1
|
Zhu H, Ni H, Yang Q, Ni J, Ji J, Yang S, Peng F. Evaluating the Bidirectional Causal Effects of Alzheimer's Disease Across Multiple Conditions: A Systematic Review and Meta-Analysis of Mendelian Randomization Studies. Int J Mol Sci 2025; 26:3589. [PMID: 40332115 PMCID: PMC12027472 DOI: 10.3390/ijms26083589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
This study systematically evaluates and meta-analyzes Mendelian randomization studies on the bidirectional causal relationship between Alzheimer's disease (AD) and systemic diseases. We searched five databases, assessed study quality, and extracted data. Diseases were classified using ICD-11, and the meta-analysis was performed with RevMan 5.4. A total of 56 studies identified genetic links between AD susceptibility and systemic diseases. Notably, genetic proxies for hip osteoarthritis (OR = 0.80; p = 0.007) and rheumatoid arthritis (OR = 0.97; p = 0.004) were inversely associated with AD risk, while gout (OR = 1.02; p = 0.049) showed a positive association. Genetic liability to depression (OR = 1.03; p = 0.001) elevated AD risk, and AD genetic risk increased susceptibility to delirium (OR = 1.32; p = 0.0005). Cardiovascular traits, including coronary artery disease (OR = 1.07; p = 0.021) and hypertension (OR = 4.30; p = 0.044), were causally linked to a higher AD risk. Other conditions, such as insomnia, chronic periodontitis, migraine, and certain cancers, exhibited significant genetic correlations. Intriguingly, herpes zoster (OR = 0.87; p = 0.005) and cataracts (OR = 0.96; p = 0.012) demonstrated inverse genetic associations with AD. These findings suggest potential therapeutic targets and preventive strategies, emphasizing the need to address comorbid systemic diseases to reduce AD risk and progression.
Collapse
Affiliation(s)
- Haoning Zhu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (H.Z.); (H.N.); (J.N.); (F.P.)
| | - Huitong Ni
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (H.Z.); (H.N.); (J.N.); (F.P.)
| | - Qiuling Yang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China;
| | - Jiaqi Ni
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (H.Z.); (H.N.); (J.N.); (F.P.)
| | - Jianguang Ji
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Shu Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (H.Z.); (H.N.); (J.N.); (F.P.)
| | - Fu Peng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (H.Z.); (H.N.); (J.N.); (F.P.)
| |
Collapse
|
2
|
Yavari M, Kalupahana NS, Harris BN, Ramalingam L, Zu Y, Kahathuduwa CN, Moustaid-Moussa N. Mechanisms Linking Obesity, Insulin Resistance, and Alzheimer's Disease: Effects of Polyphenols and Omega-3 Polyunsaturated Fatty Acids. Nutrients 2025; 17:1203. [PMID: 40218960 PMCID: PMC11990358 DOI: 10.3390/nu17071203] [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: 03/05/2025] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by progressive cognitive decline, memory loss, and behavioral changes. It poses a significant global health challenge. AD is associated with the accumulation of amyloid-β (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain, along with chronic inflammation, dysfunctional neurons, and synapse loss. While the prevalence of AD continues to rise, the current FDA-approved drugs offer only limited effectiveness. Emerging evidence suggests that obesity, insulin resistance (IR), and type 2 diabetes mellitus (T2DM) are also implicated in AD pathogenesis, with epidemiological studies and animal models confirming the impact of IR on Aβ accumulation, and high-fat diets also exacerbating Aβ accumulation. Since neuroinflammation activated by Aβ involves the nuclear factor kappa-light-chain-enhancer of the activated B cell (NF-κB) pathway, the inhibition of NF-κB and NLRP3 inflammasome activation are potential therapeutic strategies in AD. Bioactive compounds, including polyphenols (resveratrol, epigallocatechin-3-gallate, curcumin, and quercetin), and omega-3 polyunsaturated fatty acids, show promising results in animal studies and clinical trials for reducing Aβ levels, improving cognition and modulating the signaling pathways implicated in AD. This review explores the interplay between obesity, IR, inflammation, and AD pathology, emphasizing the potential of dietary compounds and their role in reducing inflammation, oxidative stress, and cognitive decline, as viable strategies for AD prevention and treatment. By integrating epidemiological findings, observational studies, and clinical trials, this review aims to provide a comprehensive understating of how metabolic dysfunctions and bioactive compounds influence AD progression.
Collapse
Affiliation(s)
- Mahsa Yavari
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.Y.); (L.R.); (Y.Z.)
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
| | - Nishan Sudheera Kalupahana
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Breanna N. Harris
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Institute for One Health Innovation, Offices of Research & Innovation, Texas Tech University, Texas Tech Health Sciences Center, Lubbock, TX 79409, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.Y.); (L.R.); (Y.Z.)
| | - Yujiao Zu
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.Y.); (L.R.); (Y.Z.)
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
| | - Chanaka Nadeeshan Kahathuduwa
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
- Department of Neurology, Texas Tech University Health Sciences Center, El Paso, TX 79409, USA
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.Y.); (L.R.); (Y.Z.)
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
- Institute for One Health Innovation, Offices of Research & Innovation, Texas Tech University, Texas Tech Health Sciences Center, Lubbock, TX 79409, USA
| |
Collapse
|
3
|
Meng X, Li X, Cao M, Dong J, Wang H, Cao W, Liu D, Wang Y. Summarizing attributable factors and evaluating risk of bias of Mendelian randomization studies for Alzheimer's dementia and cognitive status: a systematic review and meta-analysis. Syst Rev 2025; 14:61. [PMID: 40082927 PMCID: PMC11905674 DOI: 10.1186/s13643-025-02792-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 02/07/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND No effective treatment is available to delay or reverse the onset and progression of Alzheimer's dementia (AD). Mild cognitive impairment, a clinical state between normal aging and AD, may offer the proper window for AD intervention and treatment. This systematic review aimed to summarize evidence from Mendelian randomization (MR) studies exploring factors attributable to AD and related cognitive status and to assess its credibility. METHODS We searched PubMed, Embase, MEDLINE, and the Cochrane Library to identify MR studies investigating the associations between any factor and AD and related cognitive status. The risk of bias in MR studies was evaluated using nine signaling questions tailored to identify potential biases based on the STROBE-MR guidelines. RESULTS A total of 125 eligible publications were examined, including 106 AD-related MR studies reporting 674 records and 28 cognition-related MR studies reporting 141 records. We identified 185 unique causal risk factors for AD and 49 for cognitive status. More than half of the MR studies reporting AD or cognitive status outcomes exhibited poor methodological quality, with a high risk of bias observed in 59% of the AD-related studies and 64% of the cognitive-related studies. CONCLUSIONS This systematic review summarized modifiable factors and omics signatures, providing a database of MR studies on AD and related cognitive status. The evaluation of bias risk in MR studies serves to raise awareness and improve overall quality. A critical appraisal checklist for assessing the risk of bias may pave the way for the development of a standardized tool. SYSTEMATIC REVIEW REGISTRATION The review protocol was registered with the Prospective Register of Systematic Reviews (PROSPERO) under the registration number CRD42023213990.
