Medicine, Xi’an, China
| | - Gang Cao
- Department of Thyroid Surgery, Second Affiliated Hospital, Xi’an Jiaotong University School of Medicine, Xi’an, China
| | - Yang Liu
- Department of Thyroid Surgery, Second Affiliated Hospital, Xi’an Jiaotong University School of Medicine, Xi’an, China
| | - Suo Chen
- Department of Thyroid Surgery, Second Affiliated Hospital, Xi’an Jiaotong University School of Medicine, Xi’an, China
| | - Haoyu Li
- Department of Thyroid Surgery, Second Affiliated Hospital, Xi’an Jiaotong University School of Medicine, Xi’an, China
| | - Bo Zheng
- Department of General Surgery, Xi’an Gaoxin Hospital, Xi’an, China
| |
Collapse
|
12
|
Wang Z, Rixiati Y, Jia C, Xu Y, Yin Z, Huang J, Dai J, Zhang Y. Causal effect of thyroid cancer on secondary primary malignancies: findings from the UK Biobank and FinnGen cohorts. Front Immunol 2024; 15:1434737. [PMID: 39391305 PMCID: PMC11464368 DOI: 10.3389/fimmu.2024.1434737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 09/10/2024] [Indexed: 10/12/2024] Open
Abstract
Background Existing epidemiological data indicated a correlation between thyroid cancer (THCA) and the risk of secondary primary malignancies (SPMs). However, the correlation does not always imply causality. Methods The Mendelian randomization (MR) analyses were performed to investigate the causal relationships between THCA and SPMs based on international multicenter data. Odds ratios (ORs) with 95% confidence intervals (95% CIs) were calculated. The Cancer Genome Atlas (TCGA) was used to explore potential mechanisms shared by THCA and bladder cancer (BLCA). Results Summary datasets of genome-wide association studies (GWAS) on 30 types of cancers were obtained from the United Kingdom Biobank (UKB) and FinnGen database. Meta-analysis of the UKB and FinnGen results revealed that THCA was significantly positively correlated with BLCA (OR = 1.140; 95% CI, 1.072-1.212; P < 0.001). Four genes, including WNT3, FAM171A2, MLLT11, and ULBP1, were identified as key genes shared by both TCHA and BLCA. Correlation analysis indicated that THCA may increase the risk of secondary BLCA through augmentation of N2 neutrophil infiltration. Conclusions This study showed that THCA was causally related to BLCA. It is recommended to conduct more rigorous screenings for BLCA during the follow-up of THCA patients.
Collapse
Affiliation(s)
- Zhengshi Wang
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Center of Thyroid Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | | | - Chengyou Jia
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Yong Xu
- Department of Laboratory, Yueyang Hospital, Hunan Normal University, Yueyang, China
| | - Zhiqiang Yin
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Center of Thyroid Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Junwen Huang
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Center of Thyroid Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiaqi Dai
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Center of Thyroid Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yun Zhang
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Center of Thyroid Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
13
|
Niemeyer CS, Merle L, Bubak AN, Dnate' Baxter B, Polese AG, Colon-Reyes K, Vang S, Hassell JE, Bruce KD, Nagel MA, Restrepo D. Olfactory and Trigeminal Routes of HSV-1 CNS Infection with Regional Microglial Heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.614340. [PMID: 39386674 PMCID: PMC11463476 DOI: 10.1101/2024.09.22.614340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Herpes simplex virus type 1 (HSV-1) primarily targets the oral and nasal epithelia before establishing latency in the trigeminal and other peripheral ganglia (TG). HSV-1 can also infect and go latent in the central nervous system (CNS) independent of latency in the TGs. Recent studies suggest entry to the CNS via two distinct routes: the TG-brainstem connection and olfactory nerve; however, to date, there is no characterization of brain regions targeted during HSV-1 primary infection. Furthermore, the immune response by microglia may also contribute to the heterogeneity between different brain regions. However, the response to HSV-1 by microglia has not been characterized in a region-specific manner. This study investigated the time course of HSV-1 spread within the olfactory epithelium (OE) and CNS following intranasal inoculation and the corresponding macrophage/microglial response in a C57BL/6 mouse model. We found an apical to basal spread of HSV-1 within the OE and underlying tissue accompanied by an inflammatory response of macrophages. OE Infection was followed by infection of a small subset of brain regions targeted by the TG in the brainstem, as well as other cranial nerve nuclei, including the vagus and hypoglossal nerve. Furthermore, other brain regions were positive for HSV-1 antigens, such as the locus coeruleus (LC), raphe nucleus (RaN), and hypothalamus, while sparing the hippocampus and cortex. Within each brain region, microglia activation also varied widely. These findings provide critical insights into the region-specific dissemination of HSV-1 within the CNS, elucidating potential mechanisms linking viral infection to neurological and neurodegenerative diseases. Importance This study sheds light on how herpes simplex virus type 1 (HSV-1) spreads within the brain after infecting the nasal passages. Our data reveals the distinct pattern of HSV-1 through the brain during a non-encephalitic infection. Furthermore, microglial activation was also temporally and spatially specific, with some regions of the brain having sustained microglial activation even in the absence of viral antigen. Previous reports have identified specific regions of the brain found to be positive for HSV-1 infection; however, to date, there has not been a concise investigation of the anatomical spread of HSV-1 and the regions of the brain consistently vulnerable to viral entry and spread. Understanding these region-specific differences in infection and immune response is crucial because it links HSV-1 infection to potential triggers for neurological and neurodegenerative diseases.
Collapse
|
14
|
Du D, Qiu JY, Zhao J, Yuan YD. Causal relationship between immune cells and pulmonary arterial hypertension: Mendelian randomization analysis. Medicine (Baltimore) 2024; 103:e39670. [PMID: 39287266 PMCID: PMC11404942 DOI: 10.1097/md.0000000000039670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Immunity and inflammation in pulmonary arterial hypertension (PAH) has gained more attention. This research aimed to investigate the potential causal connections between 731 immunophenotypes and the likelihood of developing PAH. We obtained immunocyte data and PAH from openly accessible database and used Mendelian randomization (MR) analysis to evaluate the causal association between each immunophenotype and PAH. Various statistical methods were employed: the MR-Egger, weighted median, inverse variance weighted (IVW), simple mode, and weighted mode. In the study of 731 different types of immune cells, it was found that 9 showed a potential positive connection (IVW P < .05) with increased risk of PAH, while 19 had a possible negative link to decreased risk. Following false discovery rate (FDR) adjustment, the analysis using the IVW method demonstrated that 5 immune phenotypes were significantly associated with PAH (FDR < 0.05, OR > 1). Conversely, there was a negative correlation between PAH and 4 immune cell types (FDR < 0.05, OR < 1). Sensitivity analyses suggested the robustness of all MR findings. This research, for the first time, has revealed indicative evidence of a causal link between circulating immune cell phenotypes and PAH through genetic mechanisms. These results underscore the importance of immune cells in the pathogenesis of PAH.
Collapse
Affiliation(s)
- Dan Du
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | | | | | | |
Collapse
|
15
|
A genome-wide association meta-analysis of all-cause and vascular dementia. Alzheimers Dement 2024; 20:5973-5995. [PMID: 39046104 PMCID: PMC11497727 DOI: 10.1002/alz.14115] [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: 06/09/2023] [Revised: 04/30/2024] [Accepted: 05/20/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Dementia is a multifactorial disease with Alzheimer's disease (AD) and vascular dementia (VaD) pathologies making the largest contributions. Yet, most genome-wide association studies (GWAS) focus on AD. METHODS We conducted a GWAS of all-cause dementia (ACD) and examined the genetic overlap with VaD. Our dataset includes 800,597 individuals, with 46,902 and 8702 cases of ACD and VaD, respectively. Known AD loci for ACD and VaD were replicated. Bioinformatic analyses prioritized genes that are likely functionally relevant and shared with closely related traits and risk factors. RESULTS For ACD, novel loci identified were associated with energy transport (SEMA4D), neuronal excitability (ANO3), amyloid deposition in the brain (RBFOX1), and magnetic resonance imaging markers of small vessel disease (SVD; HBEGF). Novel VaD loci were associated with hypertension, diabetes, and neuron maintenance (SPRY2, FOXA2, AJAP1, and PSMA3). DISCUSSION Our study identified genetic risks underlying ACD, demonstrating overlap with neurodegenerative processes, vascular risk factors, and cerebral SVD. HIGHLIGHTS We conducted the largest genome-wide association study of all-cause dementia (ACD) and vascular dementia (VaD). Known genetic variants associated with AD were replicated for ACD and VaD. Functional analyses identified novel loci for ACD and VaD. Genetic risks of ACD overlapped with neurodegeneration, vascular risk factors, and cerebral small vessel disease.
