1
|
Chauhan P, Begum MY, Narapureddy BR, Gupta S, Wadhwa K, Singh G, Kumawat R, Sharma N, Ballal S, Jha SK, Abomughaid MM, B D, Ojha S, Jha NK. Unveiling the Involvement of Herpes Simplex Virus-1 in Alzheimer's Disease: Possible Mechanisms and Therapeutic Implications. Mol Neurobiol 2025; 62:5850-5874. [PMID: 39648189 DOI: 10.1007/s12035-024-04535-4] [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: 06/27/2023] [Accepted: 10/01/2024] [Indexed: 12/10/2024]
Abstract
Viruses pose a significant challenge and threat to human health, as demonstrated by the current COVID-19 pandemic. Neurodegeneration, particularly in the case of Alzheimer's disease (AD), is significantly influenced by viral infections. AD is a neurodegenerative disease that affects people of all ages and poses a significant threat to millions of individuals worldwide. The precise mechanism behind its development is not yet fully understood; however, the emergence and advancement of AD can be hastened by various environmental factors, such as bacterial and viral infections. There has been a longstanding suspicion that the herpes simplex virus-1 (HSV-1) may have a role to play in the development or advancement of AD. Reactivation of HSV-1 could potentially lead to damage to neurons, either by direct means or indirectly by triggering inflammation. This article provides an overview of the connection between HSV-1 infections and immune cells (astrocytes, microglia, and oligodendrocytes) in the progression of AD. It summarizes recent scientific research on how HSV-1 affects neurons, which could potentially shed light on the clinical features and treatment options for AD. In addition, the paper has explored the impact of HSV-1 on neurons and its role in various aspects of AD, such as Aβ secretion, tau hyperphosphorylation, metabolic dysregulation, oxidative damage, apoptosis, and autophagy. It is believed that the immune response triggered by HSV-1 reactivation plays a role in the development of neurodegeneration in AD. Despite the lack of a cure for AD, researchers have made significant efforts to study the clinical and pathological aspects of the disease, identify biomarkers, and gain insight into its underlying causes. The goal is to achieve early diagnosis and develop treatments that can modify the progression of the disease. The current article discusses the most promising therapy for combating the viral impacts, which provides additional evidence for the frequent reactivations of latent HSV-1 in the AD brain. However, further research is still required to establish the molecular and cellular mechanisms underlying the development of AD through the reactivation of HSV-1. This could potentially lead to new insights in drug development aimed at preventing HSV-1 reactivation and the subsequent development and progression of AD.
Collapse
Affiliation(s)
- Payal Chauhan
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Bayapa Reddy Narapureddy
- Department of Public Health, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Karan Wadhwa
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India.
| | - Rohit Kumawat
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajsthan, Jaipur, India
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges Jhanjeri, Mohali, 140307, Punjab, India
| | - Suhas Ballal
- Departmant of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, Delhi, 110008, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Dheepak B
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Biosciences & Technology, Galgotias University, Greater Noida, India.
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India.
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, 144411, India.
| |
Collapse
|
2
|
Ghanbarian E, Khorsand B, Petersen KK, Nallapu BT, Sajjadi SA, Lipton RB, Ezzati A. Cerebrospinal fluid inflammatory cytokines as prognostic indicators for cognitive decline across Alzheimer's disease spectrum. J Alzheimers Dis 2025:13872877251335915. [PMID: 40262111 DOI: 10.1177/13872877251335915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
BackgroundNeuroinflammation actively contributes to the pathophysiology of Alzheimer's disease (AD); however, the value of neuroinflammatory biomarkers for disease-staging or predicting disease progression remains unclear.ObjectiveTo investigate diagnostic and prognostic utility of inflammatory biomarkers in combination with conventional AD biomarkers.MethodsData from 258 participants in the Alzheimer's Disease Neuroimaging Initiative (ADNI) with cerebrospinal fluid (CSF) biomarkers of amyloid-β (Aβ), tau, and inflammation were analyzed. Clinically meaningful cognitive decline (CMCD) was defined as a ≥ 4-point increase on the Alzheimer's Disease Assessment Scale Cognitive Subscore 11. Predictor variables included demographics (D: age, sex, education), APOE4 status (A), inflammatory biomarkers (I), and classic AD biomarkers of Aβ and p-tau181 (C). Models incorporating inflammatory biomarkers assessed their contribution to improving baseline diagnostic classification and 1-year CMCD prediction.ResultsAt 1-year follow-up, 27.1% of participants experienced CMCD. Adding inflammatory biomarkers to models with D and A variables (DA model) improved classification of cognitively normal (CN) versus mild cognitive impairment (MCI) and CN versus Dementia (p < 0.001). Similarly, inflammatory markers enhanced classification in models including C (DAC model), for CN versus MCI (p < 0.01) and CN versus Dementia (p < 0.001). Predictive performance for CMCD was improved in individuals with MCI and dementia in both models (all p < 0.05). In addition, the DAI model outperformed the DAC model in predicting CMCD for MCI and Dementia groups (both p < 0.05).ConclusionsAddition of CSF inflammatory biomarkers to biomarkers of AD improves diagnostic accuracy of clinical disease stage at baseline and add incremental value to AD biomarkers for prediction of cognitive decline.
Collapse
Affiliation(s)
- Elham Ghanbarian
- Department of Neurology, University of California, Irvine, CA, USA
| | - Babak Khorsand
- Department of Neurology, University of California, Irvine, CA, USA
| | - Kellen K Petersen
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Bhargav T Nallapu
- Saul B. Korey Department of Neurology, Albert Einstein College of Medicine, New York City, NY, USA
| | - S Ahmad Sajjadi
- Department of Neurology, University of California, Irvine, CA, USA
| | - Richard B Lipton
- Saul B. Korey Department of Neurology, Albert Einstein College of Medicine, New York City, NY, USA
| | - Ali Ezzati
- Department of Neurology, University of California, Irvine, CA, USA
| |
Collapse
|
3
|
Saaoud F, Liu L, Xu K, Lu Y, Shao Y, Ben Issa M, Jiang X, Wang X, Liu X, Autieri M, Wu S, Wei J, Yu J, Bouchareb R, Gillespie A, Luo JJ, Martinez L, Vazquez-Padron R, Sun J, Zhao H, Wang H, Pratico D, Yang X. Alzheimer's disease as an auto-innate immune pathology with potential cell trans-differentiation and enhanced trained immunity in 3xTg-AD mouse model. J Alzheimers Dis 2025:13872877251329583. [PMID: 40232249 DOI: 10.1177/13872877251329583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
BackgroundAlzheimer's disease (AD) is a neurodegenerative disorder characterized by memory impairment. Neuroinflammatory processes, mediated by glial and immune cells, contribute to neuronal damage. Emerging evidence implicates innate immune mechanisms, including trained immunity and cell trans-differentiation, in AD pathogenesis, though their roles remain unclear.ObjectiveTo investigate transcriptomic changes in the 3xTg-AD mouse model, focusing on trained immunity and cell trans-differentiation in disease mechanisms.MethodsRNA-sequencing was performed on brain tissue (cortex plus hippocampus) from 11-month-old female 3xTg-AD and wild-type mice (n = 3/group). Differentially expressed genes (fold change > 1.5, p < 0.05) were identified and followed by bioinformatics and knowledge-based transcriptomic profiling. Public AD datasets were also analyzed.Results3xTg-AD mice exhibited 316 upregulated and 412 downregulated genes. Downregulated genes included those for blood-brain barrier protein, while upregulated genes related to cerebrospinal fluid. Increased expression of proinflammatory markers, as well as genes related to cell differentiation, proliferation, activation, and adhesion. Upregulation of genes associated with cell migration and trans-differentiation suggests a potential role for inflammation and cellular plasticity. Additionally, genes involved in inflammasome pathways, immunometabolism, and trained immunity were upregulated. Mechanistically, these genes were modulated by knockdown of trained immunity promoter SET-7, overexpression of trained immunity inhibitor IL-37, and knockout of inflammasome genes IL-1 receptor, caspase-1, and pattern recognition receptor CD36.ConclusionsThe finding underscore the potential role of trained immunity and cell trans-differentiation in AD, revealing a mechanistic framework in which danger-associated molecular patterns drive innate immune responses, inflammasome activation, and cell plasticity contribute to AD, offering therapeutic targets for neuroinflammation and cellular reprograming.
Collapse
Affiliation(s)
- Fatma Saaoud
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
| | - Lu Liu
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Keman Xu
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
| | - Yifan Lu
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
| | - Ying Shao
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
| | - Mohammed Ben Issa
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
| | - Xiaohua Jiang
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Xianwei Wang
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Xiaolei Liu
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
| | - Michael Autieri
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
| | - Sheng Wu
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Juncheng Wei
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Jun Yu
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Rihab Bouchareb
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Avrum Gillespie
- Section of Nephrology, Hypertension, and Kidney Transplantation, Department of Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Jin Jun Luo
- Department of Neurology, Temple University, Philadelphia, PA, USA
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Roberto Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Jianxin Sun
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Huaqing Zhao
- Department of Biomedical Education and Data Sciences, Temple University, Philadelphia, PA, USA
| | - Hong Wang
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Domenico Pratico
- Alzheimer's Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Xiaofeng Yang
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
4
|
Alami M, Morvaridzadeh M, El Khayari A, Boumezough K, El Fatimy R, Khalil A, Fulop T, Berrougui H. Reducing Alzheimer's disease risk with SGLT2 inhibitors: From glycemic control to neuroprotection. Ageing Res Rev 2025; 108:102751. [PMID: 40204129 DOI: 10.1016/j.arr.2025.102751] [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: 02/11/2025] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/11/2025]
Abstract
Recent research has established a strong link between metabolic abnormalities and an increased risk of dementia. In parallel, there is growing epidemiological evidence supporting the neuroprotective effects of antidiabetic medications against cognitive impairments. Among these, sodium-glucose co-transporter (SGLT2) inhibitors have emerged as pharmacological candidates with promising potential in alleviating the burden of age-related diseases, particularly neurodegenerative diseases (NDD). SGLT2 inhibitor therapies are FDA-approved medications routinely prescribed to manage diabetes. This novel class was initially developed to address cardiovascular disorders and to reduce the risk of hypoglycemia associated with insulin-secretagogue agents. It subsequently attracted growing interest for its beneficial effects on central nervous system (CNS) disorders. However, the molecular mechanisms through which these glucose-lowering therapies mitigate cognitive decline and limit the progression of certain brain degenerative diseases remain largely unexplored. Consequently, the neuroscientific community needs further studies that gather, analyze, and critically discuss the available mechanistic evidence regarding the neuroprotective effects of SGLT2 inhibitors. This review aims to critically examine the most relevant published findings, both in vitro and in vivo, as well as human studies evaluating the impact of SGLT2 inhibitors exposure on Alzheimer's disease (AD). It seeks to integrate the current understanding of their beneficial effects at the molecular level and their role in addressing the pathophysiology and neuropathology of AD. These insights will help extend our knowledge of how SGLT2 inhibitor therapies are associated with reduced risk of dementia and thus shed light on the link between diabetes and AD.
Collapse
Affiliation(s)
- Mehdi Alami
- Sultan Moulay Sliman University, Polydisciplinary Faculty, Department of Biology, Beni Mellal, Morocco; University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Mojgan Morvaridzadeh
- University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Abdellatif El Khayari
- Faculty of Medical Sciences, UM6P Hospitals, Mohammed VI Polytechnic University, Ben-Guerir 43150, Morocco; Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Kaoutar Boumezough
- Sultan Moulay Sliman University, Polydisciplinary Faculty, Department of Biology, Beni Mellal, Morocco; University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Rachid El Fatimy
- Faculty of Medical Sciences, UM6P Hospitals, Mohammed VI Polytechnic University, Ben-Guerir 43150, Morocco
| | - Abdelouahed Khalil
- University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Tamas Fulop
- University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Hicham Berrougui
- Sultan Moulay Sliman University, Polydisciplinary Faculty, Department of Biology, Beni Mellal, Morocco; University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada.