Collapse
Affiliation(s)
- Xiaoni Meng
- Department of Clinical Epidemiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Xiaochun Li
- School of Public Health, Capital Medical University, Beijing, 100069, China
- The Medical Department, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Meiling Cao
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Jing Dong
- Health Management Center, Xuanwu Hospital, Capital Medical University, Beijing, 100050, China
| | - Haotian Wang
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Weijie Cao
- Centre for Precision Medicine, Edith Cowan University, Perth, WA, 7027, Australia
| | - Di Liu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, University Town, 1068 Xueyuan Avenue, Nanshan District, Shenzhen, 518055, China.
| | - Youxin Wang
- Centre for Precision Medicine, Edith Cowan University, Perth, WA, 7027, Australia.
- School of Public Health, North China University of Science and Technology, 21 Bohaidadao, Caofeidian District, Tangshan, 063210, China.
| |
Collapse
|
4
|
Chen Y, Feng Y, Zhang X, Gifford KA, Elmanzalawi Y, Samuels J, Albaugh VL, English WJ, Flynn CR, Yu D, Zhang R, Ikramuddin S. Bariatric Surgery Is Associated With Reduced Incidence of Mild Cognitive Impairment and Alzheimer Disease and Related Dementias: A Retrospective Cohort Study. ANNALS OF SURGERY OPEN 2025; 6:e541. [PMID: 40134493 PMCID: PMC11932579 DOI: 10.1097/as9.0000000000000541] [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: 12/17/2024] [Indexed: 03/27/2025] Open
Abstract
Objective To evaluate the association of bariatric surgery with reduced incidence and delayed development of mild cognitive impairment (MCI) and Alzheimer disease and related dementias (ADRD) in patients with obesity. Background This retrospective longitudinal study utilized Electronic Health Records from Vanderbilt University Medical Center, covering 5303 patients who underwent bariatric surgery and 10,606 propensity score-matched obese patients who did not, from 2000 to 2023. Patients with prior MCI, ADRD, schizophrenia, alcoholism, gastric cancer, gastric ulcers, inflammatory bowel disease, coagulopathy, stroke, Parkinson disease, or brain cancer were excluded from both groups. Methods Differences in time to MCI/ADRD between surgical and control groups were analyzed using linear regression, and adjusted for confounders: demographics, medical history, and socioeconomic status. Survival probability differences for MCI and ADRD between the 2 groups over time were assessed using Kaplan-Meier curves and log-rank tests. Incidence differences of MCI and ADRD between the groups were evaluated using Fine-Gray subdistribution hazard models, accounting for the competing risk of death and confounders. Results Bariatric surgery was associated with a significantly reduced incidence of ADRD, evidenced by a subdistribution hazard ratio (SHR) of 0.37 (95% confidence interval [CI]: 0.15-0.89; P = 0.03). Similarly, the incidence of MCI was significantly lower in the surgical group, with an SHR of 0.57 (95% CI: 0.39-0.85; P = 0.01). Additionally, patients who underwent bariatric surgery experienced a delay of 2.01 years before developing MCI compared with the control group (95% CI: 0.70-3.50; P = 0.004). Conclusions These findings suggest that bariatric surgery may serve as an effective strategy to delay the onset of MCI and reduce the risk of both MCI and ADRD in patients with obesity.
Collapse
Affiliation(s)
- You Chen
- From the Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
- Department of Computer Science, Vanderbilt University, Nashville, TN
| | - Yubo Feng
- Department of Computer Science, Vanderbilt University, Nashville, TN
| | - Xinmeng Zhang
- Department of Computer Science, Vanderbilt University, Nashville, TN
| | | | | | - Jason Samuels
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN
| | - Vance L. Albaugh
- Metamor Institute, Pennington Biomedical Research Center, Baton Rouge, LA
| | - Wayne J. English
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN
| | - Charles R. Flynn
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN
| | - Danxia Yu
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Rui Zhang
- Department of Surgery, University of Minnesota, Minneapolis, MN
| | | |
Collapse
|
5
|
Li N, Wang X, Lin R, Yang F, Chang HC, Gu X, Shu J, Liu G, Yu Y, Wei W, Bao Z. ANGPTL4-mediated microglial lipid droplet accumulation: Bridging Alzheimer's disease and obesity. Neurobiol Dis 2024; 203:106741. [PMID: 39577812 DOI: 10.1016/j.nbd.2024.106741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/08/2024] [Accepted: 11/17/2024] [Indexed: 11/24/2024] Open
Abstract
Increasing evidence suggests that metabolic disorders such as obesity are implicated in the development of Alzheimer's disease (AD). The pathological buildup of lipids in microglia is regarded as a key indicator in brain aging and the progression of AD, yet the mechanisms behind this process remain uncertain. The adipokine ANGPTL4 is strongly associated with obesity and is thought to play a role in the advancement of neurodegenerative diseases. This study utilized RNA sequencing to identify differential expression in lipid-accumulating BV2 microglia and investigated the potential mechanism through ANGPTL4 overexpression in BV2. Subsequently, animal models and clinical data were employed to further explore alterations in circulating ANGPTL4 levels in AD. RNA sequencing results indicated a correlation between ANGPTL4 and microglial lipid accumulation. The overexpression of ANGPTL4 in microglia resulted in increased secretion of inflammatory factors, elevated oxidative stress levels, and diminished antiviral capacity. Furthermore, when simulating the coexistence of AD and obesity through combined treatment with Amyloid-Beta 1-42 peptide (Aβ) and Free Fatty Acids (FFA) in vitro, we observed a notable upregulation of ANGPTL4 expression, highlighting its potential role in the interplay between AD and obesity. In vivo experiments, we also observed a significant increase in ANGPTL4 expression in the hippocampus and plasma of APP/PS1 mice compared to wild-type controls. This was accompanied by heightened microglial activation and reduced expression of longevity-related genes in the hippocampus. Clinical data from the UK Biobank indicated that plasma ANGPTL4 levels are elevated in patients with AD when compared to healthy controls. Moreover, significantly higher ANGPTL4 levels were observed in obese AD patients relative to their non-obese counterparts. Our findings suggest that ANGPTL4-mediated microglial aging may serve as a crucial link between AD and obesity, proposing ANGPTL4 as a potential biomarker for AD.