Collapse
|
16
|
Kirby A, Porter T, Adewuyi EO, Laws SM. Investigating Genetic Overlap between Alzheimer's Disease, Lipids, and Coronary Artery Disease: A Large-Scale Genome-Wide Cross Trait Analysis. Int J Mol Sci 2024; 25:8814. [PMID: 39201500 PMCID: PMC11354907 DOI: 10.3390/ijms25168814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/10/2024] [Accepted: 08/11/2024] [Indexed: 09/02/2024] Open
Abstract
There is evidence to support a link between abnormal lipid metabolism and Alzheimer's disease (AD) risk. Similarly, observational studies suggest a comorbid relationship between AD and coronary artery disease (CAD). However, the intricate biological mechanisms of AD are poorly understood, and its relationship with lipids and CAD traits remains unresolved. Conflicting evidence further underscores the ongoing investigation into this research area. Here, we systematically assess the cross-trait genetic overlap of AD with 13 representative lipids (from eight classes) and seven CAD traits, leveraging robust analytical methods, well-powered large-scale genetic data, and rigorous replication testing. Our main analysis demonstrates a significant positive global genetic correlation of AD with triglycerides and all seven CAD traits assessed-angina pectoris, cardiac dysrhythmias, coronary arteriosclerosis, ischemic heart disease, myocardial infarction, non-specific chest pain, and coronary artery disease. Gene-level analyses largely reinforce these findings and highlight the genetic overlap between AD and three additional lipids: high-density lipoproteins (HDLs), low-density lipoproteins (LDLs), and total cholesterol. Moreover, we identify genome-wide significant genes (Fisher's combined p value [FCPgene] < 2.60 × 10-6) shared across AD, several lipids, and CAD traits, including WDR12, BAG6, HLA-DRA, PHB, ZNF652, APOE, APOC4, PVRL2, and TOMM40. Mendelian randomisation analysis found no evidence of a significant causal relationship between AD, lipids, and CAD traits. However, local genetic correlation analysis identifies several local pleiotropic hotspots contributing to the relationship of AD with lipids and CAD traits across chromosomes 6, 8, 17, and 19. Completing a three-way analysis, we confirm a strong genetic correlation between lipids and CAD traits-HDL and sphingomyelin demonstrate negative correlations, while LDL, triglycerides, and total cholesterol show positive correlations. These findings support genetic overlap between AD, specific lipids, and CAD traits, implicating shared but non-causal genetic susceptibility. The identified shared genes and pleiotropic hotspots are valuable targets for further investigation into AD and, potentially, its comorbidity with CAD traits.
Collapse
Affiliation(s)
- Artika Kirby
- Centre for Precision Health, Edith Cowan University, Joondalup, WA 6027, Australia; (A.K.); (T.P.)
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Tenielle Porter
- Centre for Precision Health, Edith Cowan University, Joondalup, WA 6027, Australia; (A.K.); (T.P.)
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia
| | - Emmanuel O. Adewuyi
- Centre for Precision Health, Edith Cowan University, Joondalup, WA 6027, Australia; (A.K.); (T.P.)
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Simon M. Laws
- Centre for Precision Health, Edith Cowan University, Joondalup, WA 6027, Australia; (A.K.); (T.P.)
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia
| |
Collapse
|
17
|
Teng S, Han C, Zhou J, He Z, Qian W. m 5C RNA methylation: a potential mechanism for infectious Alzheimer's disease. Front Cell Dev Biol 2024; 12:1440143. [PMID: 39175875 PMCID: PMC11338875 DOI: 10.3389/fcell.2024.1440143] [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: 05/30/2024] [Accepted: 07/30/2024] [Indexed: 08/24/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder caused by a variety of factors, including age, genetic susceptibility, cardiovascular disease, traumatic brain injury, and environmental factors. The pathogenesis of AD is largely associated with the overproduction and accumulation of amyloid-β peptides and the hyperphosphorylation of tau protein in the brain. Recent studies have identified the presence of diverse pathogens, including viruses, bacteria, and parasites, in the tissues of AD patients, underscoring the critical role of central nervous system infections in inducing pathological changes associated with AD. Nevertheless, it remains unestablished about the specific mechanism by which infections lead to the occurrence of AD. As an important post-transcriptional RNA modification, RNA 5-methylcytosine (m5C) methylation regulates a wide range of biological processes, including RNA splicing, nuclear export, stability, and translation, therefore affecting cellular function. Moreover, it has been recently demonstrated that multiple pathogenic microbial infections are associated with the m5C methylation of the host. However, the role of m5C methylation in infectious AD is still uncertain. Therefore, this review discusses the mechanisms of pathogen-induced AD and summarizes research on the molecular mechanisms of m5C methylation in infectious AD, thereby providing new insight into exploring the mechanism underlying infectious AD.
Collapse
Affiliation(s)
- Sisi Teng
- Department of Neurology, Shangjinnanfu Hospital, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Cunqiao Han
- Department of Emergency, Shangjinnanfu Hospital, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jian Zhou
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Zhenyan He
- Department of Neurosurgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Weiwei Qian
- Department of Emergency, Shangjinnanfu Hospital, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Emergency Medicine, Laboratory of Emergency Medicine, West China Hospital, and Disaster Medical Center, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
18
|
Wang N, Ren L, Li Z, Hu Y, Zhou J, Sun Q, Pei B, Li X, Peng W, Yu J, Zhao R, Huang Z, Chen Z, Huang G. The association between SII and aging: evidence from NHANES 1999-2018. Front Public Health 2024; 12:1418385. [PMID: 38993709 PMCID: PMC11236748 DOI: 10.3389/fpubh.2024.1418385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
Background The study aimed to examine the association between the systemic immune-inflammation index (SII), a contemporary metric of systemic inflammatory response, and biological aging, which are closely interconnected processes. Methods This cross-sectional study utilized 10 cycles of data from the NHANES database spanning from 1990 to 2018. The study examined the relationship between the SII index, calculated as P * N/L, where P represents preoperative peripheral platelet count, N represents neutrophil count, and L represents lymphocyte count, and biological aging. Biological aging was assessed through various methods, such as phenotypic age, phenotypic age acceleration (PhenoAgeAccel), biological age, and biological age acceleration (BioAgeAccel). Correlations were analyzed using weighted linear regression and subgroup analysis. Results Among the 7,491 participants analyzed, the average age was 45.26 ± 0.34 years, with 52.16% being female. The average phenotypic and biological ages were 40.06 ± 0.36 and 45.89 ± 0.32 years, respectively. Following adjustment for potential confounders, elevated SII scores were linked to increased phenotypic age, biological age, Phenotypic age acceleration, and Biological age acceleration. Positive correlations were observed between health behavior and health factor scores and biological aging, with stronger associations seen for health factors. In health factor-specific analyses, the β coefficient was notably higher for high BMI. The robust positive associations between SII scores and both phenotypic age and biological age in the stratified analyses were consistently observed across all strata. Conclusion The evidence from the NHANES data indicate that SII may serve as a valuable marker for assessing different facets of aging and health outcomes, such as mortality and the aging process. Additional research is warranted to comprehensively elucidate the implications of SII in the aging process and its utility as a clinical instrument for evaluating and addressing age-related ailments.