| |
Collapse
|
5
|
Hernández‐Fernández F, Martínez‐Fernández I, Barbella‐Aponte R, Vilar IF, Ayo‐Martín O, García‐García J, Collado R, Andrés A, Hernández‐Guillamón M, Pena Pardo FJ, Barrena C, de la Fuente M, Serrano‐Heras G, Melero M, Setién EL, López L, Segura T. Iatrogenic cerebral amyloid angiopathy and Alzheimer's disease co-pathology. Ann Clin Transl Neurol 2025; 12:235-241. [PMID: 39729628 PMCID: PMC11752100 DOI: 10.1002/acn3.52278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 11/06/2024] [Accepted: 11/23/2024] [Indexed: 12/29/2024] Open
Abstract
Iatrogenic cerebral amyloid angiopathy, a disease caused by contact with neurosurgical material or human growth hormone contaminated by beta-amyloid peptide (Aβ), has a prion-like transmission mechanism. We present a series of three patients under 55 years of age who underwent cranial surgery. All of them developed multiple cerebral hemorrhages, transient focal neurological deficits, and/or cognitive impairment after 3-4 decades. MRI was compatible with CAA, and Aβ deposition was confirmed. The third patient, who had a ventriculoperitoneal valve, also showed Aβ deposition in the peritoneum and diagnostic biomarkers of Alzheimer's disease. Co-pathology with Alzheimer disease and its iatrogenic transmission should be considered.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rosa Collado
- Radiology DepartmentAlbacete Universitary HospitalAlbaceteSpain
| | - Alberto Andrés
- Neurology DepartmentAlbacete Universitary HospitalAlbaceteSpain
| | | | | | - Cristina Barrena
- Neurosurgery DepartmentAlbacete Universitary HospitalAlbaceteSpain
| | | | | | - María Melero
- Internal Medicine DepartmentAlbacete Universitary HospitalAlbaceteSpain
| | | | - Luis López
- Neurology DepartmentVigo Universitary HospitalVigoSpain
| | - Tomás Segura
- Neurology DepartmentAlbacete Universitary HospitalAlbaceteSpain
- Facultad de Medicina de AlbaceteInstituto de Biomedicina, UCLMAlbaceteSpain
| |
Collapse
|
6
|
Nussinov R, Jang H, Cheng F. Ras, RhoA, and vascular pharmacology in neurodevelopment and aging. Neurochem Int 2024; 181:105883. [PMID: 39427854 PMCID: PMC11614691 DOI: 10.1016/j.neuint.2024.105883] [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: 09/12/2024] [Revised: 10/01/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024]
Abstract
Small GTPases Ras, Rac, and RhoA are crucial regulators of cellular functions. They also act in dysregulated cell proliferation and transformation. Multiple publications have focused on illuminating their roles and mechanisms, including in immune system pathologies. Their functions in neurology-related diseases, neurodegeneration and neurodevelopment, are also emerging, as well as their potential as pharmacological targets in both pathologies. Observations increasingly suggest that these pathologies may relate to activation (or suppression) of signaling by members of the Ras superfamily, especially Ras, Rho, and Rac isoforms, and components of their signaling pathways. Germline (or embryonic) mutations that they harbor are responsible for neurodevelopmental disorders, such as RASopathies, autism spectrum disorder, and dilated cardiomyopathy. In aging, they promote neurodegenerative diseases, with Rho GTPase featuring in their pharmacology, as in the case of Alzheimer's disease (AD). Significantly, drugs with observed anti-AD activity, particularly those involved in cardiovascular systems, are associated with the RhoA signaling, as well as cerebral vasculature in brain development and aging. This leads us to suggest that anti-AD drugs could inform neurodevelopmental disorders, including pediatric low-grade gliomas pharmacology. Neurodevelopmental disorders associated with RhoA, like autism, are also connected with vascular systems, thus could be targets of vascular system-connected drugs.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD, 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44106, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA; Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44195, USA; Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| |
Collapse
|
7
|
Singh R, Panghal A, Jadhav K, Thakur A, Verma RK, Singh C, Goyal M, Kumar J, Namdeo AG. Recent Advances in Targeting Transition Metals (Copper, Iron, and Zinc) in Alzheimer's Disease. Mol Neurobiol 2024; 61:10916-10940. [PMID: 38809370 DOI: 10.1007/s12035-024-04256-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
Changes in the transition metal homeostasis in the brain are closely linked with Alzheimer's disease (AD), including intraneuronal iron accumulation and extracellular copper and zinc pooling in the amyloid plague. The brain copper, zinc, and iron surplus are commonly acknowledged characteristics of AD, despite disagreements among some. This has led to the theory that oxidative stress resulting from abnormal homeostasis of these transition metals may be a causative explanation behind AD. In the nervous system, the interaction of metals with proteins appears to be an essential variable in the development or suppression of neurodegeneration. Chelation treatment may be an option for treating neurodegeneration induced by transition metal ion dyshomeostasis. Some clinicians even recommend using chelating agents as an adjunct therapy for AD. The current review also looks at the therapeutic strategies that have been attempted, primarily with metal-chelating drugs. Metal buildup in the nervous system, as reported in the AD, could be the result of compensatory mechanisms designed to improve metal availability for physiological functions.
Collapse
Affiliation(s)
- Raghuraj Singh
- Pharmaceutical Nanotechnology Lab, Institutes of Nano Science and Technology (INST), Sector 81. Mohali, Punjab, 140306, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Archna Panghal
- Department of Pharmacology and Toxicology, Facility for Risk Assessment and Intervention Studies, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Punjab, India
| | - Krishna Jadhav
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Ashima Thakur
- Faculty of Pharmaceutical Sciences, ICFAI University, Baddi, Distt. Solan, Himachal Pradesh, 174103, India
| | - Rahul Kumar Verma
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Charan Singh
- Department of Pharmaceutical Sciences Hemwati, Nandan Bahuguna Garhwal University (A Central University), Srinagar, Dist. Garhwal (Uttarakhand), 246174, India
| | - Manoj Goyal
- Department of Pharmaceutical Sciences Hemwati, Nandan Bahuguna Garhwal University (A Central University), Srinagar, Dist. Garhwal (Uttarakhand), 246174, India
| | - Jayant Kumar
- Department of Pharmaceutical Sciences Hemwati, Nandan Bahuguna Garhwal University (A Central University), Srinagar, Dist. Garhwal (Uttarakhand), 246174, India.
| | - Ajay G Namdeo
- Department of Pharmaceutical Sciences Hemwati, Nandan Bahuguna Garhwal University (A Central University), Srinagar, Dist. Garhwal (Uttarakhand), 246174, India
| |
Collapse
|
8
|
Ghanbarian E, Khorsand B, Petersen KK, Nallapu BT, Sajjadi SA, Lipton RB, Ezzati A. CSF inflammatory cytokines as prognostic indicators for cognitive decline across Alzheimer's disease spectrum. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.13.24317270. [PMID: 39606412 PMCID: PMC11601771 DOI: 10.1101/2024.11.13.24317270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background A growing body of evidence suggests that neuroinflammation contributes actively to pathophysiology of Alzheimer's disease (AD) and promotes AD progression. The predictive value of neuroinflammatory biomarkers for disease-staging or estimating disease progression is not well understood. In this study, we investigate the diagnostic and prognostic utility of inflammatory biomarkers in combination with conventional AD biomarkers. Methods We included 258 participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI) who had CSF biomarkers of β-Amyloid (Aβ), tau, and inflammation. The primary outcome of interest was clinically meaningful cognitive decline (CMCD) as defined by an increase of ≥4 on the Alzheimer's Disease Assessment Scale Cognitive Subscore 11 (ADAS-11, scores 0-70, higher scores indicate worse cognition). Predictor variables were categorized as demographics (D; age, sex, and education), genetic (APOE4 status (A)), inflammatory biomarkers (I), and classic (C) cerebrospinal fluid (CSF) biomarkers of Aβ and p-tau181. Simultaneous inclusion of eleven CSF inflammatory biomarkers as covariates in logistic regression models was examined to assess improvements in classifying baseline clinical diagnoses (cognitively normal (CN), mild cognitive impairment (MCI), Dementia) and predicting individuals with and without CMCD over one year of follow-up. Results At 1-year follow up, 27.1% of participants experienced CMCD. Inclusion of inflammatory biomarkers improved baseline classification of CN vs MCI as well as CN vs Dementia for models including D and A variables (DA; both p<0.001). Similarly, when classic CSF biomarkers of AD were included into the model (DAC model), inclusion of inflammatory markers improved classification of CN vs MCI (p<0.01) as well as CN vs Dementia (p<0.001). Addition of inflammatory biomarkers to both DA and DAC models improved predictive performance for CMCD in persons with baseline MCI and Dementia (all p<0.05), but not in the CN group. In addition, the predictive performance of the DAI model was superior to the DAC model in the MCI and Dementia groups (both p<0.05). Conclusions Addition of CSF inflammatory biomarkers to CSF biomarkers of AD can improve diagnostic accuracy of clinical disease stage at baseline and add incremental value to AD biomarkers for prediction of cognitive decline.
Collapse
Affiliation(s)
- Elham Ghanbarian
- Department of Neurology, University of California, Irvine, CA, USA
| | - Babak Khorsand
- Department of Neurology, University of California, Irvine, CA, USA
| | - Kellen K. Petersen
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Bhargav T. Nallapu
- Saul B. Korey Department of Neurology, Albert Einstein College of Medicine, New York City, NY, USA
| | - S. Ahmad Sajjadi
- Department of Neurology, University of California, Irvine, CA, USA
| | - Richard B. Lipton
- Saul B. Korey Department of Neurology, Albert Einstein College of Medicine, New York City, NY, USA
| | - Ali Ezzati
- Department of Neurology, University of California, Irvine, CA, USA
| |
Collapse
|
9
|
Sun Y, Shi Y, Li J, Qian A, Shi H. New Hypothesis on Enhancing β-Sheet Formation during the Tau Fragment Dimer Transition from a Flexible Monomer: Insights into Primary Nucleation Processes by Histidine Behaviors. J Phys Chem Lett 2024; 15:10763-10768. [PMID: 39422640 DOI: 10.1021/acs.jpclett.4c02419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Slight perturbations in pH can have significant effects on the primary nucleation processes of the tau protein. The behaviors of histidine due to its pivotal role in modulating H-bonding network interactions and electrostatic interactions have garnered considerable attention, as it can influence the structural characteristics and aggregation properties. However, the nucleation mechanisms and related intermediates are still unclear. In the current study, we performed nine independent replica exchange molecular dynamics simulations to investigate dimer formation involving R3(εδ) in conjunction with the R1, R2, and R4 monomers. Our findings substantiate that, in comparison to R1-R3(εδ) and R4-R3(εδ) systems, the R2-R3(εδ) systems consistently manifest the highest averaged β-sheet content, with the fundamental feature of R3(εδ) promoting R2 rearrangement. Our comprehensive analysis reveals that high-β-sheet-rich systems exhibit a conserved three/five β-strand structure. In these β-strand-rich systems, one chain [R1/R2/R4 or R3(εδ)] with robust intrachain H-bonding interactions coordinates with another chain through interchain H-bonding interactions, contributing to the overall stability. Furthermore, we discuss distinct histidine behaviors, including backbone/side chain interactions and donor/acceptor roles. This study provides a comprehensive understanding of the aggregation propensities of soluble tau oligomers and sheds light on the primary nucleation mechanism. It contributes to a new perspective for understanding protein folding and misfolding.
Collapse
Affiliation(s)
- Yue Sun
- School of Chemistry and Chemical Engineering, Institute of Molecular Science, Shanxi University, Taiyuan, Shanxi 030006, People's Republic of China
| | - Yaru Shi
- School of Chemistry and Chemical Engineering, Institute of Molecular Science, Shanxi University, Taiyuan, Shanxi 030006, People's Republic of China
| | - Jing Li
- School of Chemistry and Chemical Engineering, Institute of Molecular Science, Shanxi University, Taiyuan, Shanxi 030006, People's Republic of China
| | - Aniu Qian
- Institute of Resources and Environment Engineering, Shanxi University, Taiyuan, Shanxi 030006, People's Republic of China
| | - Hu Shi
- School of Chemistry and Chemical Engineering, Institute of Molecular Science, Shanxi University, Taiyuan, Shanxi 030006, People's Republic of China
| |
Collapse
|
10
|
Do Carmo S, Kautzmann MAI, Bhattacharjee S, Jun B, Steinberg C, Emmerson JT, Malcolm JC, Bonomo Q, Bazan NG, Cuello AC. Differential effect of an evolving amyloid and tau pathology on brain phospholipids and bioactive lipid mediators in rat models of Alzheimer-like pathology. J Neuroinflammation 2024; 21:185. [PMID: 39080670 PMCID: PMC11290283 DOI: 10.1186/s12974-024-03184-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Brain inflammation contributes significantly to the pathophysiology of Alzheimer's disease, and it is manifested by glial cell activation, increased production of cytokines/chemokines, and a shift in lipid mediators from a pro-homeostatic to a pro-inflammatory profile. However, whether the production of bioactive lipid mediators is affected at earlier stages, prior to the deposition of Aβ plaques and tau hyperphosphorylation, is unknown. The differential contribution of an evolving amyloid and tau pathology on the composition and abundance of membrane phospholipids and bioactive lipid mediators also remains unresolved. METHODS In this study, we examined the cortical levels of DHA- and AA-derived bioactive lipid mediators and of membrane phospholipids by liquid chromatography with tandem mass spectrometry in transgenic rat models of the Alzheimer's-like amyloid and tau pathologies at early and advanced pathological stages. RESULTS Our findings revealed a complex balance between pro-inflammatory and pro-resolving processes in which tau pathology has a more pronounced effect compared to amyloid pathology. At stages preceding tau misfolding and aggregation, there was an increase in pro-resolving lipid mediators (RVD6 and NPD1), DHA-containing phospholipids and IFN-γ levels. However, in advanced tau pathology displaying NFT-like inclusions, neuronal death, glial activation and cognitive deficits, there was an increase in cytokine and PGD2, PGE2, and PGF2α generation accompanied by a drop in IFN-γ levels. This pathology also resulted in a marked increase in AA-containing phospholipids. In comparison, pre-plaque amyloid pathology already presented high levels of cytokines and AA-containing phospholipids together with elevated RVD6 and NPD1 levels. Finally, Aβ plaque deposition was accompanied by a modest increase in prostaglandins, increased AA-containing phospholipids and reduced DHA-containing phospholipids. CONCLUSIONS Our findings suggest a dynamic trajectory of inflammatory and lipid mediators in the evolving amyloid and tau pathologies and support their differing roles on membrane properties and, consequentially, on signal transduction.
Collapse
Affiliation(s)
- Sonia Do Carmo
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William Osler, Room 1210, Montreal, H3G 1Y6, Canada.
| | - Marie-Audrey I Kautzmann
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA
| | - Surjyadipta Bhattacharjee
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA
| | - Bokkyoo Jun
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA
| | - Carolyn Steinberg
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William Osler, Room 1210, Montreal, H3G 1Y6, Canada
| | - Joshua T Emmerson
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William Osler, Room 1210, Montreal, H3G 1Y6, Canada
| | - Janice C Malcolm
- Department of Cell Anatomy and Cell Biology, McGill University, Montreal, H3A 0C7, Canada
| | - Quentin Bonomo
- Department of Neurology and Neurosurgery, McGill University, Montreal, H3G 1Y6, Canada
| | - Nicolas G Bazan
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William Osler, Room 1210, Montreal, H3G 1Y6, Canada.
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA.
| | - A Claudio Cuello
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William Osler, Room 1210, Montreal, H3G 1Y6, Canada.
- Department of Cell Anatomy and Cell Biology, McGill University, Montreal, H3A 0C7, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, H3G 1Y6, Canada.