Collapse
Affiliation(s)
- Nan Li
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Neurology, Huashan Hospital Affiliated to Fudan University, Shanghai 200032, China
| | - Xiaojun Wang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Shanghai institute of geriatric medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Ruilang Lin
- Department of Biostatistics, Key Laboratory of Public Health Safety of Ministry of Education, Key Laboratory for Health Technology Assessment, National Commission of Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Fuxia Yang
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Hung-Chen Chang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Cardiology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Shanghai institute of geriatric medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Xuchao Gu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Traditional Chinese Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Jun Shu
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Guidong Liu
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China.
| | - Yongfu Yu
- Department of Biostatistics, Key Laboratory of Public Health Safety of Ministry of Education, Key Laboratory for Health Technology Assessment, National Commission of Health, School of Public Health, Fudan University, Shanghai 200032, China.
| | - Wenshi Wei
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China.
| | - Zhijun Bao
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Shanghai institute of geriatric medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China.
| |
Collapse
|
6
|
Frank G, Gualtieri P, Cianci R, Caldarelli M, Palma R, De Santis GL, Porfilio C, Nicoletti F, Bigioni G, Di Renzo L. Body Composition and Alzheimer's Disease: A Holistic Review. Int J Mol Sci 2024; 25:9573. [PMID: 39273520 PMCID: PMC11395597 DOI: 10.3390/ijms25179573] [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: 07/25/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Alzheimer's disease (AD) represents a significant global health challenge and affects approximately 50 million people worldwide. This overview of published reviews provides a comprehensive understanding of the intricate correlations between AD and body composition, focusing particularly on obesity. We used a systematic approach to collect and analyze relevant reviews on the topic of obesity and Alzheimer's disease. A comprehensive search of electronic databases, including PubMed, MEDLINE, and Google Scholar, was conducted. We searched keywords such as "Alzheimer's disease", "body composition", "lean mass", "bone mass", and "fat mass". We considered only reviews written within the past 5 years and in English. Fifty-six relevant reviews were identified that shed light on the multiple connections between AD and body composition. The review involves several aspects, including the impact of lean mass, bone mass, and endocrinological factors related to obesity, as well as inflammation, neuroinflammation, and molecular/genetic factors. The findings highlight the complex interplay of these elements in the development of AD, underscoring the need for holistic approaches to reduce the risk of AD and to explore innovative strategies for diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Giulia Frank
- PhD School of Applied Medical-Surgical Sciences, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- School of Specialization in Food Science, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Paola Gualtieri
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Rossella Cianci
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Mario Caldarelli
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Roselisa Palma
- PhD School of Applied Medical-Surgical Sciences, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- School of Specialization in Food Science, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Gemma Lou De Santis
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Chiara Porfilio
- School of Specialization in Food Science, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Francesco Nicoletti
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Giulia Bigioni
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Laura Di Renzo
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
7
|
Serradas ML, Ding Y, Martorell PV, Kulińska I, Castro-Gomez S. Therapeutic Targets in Innate Immunity to Tackle Alzheimer's Disease. Cells 2024; 13:1426. [PMID: 39272998 PMCID: PMC11394242 DOI: 10.3390/cells13171426] [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: 07/30/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
There is an urgent need for effective disease-modifying therapeutic interventions for Alzheimer's disease (AD)-the most prevalent cause of dementia with a profound socioeconomic burden. Most clinical trials targeting the classical hallmarks of this disease-β-amyloid plaques and neurofibrillary tangles-failed, showed discrete clinical effects, or were accompanied by concerning side effects. There has been an ongoing search for novel therapeutic targets. Neuroinflammation, now widely recognized as a hallmark of all neurodegenerative diseases, has been proven to be a major contributor to AD pathology. Here, we summarize the role of neuroinflammation in the pathogenesis and progression of AD and discuss potential targets such as microglia, TREM2, the complement system, inflammasomes, and cytosolic DNA sensors. We also present an overview of ongoing studies targeting specific innate immune system components, highlighting the progress in this field of drug research while bringing attention to the delicate nature of innate immune modulations in AD.
Collapse
Affiliation(s)
- Maria L. Serradas
- Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
| | - Yingying Ding
- Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
| | - Paula V. Martorell
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Ida Kulińska
- Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
| | - Sergio Castro-Gomez
- Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
- Center for Neurology, Department of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
8
|
Lian P, Cai X, Wang C, Zhai H, Liu K, Yang X, Wu Y, Ma Z, Cao X, Xu Y. Identification and experimental validation of m7G-related molecular subtypes, immune signature, and feature genes in Alzheimer's disease. Heliyon 2024; 10:e33836. [PMID: 39027505 PMCID: PMC11255592 DOI: 10.1016/j.heliyon.2024.e33836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 06/01/2024] [Accepted: 06/27/2024] [Indexed: 07/20/2024] Open
Abstract
Background Studies has shown that N7-methylguanosine (m7G) modification plays a critical role in neurological diseases. However, the exact role and association of m7G with the immune microenvironment in Alzheimer's disease (AD) remain largely unknown and unexplored. Methods The study datasets comprised 667 AD samples and 503 control samples selected from eight datasets in the Gene Expression Omnibus database; m7G regulator genes were obtained from previous literature. The AD subtypes were identified by consensus clustering analysis according to m7G regulator genes. The clinical characteristics, immune infiltration, and biological functions of the AD subgroups were evaluated. A combination of different types of machine-learning algorithms were used for the identification of AD genes. We also assessed and validated the diagnostic performance of the identified genes via qRT-PCR, immunofluorescence, and immunohistochemical analyses. Results Two AD distinct subgroups, namely cluster A and cluster B, were identified. Cluster A had poor pathological progression and immune infiltration, representing a high-risk subgroup for AD. The differentially expressed genes of cluster A were enriched in immune and synapse-related pathways, suggesting that these genes probably contribute to AD progression by regulating immune-related pathways. Additionally, five feature genes (AEBP1, CARTPT, AK5, NPTX2, and COPG2IT1) were identified, which were used to construct a nomogram model with good ability to predict AD. The animal experiment analyses further confirmed that these feature genes were associated with AD development. Conclusion To the best of our knowledge, this is the first study to reveal close correlations among m7G RNA modification, the immune microenvironment, and the pathogenesis of AD. We also identified five feature genes associated with AD, further contributing to our understanding of the underlying mechanisms and potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Piaopiao Lian
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Cai
- Department of Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cailin Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Zhai
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoman Yang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuoran Ma
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuebing Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Zhu S, Liu Y, Xia G, Wang X, Du A, Wu J, Wang Y, Wang Y, Shen C, Wei P, Xu C. Modulation of cardiac resident macrophages immunometabolism upon high-fat-diet feeding in mice. Front Immunol 2024; 15:1371477. [PMID: 39007149 PMCID: PMC11239335 DOI: 10.3389/fimmu.2024.1371477] [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: 01/16/2024] [Accepted: 06/05/2024] [Indexed: 07/16/2024] Open
Abstract
Background A high-fat diet (HFD) contributes to various metabolic disorders and obesity, which are major contributors to cardiovascular disease. As an essential regulator for heart homeostasis, cardiac resident macrophages may go awry and contribute to cardiac pathophysiology upon HFD. Thus, to better understand how HFD induced cardiac dysfunction, this study intends to explore the transcriptional and functional changes in cardiac resident macrophages of HFD mice. Methods C57BL/6J female mice that were 6 weeks old were fed with HFD or normal chow diet (NCD) for 16 weeks. After an evaluation of cardiac functions by echocardiography, mouse hearts were harvested and cardiac resident CCR2- macrophages were sorted, followed by Smart sequencing. Bioinformatics analysis including GO, KEGG, and GSEA analyses were employed to elucidate transcriptional and functional changes. Results Hyperlipidemia and obesity were observed easily upon HFD. The mouse hearts also displayed more severe fibrosis and diastolic dysfunction in HFD mice. Smart sequencing and functional analysis revealed metabolic dysfunctions, especially lipid-related genes and pathways. Besides this, antigen-presentation-related gene such as Ctsf and inflammation, particularly for NF-κB signaling and complement cascades, underwent drastic changes in cardiac resident macrophages. GO cellular compartment analysis was also performed and showed specific organelle enrichment trends of the involved genes. Conclusion Dysregulated metabolism intertwines with inflammation in cardiac resident macrophages upon HFD feeding in mice, and further research on crosstalk among organelles could shed more light on potential mechanisms.