Collapse
Affiliation(s)
- Nanbu Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lian Ren
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Zhongnan Medical Journal Press, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ziyuan Li
- The First Clinical Medical College, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Yunhao Hu
- The First Clinical Medical College, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Jingpei Zhou
- The First Clinical Medical College, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Quan Sun
- The First Clinical Medical College, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Bin Pei
- Department of Evidence-Based Medicine Center, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Wuhan, China
| | - Xinyu Li
- The First Clinical Medical College, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Wanqing Peng
- The First Clinical Medical College, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Jinyan Yu
- The First Clinical Medical College, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Renhui Zhao
- The First Clinical Medical College, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Ziting Huang
- The First Clinical Medical College, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Zhenhu Chen
- Acupuncture and Rehabilitation Centre, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guoxin Huang
- Department of Evidence-Based Medicine Center, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Wuhan, China
| |
Collapse
|
19
|
Yang Q, Han X, Ye M, Jiang T, Wang B, Zhang Z, Li F. Association of genetically predicted 486 blood metabolites on the risk of Alzheimer's disease: a Mendelian randomization study. Front Aging Neurosci 2024; 16:1372605. [PMID: 38681667 PMCID: PMC11047179 DOI: 10.3389/fnagi.2024.1372605] [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: 01/18/2024] [Accepted: 03/27/2024] [Indexed: 05/01/2024] Open
Abstract
Background Studies have reported that metabolic disturbance exhibits in patients with Alzheimer's disease (AD). Still, the presence of definitive evidence concerning the genetic effect of metabolites on AD risk remains insufficient. A systematic exploration of the genetic association between blood metabolites and AD would contribute to the identification of new targets for AD screening and prevention. Methods We conducted an exploratory two-sample Mendelian randomization (MR) study aiming to preliminarily identify the potential metabolites involved in AD development. A genome-wide association study (GWAS) involving 7,824 participants provided information on 486 human blood metabolites. Outcome information was obtained from a large-scale GWAS meta-analysis of AD, encompassing 21,982 cases and 41,944 controls of Europeans. The primary two-sample MR analysis utilized the inverse variance weighted (IVW) model while supplementary analyses used Weighted median (WM), MR Egger, Simple mode, and Weighted mode, followed by sensitivity analyses such as the heterogeneity test, horizontal pleiotropy test, and leave-one-out analysis. For the further identification of metabolites, replication and meta-analysis with FinnGen data, steiger test, linkage disequilibrium score regression, confounding analysis, and were conducted for further evaluation. Multivariable MR was performed to assess the direct effect of metabolites on AD. Besides, an extra replication analysis with EADB data was conducted for final evaluation of the most promising findings. Results After rigorous genetic variant selection, IVW, complementary analysis, sensitivity analysis, replication and meta-analysis with the FinnGen data, five metabolites (epiandrosterone sulfate, X-12680, pyruvate, docosapentaenoate, and 1-stearoylglycerophosphocholine) were identified as being genetically associated with AD. MVMR analysis disclosed that genetically predicted these four known metabolites can directly influence AD independently of other metabolites. Only epiandrosterone sulfate and X-12680 remained suggestive significant associations with AD after replication analysis with the EADB data. Conclusion By integrating genomics with metabonomics, this study furnishes evidence substantiating the genetic association of epiandrosterone sulfate and X-12680 with AD. These findings hold significance for the screening, prevention, and treatment strategies for AD.
Collapse
Affiliation(s)
- Qiqi Yang
- Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- The First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, China
| | - Xinyu Han
- The First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, China
| | - Min Ye
- The First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, China
| | - Tianxin Jiang
- Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Baoguo Wang
- Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Zhenfeng Zhang
- Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Fei Li
- Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- Intelligent Manufacturing Institute, Hefei University of Technology, Hefei, China
| |
Collapse
|
20
|
Jaisa-aad M, Muñoz-Castro C, Serrano-Pozo A. Update on modifiable risk factors for Alzheimer's disease and related dementias. Curr Opin Neurol 2024; 37:166-181. [PMID: 38265228 PMCID: PMC10932854 DOI: 10.1097/wco.0000000000001243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
PURPOSE OF REVIEW All human beings undergo a lifelong cumulative exposure to potentially preventable adverse factors such as toxins, infections, traumatisms, and cardiovascular risk factors, collectively termed exposome. The interplay between the individual's genetics and exposome is thought to have a large impact in health outcomes such as cancer and cardiovascular disease. Likewise, a growing body of evidence is supporting the idea that preventable factors explain a sizable proportion of Alzheimer's disease and related dementia (ADRD) cases. RECENT FINDINGS Here, we will review the most recent epidemiological, experimental preclinical, and interventional clinical studies examining some of these potentially modifiable risk factors for ADRD. We will focus on new evidence regarding cardiovascular risk factors, air pollution, viral and other infectious agents, traumatic brain injury, and hearing loss. SUMMARY While greater and higher quality epidemiological and experimental evidence is needed to unequivocally confirm their causal link with ADRD and/or unravel the underlying mechanisms, these modifiable risk factors may represent a window of opportunity to reduce ADRD incidence and prevalence at the population level via health screenings, and education and health policies.
Collapse
Affiliation(s)
- Methasit Jaisa-aad
- Massachusetts General Hospital, Boston, MA 02114
- Harvard Medical School, Boston, MA 02115
| | - Clara Muñoz-Castro
- Massachusetts General Hospital, Boston, MA 02114
- Harvard Medical School, Boston, MA 02115
- Universidad de Sevilla, Sevilla (Spain)
| | - Alberto Serrano-Pozo
- Massachusetts General Hospital, Boston, MA 02114
- Harvard Medical School, Boston, MA 02115
- Massachusetts Alzheimer’s Disease Research Center
| |
Collapse
|
21
|
Gao S, Wang T, Han Z, Hu Y, Zhu P, Xue Y, Huang C, Chen Y, Liu G. Interpretation of 10 years of Alzheimer's disease genetic findings in the perspective of statistical heterogeneity. Brief Bioinform 2024; 25:bbae140. [PMID: 38711368 PMCID: PMC11074593 DOI: 10.1093/bib/bbae140] [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: 08/21/2023] [Revised: 02/22/2024] [Accepted: 03/14/2024] [Indexed: 05/08/2024] Open
Abstract
Common genetic variants and susceptibility loci associated with Alzheimer's disease (AD) have been discovered through large-scale genome-wide association studies (GWAS), GWAS by proxy (GWAX) and meta-analysis of GWAS and GWAX (GWAS+GWAX). However, due to the very low repeatability of AD susceptibility loci and the low heritability of AD, these AD genetic findings have been questioned. We summarize AD genetic findings from the past 10 years and provide a new interpretation of these findings in the context of statistical heterogeneity. We discovered that only 17% of AD risk loci demonstrated reproducibility with a genome-wide significance of P < 5.00E-08 across all AD GWAS and GWAS+GWAX datasets. We highlighted that the AD GWAS+GWAX with the largest sample size failed to identify the most significant signals, the maximum number of genome-wide significant genetic variants or maximum heritability. Additionally, we identified widespread statistical heterogeneity in AD GWAS+GWAX datasets, but not in AD GWAS datasets. We consider that statistical heterogeneity may have attenuated the statistical power in AD GWAS+GWAX and may contribute to explaining the low repeatability (17%) of genome-wide significant AD susceptibility loci and the decreased AD heritability (40-2%) as the sample size increased. Importantly, evidence supports the idea that a decrease in statistical heterogeneity facilitates the identification of genome-wide significant genetic loci and contributes to an increase in AD heritability. Collectively, current AD GWAX and GWAS+GWAX findings should be meticulously assessed and warrant additional investigation, and AD GWAS+GWAX should employ multiple meta-analysis methods, such as random-effects inverse variance-weighted meta-analysis, which is designed specifically for statistical heterogeneity.