- Department of Pharmacology, Oxford University, Oxford, OX1 3QT, UK.
| |
Collapse
|
11
|
Zhang E, Chen T, Chen Y, Long C, Tao L, Shen X, Dai F. The role of Immune cells in Alzheimer's disease: a bidirectional Mendelian randomization study. Front Aging Neurosci 2024; 16:1433691. [PMID: 39076206 PMCID: PMC11284151 DOI: 10.3389/fnagi.2024.1433691] [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: 05/16/2024] [Accepted: 06/25/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a leading cause of dementia, characterized by the accumulation of amyloid-beta (Aβ) and hyperphosphorylated tau proteins, leading to neuroinflammation and neuronal damage. The role of the immune system in AD pathogenesis is increasingly recognized, prompting an exploration of the causal relationship between immune cells and AD by using Mendelian randomization (MR) approaches. METHODS Utilizing genome-wide association study (GWAS) data from European cohorts, we conducted an MR study to investigate the causal links between immune cell phenotypes and AD. We selected single nucleotide polymorphisms (SNPs) associated with immune cell traits at a genome-wide significance threshold and applied various MR methods, including MR Egger, Weighted median, and inverse variance weighted analysis, to assess the causality between 731 immune phenotypes and AD. RESULTS Our MR analysis identified 15 immune cell types with significant causal relationships to AD pathogenesis. Notably, the absolute count of CD28-CD4-CD8- T cells and the expression of HLA DR on B cells were linked to a protective effect against AD, while 13 other immune phenotypes were identified as contributing to the risk factors for the disease. The causal effects of AD on immunophenotypic traits are predominantly negative, implying that AD may impair the functionality of immune cells. Validation through independent datasets, such as FinnGen and GCST90027158, confirmed the causal association between six specific immune cells and AD. CONCLUSION This comprehensive MR study elucidates the intricate network of causal relationships between diverse immunophenotypic traits and AD, providing novel insights into the immunopathogenesis of AD. The findings suggest potential immunological targets that could be leveraged for early diagnosis, disease monitoring, and therapeutic intervention.
Collapse
Affiliation(s)
- Erdong Zhang
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| | - Tingting Chen
- The Pharmacy Department, Guiyang Maternal and Child Health-Care Hospital, Guiyang, Guizhou, China
| | - Yanqin Chen
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| | - Chenxiang Long
- Department of Anatomy, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Ling Tao
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xiangchun Shen
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| | - Fengqiu Dai
- Department of Anatomy, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| |
Collapse
|
12
|
Weaver DF. β-Amyloid Is a Kinocidin Cytokine. ACS Chem Neurosci 2024; 15:1948-1950. [PMID: 38684014 DOI: 10.1021/acschemneuro.4c00236] [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: 05/02/2024] Open
Abstract
Although β-amyloid (Aβ) has long been studied as a key contributor to the pathology of Alzheimer's disease (AD), its physiological role (if any) remains undetermined. This Viewpoint highlights the evolving concept that if AD is an immunopathic disorder then Aβ may be an immunopeptide. The identification of Aβ as an immunopeptide is further refined to it being a kinocidin, a subtype of cytokine with antimicrobial activity.
Collapse
Affiliation(s)
- Donald F Weaver
- Krembil Research Institute, University Health Network, Dept. of Chemistry, University of Toronto, 60 Leonard Avenue, Toronto, Ontario M5T 0S8, Canada
| |
Collapse
|
13
|
Lee C, Friedman A. Generating PET scan patterns in Alzheimer's by a mathematical model. PLoS One 2024; 19:e0299637. [PMID: 38625863 PMCID: PMC11020767 DOI: 10.1371/journal.pone.0299637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/13/2024] [Indexed: 04/18/2024] Open
Abstract
Alzheimer disease (AD) is the most common form of dementia. The cause of the disease is unknown, and it has no cure. Symptoms include cognitive decline, memory loss, and impairment of daily functioning. The pathological hallmarks of the disease are aggregation of plaques of amyloid-β (Aβ) and neurofibrillary tangles of tau proteins (τ), which can be detected in PET scans of the brain. The disease can remain asymptomatic for decades, while the densities of Aβ and τ continue to grow. Inflammation is considered an early event that drives the disease. In this paper, we develop a mathematical model that can produce simulated patterns of (Aβ,τ) seen in PET scans of AD patients. The model is based on the assumption that early inflammations, R and [Formula: see text], drive the growth of Aβ and τ, respectively. Recently approved drugs can slow the progression of AD in patients, provided treatment begins early, before significant damage to the brain has occurred. In line with current longitudinal studies, we used the model to demonstrate how to assess the efficacy of such drugs when given years before the disease becomes symptomatic.
Collapse
Affiliation(s)
- Chaeyoung Lee
- Department of Mathematics, Kyonggi University, Suwon, Republic of Korea
| | - Avner Friedman
- Department of Mathematics, The Ohio State University, Columbus, OH, United States of America
| |
Collapse
|
14
|
Nouraeinejad A. The bidirectional links between coronavirus disease 2019 and Alzheimer's disease. Int J Neurosci 2024:1-15. [PMID: 38451045 DOI: 10.1080/00207454.2024.2327403] [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: 07/10/2023] [Accepted: 03/02/2024] [Indexed: 03/08/2024]
Abstract
Coronavirus disease 2019 (COVID-19) can be a critical disease, particularly in the elderly and those with comorbidities. Patients with Alzheimer's disease are more vulnerable to COVID-19 consequences. The latest results have indicated some common risk factors for both diseases. An understanding of the pathological link between COVID-19 and Alzheimer's disease will help develop timely strategies to treat both diseases. This review explores the bidirectional links between COVID-19 and Alzheimer's disease.
Collapse
Affiliation(s)
- Ali Nouraeinejad
- Faculty of Brain Sciences, Institute of Ophthalmology, University College London (UCL), London, United Kingdom
| |
Collapse
|
15
|
Weaver DF. Thirty Risk Factors for Alzheimer's Disease Unified by a Common Neuroimmune-Neuroinflammation Mechanism. Brain Sci 2023; 14:41. [PMID: 38248256 PMCID: PMC10813027 DOI: 10.3390/brainsci14010041] [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: 11/29/2023] [Revised: 12/27/2023] [Accepted: 12/30/2023] [Indexed: 01/23/2024] Open
Abstract
One of the major obstacles confronting the formulation of a mechanistic understanding for Alzheimer's disease (AD) is its immense complexity-a complexity that traverses the full structural and phenomenological spectrum, including molecular, macromolecular, cellular, neurological and behavioural processes. This complexity is reflected by the equally complex diversity of risk factors associated with AD. However, more than merely mirroring disease complexity, risk factors also provide fundamental insights into the aetiology and pathogenesis of AD as a neurodegenerative disorder since they are central to disease initiation and subsequent propagation. Based on a systematic literature assessment, this review identified 30 risk factors for AD and then extended the analysis to further identify neuroinflammation as a unifying mechanism present in all 30 risk factors. Although other mechanisms (e.g., vasculopathy, proteopathy) were present in multiple risk factors, dysfunction of the neuroimmune-neuroinflammation axis was uniquely central to all 30 identified risk factors. Though the nature of the neuroinflammatory involvement varied, the activation of microglia and the release of pro-inflammatory cytokines were a common pathway shared by all risk factors. This observation provides further evidence for the importance of immunopathic mechanisms in the aetiopathogenesis of AD.
Collapse
Affiliation(s)
- Donald F Weaver
- Krembil Research Institute, University Health Network, Departments of Medicine, Chemistry, Pharmaceutical Sciences, University of Toronto, Toronto, ON M5T 0S8, Canada
| |
Collapse
|
16
|
Hosseinzadeh S, Afshari S, Molaei S, Rezaei N, Dadkhah M. The role of genetics and gender specific differences in neurodegenerative disorders: Insights from molecular and immune landscape. J Neuroimmunol 2023; 384:578206. [PMID: 37813041 DOI: 10.1016/j.jneuroim.2023.578206] [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: 07/31/2023] [Revised: 09/09/2023] [Accepted: 09/23/2023] [Indexed: 10/11/2023]
Abstract
Neurodegenerative disorders (NDDs) are the most common neurological disorders with high prevalence and have significant socioeconomic implications. Understanding the underlying cellular and molecular mechanisms associated with the immune system can be effective in disease etiology, leading to more effective therapeutic approaches for both females and males. The central nervous system (CNS) actively participates in immune responses, both within and outside the CNS. Immune system activation is a common feature in NDDs. Gender-specific factors play a significant role in the prevalence, progression, and manifestation of NDDs. Neuroinflammation, in both inflammatory neurological and neurodegenerative conditions, is defined by the triggering of microglia and astrocyte cell activation. This results in the secretion of pro-inflammatory cytokines and chemokines. Numerous studies have documented the role of neuroinflammation in neurological diseases, highlighting the involvement of immune signaling pathways in disease development. Converging evidence support immune system involvement during neurodegeneration in NDDs. In this review, we summarize emerging evidence that reveals gender-dependent differences in immune responses related to NDDs. Also, we highlight sex differences in immune responses and discuss how these sex-specific influences can increase the risk of NDDs. Understanding the role of gender-specific factors can aid in developing targeted therapeutic strategies and improving patient outcomes. Ultimately, the better understanding of these mechanisms contributed to sex-dependent immune response in NDDs, can be critically usful in targeting of immune signaling cascades in such disorders. In this regard, sex-related immune responses in NDDs may be promising and effective targets in therapeutic strategies.
Collapse
Affiliation(s)
- Shahnaz Hosseinzadeh
- Department of Microbiology & Immunology, School of Medicine, Ardabil University of Medical Sciences, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Salva Afshari
- Students Research Committee, Pharmacy School, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Soheila Molaei
- Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran 1419733151, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education Research Network (USERN), Tehran, Iran
| | - Masoomeh Dadkhah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
17
|
Reas ET, Shadrin A, Frei O, Motazedi E, McEvoy L, Bahrami S, van der Meer D, Makowski C, Loughnan R, Wang X, Broce I, Banks SJ, Fominykh V, Cheng W, Holland D, Smeland OB, Seibert T, Selbæk G, Brewer JB, Fan CC, Andreassen OA, Dale AM. Improved multimodal prediction of progression from MCI to Alzheimer's disease combining genetics with quantitative brain MRI and cognitive measures. Alzheimers Dement 2023; 19:5151-5158. [PMID: 37132098 PMCID: PMC10620101 DOI: 10.1002/alz.13112] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/21/2023] [Accepted: 04/04/2023] [Indexed: 05/04/2023]
Abstract
INTRODUCTION There is a pressing need for non-invasive, cost-effective tools for early detection of Alzheimer's disease (AD). METHODS Using data from the Alzheimer's Disease Neuroimaging Initiative (ADNI), Cox proportional models were conducted to develop a multimodal hazard score (MHS) combining age, a polygenic hazard score (PHS), brain atrophy, and memory to predict conversion from mild cognitive impairment (MCI) to dementia. Power calculations estimated required clinical trial sample sizes after hypothetical enrichment using the MHS. Cox regression determined predicted age of onset for AD pathology from the PHS. RESULTS The MHS predicted conversion from MCI to dementia (hazard ratio for 80th versus 20th percentile: 27.03). Models suggest that application of the MHS could reduce clinical trial sample sizes by 67%. The PHS alone predicted age of onset of amyloid and tau. DISCUSSION The MHS may improve early detection of AD for use in memory clinics or for clinical trial enrichment. HIGHLIGHTS A multimodal hazard score (MHS) combined age, genetics, brain atrophy, and memory. The MHS predicted time to conversion from mild cognitive impairment to dementia. MHS reduced hypothetical Alzheimer's disease (AD) clinical trial sample sizes by 67%. A polygenic hazard score predicted age of onset of AD neuropathology.
Collapse
Affiliation(s)
- Emilie T. Reas
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Alexey Shadrin
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Oleksandr Frei
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
- Center for Bioinformatics, Department of Informatics, University of Oslo, PO box 1080, Blindern, 0316 Oslo, Norway
| | - Ehsan Motazedi
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Linda McEvoy
- Department of Radiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shahram Bahrami
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Dennis van der Meer
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Carolina Makowski
- Department of Radiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Robert Loughnan
- University of California, San Diego, La Jolla, California, USA
| | - Xin Wang
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Iris Broce
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Sarah J. Banks
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Vera Fominykh
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Weiqiu Cheng
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Dominic Holland
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Olav B. Smeland
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Tyler Seibert
- Department of Radiology, University of California, San Diego, La Jolla, CA 92093, USA
| | | | - James B. Brewer
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Chun C. Fan
- Population Neuroscience and Genetics Lab, University of California, La Jolla, CA 92093, USA
- Center for Human Development, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Radiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ole A. Andreassen
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Anders M. Dale
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
- Population Neuroscience and Genetics Lab, University of California, La Jolla, CA 92093, USA
- Department of Radiology, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
18
|
Widjaya MA, Liu CH, Lee SD, Cheng WC. Transcriptomics Meta-Analysis Reveals Phagosome and Innate Immune System Dysfunction as Potential Mechanisms in the Cortex of Alzheimer's Disease Mouse Strains. J Mol Neurosci 2023; 73:773-786. [PMID: 37733230 DOI: 10.1007/s12031-023-02152-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/30/2023] [Indexed: 09/22/2023]
Abstract
Immune-related pathways can affect the immune system directly, such as the chemokine signaling pathway, or indirectly, such as the phagosome pathway. Alzheimer's disease (AD) is reportedly associated with several immune-related pathways. However, exploring its underlying mechanism is challenging in animal studies because AD mouse strains differentially express immune-related pathway characteristics. To overcome this problem, we performed a meta-analysis to identify significant and consistent immune-related AD pathways that are expressed in different AD mouse strains. Next-generation RNA sequencing (RNA-seq) and microarray datasets for the cortex of AD mice from different strains such as APP/PSEN1, APP/PS2, 3xTg, TREM, and 5xFAD were collected from the NCBI GEO database. Each dataset's quality control and normalization were already processed from each original study source using various methods depending on the high-throughput analysis platform (FastQC, median of ratios, RMA, between array normalization). Datasets were analyzed using DESeq2 for RNA-seq and GEO2R for microarray to identify differentially expressed (DE) genes. Significantly DE genes were meta-analyzed using Stouffer's method, with significant genes further analyzed for functional enrichment. Ten datasets representing 20 conditions were obtained from the NCBI GEO database, comprising 116 control and 120 AD samples. The DE analysis identified 284 significant DE genes. The meta-analysis identified three significantly enriched immune-related AD pathways: phagosome, the complement and coagulation cascade, and chemokine signaling. Phagosomes-related genes correlated with complement and immune system. Meanwhile, phagosomes and chemokine signaling genes overlapped with B cells receptors pathway genes indicating potential correlation between phagosome, chemokines, and adaptive immune system as well. The transcriptomic meta-analysis showed that AD is associated with immune-related pathways in the brain's cortex through the phagosome, complement and coagulation cascade, and chemokine signaling pathways. Interestingly, phagosome and chemokine signaling pathways had potential correlation with B cells receptors pathway.