Collapse
Affiliation(s)
- Simeng Zhu
- Department of Cardiology, Sixth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yujia Liu
- Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guofang Xia
- Department of Cardiology, Sixth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaoqing Wang
- Department of Cardiology, Sixth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ailian Du
- Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jin Wu
- Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yanpeng Wang
- Department of Cardiology, Sixth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuanlong Wang
- Department of Cardiology, Sixth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chengxing Shen
- Department of Cardiology, Sixth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Peng Wei
- Department of Cardiology, Sixth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Congfeng Xu
- Department of Cardiology, Sixth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Campolim CM, Schimenes BC, Veras MM, Kim YB, Prada PO. Air pollution accelerates the development of obesity and Alzheimer's disease: the role of leptin and inflammation - a mini-review. Front Immunol 2024; 15:1401800. [PMID: 38933275 PMCID: PMC11199417 DOI: 10.3389/fimmu.2024.1401800] [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: 03/16/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Air pollution is an urgent concern linked to numerous health problems in low- and middle-income countries, where 92% of air pollution-related deaths occur. Particulate matter 2.5 (PM2.5) is the most harmful component of air pollutants, increasing inflammation and changing gut microbiota, favoring obesity, type 2 diabetes, and Alzheimer's Disease (AD). PM2.5 contains lipopolysaccharides (LPS), which can activate the Toll-like receptor 4 (TLR4) signaling pathway. This pathway can lead to the release of pro-inflammatory markers, including interleukins, and suppressor of cytokine signaling-3 (SOCS3), which inhibits leptin action, a hormone that keeps the energy homeostasis. Leptin plays a role in preventing amyloid plaque deposition and hyperphosphorylation of tau-protein (p-tau), mechanisms involved in the neurodegeneration in AD. Approximately 50 million people worldwide are affected by dementia, with a significant proportion living in low-and middle-income countries. This number is expected to triple by 2050. This mini-review focuses on the potential impact of PM2.5 exposure on the TLR4 signaling pathway, its contribution to leptin resistance, and dysbiosis that exacerbates the link between obesity and AD.
Collapse
Affiliation(s)
- Clara Machado Campolim
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, MA, United States
| | | | - Mariana Matera Veras
- Laboratory of Environmental and Experimental Pathology LIM05, Department of Pathology, School of Medicine, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, MA, United States
| | - Patricia Oliveira Prada
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
- Obesity and Comorbidities Research Center, Campinas, SP, Brazil
- Department of Structural and Functional Biology, Institute of Biology (IB), University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
11
|
Lian P, Cai X, Yang X, Ma Z, Wang C, Liu K, Wu Y, Cao X, Xu Y. Analysis and experimental validation of necroptosis-related molecular classification, immune signature and feature genes in Alzheimer's disease. Apoptosis 2024; 29:726-742. [PMID: 38478169 PMCID: PMC11055779 DOI: 10.1007/s10495-024-01943-8] [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: 02/04/2024] [Indexed: 04/28/2024]
Abstract
Necroptosis, a programmed cell death pathway, has been demonstrated to be activated in Alzheimer's disease (AD). However, the precise role of necroptosis and its correlation with immune cell infiltration in AD remains unclear. In this study, we conducted non-negative matrix factorization clustering analysis to identify three subtypes of AD based on necroptosis-relevant genes. Notably, these subtypes exhibited varying necroptosis scores, clinical characteristics and immune infiltration signatures. Cluster B, characterized by high necroptosis scores, showed higher immune cell infiltration and was associated with a more severe pathology, potentially representing a high-risk subgroup. To identify potential biomarkers for AD within cluster B, we employed two machine learning algorithms: the least absolute shrinkage and selection operator regression and Random Forest. Subsequently, we identified eight feature genes (CARTPT, KLHL35, NRN1, NT5DC3, PCYOX1L, RHOQ, SLC6A12, and SLC38A2) that were utilized to develop a diagnosis model with remarkable predictive capacity for AD. Moreover, we conducted validation using bulk RNA-seq, single-nucleus RNA-seq, and in vivo experiments to confirm the expression of these feature genes. In summary, our study identified a novel necroptosis-related subtype of AD and eight diagnostic biomarkers, explored the roles of necroptosis in AD progression and shed new light for the clinical diagnosis and treatment of this disease.
Collapse
Affiliation(s)
- Piaopiao Lian
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Cai
- Department of Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoman Yang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhuoran Ma
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cailin Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuebing Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
12
|
Reich N, Hölscher C. Cholecystokinin (CCK): a neuromodulator with therapeutic potential in Alzheimer's and Parkinson's disease. Front Neuroendocrinol 2024; 73:101122. [PMID: 38346453 DOI: 10.1016/j.yfrne.2024.101122] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/04/2024] [Accepted: 01/25/2024] [Indexed: 02/16/2024]
Abstract
Cholecystokinin (CCK) is a neuropeptide modulating digestion, glucose levels, neurotransmitters and memory. Recent studies suggest that CCK exhibits neuroprotective effects in Alzheimer's disease (AD) and Parkinson's disease (PD). Thus, we review the physiological function and therapeutic potential of CCK. The neuropeptide facilitates hippocampal glutamate release and gates GABAergic basket cell activity, which improves declarative memory acquisition, but inhibits consolidation. Cortical CCK alters recognition memory and enhances audio-visual processing. By stimulating CCK-1 receptors (CCK-1Rs), sulphated CCK-8 elicits dopamine release in the substantia nigra and striatum. In the mesolimbic pathway, CCK release is triggered by dopamine and terminates reward responses via CCK-2Rs. Importantly, activation of hippocampal and nigral CCK-2Rs is neuroprotective by evoking AMPK activation, expression of mitochondrial fusion modulators and autophagy. Other benefits include vagus nerve/CCK-1R-mediated expression of brain-derived neurotrophic factor, intestinal protection and suppression of inflammation. We also discuss caveats and the therapeutic combination of CCK with other peptide hormones.