Collapse
Affiliation(s)
- Shan Gao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, No. 10, Xitoutiao, You’an Men Wai, Fengtai District, Beijing 100069, China
| | - Tao Wang
- Chinese Institute for Brain Research, No. 26, Kexueyuan Road, Changping District, Beijing 102206, China
| | - Zhifa Han
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, No. 5, Dongdan Santichao, Dongcheng District, Beijing 100193, China
| | - Yang Hu
- School of Computer Science and Technology, Harbin Institute of Technology, No. 92, Xidazhi Street, Nangang District, Harbin 150006, China
| | - Ping Zhu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, No. 10, Xitoutiao, You’an Men Wai, Fengtai District, Beijing 100069, China
| | - Yanli Xue
- School of Biomedical Engineering, Capital Medical University, No. 10 Xitoutiao, You'an Men Wai, Fengtai District, Beijing 100069, China
| | - Chen Huang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida WaiLong, Taipa 999078, Macao SAR, China
| | - Yan Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, No. 22, Wenchang Road, Wuhu 241002, Anhui, China
- Institute of Chronic Disease Prevention and Control, Wannan Medical College, No. 22, Wenchang Road, Wuhu 241002, Anhui, China
| | - Guiyou Liu
- Chinese Institute for Brain Research, No. 26, Kexueyuan Road, Changping District, Beijing 102206, China
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, No. 22, Wenchang Road, Wuhu 241002, Anhui, China
- Institute of Chronic Disease Prevention and Control, Wannan Medical College, No. 22, Wenchang Road, Wuhu 241002, Anhui, China
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong, Department of Neurology, Second Affiliated Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian 271000, Shandong, China
- Beijing Key Laboratory of Hypoxia Translational Medicine, National Engineering Laboratory of Internet Medical Diagnosis and Treatment Technology, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Road, Xicheng District, Beijing 100053, China
| |
Collapse
|
22
|
Han S, Yao J, Yamazaki H, Streicher SA, Rao J, Nianogo RA, Zhang Z, Huang BZ. Genetically Determined Circulating Lactase/Phlorizin Hydrolase Concentrations and Risk of Colorectal Cancer: A Two-Sample Mendelian Randomization Study. Nutrients 2024; 16:808. [PMID: 38542719 PMCID: PMC10975724 DOI: 10.3390/nu16060808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/06/2024] [Accepted: 03/10/2024] [Indexed: 04/01/2024] Open
Abstract
Previous research has found that milk is associated with a decreased risk of colorectal cancer (CRC). However, it is unclear whether the milk digestion by the enzyme lactase-phlorizin hydrolase (LPH) plays a role in CRC susceptibility. Our study aims to investigate the direct causal relationship of CRC risk with LPH levels by applying a two-sample Mendelian Randomization (MR) strategy. Genetic instruments for LPH were derived from the Fenland Study, and CRC-associated summary statistics for these instruments were extracted from the FinnGen Study, PLCO Atlas Project, and Pan-UK Biobank. Primary MR analyses focused on a cis-variant (rs4988235) for LPH levels, with results integrated via meta-analysis. MR analyses using all variants were also undertaken. This analytical approach was further extended to assess CRC subtypes (colon and rectal). Meta-analysis across the three datasets illustrated an inverse association between genetically predicted LPH levels and CRC risk (OR: 0.92 [95% CI, 0.89-0.95]). Subtype analyses revealed associations of elevated LPH levels with reduced risks for both colon (OR: 0.92 [95% CI, 0.89-0.96]) and rectal cancer (OR: 0.92 [95% CI, 0.87, 0.98]). Consistency was observed across varied analytical methods and datasets. Further exploration is warranted to unveil the underlying mechanisms and validate LPH's potential role in CRC prevention.
Collapse
Affiliation(s)
- Sihao Han
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, CA 90095, USA; (J.Y.); (J.R.); (R.A.N.); (Z.Z.)
| | - Jiemin Yao
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, CA 90095, USA; (J.Y.); (J.R.); (R.A.N.); (Z.Z.)
| | - Hajime Yamazaki
- Section of Clinical Epidemiology, Department of Community Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8303, Japan;
- Center for Innovative Research for Communities and Clinical Excellence (CiRC2LE), Fukushima Medical University, Fukushima 960-1295, Japan
| | - Samantha A. Streicher
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI 96813, USA;
| | - Jianyu Rao
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, CA 90095, USA; (J.Y.); (J.R.); (R.A.N.); (Z.Z.)
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Roch A. Nianogo
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, CA 90095, USA; (J.Y.); (J.R.); (R.A.N.); (Z.Z.)
| | - Zuofeng Zhang
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, CA 90095, USA; (J.Y.); (J.R.); (R.A.N.); (Z.Z.)
| | - Brian Z. Huang
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA;
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
23
|
Xu F, Dirsch O, Dahmen U. Causal relationship between psychological factors and hepatocellular carcinoma as revealed by Mendelian randomization. J Cancer Res Clin Oncol 2024; 150:100. [PMID: 38383696 PMCID: PMC10881603 DOI: 10.1007/s00432-024-05617-5] [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: 12/21/2023] [Accepted: 01/09/2024] [Indexed: 02/23/2024]
Abstract
PURPOSE The impact of psychological factors on the incidence of hepatocellular carcinoma (HCC) in humans remains unclear. Mendelian randomization (MR) study is a novel approach aimed at unbiased detection of causal effects. Therefore, we conducted a two-sample MR to determine if there is a causal relationship between psychological distress (PD), participation in leisure/social activities of religious groups (LARG), and HCC. METHODS The genetic summary data of exposures and outcome were retrieved from genome-wide association studies (GWAS). We used PD and LARG as exposures and HCC as outcome. Five MR methods were used to investigate the causal relationship between PD, LARG, and HCC. The result of inverse variance weighted (IVW) method was deemed as principal result. Besides, we performed a comprehensive sensitivity analysis to verify the robustness of the results. RESULTS The IVW results showed that PD [odds ratio (OR) 1.006, 95% confidence interval (CI) 1.000-1.011, P = 0.033] and LARG (OR 0.994, 95% CI 0.988-1.000, P = 0.035) were causally associated with the incidence of HCC. Sensitivity analysis did not identify any bias in the results. CONCLUSION PD turned out to be a mild risk factor for HCC. In contrast, LARG is a protective factor for HCC. Therefore, it is highly recommended that people with PD are seeking positive leisure activities such as participation in formal religious social activities, which may help them reduce the risk of HCC.
Collapse
Affiliation(s)
- Fengming Xu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, China
- Else Kröner Graduate School for Medical Students "JSAM", Jena University Hospital, 07747, Jena, Germany
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747, Jena, Germany
| | - Olaf Dirsch
- Institute of Pathology, Klinikum Chemnitz gGmbH, 09111, Chemnitz, Germany
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747, Jena, Germany.
| |
Collapse
|
24
|
Liu Y, Tan Y, Zhang Z, Yi M, Zhu L, Peng W. The interaction between ageing and Alzheimer's disease: insights from the hallmarks of ageing. Transl Neurodegener 2024; 13:7. [PMID: 38254235 PMCID: PMC10804662 DOI: 10.1186/s40035-024-00397-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Ageing is a crucial risk factor for Alzheimer's disease (AD) and is characterised by systemic changes in both intracellular and extracellular microenvironments that affect the entire body instead of a single organ. Understanding the specific mechanisms underlying the role of ageing in disease development can facilitate the treatment of ageing-related diseases, such as AD. Signs of brain ageing have been observed in both AD patients and animal models. Alleviating the pathological changes caused by brain ageing can dramatically ameliorate the amyloid beta- and tau-induced neuropathological and memory impairments, indicating that ageing plays a crucial role in the pathophysiological process of AD. In this review, we summarize the impact of several age-related factors on AD and propose that preventing pathological changes caused by brain ageing is a promising strategy for improving cognitive health.
Collapse
Affiliation(s)
- Yuqing Liu
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China
| | - Yejun Tan
- School of Mathematics, University of Minnesota Twin Cities, Minneapolis, MN, 55455, USA
| | - Zheyu Zhang
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China
| | - Min Yi
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China
| | - Lemei Zhu
- Academician Workstation, Changsha Medical University, Changsha, 410219, People's Republic of China
| | - Weijun Peng
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China.
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China.
| |
Collapse
|
25
|
Newby D, Orgeta V, Marshall CR, Lourida I, Albertyn CP, Tamburin S, Raymont V, Veldsman M, Koychev I, Bauermeister S, Weisman D, Foote IF, Bucholc M, Leist AK, Tang EYH, Tai XY, Llewellyn DJ, Ranson JM. Artificial intelligence for dementia prevention. Alzheimers Dement 2023; 19:5952-5969. [PMID: 37837420 PMCID: PMC10843720 DOI: 10.1002/alz.13463] [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: 04/12/2023] [Revised: 08/01/2023] [Accepted: 08/07/2023] [Indexed: 10/16/2023]
Abstract
INTRODUCTION A wide range of modifiable risk factors for dementia have been identified. Considerable debate remains about these risk factors, possible interactions between them or with genetic risk, and causality, and how they can help in clinical trial recruitment and drug development. Artificial intelligence (AI) and machine learning (ML) may refine understanding. METHODS ML approaches are being developed in dementia prevention. We discuss exemplar uses and evaluate the current applications and limitations in the dementia prevention field. RESULTS Risk-profiling tools may help identify high-risk populations for clinical trials; however, their performance needs improvement. New risk-profiling and trial-recruitment tools underpinned by ML models may be effective in reducing costs and improving future trials. ML can inform drug-repurposing efforts and prioritization of disease-modifying therapeutics. DISCUSSION ML is not yet widely used but has considerable potential to enhance precision in dementia prevention. HIGHLIGHTS Artificial intelligence (AI) is not widely used in the dementia prevention field. Risk-profiling tools are not used in clinical practice. Causal insights are needed to understand risk factors over the lifespan. AI will help personalize risk-management tools for dementia prevention. AI could target specific patient groups that will benefit most for clinical trials.