Collapse
Affiliation(s)
- Michael Anekson Widjaya
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Chia-Hsin Liu
- Cancer Biology and Precision Therapeutics Center, China Medical University and Academia Sinica China Medical University, Taichung, 40403, Taiwan
| | - Shin-Da Lee
- Department of Physical Therapy, PhD program in Healthcare Science, China Medical University, Taichung, 406040, Taiwan.
| | - Wei-Chung Cheng
- Cancer Biology and Precision Therapeutics Center, China Medical University and Academia Sinica China Medical University, Taichung, 40403, Taiwan.
- Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, Taiwan.
| |
Collapse
|
19
|
Clement M. The association of microbial infection and adaptive immune cell activation in Alzheimer's disease. DISCOVERY IMMUNOLOGY 2023; 2:kyad015. [PMID: 38567070 PMCID: PMC10917186 DOI: 10.1093/discim/kyad015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/31/2023] [Accepted: 09/04/2023] [Indexed: 04/04/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common form of dementia. Early symptoms include the loss of memory and mild cognitive ability; however, as the disease progresses, these symptoms can present with increased severity manifesting as mood and behaviour changes, disorientation, and a loss of motor/body control. AD is one of the leading causes of death in the UK, and with an ever-increasing ageing society, patient numbers are predicted to rise posing a significant global health emergency. AD is a complex neurophysiological disorder where pathology is characterized by the deposition and aggregation of misfolded amyloid-beta (Aβ)-protein that in-turn promotes excessive tau-protein production which together drives neuronal cell dysfunction, neuroinflammation, and neurodegeneration. It is widely accepted that AD is driven by a combination of both genetic and immunological processes with recent data suggesting that adaptive immune cell activity within the parenchyma occurs throughout disease. The mechanisms behind these observations remain unclear but suggest that manipulating the adaptive immune response during AD may be an effective therapeutic strategy. Using immunotherapy for AD treatment is not a new concept as the only two approved treatments for AD use antibody-based approaches to target Aβ. However, these have been shown to only temporarily ease symptoms or slow progression highlighting the urgent need for newer treatments. This review discusses the role of the adaptive immune system during AD, how microbial infections may be contributing to inflammatory immune activity and suggests how adaptive immune processes can pose as therapeutic targets for this devastating disease.
Collapse
Affiliation(s)
- Mathew Clement
- Division of Infection and Immunity, Systems Immunity University Research Institute, Cardiff University, Cardiff, UK
| |
Collapse
|
20
|
Temmerman J, Engelborghs S, Bjerke M, D’haeseleer M. Cerebrospinal fluid inflammatory biomarkers for disease progression in Alzheimer's disease and multiple sclerosis: a systematic review. Front Immunol 2023; 14:1162340. [PMID: 37520580 PMCID: PMC10374015 DOI: 10.3389/fimmu.2023.1162340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/12/2023] [Indexed: 08/01/2023] Open
Abstract
Inflammatory processes are involved in the pathophysiology of both Alzheimer's disease (AD) and multiple sclerosis (MS) but their exact contribution to disease progression remains to be deciphered. Biomarkers are needed to define pathophysiological processes of these disorders, who may increasingly co-exist in the elderly generations of the future, due to the rising prevalence in both and ameliorated treatment options with improved life expectancy in MS. The purpose of this review was to provide a systematic overview of inflammatory biomarkers, as measured in the cerebrospinal fluid (CSF), that are associated with clinical disease progression. International peer-reviewed literature was screened using the PubMed and Web of Science databases. Disease progression had to be measured using clinically validated tests representing baseline functional and/or cognitive status, the evolution of such clinical scores over time and/or the transitioning from one disease stage to a more severe stage. The quality of included studies was systematically evaluated using a set of questions for clinical, neurochemical and statistical characteristics of the study. A total of 84 papers were included (twenty-five for AD and 59 for MS). Elevated CSF levels of chitinase-3-like protein 1 (YKL-40) were associated with disease progression in both AD and MS. Osteopontin and monocyte chemoattractant protein-1 were more specifically related to disease progression in AD, whereas the same was true for interleukin-1 beta, tumor necrosis factor alpha, C-X-C motif ligand 13, glial fibrillary acidic protein and IgG oligoclonal bands in MS. We observed a broad heterogeneity of studies with varying cohort characterization, non-disclosure of quality measures for neurochemical analyses and a lack of adequate longitudinal designs. Most of the retrieved biomarkers are related to innate immune system activity, which seems to be an important mediator of clinical disease progression in AD and MS. Overall study quality was limited and we have framed some recommendations for future biomarker research in this field. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42021264741.
Collapse
Affiliation(s)
- Joke Temmerman
- Vrije Universiteit Brussel, Center for Neurosciences (C4N), Jette, Brussels, Belgium
- Universiteit Antwerpen, Department of Biomedical Sciences and Institute Born-Bunge, Reference Center for Biological Markers of Dementia (BIODEM), Wilrijk, Antwerp, Belgium
- Universitair Ziekenhuis Brussel, Department of Neurology, Jette, Brussels, Belgium
| | - Sebastiaan Engelborghs
- Vrije Universiteit Brussel, Center for Neurosciences (C4N), Jette, Brussels, Belgium
- Universiteit Antwerpen, Department of Biomedical Sciences and Institute Born-Bunge, Reference Center for Biological Markers of Dementia (BIODEM), Wilrijk, Antwerp, Belgium
- Universitair Ziekenhuis Brussel, Department of Neurology, Jette, Brussels, Belgium
| | - Maria Bjerke
- Vrije Universiteit Brussel, Center for Neurosciences (C4N), Jette, Brussels, Belgium
- Universiteit Antwerpen, Department of Biomedical Sciences and Institute Born-Bunge, Reference Center for Biological Markers of Dementia (BIODEM), Wilrijk, Antwerp, Belgium
- Universitair Ziekenhuis Brussel, Department of Neurology, Jette, Brussels, Belgium
- Universitair Ziekenhuis Brussel, Department of Clinical Biology, Laboratory of Clinical Neurochemistry, Jette, Brussels, Belgium
| | - Miguel D’haeseleer
- Vrije Universiteit Brussel, Center for Neurosciences (C4N), Jette, Brussels, Belgium
- Universitair Ziekenhuis Brussel, Department of Neurology, Jette, Brussels, Belgium
- National MS Center (NMSC), Neurology, Melsbroek, Steenokkerzeel, Belgium
| |
Collapse
|
21
|
Johnson AM, Lukens JR. The innate immune response in tauopathies. Eur J Immunol 2023; 53:e2250266. [PMID: 36932726 PMCID: PMC10247424 DOI: 10.1002/eji.202250266] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/23/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023]
Abstract
Tauopathies, which include frontotemporal dementia, Alzheimer's disease, and chronic traumatic encephalopathy, are a class of neurological disorders resulting from pathogenic tau aggregates. These aggregates disrupt neuronal health and function leading to the cognitive and physical decline of tauopathy patients. Genome-wide association studies and clinical evidence have brought to light the large role of the immune system in inducing and driving tau-mediated pathology. More specifically, innate immune genes are found to harbor tauopathy risk alleles, and innate immune pathways are upregulated throughout the course of disease. Experimental evidence has expanded on these findings by describing pivotal roles for the innate immune system in the regulation of tau kinases and tau aggregates. In this review, we summarize the literature implicating innate immune pathways as drivers of tauopathy.
Collapse
Affiliation(s)
- Alexis M. Johnson
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA
- Neuroscience Graduate Program, UVA, Charlottesville, VA 22908, USA
- BIG Training Graduate Program, UVA, Charlottesville, VA 22908, USA
| | - John R. Lukens
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA
- Neuroscience Graduate Program, UVA, Charlottesville, VA 22908, USA
- BIG Training Graduate Program, UVA, Charlottesville, VA 22908, USA
| |
Collapse
|
22
|
Sobue A, Komine O, Yamanaka K. Neuroinflammation in Alzheimer's disease: microglial signature and their relevance to disease. Inflamm Regen 2023; 43:26. [PMID: 37165437 PMCID: PMC10170691 DOI: 10.1186/s41232-023-00277-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/26/2023] [Indexed: 05/12/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, pathologically characterized by senile plaques and neurofibrillary tangles (NFTs), resulting in neurodegeneration. Neuroinflammation, defined as the activation of glial cells such as microglia and astrocytes, is observed surrounding senile plaques and affected neurons in AD. Recently conducted genome-wide association studies (GWAS) indicate that a large section of identified AD risk genes are involved in immune responses and are enriched in microglia. Microglia are innate immune cells in the central nervous system (CNS), which are involved in immune surveillance and maintenance of homeostasis in the CNS. Recently, a novel subpopulation of activated microglia named as disease-associated microglia (DAM), also known as activated response microglia (ARM) or microglial neurodegenerative phenotype (MGnD), was identified in AD model mice. These microglia closely associate with β-amyloid (Aβ) plaques and exhibit characteristic gene expression profiles accompanied with reduced expressions of homeostatic microglial genes. However, it remains unclear whether decreased homeostatic microglia functions or increased DAM/ARM/MGnD functions correlate with the degree of neuronal loss in AD. To translate the results of rodent studies to human AD, precuneus, the brain region vulnerable to β-amyloid accumulation in preclinical AD, is of high interest, as it can provide novel insights into the mechanisms of microglia response to Aβ in early AD. In this study, we performed comparative analyses of gene expression profiles of microglia among three representative neurodegenerative mouse models and the human precunei with early AD pathology. We proceeded to evaluate the identified genes as potential therapeutic targets for AD. We believe that our findings will provide important resources to better understand the role of glial dysfunction in AD.
Collapse
Affiliation(s)
- Akira Sobue
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Aichi, 464-8601, Japan.
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Aichi, 466-8550, Japan.
- Medical Interactive Research and Academia Industry Collaboration Center, Research Institute of Environmental Medicine, Nagoya University, Aichi, 464-8601, Japan.
| | - Okiru Komine
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Aichi, 464-8601, Japan.
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Aichi, 466-8550, Japan.
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Aichi, 464-8601, Japan.
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Aichi, 466-8550, Japan.
- Institute for Glyco-core Research (iGCORE), Nagoya University, Aichi, Japan.
- Center for One Medicine Innovative Translational Research (COMIT), Nagoya University, Aichi, Japan.
| |
Collapse
|
23
|
Schreiner TG, Schreiner OD, Adam M, Popescu BO. The Roles of the Amyloid Beta Monomers in Physiological and Pathological Conditions. Biomedicines 2023; 11:1411. [PMID: 37239082 PMCID: PMC10216198 DOI: 10.3390/biomedicines11051411] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/30/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Amyloid beta peptide is an important biomarker in Alzheimer's disease, with the amyloidogenic hypothesis as one of the central hypotheses trying to explain this type of dementia. Despite numerous studies, the etiology of Alzheimer's disease remains incompletely known, as the pathological accumulation of amyloid beta aggregates cannot fully explain the complex clinical picture of the disease. Or, for the development of effective therapies, it is mandatory to understand the roles of amyloid beta at the brain level, from its initial monomeric stage prior to aggregation in the form of senile plaques. In this sense, this review aims to bring new, clinically relevant data on a subject intensely debated in the literature in the last years. In the first part, the amyloidogenic cascade is reviewed and the possible subtypes of amyloid beta are differentiated. In the second part, the roles played by the amyloid beta monomers in physiological and pathological (neurodegenerative) conditions are illustrated based on the most relevant and recent studies published on this topic. Finally, considering the importance of amyloid beta monomers in the pathophysiology of Alzheimer's disease, new research directions with diagnostic and therapeutic impacts are suggested.
Collapse
Affiliation(s)
- Thomas Gabriel Schreiner
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania;
- Faculty of Electrical Engineering and Information Technology, Gheorghe Asachi Technical University of Iasi, 21–23 Professor Dimitrie Mangeron Blvd., 700050 Iasi, Romania;
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania;
| | - Oliver Daniel Schreiner
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania;
- Medical Oncology Department, Regional Institute of Oncology, 700483 Iași, Romania
| | - Maricel Adam
- Faculty of Electrical Engineering and Information Technology, Gheorghe Asachi Technical University of Iasi, 21–23 Professor Dimitrie Mangeron Blvd., 700050 Iasi, Romania;
| | - Bogdan Ovidiu Popescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania;
- Neurology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
- Laboratory of Cell Biology, Neurosciences and Experimental Myology, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
24
|
Bianco A, Antonacci Y, Liguori M. Sex and Gender Differences in Neurodegenerative Diseases: Challenges for Therapeutic Opportunities. Int J Mol Sci 2023; 24:6354. [PMID: 37047320 PMCID: PMC10093984 DOI: 10.3390/ijms24076354] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
The term "neurodegenerative diseases" (NDs) identifies a group of heterogeneous diseases characterized by progressive loss of selectively vulnerable populations of neurons, which progressively deteriorates over time, leading to neuronal dysfunction. Protein aggregation and neuronal loss have been considered the most characteristic hallmarks of NDs, but growing evidence confirms that significant dysregulation of innate immune pathways plays a crucial role as well. NDs vary from multiple sclerosis, in which the autoimmune inflammatory component is predominant, to more "classical" NDs, such as Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, and spinal muscular atrophy. Of interest, many of the clinical differences reported in NDs seem to be closely linked to sex, which may be justified by the significant changes in immune mechanisms between affected females and males. In this review, we examined some of the most studied NDs by looking at their pathogenic and phenotypical features to highlight sex-related discrepancies, if any, with particular interest in the individuals' responses to treatment. We believe that pointing out these differences in clinical practice may help achieve more successful precision and personalized care.