Collapse
Affiliation(s)
- Niklas Reich
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, UK; Faculty of Health and Medicine, Biomedical & Life Sciences Division, Lancaster University, Lancaster LA1 4YQ, UK.
| | - Christian Hölscher
- Second associated Hospital, Neurology Department, Shanxi Medical University, Taiyuan, Shanxi, China; Henan Academy of Innovations in Medical Science, Neurodegeneration research group, Xinzhen, Henan province, China
| |
Collapse
|
13
|
Tao M, Guo HY, Ji X, Wang W, Yuan H, Peng H. Long-term trends in Alzheimer's disease and other dementias deaths with high body mass index in China from 1990 to 2019, and projections up to 2042. Arch Public Health 2024; 82:42. [PMID: 38528579 DOI: 10.1186/s13690-024-01273-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 03/18/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND In China, the rising prevalence of high Body Mass Index (BMI) is linked to increasing health issues, including Alzheimer's disease (AD). This study analyzes mortality trends related to AD and other dementias associated with high BMI from 1990 to 2019, considering age, period, and birth cohort effects, and forecasts future trends. METHODS We analyzed mortality data for AD and other dementias linked to high BMI in Chinese residents from the Global Burden of Disease 2019 database. Using Joinpoint regression, we examined age-standardized mortality rate (ASMR) trends and calculated annual and average annual percentage changes (APC and AAPC). Age-period-cohort models provided deeper insights, with Bayesian models used to project future ASMR trends to 2042. RESULTS From 1990 to 2019, the ASMR for AD and other dementias associated with high BMI in China showed an overall increasing trend. Females had a lower increase rate than males, yet their overall levels remained higher. Specifically, the ASMR for males increased by an average of 2.70% per year, peaking between 2006 and 2010, while for females, it increased by an average of 2.29% per year, also peaking in the same period. Age-period-cohort analysis revealed increasing mortality relative risk with age and period, but a decrease with birth cohort. Projections suggest a continued rise in ASMR by 2042, with rates for males and females expected to be 2.48/100,000 and 2.94/100,000, respectively. CONCLUSION The increasing mortality trend from AD and other dementias associated with high BMI highlights the urgent need for policy interventions focused on overweight prevention, particularly vital for addressing the health challenges in China's aging population.
Collapse
Affiliation(s)
- Mengjun Tao
- Health management center, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Hao-Yang Guo
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China
| | - Xincan Ji
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China
| | - Wei Wang
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China
| | - Hui Yuan
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China.
| | - Hui Peng
- Department of Science and Technology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China.
| |
Collapse
|
14
|
Anderson EL, Davies NM, Korologou-Linden R, Kivimäki M. Dementia prevention: the Mendelian randomisation perspective. J Neurol Neurosurg Psychiatry 2024; 95:384-390. [PMID: 37967935 DOI: 10.1136/jnnp-2023-332293] [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: 07/24/2023] [Accepted: 10/25/2023] [Indexed: 11/17/2023]
Abstract
Understanding the causes of Alzheimer's disease and related dementias remains a challenge. Observational studies investigating dementia risk factors are limited by the pervasive issues of confounding, reverse causation and selection biases. Conducting randomised controlled trials for dementia prevention is often impractical due to the long prodromal phase and the inability to randomise many potential risk factors. In this essay, we introduce Mendelian randomisation as an alternative approach to examine factors that may prevent or delay Alzheimer's disease. Mendelian randomisation is a causal inference method that has successfully identified risk factors and treatments in various other fields. However, applying this method to dementia risk factors has yielded unexpected findings. Here, we consider five potential explanations and provide recommendations to enhance causal inference from Mendelian randomisation studies on dementia. By employing these strategies, we can better understand factors affecting dementia risk.
Collapse
Affiliation(s)
- Emma Louise Anderson
- Mental Health of Older People, Division of Psychiatry, University College London, London, UK
| | - Neil M Davies
- Epidemiology & Applied Clinical Research, Division of Psychiatry, University College London, London, UK
- Department of Statistical Sciences, University College London, London, UK
| | | | - Mika Kivimäki
- Mental Health of Older People, Division of Psychiatry, University College London, London, UK
| |
Collapse
|
15
|
Lu P, Gao CX, Luo FJ, Huang YT, Gao MM, Long YS. Hippocampal proteomic changes in high-fat diet-induced obese mice associated with memory decline. J Nutr Biochem 2024; 125:109554. [PMID: 38142716 DOI: 10.1016/j.jnutbio.2023.109554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/24/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Substantial evidence suggest that chronic consumption of high-fat diets (HFDs) can lead to obesity, abnormal metabolism, as well as cognitive impairment. Molecular and cellular changes regarding hippocampal dysfunctions have been identified in multiple HFD animal models. Therefore, in-depth identification of expression changes of hippocampal proteins is critical for understanding the mechanism of HFD-induced cognitive deficits. In this study, we fed 3-week-old male mice with HFD for 3 months to generate obese mice who exhibit systemic metabolic abnormality and learning and memory decline. Using an iTRAQ-labeled proteomic analysis, we identified a total of 82 differentially expressed proteins (DEPs) in the hippocampus upon HFD with 35 up-regulated proteins and 47 down-regulated proteins. Functional enrichment indicated that these DEPs were predominantly enriched in regulation of catabolic process, dendritic shaft, neuron projection morphogenesis and GTPase regulator activity. Protein-protein interaction enrichment showed that the DEPs are mostly enriched in postsynaptic functions; and of them, six proteins (i.e., DLG3, SYNGAP1, DCLK1, GRIA4, GRIP1, and ARHGAP32) were involved in several functional assemblies of the postsynaptic density including G-protein signaling, scaffolding and adaptor, kinase and AMPA signaling, respectively. Collectively, our findings suggest that these DEPs upon HFD might contribute to memory decline by disturbing neuronal and postsynaptic functions in the hippocampus.
Collapse
Affiliation(s)
- Ping Lu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Cun-Xiu Gao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Fei-Jian Luo
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Yu-Ting Huang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Mei-Mei Gao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Yue-Sheng Long
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China.
| |
Collapse
|
16
|
Cai X, Wang Y, Li Y, Du Z, Wang Z. The Causal Associations between Adipokines and Alzheimer's Disease: A Two-Sample Mendelian Randomization Study. J Alzheimers Dis Rep 2024; 8:75-83. [PMID: 38312531 PMCID: PMC10836602 DOI: 10.3233/adr-230110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/13/2023] [Indexed: 02/06/2024] Open
Abstract
Background Observational studies have indicated the association of alteration of adipokines with Alzheimer's disease (AD). However, it remains unclear whether the associations are causal. Objective To determine the causal associations between adipokines and AD. Methods A Mendelian randomization (MR) method was applied to investigate the causal relationships of adipokines, including adiponectin and resistin, with risk of AD. Genetic proxies from genome-wide association studies (GWAS) of adiponectin and resistin were selected as instrumental variables. GWAS summary statistics for AD were extracted as outcome. Results In this study, we found evidence of the causal effects of adiponectin on AD (OR: 0.850, 95% CI: 0.731-0.990, p = 0.037). However, no relationship between resistin and AD (OR: 0.936, 95% CI: 0.851-1.029, p = 0.171) was detected. In the reverse causation analysis, null associations of AD were found for adiponectin and resistin (all p > 0.05). Conclusions This study provides evidence of causality between adiponectin and risk of AD. However, no genetic susceptibility of resistin was discovered for AD.