Collapse
Affiliation(s)
- Danielle Newby
- University of Oxford, Department of Psychiatry, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Vasiliki Orgeta
- Division of Psychiatry, University College London, London, W1T 7BN, UK
| | - Charles R Marshall
- Preventive Neurology Unit, Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, E1 4NS, UK
- Department of Neurology, Royal London Hospital, London, E1 1BB, UK
| | - Ilianna Lourida
- Population Health Sciences Institute, Newcastle University, Newcastle, NE2 4AX, UK
- University of Exeter Medical School, Exeter, EX1 2HZ, UK
| | - Christopher P Albertyn
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, SE5 8AF, UK
| | - Stefano Tamburin
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, 37129, Italy
| | - Vanessa Raymont
- University of Oxford, Department of Psychiatry, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Michele Veldsman
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, OX3 9DU, UK
- Department of Experimental Psychology, University of Oxford, Oxford, OX2 6GG, UK
| | - Ivan Koychev
- University of Oxford, Department of Psychiatry, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Sarah Bauermeister
- University of Oxford, Department of Psychiatry, Warneford Hospital, Oxford, OX3 7JX, UK
| | - David Weisman
- Abington Neurological Associates, Abington, PA 19001, USA
| | - Isabelle F Foote
- Preventive Neurology Unit, Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, E1 4NS, UK
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Magda Bucholc
- Cognitive Analytics Research Lab, School of Computing, Engineering & Intelligent Systems, Ulster University, Derry, BT48 7JL, UK
| | - Anja K Leist
- Institute for Research on Socio-Economic Inequality (IRSEI), Department of Social Sciences, University of Luxembourg, L-4365, Luxembourg
| | - Eugene Y H Tang
- Population Health Sciences Institute, Newcastle University, Newcastle, NE2 4AX, UK
| | - Xin You Tai
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, OX3 9DU, UK
- Division of Clinical Neurology, John Radcliffe Hospital, Oxford University Hospitals Trust, Oxford, OX3 9DU, UK
| | | | - David J. Llewellyn
- University of Exeter Medical School, Exeter, EX1 2HZ, UK
- The Alan Turing Institute, London, NW1 2DB, UK
| | | |
Collapse
|
26
|
Kjeldsen EW, Luo J, Nordestgaard LT, Sandau N, Frikke-Schmidt R. Reevaluating the Role of High-Density Lipoprotein Cholesterol: New Perspectives on Cardiovascular Disease and Alzheimer Disease. Clin Chem 2023; 69:1329-1332. [PMID: 37700571 DOI: 10.1093/clinchem/hvad126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 07/17/2023] [Indexed: 09/14/2023]
Affiliation(s)
- Emilie Westerlin Kjeldsen
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen 2100, Denmark
| | - Jiao Luo
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen 2100, Denmark
| | - Liv Tybjærg Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen 2100, Denmark
| | - Nicolai Sandau
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen 2100, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen 2100, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| |
Collapse
|
27
|
Ferguson EL, Zimmerman SC, Jiang C, Choi M, Swinnerton K, Choudhary V, Meyers TJ, Hoffmann TJ, Gilsanz P, Oni-Orisan A, Whitmer RA, Risch N, Krauss RM, Schaefer CA, Glymour MM. Low- and High-Density Lipoprotein Cholesterol and Dementia Risk Over 17 Years of Follow-up Among Members of a Large Health Care Plan. Neurology 2023; 101:e2172-e2184. [PMID: 37793911 PMCID: PMC10663022 DOI: 10.1212/wnl.0000000000207876] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/24/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND AND OBJECTIVES The associations of high-density lipoprotein cholesterol (HDL-C) and low-density lipoprotein cholesterol (LDL-C) with dementia risk in later life may be complex, and few studies have sufficient data to model nonlinearities or adequately adjust for statin use. We evaluated the observational associations of HDL-C and LDL-C with incident dementia in a large and well-characterized cohort with linked survey and electronic health record (EHR) data. METHODS Kaiser Permanente Northern California health plan members aged 55 years and older who completed a health behavior survey between 2002 and 2007, had no history of dementia before the survey, and had laboratory measurements of cholesterol within 2 years after survey completion were followed up through December 2020 for incident dementia (Alzheimer disease-related dementia [ADRD]; Alzheimer disease, vascular dementia, and/or nonspecific dementia) based on ICD-9 or ICD-10 codes in EHRs. We used Cox models for incident dementia with follow-up time beginning 2 years postsurvey (after cholesterol measurement) and censoring at end of membership, death, or end of study period. We evaluated nonlinearities using B-splines, adjusted for demographic, clinical, and survey confounders, and tested for effect modification by baseline age or prior statin use. RESULTS A total of 184,367 participants [mean age at survey = 69.5 years, mean HDL-C = 53.7 mg/dL (SD = 15.0), mean LDL-C = 108 mg/dL (SD = 30.6)] were included. Higher and lower HDL-C values were associated with elevated ADRD risk compared with the middle quantile: HDL-C in the lowest quintile was associated with an HR of 1.07 (95% CI 1.03-1.11), and HDL-C in the highest quintile was associated with an HR of 1.15 (95% CI 1.11-1.20). LDL-C was not associated with dementia risk overall, but statin use qualitatively modified the association. Higher LDL-C was associated with a slightly greater risk of ADRD for statin users (53% of the sample, HR per 10 mg/dL increase = 1.01, 95% CI 1.01-1.02) and a lower risk for nonusers (HR per 10 mg/dL increase = 0.98; 95% CI 0.97-0.99). There was evidence for effect modification by age with linear HDL-C (p = 0.003) but not LDL-C (p = 0.59). DISCUSSION Both low and high levels of HDL-C were associated with elevated dementia risk. The association between LDL-C and dementia risk was modest.
Collapse
Affiliation(s)
- Erin L Ferguson
- From the Department of Epidemiology and Biostatistics (E.L.F., S.C.Z., M.C., K.S., T.J.H., N.R., M.M.G.), University of California, San Francisco; Kaiser Permanente Division of Research (C.J., V.C., T.J.M., P.G., C.A.S.), Oakland; Institute for Human Genetics (A.O.-O., N.R.); Department of Clinical Pharmacy (A.O.-O.), University of California, San Francisco; Department of Public Health Sciences (R.A.W.), University of California, Davis; and Departments of Pediatrics and Medicine (R.M.K.), University of California, San Francisco. K.S. is currently affiliated with the VA Boston Healthcare System, MA. T.J.H. is currently affiliated with the Department of Epidemiology and Biostatistics, University of California, San Francisco, and the Institute for Human Genetics, University of California, San Francisco. P.G. is currently affiliated with the Kaiser Permanente Division of Research, Oakland, CA, and the Department of Epidemiology and Biostatistics, University of California, San Francisco. N.R. is currently affiliated with the Department of Epidemiology and Biostatistics, the Institute for Human Genetics, University of California, San Francisco, and the Kaiser Permanente Division of Research, Oakland, CA. M.M.G. is currently affiliated with the Department of Epidemiology, Boston University School of Public Health, MA.