Collapse
Affiliation(s)
| | | | - Maria Liguori
- National Research Council (CNR), Institute of Biomedical Technologies, Bari Unit, 70125 Bari, Italy
| |
Collapse
|
25
|
Karahan H, Smith DC, Kim B, McCord B, Mantor J, John SK, Al-Amin MM, Dabin LC, Kim J. The effect of Abi3 locus deletion on the progression of Alzheimer's disease-related pathologies. Front Immunol 2023; 14:1102530. [PMID: 36895556 PMCID: PMC9988916 DOI: 10.3389/fimmu.2023.1102530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 02/06/2023] [Indexed: 02/23/2023] Open
Abstract
Human genetics studies of Alzheimer's disease (AD) have identified the ABI3 gene as a candidate risk gene for AD. Because ABI3 is highly expressed in microglia, the brain's immune cells, it was suggested that ABI3 might impact AD pathogenesis by regulating the immune response. Recent studies suggest that microglia have multifaceted roles in AD. Their immune response and phagocytosis functions can have beneficial effects in the early stages of AD by clearing up amyloid-beta (Aβ) plaques. However, they can be harmful at later stages due to their continuous inflammatory response. Therefore, it is important to understand the role of genes in microglia functions and their impact on AD pathologies along the progression of the disease. To determine the role of ABI3 at the early stage of amyloid pathology, we crossed Abi3 knock-out mice with the 5XFAD Aβ-amyloidosis mouse model and aged them until 4.5-month-old. Here, we demonstrate that deletion of the Abi3 locus increased Aβ plaque deposition, while there was no significant change in microgliosis and astrogliosis. Transcriptomic analysis indicates alterations in the expression of immune genes, such as Tyrobp, Fcer1g, and C1qa. In addition to the transcriptomic changes, we found elevated cytokine protein levels in Abi3 knock-out mouse brains, strengthening the role of ABI3 in neuroinflammation. These findings suggest that loss of ABI3 function may exacerbate AD progression by increasing Aβ accumulation and inflammation starting from earlier stages of the pathology.
Collapse
Affiliation(s)
- 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
| | - Daniel C. Smith
- 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
| | - Byungwook Kim
- 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
| | - Jordan Mantor
- 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
| | - Sutha K. John
- 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
| | - Md 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
| | - Luke C. Dabin
- 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
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
26
|
CD36 neutralisation blunts TLR2-IRF7 but not IRF3 pathway in neonatal mouse brain and immature human microglia following innate immune challenge. Sci Rep 2023; 13:2304. [PMID: 36759676 PMCID: PMC9911392 DOI: 10.1038/s41598-023-29423-0] [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: 07/19/2022] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Innate immune response in neonatal brain is associated with a robust microglial activation and induction of Toll-like Receptors (TLRs). To date, the role of the scavenger receptor CD36 in TLRs modulation, particularly TLR2 signaling, has been well established in adult brain. However, the crosstalk between TLR4, TLR2 and CD36 and its immunogenic influence in the neonatal brain remains unclear. In this study, using a CD36 blocking antibody (anti-CD36) at post-natal day 8, we evaluated the response of neonates to systemic endotoxin (lipopolysaccharide; LPS) challenge. We visualized the TLR2 response by bioluminescence imaging using the transgenic mouse model bearing the dual reporter system luciferase/green fluorescent protein under transcriptional control of a murine TLR2 promoter. The anti-CD36 treatment modified the LPS induced inflammatory profile in neonatal brains, causing a significant decrease in inflammatory cytokine levels and the TLR2 and TLR3 mediated signalling.The interferon regulatory factor 3 (IRF3) pathway remained unaffected. Treatment of the LPS-challenged human immature microglia with anti-CD36 induced a marked decrease in TLR2/TLR3 expression levels while TLR4 and IRF3 expression was not affected, suggesting the shared CD36 regulatory mechanisms in human and mouse microglia. Collectively, our results indicate that blocking CD36 alters LPS-induced inflammatory profile of mouse and human microglia, suggesting its role in fine-tuning of neuroinflammation.
Collapse
|
27
|
Vogrinc D, Gregorič Kramberger M, Emeršič A, Čučnik S, Goričar K, Dolžan V. Genetic Polymorphisms in Oxidative Stress and Inflammatory Pathways as Potential Biomarkers in Alzheimer's Disease and Dementia. Antioxidants (Basel) 2023; 12:antiox12020316. [PMID: 36829875 PMCID: PMC9952323 DOI: 10.3390/antiox12020316] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/12/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Oxidative stress and neuroinflammation are important processes involved in Alzheimer's disease (AD) and mild cognitive impairment (MCI). Numerous risk factors, including genetic background, can affect the complex interplay between those mechanisms in the aging brain and can also affect typical AD hallmarks: amyloid plaques and neurofibrillary tangles. Our aim was to evaluate the association of polymorphisms in oxidative stress- and inflammation-related genes with cerebrospinal fluid (CSF) biomarker levels and cognitive test results. The study included 54 AD patients, 14 MCI patients with pathological CSF biomarker levels, 20 MCI patients with normal CSF biomarker levels and 62 controls. Carriers of two polymorphic IL1B rs16944 alleles had higher CSF Aβ1-42 levels (p = 0.025), while carriers of at least one polymorphic NFE2L2 rs35652124 allele had lower CSF Aβ1-42 levels (p = 0.040). Association with IL1B rs16944 remained significant in the AD group (p = 0.029). Additionally, MIR146A rs2910164 was associated with Aβ42/40 ratio (p = 0.043) in AD. Significant associations with cognitive test scores were observed for CAT rs1001179 (p = 0.022), GSTP1 rs1138272 (p = 0.005), KEAP1 rs1048290 and rs9676881 (both p = 0.019), as well as NFE2L2 rs35652124 (p = 0.030). In the AD group, IL1B rs1071676 (p = 0.004), KEAP1 rs1048290 and rs9676881 (both p = 0.035) remained associated with cognitive scores. Polymorphisms in antioxidative and inflammation genes might be associated with CSF biomarkers and cognitive test scores and could serve as additional biomarkers contributing to early diagnosis of dementia.
Collapse
Affiliation(s)
- David Vogrinc
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Milica Gregorič Kramberger
- Department of Neurology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Andreja Emeršič
- Department of Neurology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Saša Čučnik
- Department of Neurology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Rheumatology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Katja Goričar
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Vita Dolžan
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Correspondence:
| |
Collapse
|
28
|
Zalewska T, Pawelec P, Ziabska K, Ziemka-Nalecz M. Sexual Dimorphism in Neurodegenerative Diseases and in Brain Ischemia. Biomolecules 2022; 13:26. [PMID: 36671411 PMCID: PMC9855831 DOI: 10.3390/biom13010026] [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: 11/22/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
Epidemiological studies and clinical observations show evidence of sexual dimorphism in brain responses to several neurological conditions. It is suggested that sex-related differences between men and women may have profound effects on disease susceptibility, pathophysiology, and progression. Sexual differences of the brain are achieved through the complex interplay of several factors contributing to this phenomenon, such as sex hormones, as well as genetic and epigenetic differences. Despite recent advances, the precise link between these factors and brain disorders is incompletely understood. This review aims to briefly outline the most relevant aspects that differ between men and women in ischemia and neurodegenerative disorders (AD, PD, HD, ALS, and SM). Recognition of disparities between both sexes could aid the development of individual approaches to ameliorate or slow the progression of intractable disorders.
Collapse
Affiliation(s)
- Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 A. Pawinskiego Str., 02-106 Warsaw, Poland
| | | | | | | |
Collapse
|
29
|
Immunosenescence and Aging: Neuroinflammation Is a Prominent Feature of Alzheimer's Disease and Is a Likely Contributor to Neurodegenerative Disease Pathogenesis. J Pers Med 2022; 12:jpm12111817. [PMID: 36579548 PMCID: PMC9698256 DOI: 10.3390/jpm12111817] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/25/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Alzheimer's disease (AD) is a chronic multifactorial and complex neuro-degenerative disorder characterized by memory impairment and the loss of cognitive ability, which is a problem affecting the elderly. The pathological intracellular accumulation of abnormally phosphorylated Tau proteins, forming neurofibrillary tangles, and extracellular amyloid-beta (Aβ) deposition, forming senile plaques, as well as neural disconnection, neural death and synaptic dysfunction in the brain, are hallmark pathologies that characterize AD. The prevalence of the disease continues to increase globally due to the increase in longevity, quality of life, and medical treatment for chronic diseases that decreases the mortality and enhance the survival of elderly. Medical awareness and the accurate diagnosis of the disease also contribute to the high prevalence observed globally. Unfortunately, no definitive treatment exists that can be used to modify the course of AD, and no available treatment is capable of mitigating the cognitive decline or reversing the pathology of the disease as of yet. A plethora of hypotheses, ranging from the cholinergic theory and dominant Aβ cascade hypothesis to the abnormally excessive phosphorylated Tau protein hypothesis, have been reported. Various explanations for the pathogenesis of AD, such as the abnormal excitation of the glutamate system and mitochondrial dysfunction, have also been suggested. Despite the continuous efforts to deliver significant benefits and an effective treatment for this distressing, globally attested aging illness, multipronged approaches and strategies for ameliorating the disease course based on knowledge of the underpinnings of the pathogenesis of AD are urgently needed. Immunosenescence is an immune deficit process that appears with age (inflammaging process) and encompasses the remodeling of the lymphoid organs, leading to alterations in the immune function and neuroinflammation during advanced aging, which is closely linked to the outgrowth of infections, autoimmune diseases, and malignant cancers. It is well known that long-standing inflammation negatively influences the brain over the course of a lifetime due to the senescence of the immune system. Herein, we aim to trace the role of the immune system in the pathogenesis of AD. Thus, we explore alternative avenues, such as neuroimmune involvement in the pathogenesis of AD. We determine the initial triggers of neuroinflammation, which is an early episode in the pre-symptomatic stages of AD and contributes to the advancement of the disease, and the underlying key mechanisms of brain damage that might aid in the development of therapeutic strategies that can be used to combat this devastating disease. In addition, we aim to outline the ways in which different aspects of the immune system, both in the brain and peripherally, behave and thus to contribute to AD.
Collapse
|
30
|
Blood Analytes as Biomarkers of Mechanisms Involved in Alzheimer’s Disease Progression. Int J Mol Sci 2022; 23:ijms232113289. [DOI: 10.3390/ijms232113289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/25/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Alzheimer’s disease (AD) is the leading cause of dementia, but the pathogenetic factors are not yet well known, and the relationships between brain and systemic biochemical derangements and disease onset and progression are unclear. We aim to focus on blood biomarkers for an accurate prognosis of the disease. We used a dataset characterized by longitudinal findings collected over the past 10 years from 90 AD patients. The dataset included 277 observations (both clinical and biochemical ones, encompassing blood analytes encompassing routine profiles for different organs, together with immunoinflammatory and oxidative markers). Subjects were grouped into four severity classes according to the Clinical Dementia Rating (CDR) Scale: mild (CDR = 0.5 and CDR = 1), moderate (CDR = 2), severe (CDR = 3) and very severe (CDR = 4 and CDR = 5). Statistical models were used for the identification of potential blood markers of AD progression. Moreover, we employed the Pathfinder tool of the Reactome database to investigate the biological pathways in which the analytes of interest could be involved. Statistical results reveal an inverse significant relation between four analytes (high-density cholesterol, total cholesterol, iron and ferritin) with AD severity. In addition, the Reactome database suggests that such analytes could be involved in pathways that are altered in AD progression. Indeed, the identified blood markers include molecules that reflect the heterogeneous pathogenetic mechanisms of AD. The combination of such blood analytes might be an early indicator of AD progression and constitute useful therapeutic targets.
Collapse
|
31
|
In Vitro and In Vivo Neuroprotective Effects of Sarcosine. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5467498. [PMID: 36281465 PMCID: PMC9587910 DOI: 10.1155/2022/5467498] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/31/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by behavioral and psychological symptoms in addition to cognitive impairment and loss of memory. The exact pathogenesis and genetic background of AD are unclear and there remains no effective treatment option. Sarcosine, an n-methyl derivative of glycine, showed a promising therapeutic strategy for some cognitive disorders. To our knowledge, the impacts of sarcosine supplementation against AD have not yet been elucidated. Therefore, we aimed to determine the neuroprotective potential of sarcosine in in vitro and in vivo AD model. In vitro studies have demonstrated that sarcosine increased the percentage of viable cells against aluminum induced neurotoxicity. In AlCl3-induced rat model of AD, the level of antioxidant capacity was significantly decreased and expression levels of APP, BACE1, TNF-α, APH1A, and PSENEN genes were elevated compared to the control group. Additionally, histopathological examinations of the hippocampus of AlCl3-induced rat brains showed the presence of neurofibrillary tangles (NFTs). However, the administration of sarcosine produced marked improvement and protection of AD-associated pathologies induced by AlCl3 in experimental rats. Therefore, this investigation may contribute to design novel therapeutic strategies using sarcosine for the management of AD pathologies.
Collapse
|
32
|
Xia Y, Chen C, Chen J, Hu C, Yang W, Wang L, Liu L, Gao LP, Wu YZ, Chen DD, Shi Q, Chen ZB, Dong XP. Enhanced M-CSF/CSF1R Signaling Closely Associates with PrP Sc Accumulation in the Scrapie-Infected Cell Line and the Brains of Scrapie-Infected Experimental Rodents. Mol Neurobiol 2022; 59:6534-6551. [PMID: 35970974 DOI: 10.1007/s12035-022-02989-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 08/06/2022] [Indexed: 12/17/2022]
Abstract
Activation and proliferation of microglia are one of the hallmarks of prion disease and is usually accompanied by increased levels of various cytokines and chemokines. Our previous study demonstrated that the level of brain macrophage colony-stimulating factor (M-CSF) was abnormally elevated during prion infection, but its association with PrPSc is not completely clear. In this study, colocalization of the increased M-CSF with accumulated PrPSc was observed by IHC with serial brain sections. Reliable molecular interaction between total PrP and M-CSF was observed in the brain of 263 K-infected hamsters and in cultured prion-infected cell line. Immunofluorescent assays showed that morphological colocalization of M-CSF with neurons and microglia, but not with astrocytes in brains of scrapie-infected animals. The transcriptional and expressing levels of CSF1R were also significantly increased in prion-infected cell line and mice, and colocalization of CSF1R with neurons and microglia was observed in the brains of prion-infected mouse models. Removal of PrPSc replication by resveratrol in SMB-S15 cells induced limited reductions of cellular levels of M-CSF and CSF1R. In addition, we found that the level of IL-34, another ligand of CSF1R, did not change significantly after prion infection, but its distribution on the cell types in the brains shifted from neurons in healthy mice to the proliferated astrocytes and microglia in scrapie-infected mice. Our data demonstrate activation of M-CSF/IL-34/CSF1R signaling in the microenvironment of prion infection, strongly indicating its vital role in the pathophysiology of prions. It provides solid scientific evidence for the therapeutic potential of inhibiting M-CSF/CSF1R signaling in prion diseases.