Collapse
Affiliation(s)
- Xiaoying Cai
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yaqing Wang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying Li
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhanxin Du
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhongxing Wang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
17
|
Hao N, Bai X, Hu A, Zhao G, Chen Y, Zhao J, Ling Q, Li X, Cai C, Wang Q, Wang Z, Fang J. Assessing the Global, Regional, and National Impact of High Body Mass Index on Alzheimer's Disease and Other Dementias Between 1990 and 2019. J Alzheimers Dis 2024; 97:293-307. [PMID: 38043013 DOI: 10.3233/jad-230827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2023]
Abstract
BACKGROUND Obesity significantly increases Alzheimer's disease (AD) and dementia risk. Understanding the link between a high body mass index (BMI) and these conditions is crucial for effective management and prevention. OBJECTIVE We aimed to estimate the burden of AD and other dementias attributed to high BMI from 1990 to 2019 based on sex, age, and socio-demographic indicators (SDI) at global, regional, and national levels. METHODS We collected data on deaths, disability-adjusted life years (DALYs), age-standardized mortality rates (ASMR), and age-standardized DALY rates (ASDR) from the 2019 Global Burden of Disease study for AD and dementia attributed to high BMI. We explored the correlation between SDI levels and ASDR. RESULTS In 2019, there were 198,476.2 deaths (95% UI: 32,695.4-593,366.4) and 3,159,912.4 DALYs (848,330.5-8,042,531) attributed to high BMI. Numbers of deaths, DALYs, ASMR, and ASDR increased since 1990. Females had higher deaths, ASMR, and ASDR than males. Mortality and DALYs rates increased with age. ASMR and ASDR increased across five SDI levels, with the highest rise in Low-middle SDI. High-income North America had the most deaths [30,993.9 (5,101.7-89,912.9)], while North Africa and the Middle East had the highest ASMR [4.61 (0.79-13.64)] and ASDR [72.56 (20.98-181.16)] in 2019. CONCLUSIONS The burden of AD and other dementias attributed to high BMI increased since 1990 globally and is still heaviest in developed regions. Females accounted predominantly for the burden than males. Timely measures are needed to against high BMI.
Collapse
Affiliation(s)
- Ning Hao
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xue Bai
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - An Hu
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Gaofeng Zhao
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yansheng Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jianhe Zhao
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Qiong Ling
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiangyu Li
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Chuipu Cai
- College of Engineering, Shantou University, Shantou, China
| | - Qi Wang
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhaojun Wang
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jiansong Fang
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
18
|
Chen X, Chen S, Li Z, Zhu R, Jia Z, Ban J, Zhen R, Chen X, Pan X, Ren Q, Yue L, Niu S. Effect of semaglutide and empagliflozin on cognitive function and hippocampal phosphoproteomic in obese mice. Front Pharmacol 2023; 14:975830. [PMID: 37007007 PMCID: PMC10063902 DOI: 10.3389/fphar.2023.975830] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 03/03/2023] [Indexed: 03/19/2023] Open
Abstract
Objective: Based on the 4D label-free phosphoproteomic technique, we examined the differences in cognitive function and hippocampal phosphorylated protein expression in high-fat diet-induced obese mice after the intervention of semaglutide and empagliflozin, as well as the effects of both on protein activity and function in obese mice’s hippocampal tissues and the signaling pathways involved.Methods: Thirty-two C57BL/6JC male mice were assigned to two groups randomly: A control group (group C, 10% of energy is from fat, n = 8) and a high-fat diet group (group H, 60% of energy is from fat, n = 24). The high-fat diet-induced obese mice were screened after 12 weeks of feeding based on the criterion that the bodyweight of mice in fat rich diet group was greater than or equal to 20% of the average body weight of the mice in the blank control group. Group H separate into group H (n = 8), group Semaglutide (group S, n = 8), and group empagliflozin (group E, n = 8). For a total of 12 weeks, group S received 30 nmol/kg/d bodyweight of semaglutide intraperitoneally, group E received 10 mg/kg/d bodyweight of empagliflozin via gavage, and groups C and H received equal amounts of saline by intraperitoneal injection and gavage. At the end of treatment, the mice were appraised for cognitive function employing the Morris water maze (MWM), and serum fasting glucose, lipids, and inflammatory parameters were measured. The 4D label-free phosphoproteomics method was employed to screen the differential phosphoproteins and loci in hippocampal tissues of mice in different treatment groups, and bioinformatics was used to analyze the biological processes, signaling pathways, and related protein–protein interaction (PPI) network analysis of these differentially phosphorylated proteins.Results: In comparison to normal controls, The escape latency of obese mice induced by high-fat diet was prolonged, the percentage of swimming time in the target quadrant was reduced, and the number of times of crossing the platform was reduced, whereas semaglutide and empagliflozin treatment reduced escape latency, increase the percentage of swim time in the target quadrant and increase the frequency of passing through the platform area, although there is little difference in the effect of the two drugs. The phosphoproteomic results showed 20,493 unique phosphorylated peptides, representing 21,239 phosphorylation sites and 4,290 phosphorylated proteins. Further analysis revealed that the proteins corresponding to these differentially phosphorylated sites are jointly distributed in signaling pathways such as dopaminergic synapses and axon guidance, and are involved in biological processes such as neuronal projection development, synaptic plasticity, and axonogenesis. Notably, the key factors voltage-dependent L-type calcium channel subunit alpha-1D (CACNA1D), voltage-dependent P/Q-type calcium channel subunit alpha-1A (CACNA1A), and voltage-dependent N-type calcium channel subunit alpha-1B (CACNA1B) were all found to be involved in the dopaminergic synapse pathway, and their expression was upregulated by semaglutide and empagliflozin.Conclusion: We found for the first time that a high-fat diet decreased CACNA1D, CACNA1A, and CACNA1B protein serine phosphorylation, which may affect neuronal development, synaptic plasticity, and cognitive function in mice. Notably, semaglutide and empagliflozin increased the phosphorylation of these proteins.