| | - Scott C Zimmerman
- From the Department of Epidemiology and Biostatistics (E.L.F., S.C.Z., M.C., K.S., T.J.H., N.R., M.M.G.), University of California, San Francisco; Kaiser Permanente Division of Research (C.J., V.C., T.J.M., P.G., C.A.S.), Oakland; Institute for Human Genetics (A.O.-O., N.R.); Department of Clinical Pharmacy (A.O.-O.), University of California, San Francisco; Department of Public Health Sciences (R.A.W.), University of California, Davis; and Departments of Pediatrics and Medicine (R.M.K.), University of California, San Francisco. K.S. is currently affiliated with the VA Boston Healthcare System, MA. T.J.H. is currently affiliated with the Department of Epidemiology and Biostatistics, University of California, San Francisco, and the Institute for Human Genetics, University of California, San Francisco. P.G. is currently affiliated with the Kaiser Permanente Division of Research, Oakland, CA, and the Department of Epidemiology and Biostatistics, University of California, San Francisco. N.R. is currently affiliated with the Department of Epidemiology and Biostatistics, the Institute for Human Genetics, University of California, San Francisco, and the Kaiser Permanente Division of Research, Oakland, CA. M.M.G. is currently affiliated with the Department of Epidemiology, Boston University School of Public Health, MA
| | - Chen Jiang
- From the Department of Epidemiology and Biostatistics (E.L.F., S.C.Z., M.C., K.S., T.J.H., N.R., M.M.G.), University of California, San Francisco; Kaiser Permanente Division of Research (C.J., V.C., T.J.M., P.G., C.A.S.), Oakland; Institute for Human Genetics (A.O.-O., N.R.); Department of Clinical Pharmacy (A.O.-O.), University of California, San Francisco; Department of Public Health Sciences (R.A.W.), University of California, Davis; and Departments of Pediatrics and Medicine (R.M.K.), University of California, San Francisco. K.S. is currently affiliated with the VA Boston Healthcare System, MA. T.J.H. is currently affiliated with the Department of Epidemiology and Biostatistics, University of California, San Francisco, and the Institute for Human Genetics, University of California, San Francisco. P.G. is currently affiliated with the Kaiser Permanente Division of Research, Oakland, CA, and the Department of Epidemiology and Biostatistics, University of California, San Francisco. N.R. is currently affiliated with the Department of Epidemiology and Biostatistics, the Institute for Human Genetics, University of California, San Francisco, and the Kaiser Permanente Division of Research, Oakland, CA. M.M.G. is currently affiliated with the Department of Epidemiology, Boston University School of Public Health, MA
| | - Minhyuk Choi
- From the Department of Epidemiology and Biostatistics (E.L.F., S.C.Z., M.C., K.S., T.J.H., N.R., M.M.G.), University of California, San Francisco; Kaiser Permanente Division of Research (C.J., V.C., T.J.M., P.G., C.A.S.), Oakland; Institute for Human Genetics (A.O.-O., N.R.); Department of Clinical Pharmacy (A.O.-O.), University of California, San Francisco; Department of Public Health Sciences (R.A.W.), University of California, Davis; and Departments of Pediatrics and Medicine (R.M.K.), University of California, San Francisco. K.S. is currently affiliated with the VA Boston Healthcare System, MA. T.J.H. is currently affiliated with the Department of Epidemiology and Biostatistics, University of California, San Francisco, and the Institute for Human Genetics, University of California, San Francisco. P.G. is currently affiliated with the Kaiser Permanente Division of Research, Oakland, CA, and the Department of Epidemiology and Biostatistics, University of California, San Francisco. N.R. is currently affiliated with the Department of Epidemiology and Biostatistics, the Institute for Human Genetics, University of California, San Francisco, and the Kaiser Permanente Division of Research, Oakland, CA. M.M.G. is currently affiliated with the Department of Epidemiology, Boston University School of Public Health, MA
| | - Kaitlin Swinnerton
- From the Department of Epidemiology and Biostatistics (E.L.F., S.C.Z., M.C., K.S., T.J.H., N.R., M.M.G.), University of California, San Francisco; Kaiser Permanente Division of Research (C.J., V.C., T.J.M., P.G., C.A.S.), Oakland; Institute for Human Genetics (A.O.-O., N.R.); Department of Clinical Pharmacy (A.O.-O.), University of California, San Francisco; Department of Public Health Sciences (R.A.W.), University of California, Davis; and Departments of Pediatrics and Medicine (R.M.K.), University of California, San Francisco. K.S. is currently affiliated with the VA Boston Healthcare System, MA. T.J.H. is currently affiliated with the Department of Epidemiology and Biostatistics, University of California, San Francisco, and the Institute for Human Genetics, University of California, San Francisco. P.G. is currently affiliated with the Kaiser Permanente Division of Research, Oakland, CA, and the Department of Epidemiology and Biostatistics, University of California, San Francisco. N.R. is currently affiliated with the Department of Epidemiology and Biostatistics, the Institute for Human Genetics, University of California, San Francisco, and the Kaiser Permanente Division of Research, Oakland, CA. M.M.G. is currently affiliated with the Department of Epidemiology, Boston University School of Public Health, MA
| | - Vidhu Choudhary
- From the Department of Epidemiology and Biostatistics (E.L.F., S.C.Z., M.C., K.S., T.J.H., N.R., M.M.G.), University of California, San Francisco; Kaiser Permanente Division of Research (C.J., V.C., T.J.M., P.G., C.A.S.), Oakland; Institute for Human Genetics (A.O.-O., N.R.); Department of Clinical Pharmacy (A.O.-O.), University of California, San Francisco; Department of Public Health Sciences (R.A.W.), University of California, Davis; and Departments of Pediatrics and Medicine (R.M.K.), University of California, San Francisco. K.S. is currently affiliated with the VA Boston Healthcare System, MA. T.J.H. is currently affiliated with the Department of Epidemiology and Biostatistics, University of California, San Francisco, and the Institute for Human Genetics, University of California, San Francisco. P.G. is currently affiliated with the Kaiser Permanente Division of Research, Oakland, CA, and the Department of Epidemiology and Biostatistics, University of California, San Francisco. N.R. is currently affiliated with the Department of Epidemiology and Biostatistics, the Institute for Human Genetics, University of California, San Francisco, and the Kaiser Permanente Division of Research, Oakland, CA. M.M.G. is currently affiliated with the Department of Epidemiology, Boston University School of Public Health, MA
| | - Travis J Meyers
- From the Department of Epidemiology and Biostatistics (E.L.F., S.C.Z., M.C., K.S., T.J.H., N.R., M.M.G.), University of California, San Francisco; Kaiser Permanente Division of Research (C.J., V.C., T.J.M., P.G., C.A.S.), Oakland; Institute for Human Genetics (A.O.-O., N.R.); Department of Clinical Pharmacy (A.O.-O.), University of California, San Francisco; Department of Public Health Sciences (R.A.W.), University of California, Davis; and Departments of Pediatrics and Medicine (R.M.K.), University of California, San Francisco. K.S. is currently affiliated with the VA Boston Healthcare System, MA. T.J.H. is currently affiliated with the Department of Epidemiology and Biostatistics, University of California, San Francisco, and the Institute for Human Genetics, University of California, San Francisco. P.G. is currently affiliated with the Kaiser Permanente Division of Research, Oakland, CA, and the Department of Epidemiology and Biostatistics, University of California, San Francisco. N.R. is currently affiliated with the Department of Epidemiology and Biostatistics, the Institute for Human Genetics, University of California, San Francisco, and the Kaiser Permanente Division of Research, Oakland, CA. M.M.G. is currently affiliated with the Department of Epidemiology, Boston University School of Public Health, MA
| | - Thomas J Hoffmann
- From the Department of Epidemiology and Biostatistics (E.L.F., S.C.Z., M.C., K.S., T.J.H., N.R., M.M.G.), University of California, San Francisco; Kaiser Permanente Division of Research (C.J., V.C., T.J.M., P.G., C.A.S.), Oakland; Institute for Human Genetics (A.O.-O., N.R.); Department of Clinical Pharmacy (A.O.-O.), University of California, San Francisco; Department of Public Health Sciences (R.A.W.), University of California, Davis; and Departments of Pediatrics and Medicine (R.M.K.), University of California, San Francisco. K.S. is currently affiliated with the VA Boston Healthcare System, MA. T.J.H. is currently affiliated with the Department of Epidemiology and Biostatistics, University of California, San Francisco, and the Institute for Human Genetics, University of California, San Francisco. P.G. is currently affiliated with the Kaiser Permanente Division of Research, Oakland, CA, and the Department of Epidemiology and Biostatistics, University of California, San Francisco. N.R. is currently affiliated with the Department of Epidemiology and Biostatistics, the Institute for Human Genetics, University of California, San Francisco, and the Kaiser Permanente Division of Research, Oakland, CA. M.M.G. is currently affiliated with the Department of Epidemiology, Boston University School of Public Health, MA