Collapse
Affiliation(s)
- Ying Xia
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Cao Chen
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China.
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.
| | - Jia Chen
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Chao Hu
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Wei Yang
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Lin Wang
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Lian Liu
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Li-Ping Gao
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Yue-Zhang Wu
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Dong-Dong Chen
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Qi Shi
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhi-Bao Chen
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China.
| | - Xiao-Ping Dong
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China.
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China.
- Shanghai Institute of Infectious Disease and Biosafety, Shanghai, China.
| |
Collapse
|
33
|
Kosyreva AM, Sentyabreva AV, Tsvetkov IS, Makarova OV. Alzheimer’s Disease and Inflammaging. Brain Sci 2022; 12:brainsci12091237. [PMID: 36138973 PMCID: PMC9496782 DOI: 10.3390/brainsci12091237] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/22/2022] [Accepted: 09/10/2022] [Indexed: 11/23/2022] Open
Abstract
Alzheimer’s disease is one of the most common age-related neurodegenerative disorders. The main theory of Alzheimer’s disease progress is the amyloid-β cascade hypothesis. However, the initial mechanisms of insoluble forms of amyloid-β formation and hyperphosphorylated tau protein in neurons remain unclear. One of the factors, which might play a key role in senile plaques and tau fibrils generation due to Alzheimer’s disease, is inflammaging, i.e., systemic chronic low-grade age-related inflammation. The activation of the proinflammatory cell phenotype is observed during aging, which might be one of the pivotal mechanisms for the development of chronic inflammatory diseases, e.g., atherosclerosis, metabolic syndrome, type 2 diabetes mellitus, and Alzheimer’s disease. This review discusses the role of the inflammatory processes in developing neurodegeneration, activated during physiological aging and due to various diseases such as atherosclerosis, obesity, type 2 diabetes mellitus, and depressive disorders.
Collapse
|
34
|
Asadi MR, Talebi M, Gharesouran J, Sabaie H, Jalaiei A, Arsang-Jang S, Taheri M, Sayad A, Rezazadeh M. Analysis of ROQUIN, Tristetraprolin (TTP), and BDNF/miR-16/TTP regulatory axis in late onset Alzheimer’s disease. Front Aging Neurosci 2022; 14:933019. [PMID: 36016853 PMCID: PMC9397504 DOI: 10.3389/fnagi.2022.933019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/14/2022] [Indexed: 12/25/2022] Open
Abstract
Alzheimer’s disease (AD) is a heterogeneous degenerative disorder of the brain that is on the rise worldwide. One of the critical processes that might be disturbed in AD is gene expression regulation. Tristetraprolin (TTP) and RC3H1 gene (ROQUIN) are two RNA-binding proteins (RBPs) that target AU-rich elements (AREs) and constitutive decay elements (CDEs), respectively. TTP and ROQUIN, members of the CCCH zinc-finger protein family, have been demonstrated to fine-tune numerous inflammatory factors. In addition, miR-16 has distinct characteristics and may influence the target mRNA through the ARE site. Interestingly, BDNF mRNA has ARE sites in the 3’ untranslated region (UTR) and can be targeted by regulatory factors, such as TTP and miR-16 on MRE sequences, forming BDNF/miR-16/TTP regulatory axis. A number of two microarray datasets were downloaded, including information on mRNAs (GSE106241) and miRNAs (GSE157239) from individuals with AD and corresponding controls. R software was used to identify BDNF, TTP, ROQUIN, and miR-16 expression levels in temporal cortex (TC) tissue datasets. Q-PCR was also used to evaluate the expression of these regulatory factors and the expression of BDNF in the blood of 50 patients with AD and 50 controls. Bioinformatic evaluation showed that TTP and miR-16 overexpression might act as post-transcriptional regulatory factors to control BDNF expression in AD in TC samples. Instead, this expression pattern was not found in peripheral blood samples from patients with AD compared to normal controls. ROQUIN expression was increased in the peripheral blood of patients with AD. Hsa-miR-16-5p levels did not show significant differences in peripheral blood samples. Finally, it was shown that TTP and BDNF, based on evaluating the receiver operating characteristic (ROC), effectively identify patients with AD from healthy controls. This study could provide a new perspective on the molecular regulatory processes associated with AD pathogenic mechanisms linked to the BDNF growth factor, although further research is needed on the possible roles of these factors in AD.
Collapse
Affiliation(s)
- Mohammad Reza Asadi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahnaz Talebi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Gharesouran
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hani Sabaie
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Jalaiei
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahram Arsang-Jang
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Arezou Sayad
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Arezou Sayad,
| | - Maryam Rezazadeh
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
- Maryam Rezazadeh,
| |
Collapse
|
35
|
Guo Y, Chomiak A, Hong Y, Lowe CC, Kopsidas CA, Chan WC, Andrade J, Pan H, Zhou X, Monuki ES, Feng Y. Histone H2A ubiquitination resulting from Brap loss of function connects multiple aging hallmarks and accelerates neurodegeneration. iScience 2022; 25:104519. [PMID: 35754718 PMCID: PMC9213774 DOI: 10.1016/j.isci.2022.104519] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 04/06/2022] [Accepted: 05/29/2022] [Indexed: 01/10/2023] Open
Abstract
Aging is an intricate process characterized by multiple hallmarks including stem cell exhaustion, genome instability, epigenome alteration, impaired proteostasis, and cellular senescence. Whereas each of these traits is detrimental at the cellular level, it remains unclear how they are interconnected to cause systemic organ deterioration. Here we show that abrogating Brap, a BRCA1-associated protein essential for neurogenesis, results in persistent DNA double-strand breaks and elevation of histone H2A mono- and poly-ubiquitination (H2Aub). These defects extend to cellular senescence and proteasome-mediated histone H2A proteolysis with alterations in cells' proteomic and epigenetic states. Brap deletion in the mouse brain causes neuroinflammation, impaired proteostasis, accelerated neurodegeneration, and substantially shortened the lifespan. We further show the elevation of H2Aub also occurs in human brain tissues with Alzheimer's disease. These data together suggest that chromatin aberrations mediated by H2Aub may act as a nexus of multiple aging hallmarks and promote tissue-wide degeneration.
Collapse
Affiliation(s)
- Yan Guo
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E. Superior Street, Chicago, IL 60611, USA
| | - Alison.A. Chomiak
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E. Superior Street, Chicago, IL 60611, USA
| | - Ye Hong
- University of Turku, Turku 20500, Finland
| | - Clara C. Lowe
- Department of Biochemistry and Molecular Biology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Caroline A. Kopsidas
- Department of Biochemistry and Molecular Biology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Wen-Ching Chan
- Center for Research Informatics, University of Chicago, Chicago, IL 60637, USA
| | - Jorge Andrade
- Center for Research Informatics, University of Chicago, Chicago, IL 60637, USA
| | - Hongna Pan
- Department of Biochemistry and Molecular Biology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Xiaoming Zhou
- Department of Medicine, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Edwin S. Monuki
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA 92697, USA
| | - Yuanyi Feng
- Department of Biochemistry and Molecular Biology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| |
Collapse
|
36
|
Pyun J, Park YH, Hodges A, Jang J, Bice PJ, Kim S, Saykin AJ, Nho K. Immunity gene IFITM3 variant: Relation to cognition and Alzheimer's disease pathology. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12317. [PMID: 35769874 PMCID: PMC9212215 DOI: 10.1002/dad2.12317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/31/2022] [Accepted: 04/02/2022] [Indexed: 01/29/2023]
Abstract
Introduction We investigated single-nucleotide polymorphisms (SNPs) in IFITM3, an innate immunity gene and modulator of amyloid beta in Alzheimer's disease (AD), for association with cognition and AD biomarkers. Methods We used data from the Alzheimer's Disease Neuroimaging Initiative (ADNI; N = 1565) and AddNeuroMed (N = 633) as discovery and replication samples, respectively. We performed gene-based association analysis of SNPs in IFITM3 with cognitive performance and SNP-based association analysis with cognitive decline and amyloid, tau, and neurodegeneration biomarkers for AD. Results Gene-based association analysis showed that IFITM3 was significantly associated with cognitive performance. Particularly, rs10751647 in IFITM3 was associated with less cognitive decline, less amyloid and tau burden, and less brain atrophy in ADNI. The association of rs10751647 with cognitive decline and brain atrophy was replicated in AddNeuroMed. Discussion This suggests that rs10751647 in IFITM3 is associated with less vulnerability for cognitive decline and AD biomarkers, providing mechanistic insight regarding involvement of immunity and infection in AD. Highlights IFITM3 is significantly associated with cognitive performance.rs10751647 in IFITM3 is associated with cognitive decline rates with replication.rs10751647 is associated with amyloid beta load, cerebrospinal fluid phosphorylated tau levels, and brain atrophy.rs10751647 is associated with IFITM3 expression levels in blood and brain.rs10751647 in IFITM3 is related to less vulnerability to Alzheimer's disease pathogenesis.
Collapse
Affiliation(s)
- Jung‐Min Pyun
- Department of NeurologySeoul National University Bundang Hospital and Seoul National University College of MedicineSeongnamRepublic of Korea
- Department of NeurologySoonchunhyang University Seoul HospitalSoonchunhyang University College of MedicineSeoulRepublic of Korea
| | - Young Ho Park
- Department of NeurologySeoul National University Bundang Hospital and Seoul National University College of MedicineSeongnamRepublic of Korea
| | - Angela Hodges
- Institute of PsychiatryPsychology & NeuroscienceKing's College LondonLondonUK
| | - Jae‐Won Jang
- Department of NeurologyKangwon National University HospitalChuncheonRepublic of Korea
| | - Paula J. Bice
- Department of Radiology and Imaging Sciences, and the Indiana Alzheimer Disease CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - SangYun Kim
- Department of NeurologySeoul National University Bundang Hospital and Seoul National University College of MedicineSeongnamRepublic of Korea
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences, and the Indiana Alzheimer Disease CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, and the Indiana Alzheimer Disease CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for Computational Biology and BioinformaticsIndiana University School of MedicineIndianapolisIndianaUSA
| | | |
Collapse
|
37
|
Taylor MK, Sullivan DK, Keller JE, Burns JM, Swerdlow RH. Potential for Ketotherapies as Amyloid-Regulating Treatment in Individuals at Risk for Alzheimer’s Disease. Front Neurosci 2022; 16:899612. [PMID: 35784855 PMCID: PMC9243383 DOI: 10.3389/fnins.2022.899612] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/30/2022] [Indexed: 12/27/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative condition characterized by clinical decline in memory and other cognitive functions. A classic AD neuropathological hallmark includes the accumulation of amyloid-β (Aβ) plaques, which may precede onset of clinical symptoms by over a decade. Efforts to prevent or treat AD frequently emphasize decreasing Aβ through various mechanisms, but such approaches have yet to establish compelling interventions. It is still not understood exactly why Aβ accumulates in AD, but it is hypothesized that Aβ and other downstream pathological events are a result of impaired bioenergetics, which can also manifest prior to cognitive decline. Evidence suggests that individuals with AD and at high risk for AD have functional brain ketone metabolism and ketotherapies (KTs), dietary approaches that produce ketone bodies for energy metabolism, may affect AD pathology by targeting impaired brain bioenergetics. Cognitively normal individuals with elevated brain Aβ, deemed “preclinical AD,” and older adults with peripheral metabolic impairments are ideal candidates to test whether KTs modulate AD biology as they have impaired mitochondrial function, perturbed brain glucose metabolism, and elevated risk for rapid Aβ accumulation and symptomatic AD. Here, we discuss the link between brain bioenergetics and Aβ, as well as the potential for KTs to influence AD risk and progression.