Collapse
Affiliation(s)
- Xiaoyi Chen
- Department of Internal Medicine, Hebei North University, Zhangjiakou, China
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Shuchun Chen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- *Correspondence: Shuchun Chen,
| | - Zelin Li
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Ruiyi Zhu
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Zhuoya Jia
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Jiangli Ban
- Department of Internal Medicine, Hebei North University, Zhangjiakou, China
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Ruoxi Zhen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Xing Chen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Xiaoyu Pan
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Qingjuan Ren
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Lin Yue
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Shu Niu
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
19
|
Toshida K, Itoh S, Harada N, Morinaga A, Yugawa K, Tomiyama T, Kosai-Fujimoto Y, Tomino T, Kurihara T, Nagao Y, Morita K, Oda Y, Yoshizumi T. Cancer-associated fibroblasts promote tumor cell growth via miR-493-5p in intrahepatic cholangiocarcinoma. Cancer Sci 2023; 114:937-947. [PMID: 36369960 PMCID: PMC9986089 DOI: 10.1111/cas.15644] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/18/2022] [Accepted: 11/09/2022] [Indexed: 11/14/2022] Open
Abstract
The association between tumor microenvironment (TME) and cancer-associated fibroblasts (CAFs) in intrahepatic cholangiocarcinoma (ICC) progression is poorly understood. This study aimed to reveal whether specific microRNAs (miRNAs) in extracellular vesicles (EVs) derived from CAFs were involved in ICC progression. Conditioned medium (CM) and EVs in the CM of CAFs and normal fibroblasts (NFs) derived from ICC specimens were used to investigate the effects on tumor cell lines. miRNA microarray assay was used to examine the miRNAs of EVs derived from CAFs and NFs in ICC, and the effects of miR-493-5p on tumor cell lines were examined. Additionally, databases were used to identify miR-493-5p targets, and the relationship between prognosis of ICC patients and cocaine- and amphetamine-regulated transcript propeptide (CARTPT), one of the targets of miR-493-5p, expression in ICC tissues was retrospectively analyzed. Compared with NF-derived CM and EVs, CAF-derived CM and EVs promoted cell lines in proliferation, scratch, migration, and invasion assays. miRNA microarray analysis revealed that miR-493-5p was significantly increased in CAF-derived EVs compared to NF-derived EVs. Tumor cell lines transfected with miR-493-5p were promoted in proliferation and scratch assays. Immunohistochemical staining was performed on 76 ICC specimens; both overall and recurrence-free survival rates were significantly worse in the CARTPT-negative group. Univariate and multivariate analyses showed that low CARTPT expression was an independent poor prognostic factor for overall and recurrence-free survival. Overall, our data suggest that CAFs in the ICC TME suppress CARTPT in tumor cells and promote tumor cells via miR-493-5p in EVs.
Collapse
Affiliation(s)
- Katsuya Toshida
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Shinji Itoh
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Noboru Harada
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Akinari Morinaga
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Kyohei Yugawa
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan.,Department of Anatomic Pathology, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Takahiro Tomiyama
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Yukiko Kosai-Fujimoto
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Takahiro Tomino
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Takeshi Kurihara
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Nagao
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Kazutoyo Morita
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| |
Collapse
|
20
|
Pino JMV, Silva VF, Mônico-Neto M, Seva DC, Kato MY, Alves JN, Pereira GC, Antunes HKM, Galvao TD, Bitterncourt LRA, Tufik S, Zambrano LI, Dâmaso AR, Oyama LM, Thivel D, Campos RMS, Lee KS. Severe Obesity in Women Can Lead to Worse Memory Function and Iron Dyshomeostasis Compared to Lower Grade Obesity. Int J Endocrinol 2023; 2023:7625720. [PMID: 37101600 PMCID: PMC10125754 DOI: 10.1155/2023/7625720] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 03/07/2023] [Accepted: 03/25/2023] [Indexed: 04/28/2023] Open
Abstract
Objective Obesity is one of the modifiable risk factors for dementia. Insulin resistance, the abundance of advanced glycated end-products, and inflammation are some of the mechanisms associated with the lower cognitive performance observed in obesity. This study aims to evaluate the cognitive function of subjects with distinct degrees of obesity, comparing class I and II obesity (OBI/II) to class III obesity (OBIII), and to investigate metabolic markers that can distinguish OBIII from OBI/II. Study Design. This is a cross-sectional study, in which 45 females with BMI varying from 32.8 to 51.9 kg/m2 completed a set of 4 cognitive tests (verbal paired-associate test, stroop color, digit span, and Toulouse-Pieron cancellation test) and their plasma metabolites, enzymes, and hormones related to glycemia, dyslipidemia, and liver function, as well as the biomarkers of iron status, were concomitantly analyzed. Results OBIII showed lower scores in the verbal paired-associate test compared to OBI/II. In other cognitive tests, both groups showed similar performance. OBIII presented a lower iron status compared to OBI/II based on total iron binding capacity, degree of transferrin saturation, hemoglobin, mean corpuscular volume, and mean corpuscular hemoglobin. The levels of indicators for glycemia, liver function, and lipid metabolism were similar in both groups. Analysis of plasma metabolites showed that OBIII had lower levels of pyroglutamic acid, myoinositol, and aspartic acid and higher levels of D-ribose than OBI/II. Conclusion Iron is an essential micronutrient for several metabolic pathways. Thus, iron dyshomeostasis observed in severe obesity may aggravate the cognitive impairment by altering metabolic homeostasis and enhancing oxidative stress. These findings can contribute to searching for biomarkers that indicate cognitive performance in the population with obesity.
Collapse
Affiliation(s)
- Jessica M. V. Pino
- Department of Biochemistry, Universidade Federal de São Paulo, Sao Paulo, Brazil
| | - Vitória F. Silva
- Department of Biochemistry, Universidade Federal de São Paulo, Sao Paulo, Brazil
| | - Marcos Mônico-Neto
- Post Graduated Program of Interdisciplinary Health Sciences, Universidade Federal de São Paulo, Santos, Brazil
- BariMais Clinic-Integrated Medicine, Sao Paulo, Brazil
| | - Danielle C. Seva
- Post Graduated Program of Interdisciplinary Health Sciences, Universidade Federal de São Paulo, Santos, Brazil
| | - Melissa Y. Kato
- Department of Biochemistry, Universidade Federal de São Paulo, Sao Paulo, Brazil
| | - July N. Alves
- Department of Biochemistry, Universidade Federal de São Paulo, Sao Paulo, Brazil
| | - Gabriela C. Pereira
- Department of Biochemistry, Universidade Federal de São Paulo, Sao Paulo, Brazil
| | - Hanna Karen M. Antunes
- Post Graduated Program of Interdisciplinary Health Sciences, Universidade Federal de São Paulo, Santos, Brazil
| | | | | | - Sergio Tufik
- Department of Psychobiology, Universidade Federal de São Paulo, Sao Paulo, Brazil
| | - Lysien I. Zambrano
- Institute for Research in Medical Sciences and Right to Health (ICIMEDES)/Scientific Research Unit (UIC), Faculty of Medical Sciences (FCM), National Autonomous University of Honduras (UNAH). Tegucigalpa, Honduras, Honduras
| | - Ana R. Dâmaso
- Post Graduate Program of Nutrition, Universidade Federal de São Paulo, Sao Paulo, Brazil
| | - Lila M. Oyama
- Post Graduate Program of Nutrition, Universidade Federal de São Paulo, Sao Paulo, Brazil
| | - David Thivel
- Clermont Auvergne University, EA 3533, Laboratory of the Metabolic Adaptations to Exercise under Physiological and Pathological Conditions (AME2P), CRNH-Auvergne, Clermont-Ferrand, France
| | - Raquel M. S. Campos
- Post Graduated Program of Interdisciplinary Health Sciences, Universidade Federal de São Paulo, Santos, Brazil
| | - Kil S. Lee
- Department of Biochemistry, Universidade Federal de São Paulo, Sao Paulo, Brazil
| |
Collapse
|
21
|
Smith DC, Karahan H, Wijeratne HRS, Al-Amin M, McCord B, Moon Y, Kim J. Deletion of the Alzheimer's disease risk gene Abi3 locus results in obesity and systemic metabolic disruption in mice. Front Aging Neurosci 2022; 14:1035572. [PMID: 36620768 PMCID: PMC9813750 DOI: 10.3389/fnagi.2022.1035572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease (AD) genetics studies have identified a coding variant within ABI3 gene that increases the risk of developing AD. Recently, we demonstrated that deletion of the Abi3 gene locus dramatically exacerbates AD neuropathology in a transgenic mouse model of amyloidosis. In the course of this AD project, we unexpectedly found that deletion of the Abi3 gene locus resulted in a dramatic obese phenotype in non-transgenic mice. Here, we report our investigation into this serendipitous metabolic finding. Specifically, we demonstrate that mice with deletion of the Abi3 gene locus (Abi3-/- ) have dramatically increased body weight and body fat. Further, we determined that Abi3-/- mice have impaired energy expenditure. Additionally, we found that deletion of the Abi3 gene locus altered gene expression within the hypothalamus, particularly within immune-related pathways. Subsequent immunohistological analysis of the central nervous system (CNS) revealed that microglia number and area were decreased specifically within the mediobasal hypothalamus of Abi3-/- mice. Altogether, this investigation establishes the functional importance of the Abi3 gene locus in the regulation of systemic metabolism and maintenance of healthy body weight. While our previous findings indicated the importance of Abi3 in neurodegeneration, this study indicates that Abi3 related functions are also essential for metabolic regulation.