| | - Paola Gilsanz
- From the Department of Epidemiology and Biostatistics (E.L.F., S.C.Z., M.C., K.S., T.J.H., N.R., M.M.G.), University of California, San Francisco; Kaiser Permanente Division of Research (C.J., V.C., T.J.M., P.G., C.A.S.), Oakland; Institute for Human Genetics (A.O.-O., N.R.); Department of Clinical Pharmacy (A.O.-O.), University of California, San Francisco; Department of Public Health Sciences (R.A.W.), University of California, Davis; and Departments of Pediatrics and Medicine (R.M.K.), University of California, San Francisco. K.S. is currently affiliated with the VA Boston Healthcare System, MA. T.J.H. is currently affiliated with the Department of Epidemiology and Biostatistics, University of California, San Francisco, and the Institute for Human Genetics, University of California, San Francisco. P.G. is currently affiliated with the Kaiser Permanente Division of Research, Oakland, CA, and the Department of Epidemiology and Biostatistics, University of California, San Francisco. N.R. is currently affiliated with the Department of Epidemiology and Biostatistics, the Institute for Human Genetics, University of California, San Francisco, and the Kaiser Permanente Division of Research, Oakland, CA. M.M.G. is currently affiliated with the Department of Epidemiology, Boston University School of Public Health, MA
| | - Akinyemi Oni-Orisan
- From the Department of Epidemiology and Biostatistics (E.L.F., S.C.Z., M.C., K.S., T.J.H., N.R., M.M.G.), University of California, San Francisco; Kaiser Permanente Division of Research (C.J., V.C., T.J.M., P.G., C.A.S.), Oakland; Institute for Human Genetics (A.O.-O., N.R.); Department of Clinical Pharmacy (A.O.-O.), University of California, San Francisco; Department of Public Health Sciences (R.A.W.), University of California, Davis; and Departments of Pediatrics and Medicine (R.M.K.), University of California, San Francisco. K.S. is currently affiliated with the VA Boston Healthcare System, MA. T.J.H. is currently affiliated with the Department of Epidemiology and Biostatistics, University of California, San Francisco, and the Institute for Human Genetics, University of California, San Francisco. P.G. is currently affiliated with the Kaiser Permanente Division of Research, Oakland, CA, and the Department of Epidemiology and Biostatistics, University of California, San Francisco. N.R. is currently affiliated with the Department of Epidemiology and Biostatistics, the Institute for Human Genetics, University of California, San Francisco, and the Kaiser Permanente Division of Research, Oakland, CA. M.M.G. is currently affiliated with the Department of Epidemiology, Boston University School of Public Health, MA
| | - Rachel A Whitmer
- From the Department of Epidemiology and Biostatistics (E.L.F., S.C.Z., M.C., K.S., T.J.H., N.R., M.M.G.), University of California, San Francisco; Kaiser Permanente Division of Research (C.J., V.C., T.J.M., P.G., C.A.S.), Oakland; Institute for Human Genetics (A.O.-O., N.R.); Department of Clinical Pharmacy (A.O.-O.), University of California, San Francisco; Department of Public Health Sciences (R.A.W.), University of California, Davis; and Departments of Pediatrics and Medicine (R.M.K.), University of California, San Francisco. K.S. is currently affiliated with the VA Boston Healthcare System, MA. T.J.H. is currently affiliated with the Department of Epidemiology and Biostatistics, University of California, San Francisco, and the Institute for Human Genetics, University of California, San Francisco. P.G. is currently affiliated with the Kaiser Permanente Division of Research, Oakland, CA, and the Department of Epidemiology and Biostatistics, University of California, San Francisco. N.R. is currently affiliated with the Department of Epidemiology and Biostatistics, the Institute for Human Genetics, University of California, San Francisco, and the Kaiser Permanente Division of Research, Oakland, CA. M.M.G. is currently affiliated with the Department of Epidemiology, Boston University School of Public Health, MA
| | - Neil Risch
- From the Department of Epidemiology and Biostatistics (E.L.F., S.C.Z., M.C., K.S., T.J.H., N.R., M.M.G.), University of California, San Francisco; Kaiser Permanente Division of Research (C.J., V.C., T.J.M., P.G., C.A.S.), Oakland; Institute for Human Genetics (A.O.-O., N.R.); Department of Clinical Pharmacy (A.O.-O.), University of California, San Francisco; Department of Public Health Sciences (R.A.W.), University of California, Davis; and Departments of Pediatrics and Medicine (R.M.K.), University of California, San Francisco. K.S. is currently affiliated with the VA Boston Healthcare System, MA. T.J.H. is currently affiliated with the Department of Epidemiology and Biostatistics, University of California, San Francisco, and the Institute for Human Genetics, University of California, San Francisco. P.G. is currently affiliated with the Kaiser Permanente Division of Research, Oakland, CA, and the Department of Epidemiology and Biostatistics, University of California, San Francisco. N.R. is currently affiliated with the Department of Epidemiology and Biostatistics, the Institute for Human Genetics, University of California, San Francisco, and the Kaiser Permanente Division of Research, Oakland, CA. M.M.G. is currently affiliated with the Department of Epidemiology, Boston University School of Public Health, MA
| | - Ronald M Krauss
- From the Department of Epidemiology and Biostatistics (E.L.F., S.C.Z., M.C., K.S., T.J.H., N.R., M.M.G.), University of California, San Francisco; Kaiser Permanente Division of Research (C.J., V.C., T.J.M., P.G., C.A.S.), Oakland; Institute for Human Genetics (A.O.-O., N.R.); Department of Clinical Pharmacy (A.O.-O.), University of California, San Francisco; Department of Public Health Sciences (R.A.W.), University of California, Davis; and Departments of Pediatrics and Medicine (R.M.K.), University of California, San Francisco. K.S. is currently affiliated with the VA Boston Healthcare System, MA. T.J.H. is currently affiliated with the Department of Epidemiology and Biostatistics, University of California, San Francisco, and the Institute for Human Genetics, University of California, San Francisco. P.G. is currently affiliated with the Kaiser Permanente Division of Research, Oakland, CA, and the Department of Epidemiology and Biostatistics, University of California, San Francisco. N.R. is currently affiliated with the Department of Epidemiology and Biostatistics, the Institute for Human Genetics, University of California, San Francisco, and the Kaiser Permanente Division of Research, Oakland, CA. M.M.G. is currently affiliated with the Department of Epidemiology, Boston University School of Public Health, MA
| | - Catherine A Schaefer
- From the Department of Epidemiology and Biostatistics (E.L.F., S.C.Z., M.C., K.S., T.J.H., N.R., M.M.G.), University of California, San Francisco; Kaiser Permanente Division of Research (C.J., V.C., T.J.M., P.G., C.A.S.), Oakland; Institute for Human Genetics (A.O.-O., N.R.); Department of Clinical Pharmacy (A.O.-O.), University of California, San Francisco; Department of Public Health Sciences (R.A.W.), University of California, Davis; and Departments of Pediatrics and Medicine (R.M.K.), University of California, San Francisco. K.S. is currently affiliated with the VA Boston Healthcare System, MA. T.J.H. is currently affiliated with the Department of Epidemiology and Biostatistics, University of California, San Francisco, and the Institute for Human Genetics, University of California, San Francisco. P.G. is currently affiliated with the Kaiser Permanente Division of Research, Oakland, CA, and the Department of Epidemiology and Biostatistics, University of California, San Francisco. N.R. is currently affiliated with the Department of Epidemiology and Biostatistics, the Institute for Human Genetics, University of California, San Francisco, and the Kaiser Permanente Division of Research, Oakland, CA. M.M.G. is currently affiliated with the Department of Epidemiology, Boston University School of Public Health, MA
| | - M Maria Glymour
- From the Department of Epidemiology and Biostatistics (E.L.F., S.C.Z., M.C., K.S., T.J.H., N.R., M.M.G.), University of California, San Francisco; Kaiser Permanente Division of Research (C.J., V.C., T.J.M., P.G., C.A.S.), Oakland; Institute for Human Genetics (A.O.-O., N.R.); Department of Clinical Pharmacy (A.O.-O.), University of California, San Francisco; Department of Public Health Sciences (R.A.W.), University of California, Davis; and Departments of Pediatrics and Medicine (R.M.K.), University of California, San Francisco. K.S. is currently affiliated with the VA Boston Healthcare System, MA. T.J.H. is currently affiliated with the Department of Epidemiology and Biostatistics, University of California, San Francisco, and the Institute for Human Genetics, University of California, San Francisco. P.G. is currently affiliated with the Kaiser Permanente Division of Research, Oakland, CA, and the Department of Epidemiology and Biostatistics, University of California, San Francisco. N.R. is currently affiliated with the Department of Epidemiology and Biostatistics, the Institute for Human Genetics, University of California, San Francisco, and the Kaiser Permanente Division of Research, Oakland, CA. M.M.G. is currently affiliated with the Department of Epidemiology, Boston University School of Public Health, MA