Collapse
Affiliation(s)
- Matthew K. Taylor
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States
- *Correspondence: Matthew K. Taylor,
| | - Debra K. Sullivan
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States
| | - Jessica E. Keller
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States
| | - Jeffrey M. Burns
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Russell H. Swerdlow
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
38
|
Xie J, Van Hoecke L, Vandenbroucke RE. The Impact of Systemic Inflammation on Alzheimer's Disease Pathology. Front Immunol 2022; 12:796867. [PMID: 35069578 PMCID: PMC8770958 DOI: 10.3389/fimmu.2021.796867] [Citation(s) in RCA: 142] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating age-related neurodegenerative disorder with an alarming increasing prevalence. Except for the recently FDA-approved Aducanumab of which the therapeutic effect is not yet conclusively proven, only symptomatic medication that is effective for some AD patients is available. In order to be able to design more rational and effective treatments, our understanding of the mechanisms behind the pathogenesis and progression of AD urgently needs to be improved. Over the last years, it became increasingly clear that peripheral inflammation is one of the detrimental factors that can contribute to the disease. Here, we discuss the current understanding of how systemic and intestinal (referred to as the gut-brain axis) inflammatory processes may affect brain pathology, with a specific focus on AD. Moreover, we give a comprehensive overview of the different preclinical as well as clinical studies that link peripheral Inflammation to AD initiation and progression. Altogether, this review broadens our understanding of the mechanisms behind AD pathology and may help in the rational design of further research aiming to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Junhua Xie
- VIB Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Lien Van Hoecke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
39
|
Chen JJ, Thiyagarajah M, Song J, Chen C, Herrmann N, Gallagher D, Rapoport MJ, Black SE, Ramirez J, Andreazza AC, Oh P, Marzolini S, Graham SJ, Lanctôt KL. Altered central and blood glutathione in Alzheimer's disease and mild cognitive impairment: a meta-analysis. Alzheimers Res Ther 2022; 14:23. [PMID: 35123548 PMCID: PMC8818133 DOI: 10.1186/s13195-022-00961-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 01/06/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND Increasing evidence implicates oxidative stress (OS) in Alzheimer disease (AD) and mild cognitive impairment (MCI). Depletion of the brain antioxidant glutathione (GSH) may be important in OS-mediated neurodegeneration, though studies of post-mortem brain GSH changes in AD have been inconclusive. Recent in vivo measurements of the brain and blood GSH may shed light on GSH changes earlier in the disease. AIM To quantitatively review in vivo GSH in AD and MCI compared to healthy controls (HC) using meta-analyses. METHOD Studies with in vivo brain or blood GSH levels in MCI or AD with a HC group were identified using MEDLINE, PsychInfo, and Embase (1947-June 2020). Standardized mean differences (SMD) and 95% confidence intervals (CI) were calculated for outcomes using random effects models. Outcome measures included brain GSH (Meshcher-Garwood Point Resolved Spectroscopy (MEGA-PRESS) versus non-MEGA-PRESS) and blood GSH (intracellular versus extracellular) in AD and MCI. The Q statistic and Egger's test were used to assess heterogeneity and risk of publication bias, respectively. RESULTS For brain GSH, 4 AD (AD=135, HC=223) and 4 MCI (MCI=213, HC=211) studies were included. For blood GSH, 26 AD (AD=1203, HC=1135) and 7 MCI (MCI=434, HC=408) studies were included. Brain GSH overall did not differ in AD or MCI compared to HC; however, the subgroup of studies using MEGA-PRESS reported lower brain GSH in AD (SMD [95%CI] -1.45 [-1.83, -1.06], p<0.001) and MCI (-1.15 [-1.71, -0.59], z=4.0, p<0.001). AD had lower intracellular and extracellular blood GSH overall (-0.87 [-1. 30, -0.44], z=3.96, p<0.001). In a subgroup analysis, intracellular GSH was lower in MCI (-0.66 [-1.11, -0.21], p=0.025). Heterogeneity was observed throughout (I2 >85%) and not fully accounted by subgroup analysis. Egger's test indicated risk of publication bias. CONCLUSION Blood intracellular GSH decrease is seen in MCI, while both intra- and extracellular decreases were seen in AD. Brain GSH is decreased in AD and MCI in subgroup analysis. Potential bias and heterogeneity suggest the need for measurement standardization and additional studies to explore sources of heterogeneity.
Collapse
Affiliation(s)
- Jinghan Jenny Chen
- Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Room FG52, Toronto, ON, M4N 3M5, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Mathura Thiyagarajah
- Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Room FG52, Toronto, ON, M4N 3M5, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Jianmeng Song
- Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Room FG52, Toronto, ON, M4N 3M5, Canada
| | - Clara Chen
- Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Room FG52, Toronto, ON, M4N 3M5, Canada
| | - Nathan Herrmann
- Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Room FG52, Toronto, ON, M4N 3M5, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Geriatric Psychiatry, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Damien Gallagher
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Geriatric Psychiatry, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Mark J Rapoport
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Geriatric Psychiatry, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Sandra E Black
- Evaluative Clinical Sciences, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
- KITE-Toronto Rehabilitation Institute, University Health Network, Toronto, ON, Canada
| | - Joel Ramirez
- Evaluative Clinical Sciences, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Ana C Andreazza
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Paul Oh
- KITE-Toronto Rehabilitation Institute, University Health Network, Toronto, ON, Canada
| | - Susan Marzolini
- KITE-Toronto Rehabilitation Institute, University Health Network, Toronto, ON, Canada
| | - Simon J Graham
- Physical Sciences, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Krista L Lanctôt
- Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Room FG52, Toronto, ON, M4N 3M5, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
- Geriatric Psychiatry, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.
- Evaluative Clinical Sciences, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada.
| |
Collapse
|
40
|
Decourt B, D’Souza GX, Shi J, Ritter A, Suazo J, Sabbagh MN. The Cause of Alzheimer's Disease: The Theory of Multipathology Convergence to Chronic Neuronal Stress. Aging Dis 2022; 13:37-60. [PMID: 35111361 PMCID: PMC8782548 DOI: 10.14336/ad.2021.0529] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 05/28/2021] [Indexed: 12/18/2022] Open
Abstract
The field of Alzheimer's disease (AD) research critically lacks an all-inclusive etiology theory that would integrate existing hypotheses and explain the heterogeneity of disease trajectory and pathologies observed in each individual patient. Here, we propose a novel comprehensive theory that we named: the multipathology convergence to chronic neuronal stress. Our new theory reconsiders long-standing dogmas advanced by previous incomplete theories. Firstly, while it is undeniable that amyloid beta (Aβ) is involved in AD, in the seminal stage of the disease Aβ is unlikely pathogenic. Instead, we hypothesize that the root cause of AD is neuronal stress in the central nervous system (CNS), and Aβ is expressed as part of the physiological response to protect CNS neurons from stress. If there is no return to homeostasis, then Aβ becomes overexpressed, and this includes the generation of longer forms that are more toxic and prone to oligomerization. Secondly, AD etiology is plausibly not strictly compartmentalized within the CNS but may also result from the dysfunction of other physiological systems in the entire body. This view implies that AD may not have a single cause, but rather needs to be considered as a spectrum of multiple chronic pathological modalities converging to the persistent stressing of CNS neurons. These chronic pathological modalities, which include cardiovascular disease, metabolic disorders, and CNS structural changes, often start individually, and over time combine with other chronic modalities to incrementally escalate the amount of stress applied to CNS neurons. We present the case for considering Aβ as a marker of neuronal stress in response to hypoxic, toxic, and starvation events, rather than solely a marker of AD. We also detail numerous human chronic conditions that can lead to neuronal stress in the CNS, making the link with co-morbidities encountered in daily clinical AD practice. Finally, we explain how our theory could be leveraged to improve clinical care for AD and related dementia in personalized medicine paradigms in the near future.
Collapse
Affiliation(s)
- Boris Decourt
- Translational Neurodegenerative Research Laboratory, Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV 89106, USA.
| | - Gary X D’Souza
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA.
| | - Jiong Shi
- Translational Neurodegenerative Research Laboratory, Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV 89106, USA.
- Cleveland Clinic Nevada and Lou Ruvo Center for Brain Health, Las Vegas, NV 89106, USA.
| | - Aaron Ritter
- Cleveland Clinic Nevada and Lou Ruvo Center for Brain Health, Las Vegas, NV 89106, USA.
| | - Jasmin Suazo
- Translational Neurodegenerative Research Laboratory, Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV 89106, USA.
| | - Marwan N Sabbagh
- Translational Neurodegenerative Research Laboratory, Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV 89106, USA.
- Cleveland Clinic Nevada and Lou Ruvo Center for Brain Health, Las Vegas, NV 89106, USA.
| |
Collapse
|
41
|
Kendall LV, Boyd TD, Sillau SH, Bosco-Lauth A, Markham N, Fong D, Clarke P, Tyler KL, Potter H. GM-CSF Promotes Immune Response and Survival in a Mouse Model of COVID-19. RESEARCH SQUARE 2022:rs.3.rs-1213395. [PMID: 35118463 PMCID: PMC8811947 DOI: 10.21203/rs.3.rs-1213395/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
COVID-19 results in increased expression of inflammatory cytokines, but inflammation-targeting clinical trials have yielded poor to mixed results. Our studies of other disorders with an inflammatory component, including Alzheimer's disease, chemobrain, Down syndrome, normal aging, and West Nile Virus infection, showed that treatment with the 'pro-inflammatory' cytokine granulocyte-macrophage colony stimulating factor (GM-CSF) in humans or mouse models alleviated clinical, behavioral, and pathological features. We proposed that human recombinant GM-CSF (sargramostim) be repurposed to promote both the innate and adaptive immune responses in COVID-19 to reduce viral load and mortality1. Here, we report the results of a placebo-controlled study of GM-CSF in human ACE2 transgenic mice inoculated intranasally with SARS-CoV2 virus, a model of COVID-19. Infection resulted in high viral titers in lungs and brains and over 85% mortality. GM-CSF treatment beginning one day after infection increased anti-viral antibody titers, lowered mean lung viral titers proportionately (p=0.0020) and increased the odds of long-term survival by up to 5.8-fold (p=0.0358), compared to placebo. These findings suggest that, as an activator of both the innate and adaptive immune systems, GM-CSF/sargramostim may be an effective COVID-19 therapy with the potential to protect from re-infection more effectively than treatment with antiviral drugs or monoclonal antibodies.
Collapse
Affiliation(s)
- L V Kendall
- Colorado State University, Department of Microbiology, Immunology and Pathology, Fort Collins, CO
| | - T D Boyd
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO
| | - S H Sillau
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO
| | - A Bosco-Lauth
- Colorado State University, Department of Biomedical Sciences, Fort Collins, CO
| | - N Markham
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO
| | - D Fong
- Department of Pathology, University of Colorado Anschutz School of Medicine, Aurora, CO
| | - P Clarke
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO
| | - K L Tyler
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO
- Denver VA Medical Center, Denver CO
- Departments of Immunology and Microbiology, and Medicine, University of Colorado School of Medicine, Aurora, CO
| | - H Potter
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
42
|
Wang L, Davis PB, Volkow ND, Berger NA, Kaelber DC, Xu R. Association of COVID-19 with New-Onset Alzheimer's Disease. J Alzheimers Dis 2022; 89:411-414. [PMID: 35912749 PMCID: PMC10361652 DOI: 10.3233/jad-220717] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
An infectious etiology of Alzheimer's disease has been postulated for decades. It remains unknown whether SARS-CoV-2 viral infection is associated with increased risk for Alzheimer's disease. In this retrospective cohort study of 6,245,282 older adults (age ≥65 years) who had medical encounters between 2/2020-5/2021, we show that people with COVID-19 were at significantly increased risk for new diagnosis of Alzheimer's disease within 360 days after the initial COVID-19 diagnosis (hazard ratio or HR:1.69, 95% CI: 1.53-1.72), especially in people age ≥85 years and in women. Our findings call for research to understand the underlying mechanisms and for continuous surveillance of long-term impacts of COVID-19 on Alzheimer's disease.
Collapse
Affiliation(s)
- Lindsey Wang
- Center for Science, Health, and Society, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Pamela B. Davis
- Center for Community Health Integration, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Nora D. Volkow
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Nathan A. Berger
- Center for Science, Health, and Society, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - David C. Kaelber
- The Center for Clinical Informatics Research and Education, The MetroHealth System, Cleveland, OH, USA
| | - Rong Xu
- Center for Artificial Intelligence in Drug Discovery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
43
|
Jonas LA, Jain T, Li YM. Functional insight into LOAD-associated microglial response genes. Open Biol 2022; 12:210280. [PMID: 35078351 PMCID: PMC8790339 DOI: 10.1098/rsob.210280] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/01/2021] [Indexed: 01/07/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by the presence of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs), neuronal and synaptic loss and inflammation of the central nervous system (CNS). The majority of AD research has been dedicated to the understanding of two major AD hallmarks (i.e. Aβ and NFTs); however, recent genome-wide association studies (GWAS) data indicate neuroinflammation as having a critical role in late-onset AD (LOAD) development, thus unveiling a novel avenue for AD therapeutics. Recent evidence has provided much support to the innate immune system's involvement with AD progression; however, much remains to be uncovered regarding the role of glial cells, specifically microglia, in AD. Moreover, numerous variants in immune and/or microglia-related genes have been identified in whole-genome sequencing and GWAS analyses, including such genes as TREM2, CD33, APOE, API1, MS4A, ABCA7, BIN1, CLU, CR1, INPP5D, PICALM and PLCG2. In this review, we aim to provide an insight into the function of the major LOAD-associated microglia response genes.
Collapse
Affiliation(s)
- Lauren A. Jonas
- Weill Cornell, Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10065, USA
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tanya Jain
- Weill Cornell, Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10065, USA
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yue-Ming Li
- Weill Cornell, Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10065, USA
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
44
|
Potjewyd FM, Axtman AD. Exploration of Aberrant E3 Ligases Implicated in Alzheimer's Disease and Development of Chemical Tools to Modulate Their Function. Front Cell Neurosci 2021; 15:768655. [PMID: 34867205 PMCID: PMC8637409 DOI: 10.3389/fncel.2021.768655] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/28/2021] [Indexed: 11/24/2022] Open
Abstract
The Ubiquitin Proteasome System (UPS) is responsible for the degradation of misfolded or aggregated proteins via a multistep ATP-dependent proteolytic mechanism. This process involves a cascade of ubiquitin (Ub) transfer steps from E1 to E2 to E3 ligase. The E3 ligase transfers Ub to a targeted protein that is brought to the proteasome for degradation. The inability of the UPS to remove misfolded or aggregated proteins due to UPS dysfunction is commonly observed in neurodegenerative diseases, such as Alzheimer's disease (AD). UPS dysfunction in AD drives disease pathology and is associated with the common hallmarks such as amyloid-β (Aβ) accumulation and tau hyperphosphorylation, among others. E3 ligases are key members of the UPS machinery and dysfunction or changes in their expression can propagate other aberrant processes that accelerate AD pathology. The upregulation or downregulation of expression or activity of E3 ligases responsible for these processes results in changes in protein levels of E3 ligase substrates, many of which represent key proteins that propagate AD. A powerful way to better characterize UPS dysfunction in AD and the role of individual E3 ligases is via the use of high-quality chemical tools that bind and modulate specific E3 ligases. Furthermore, through combining gene editing with recent advances in 3D cell culture, in vitro modeling of AD in a dish has become more relevant and possible. These cell-based models of AD allow for study of specific pathways and mechanisms as well as characterization of the role E3 ligases play in driving AD. In this review, we outline the key mechanisms of UPS dysregulation linked to E3 ligases in AD and highlight the currently available chemical modulators. We present several key approaches for E3 ligase ligand discovery being employed with respect to distinct classes of E3 ligases. Where possible, specific examples of the use of cultured neurons to delineate E3 ligase biology have been captured. Finally, utilizing the available ligands for E3 ligases in the design of proteolysis targeting chimeras (PROTACs) to degrade aberrant proteins is a novel strategy for AD, and we explore the prospects of PROTACs as AD therapeutics.