Collapse
Affiliation(s)
- Daniel C. Smith
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Hande Karahan
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - H. R. Sagara Wijeratne
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Mamun Al-Amin
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Brianne McCord
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Younghye Moon
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jungsu Kim
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
22
|
Identification and Experimental Validation of Marker Genes between Diabetes and Alzheimer’s Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8122532. [PMID: 35996379 PMCID: PMC9391608 DOI: 10.1155/2022/8122532] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/15/2022] [Accepted: 08/01/2022] [Indexed: 11/21/2022]
Abstract
Currently, Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) are widely prevalent in the elderly population, and accumulating evidence implies a strong link between them. For example, patients with T2DM have a higher risk of developing neurocognitive disorders, including AD, but the exact mechanisms are still unclear. This time, by combining bioinformatics analysis and in vivo experimental validation, we attempted to find a common biological link between AD and T2DM. We firstly downloaded the gene expression profiling (AD: GSE122063; T2DM: GSE161355) derived from the temporal cortex. To find the associations, differentially expressed genes (DEGs) of the two datasets were filtered and intersected. Based on them, enrichment analysis was carried out, and the least absolute shrinkage and selection operator (LASSO) logistic regression and support vector machine-recursive feature elimination (SVM-RFE) algorithms were used to identify the specific genes. After verifying in the external dataset and in the samples from the AD and type 2 diabetes animals, the shared targets of the two diseases were finally determined. Based on them, the ceRNA networks were constructed. Besides, the logistic regression and single-sample gene set enrichment analysis (ssGSEA) were performed. As a result, 62 DEGs were totally identified between AD and T2DM, and the enrichment analysis indicated that they were much related to the function of synaptic vesicle and MAPK signaling pathway. Based on the evidence from external dataset and RT-qPCR, CARTPT, EPHA5, and SERPINA3 were identified as the marker genes in both diseases, and their clinical significance and biological functions were further analyzed. In conclusion, discovering and exploring the marker genes that are dysregulated in both 2 diseases could help us better comprehend the intrinsic relationship between T2DM and AD, which may inspire us to develop new strategies for facing the dilemmas of clinical or basic research in cognitive dysfunction.
Collapse
|
23
|
Kumari A, Rahaman A, Zeng XA, Farooq MA, Huang Y, Yao R, Ali M, Ishrat R, Ali R. Temporal Cortex Microarray Analysis Revealed Impaired Ribosomal Biogenesis and Hyperactivity of the Glutamatergic System: An Early Signature of Asymptomatic Alzheimer's Disease. Front Neurosci 2022; 16:966877. [PMID: 35958988 PMCID: PMC9359077 DOI: 10.3389/fnins.2022.966877] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 06/23/2022] [Indexed: 11/21/2022] Open
Abstract
Pathogenic aging is regarded as asymptomatic AD when there is no cognitive deficit except for neuropathology consistent with Alzheimer's disease. These individuals are highly susceptible to developing AD. Braak and Braak's theory specific to tau pathology illustrates that the brain's temporal cortex region is an initiation site for early AD progression. So, the hub gene analysis of this region may reveal early altered biological cascades that may be helpful to alleviate AD in an early stage. Meanwhile, cognitive processing also drags its attention because cognitive impairment is the ultimate result of AD. Therefore, this study aimed to explore changes in gene expression of aged control, asymptomatic AD (AsymAD), and symptomatic AD (symAD) in the temporal cortex region. We used microarray data sets to identify differentially expressed genes (DEGs) with the help of the R programming interface. Further, we constructed the protein-protein interaction (PPI) network by performing the STRING plugin in Cytoscape and determined the hub genes via the CytoHubba plugin. Furthermore, we conducted Gene Ontology (GO) enrichment analysis via Bioconductor's cluster profile package. Resultant, the AsymAD transcriptome revealed the early-stage changes of glutamatergic hyperexcitability. Whereas the connectivity of major hub genes in this network indicates a shift from initially reduced rRNA biosynthesis in the AsymAD group to impaired protein synthesis in the symAD group. Both share the phenomenon of breaking tight junctions and others. In conclusion, this study offers new understandings of the early biological vicissitudes that occur in the brain before the manifestation of symAD and gives new promising therapeutic targets for early AD intervention.
Collapse
Affiliation(s)
- Ankita Kumari
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, China
- Overseas Expertise Introduction Centre for Discipline Innovation of Food Nutrition and Human Health (111 Centre), Guangzhou, China
| | - Abdul Rahaman
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, China
- Overseas Expertise Introduction Centre for Discipline Innovation of Food Nutrition and Human Health (111 Centre), Guangzhou, China
- Abdul Rahaman
| | - Xin-An Zeng
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, China
- Overseas Expertise Introduction Centre for Discipline Innovation of Food Nutrition and Human Health (111 Centre), Guangzhou, China
- *Correspondence: Xin-An Zeng
| | - Muhammad Adil Farooq
- Institute of Food Science and Technology, Khwaja Fareed University of Engineering and Information Technology, Rahim Yar Khan, Pakistan
| | - Yanyan Huang
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, China
| | - Runyu Yao
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Overseas Expertise Introduction Centre for Discipline Innovation of Food Nutrition and Human Health (111 Centre), Guangzhou, China
| | - Murtaza Ali
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, China
- Overseas Expertise Introduction Centre for Discipline Innovation of Food Nutrition and Human Health (111 Centre), Guangzhou, China
| | - Romana Ishrat
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
- Romana Ishrat
| | - Rafat Ali
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|