| |
Collapse
|
28
|
Poliakova T, Wellington CL. Roles of peripheral lipoproteins and cholesteryl ester transfer protein in the vascular contributions to cognitive impairment and dementia. Mol Neurodegener 2023; 18:86. [PMID: 37974180 PMCID: PMC10652636 DOI: 10.1186/s13024-023-00671-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023] Open
Abstract
This narrative review focuses on the role of cholesteryl ester transfer protein (CETP) and peripheral lipoproteins in the vascular contributions to cognitive impairment and dementia (VCID). Humans have a peripheral lipoprotein profile where low-density lipoproteins (LDL) represent the dominant lipoprotein fraction and high-density lipoproteins (HDL) represent a minor lipoprotein fraction. Elevated LDL-cholesterol (LDL-C) levels are well-established to cause cardiovascular disease and several LDL-C-lowering therapies are clinically available to manage this vascular risk factor. The efficacy of LDL-C-lowering therapies to reduce risk of all-cause dementia and AD is now important to address as recent studies demonstrate a role for LDL in Alzheimer's Disease (AD) as well as in all-cause dementia. The LDL:HDL ratio in humans is set mainly by CETP activity, which exchanges cholesteryl esters for triglycerides across lipoprotein fractions to raise LDL and lower HDL as CETP activity increases. Genetic and pharmacological studies support the hypothesis that CETP inhibition reduces cardiovascular risk by lowering LDL, which, by extension, may also lower VCID. Unlike humans, wild-type mice do not express catalytically active CETP and have HDL as their major lipoprotein fraction. As HDL has potent beneficial effects on endothelial cells, the naturally high HDL levels in mice protect them from vascular disorders, likely including VCID. Genetic restoration of CETP expression in mice to generate a more human-like lipid profile may increase the relevance of murine models for VCID studies. The therapeutic potential of existing and emerging LDL-lowering therapies for VCID will be discussed. Figure Legend. Cholesteryl Ester Transfer Protein in Alzheimer's Disease. CETP is mainly produced by the liver, and exchanges cholesteryl esters for triglycerides across lipoprotein fractions to raise circulating LDL and lower HDL as CETP activity increases. Low CETP activity is associated with better cardiovascular health, due to decreased LDL and increased HDL, which may also improve brain health. Although most peripheral lipoproteins cannot enter the brain parenchyma due to the BBB, it is increasingly appreciated that direct access to the vascular endothelium may enable peripheral lipoproteins to have indirect effects on brain health. Thus, lipoproteins may affect the cerebrovasculature from both sides of the BBB. Recent studies show an association between elevated plasma LDL, a well-known cardiovascular risk factor, and a higher risk of AD, and considerable evidence suggests that high HDL levels are associated with reduced CAA and lower neuroinflammation. Considering the potential detrimental role of LDL in AD and the importance of HDL's beneficial effects on endothelial cells, high CETP activity may lead to compromised BBB integrity, increased CAA deposits and greater neuroinflammation. Abbreviations: CETP - cholesteryl transfer ester protein; LDL - low-density lipoproteins; HDL - high-density lipoproteins; BBB - blood-brain barrier; CAA - cerebral amyloid angiopathy, SMC - smooth muscle cells, PVM - perivascular macrophages, RBC - red blood cells.
Collapse
Affiliation(s)
- Tetiana Poliakova
- Department of Pathology and Laboratory Medicine, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafagian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Cheryl L Wellington
- Department of Pathology and Laboratory Medicine, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- Djavad Mowafagian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
- International Collaboration On Repair Discoveries, Vancouver, BC, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
29
|
Wu Y, Sun Z, Zheng Q, Miao J, Dorn S, Mukherjee S, Fletcher JM, Lu Q. Pervasive biases in proxy GWAS based on parental history of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.13.562272. [PMID: 37904974 PMCID: PMC10614766 DOI: 10.1101/2023.10.13.562272] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Almost every recent Alzheimer's disease (AD) genome-wide association study (GWAS) has performed meta-analysis to combine studies with clinical diagnosis of AD with studies that use proxy phenotypes based on parental disease history. Here, we report major limitations in current GWAS-by-proxy (GWAX) practices due to uncorrected survival bias and non-random participation of parental illness survey, which cause substantial discrepancies between AD GWAS and GWAX results. We demonstrate that current AD GWAX provide highly misleading genetic correlations between AD risk and higher education which subsequently affects a variety of genetic epidemiologic applications involving AD and cognition. Our study sheds important light on the design and analysis of mid-aged biobank cohorts and underscores the need for caution when interpreting genetic association results based on proxy-reported parental disease history.
Collapse
Affiliation(s)
- Yuchang Wu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
- Center for Demography of Health and Aging, University of Wisconsin-Madison, Madison, WI, USA
| | - Zhongxuan Sun
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Qinwen Zheng
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Jiacheng Miao
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Stephen Dorn
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Jason M. Fletcher
- Center for Demography of Health and Aging, University of Wisconsin-Madison, Madison, WI, USA
- Department of Population Health Sciences, University of Wisconsin-Madison, Madison, WI, USA
- La Follette School of Public Affairs, University of Wisconsin-Madison, Madison, WI, USA
| | - Qiongshi Lu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
- Center for Demography of Health and Aging, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
30
|
Errors in Author Affiliations. JAMA Netw Open 2023; 6:e2321189. [PMID: 37273211 PMCID: PMC10242419 DOI: 10.1001/jamanetworkopen.2023.21189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/06/2023] Open
|
31
|
Pacheco Pachado M, Casas AI, Elbatreek MH, Nogales C, Guney E, Espay AJ, Schmidt HH. Re-Addressing Dementia by Network Medicine and Mechanism-Based Molecular Endotypes. J Alzheimers Dis 2023; 96:47-56. [PMID: 37742653 PMCID: PMC10657714 DOI: 10.3233/jad-230694] [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] [Accepted: 08/22/2023] [Indexed: 09/26/2023]
Abstract
Alzheimer's disease (AD) and other forms of dementia are together a leading cause of disability and death in the aging global population, imposing a high personal, societal, and economic burden. They are also among the most prominent examples of failed drug developments. Indeed, after more than 40 AD trials of anti-amyloid interventions, reduction of amyloid-β (Aβ) has never translated into clinically relevant benefits, and in several cases yielded harm. The fundamental problem is the century-old, brain-centric phenotype-based definitions of diseases that ignore causal mechanisms and comorbidities. In this hypothesis article, we discuss how such current outdated nosology of dementia is a key roadblock to precision medicine and articulate how Network Medicine enables the substitution of clinicopathologic phenotypes with molecular endotypes and propose a new framework to achieve precision and curative medicine for patients with neurodegenerative disorders.
Collapse
Affiliation(s)
- Mayra Pacheco Pachado
- Department of Pharmacology and Personalised Medicine, School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Ana I. Casas
- Department of Pharmacology and Personalised Medicine, School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Universitätsklinikum Essen, Klinik für Neurologie, Essen, Germany
| | - Mahmoud H. Elbatreek
- Department of Pharmacology and Personalised Medicine, School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Cristian Nogales
- Department of Pharmacology and Personalised Medicine, School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Emre Guney
- Discovery and Data Science (DDS) Unit, STALICLA R&D SL, Barcelona, Spain
| | - Alberto J. Espay
- James J. and Joan A. Gardner Family Center for Parkinson’s Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, USA
| | - Harald H.H.W. Schmidt
- Department of Pharmacology and Personalised Medicine, School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|