Collapse
|
45
|
Velásquez E, Szeitz B, Gil J, Rodriguez J, Palkovits M, Renner É, Hortobágyi T, Döme P, Nogueira FC, Marko-Varga G, Domont GB, Rezeli M. Topological Dissection of Proteomic Changes Linked to the Limbic Stage of Alzheimer's Disease. Front Immunol 2021; 12:750665. [PMID: 34712240 PMCID: PMC8546208 DOI: 10.3389/fimmu.2021.750665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/24/2021] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder and the most common cause of dementia worldwide. In AD, neurodegeneration spreads throughout different areas of the central nervous system (CNS) in a gradual and predictable pattern, causing progressive memory decline and cognitive impairment. Deposition of neurofibrillary tangles (NFTs) in specific CNS regions correlates with the severity of AD and constitutes the basis for disease classification into different Braak stages (I-VI). Early clinical symptoms are typically associated with stages III-IV (i.e., limbic stages) when the involvement of the hippocampus begins. Histopathological changes in AD have been linked to brain proteome alterations, including aberrant posttranslational modifications (PTMs) such as the hyperphosphorylation of Tau. Most proteomic studies to date have focused on AD progression across different stages of the disease, by targeting one specific brain area at a time. However, in AD vulnerable regions, stage-specific proteomic alterations, including changes in PTM status occur in parallel and remain poorly characterized. Here, we conducted proteomic, phosphoproteomic, and acetylomic analyses of human postmortem tissue samples from AD (Braak stage III-IV, n=11) and control brains (n=12), covering all anatomical areas affected during the limbic stage of the disease (total hippocampus, CA1, entorhinal and perirhinal cortices). Overall, ~6000 proteins, ~9000 unique phosphopeptides and 221 acetylated peptides were accurately quantified across all tissues. Our results reveal significant proteome changes in AD brains compared to controls. Among others, we have observed the dysregulation of pathways related to the adaptive and innate immune responses, including several altered antimicrobial peptides (AMPs). Notably, some of these changes were restricted to specific anatomical areas, while others altered according to disease progression across the regions studied. Our data highlights the molecular heterogeneity of AD and the relevance of neuroinflammation as a major player in AD pathology. Data are available via ProteomeXchange with identifier PXD027173.
Collapse
Affiliation(s)
- Erika Velásquez
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, Malmö, Sweden
| | - Beáta Szeitz
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Jeovanis Gil
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, Malmö, Sweden.,Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Jimmy Rodriguez
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Miklós Palkovits
- Human Brain Tissue Bank, Semmelweis University, Budapest, Hungary
| | - Éva Renner
- Human Brain Tissue Bank, Semmelweis University, Budapest, Hungary
| | - Tibor Hortobágyi
- Institute of Pathology, Faculty of Medicine, University of Szeged, Szeged, Hungary.,ELKH-DE Cerebrovascular and Neurodegenerative Research Group, Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Döme
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary.,National Institute of Mental Health, Neurology and Neurosurgery, Budapest, Hungary
| | - Fábio Cs Nogueira
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratory of Proteomics, Laboratório de Apoio ao Desenvolvimento Tecnológico (LADETEC), Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - György Marko-Varga
- Division of Clinical Protein Science & Imaging, Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - Gilberto B Domont
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Melinda Rezeli
- Division of Clinical Protein Science & Imaging, Department of Biomedical Engineering, Lund University, Lund, Sweden
| |
Collapse
|
46
|
Paranjpe MD, Belonwu S, Wang JK, Oskotsky T, Gupta A, Taubes A, Zalocusky KA, Paranjpe I, Glicksberg BS, Huang Y, Sirota M. Sex-Specific Cross Tissue Meta-Analysis Identifies Immune Dysregulation in Women With Alzheimer's Disease. Front Aging Neurosci 2021; 13:735611. [PMID: 34658838 PMCID: PMC8515049 DOI: 10.3389/fnagi.2021.735611] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/01/2021] [Indexed: 01/09/2023] Open
Abstract
Background: Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia in the United States. In spite of evidence of females having a greater lifetime risk of developing Alzheimer's Disease (AD) and greater apolipoprotein E4-related (APOE ε4) AD risk compared to males, molecular signatures underlying these differences remain elusive. Methods: We took a meta-analysis approach to study gene expression in the brains of 1,084 AD patients and age-matched controls and whole blood from 645 AD patients and age-matched controls in seven independent datasets. Sex-specific gene expression patterns were investigated through use of gene-based, pathway-based and network-based approaches. The ability of a sex-specific AD gene expression signature to distinguish Alzheimer's disease from healthy controls was assessed using a linear support vector machine model. Cell type deconvolution from whole blood gene expression data was performed to identify differentially regulated cells in males and females with AD. Results: Strikingly gene-expression, network-based analysis and cell type deconvolution approaches revealed a consistent immune signature in the brain and blood of female AD patients that was absent in males. In females, network-based analysis revealed a coordinated program of gene expression involving several zinc finger nuclease genes related to Herpes simplex viral infection whose expression was modulated by the presence of the APOE ε4 allele. Interestingly, this gene expression program was missing in the brains of male AD patients. Cell type deconvolution identified an increase in neutrophils and naïve B cells and a decrease in M2 macrophages, memory B cells, and CD8+ T cells in AD samples compared to controls in females. Interestingly, among males with AD, no significant differences in immune cell proportions compared to controls were observed. Machine learning-based classification of AD using gene expression from whole blood in addition to clinical features produced an improvement in classification accuracy upon stratifying by sex, achieving an AUROC of 0.91 for females and 0.80 for males. Conclusion: These results help identify sex and APOE ε4 genotype-specific transcriptomic signatures of AD and underscore the importance of considering sex in the development of biomarkers and therapeutic strategies for AD.
Collapse
Affiliation(s)
- Manish D Paranjpe
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States.,Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, United States
| | - Stella Belonwu
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States.,Pharmaceutical Sciences and Pharmacogenomics Graduate Program, University of California, San Francisco, San Francisco, CA, United States
| | - Jason K Wang
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, United States
| | - Tomiko Oskotsky
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Aarzu Gupta
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Alice Taubes
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States.,The Gladstone Institute of Neurological Disease, San Francisco, CA, United States
| | - Kelly A Zalocusky
- The Gladstone Institute of Neurological Disease, San Francisco, CA, United States.,Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Ishan Paranjpe
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States.,Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Benjamin S Glicksberg
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Yadong Huang
- The Gladstone Institute of Neurological Disease, San Francisco, CA, United States.,Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
47
|
Xie J, Gorlé N, Vandendriessche C, Van Imschoot G, Van Wonterghem E, Van Cauwenberghe C, Parthoens E, Van Hamme E, Lippens S, Van Hoecke L, Vandenbroucke RE. Low-grade peripheral inflammation affects brain pathology in the App NL-G-Fmouse model of Alzheimer's disease. Acta Neuropathol Commun 2021; 9:163. [PMID: 34620254 PMCID: PMC8499584 DOI: 10.1186/s40478-021-01253-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/01/2021] [Indexed: 12/22/2022] Open
Abstract
Alzheimer’s disease (AD) is a chronic neurodegenerative disease characterized by the accumulation of amyloid β (Aβ) and neurofibrillary tangles. The last decade, it became increasingly clear that neuroinflammation plays a key role in both the initiation and progression of AD. Moreover, also the presence of peripheral inflammation has been extensively documented. However, it is still ambiguous whether this observed inflammation is cause or consequence of AD pathogenesis. Recently, this has been studied using amyloid precursor protein (APP) overexpression mouse models of AD. However, the findings might be confounded by APP-overexpression artifacts. Here, we investigated the effect of low-grade peripheral inflammation in the APP knock-in (AppNL-G-F) mouse model. This revealed that low-grade peripheral inflammation affects (1) microglia characteristics, (2) blood-cerebrospinal fluid barrier integrity, (3) peripheral immune cell infiltration and (4) Aβ deposition in the brain. Next, we identified mechanisms that might cause this effect on AD pathology, more precisely Aβ efflux, persistent microglial activation and insufficient Aβ clearance, neuronal dysfunction and promotion of Aβ aggregation. Our results further strengthen the believe that even low-grade peripheral inflammation has detrimental effects on AD progression and may further reinforce the idea to modulate peripheral inflammation as a therapeutic strategy for AD.![]()
Collapse
|
48
|
Oxidative Stress and Beta Amyloid in Alzheimer's Disease. Which Comes First: The Chicken or the Egg? Antioxidants (Basel) 2021; 10:antiox10091479. [PMID: 34573112 PMCID: PMC8468973 DOI: 10.3390/antiox10091479] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023] Open
Abstract
The pathogenesis of Alzheimer's disease involves β amyloid (Aβ) accumulation known to induce synaptic dysfunction and neurodegeneration. The brain's vulnerability to oxidative stress (OS) is considered a crucial detrimental factor in Alzheimer's disease. OS and Aβ are linked to each other because Aβ induces OS, and OS increases the Aβ deposition. Thus, the answer to the question "which comes first: the chicken or the egg?" remains extremely difficult. In any case, the evidence for the primary occurrence of oxidative stress in AD is attractive. Thus, evidence indicates that a long period of gradual oxidative damage accumulation precedes and results in the appearance of clinical and pathological AD symptoms, including Aβ deposition, neurofibrillary tangle formation, metabolic dysfunction, and cognitive decline. Moreover, oxidative stress plays a crucial role in the pathogenesis of many risk factors for AD. Alzheimer's disease begins many years before its symptoms, and antioxidant treatment can be an important therapeutic target for attacking the disease.
Collapse
|
49
|
Comparative Transcriptome Analysis of the Expression of Antioxidant and Immunity Genes in the Spleen of a Cyanidin 3-O-Glucoside-Treated Alzheimer's Mouse Model. Antioxidants (Basel) 2021; 10:antiox10091435. [PMID: 34573067 PMCID: PMC8472539 DOI: 10.3390/antiox10091435] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 12/13/2022] Open
Abstract
Cyanidin 3-O-glucoside (C3G) is a well-known antioxidant found as a dietary anthocyanin in different fruits and vegetables. It has protective and therapeutic effects on various diseases. It can reduce neuronal death from amyloid-beta (Aβ)-induced toxicity and promote the inhibition of Aβ fibrillization. Antioxidant and immune modulation might play a critical role in the properties of C3G against Alzheimer's disease (AD) and other diseases. However, limited studies have been performed on the mechanism involved in the effect of C3G through transcriptome analysis. Thus, the objective of this study was to perform comparative transcriptome analysis of the spleen to determine gene expression profiles of wild-type mice (C57BL/6J Jms), an Alzheimer's mouse model (APPswe/PS1dE9 mice), and a C3G-treated Alzheimer's mouse model. Differentially expressed antioxidant, immune-related, and AD pathways genes were identified in the treated group. The validation of gene expression data via RT-PCR studies further supported the current findings. Six important antioxidant genes (S100a8, S100a9, Prdx2, Hp, Mpst, and Prxl2a) and a high number of immune-related genes were found to be upregulated in the treatment groups, suggesting the possible antioxidant and immunomodulatory mechanisms of C3G, respectively. Further studies are strongly recommended to elucidate the precise role of these essential genes and optimize the therapeutic function of C3G in AD and other disease conditions.
Collapse
|
50
|
Gao J, Wang L, Zhao C, Wu Y, Lu Z, Gu Y, Ba Z, Wang X, Wang J, Xu Y. Peony seed oil ameliorates neuroinflammation-mediated cognitive deficits by suppressing microglial activation through inhibition of NF-κB pathway in presenilin 1/2 conditional double knockout mice. J Leukoc Biol 2021; 110:1005-1022. [PMID: 34494312 DOI: 10.1002/jlb.3ma0821-639rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 08/05/2021] [Accepted: 08/13/2021] [Indexed: 12/27/2022] Open
Abstract
Chronic neuroinflammation has been shown to exert adverse influences on the pathology of Alzheimer's disease (AD), associated with the release of abundant proinflammatory mediators by excessively activated microglia, causing synaptic dysfunction, neuronal degeneration, and memory deficits. Thus, the prevention of microglial activation-associated neuroinflammation is important target for deterring neurodegenerative disorders. Peony seed oil (PSO) is a new food resource, rich in α-linolenic acid, the precursor of long chain omega-3 polyunsaturated fatty acids, including docosahexaenoic acid and eicosapentaenoic acid, which exhibit anti-inflammatory properties by altering cell membrane phospholipid fatty acid compositions, disrupting lipid rafts, and inhibiting the activation of the proinflammatory transcription factor NF-κB. However, few studies have examined the anti-neuroinflammatory effects of PSO in AD, and the relevant molecular mechanisms remain unclear. Presenilin1/2 conditional double knockout (PS cDKO) mice display obvious AD-like phenotypes, such as neuroinflammatory responses, synaptic dysfunction, and cognitive deficits. Here, we assessed the potential neuroprotective effects of PSO against neuroinflammation-mediated cognitive deficits in PS cDKO using behavioral tests and molecular biologic analyses. Our study demonstrated that PSO suppressed microglial activation and neuroinflammation through the down-regulation of proinflammatory mediators, such as inducible NOS, COX-2, IL-1β, and TNF-α, in the prefrontal cortex and hippocampus of PS cDKO mice. Further, PSO significantly lessened memory impairment by reversing hyperphosphorylated tau and synaptic proteins deficits in PS cDKO mice. Importantly, PSO's therapeutic effects on cognitive deficits were due to inhibiting neuroinflammatory responses mediated by NF-κB signaling pathway. Taken together, PSO may represent an effective dietary supplementation to restrain the neurodegenerative processes of AD.
Collapse
Affiliation(s)
- Jie Gao
- Department of Physiology, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Rehabilitation Science, University of Traditional Chinese Medicine, Shanghai, China.,Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Lijun Wang
- Department of Physiology, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Rehabilitation Science, University of Traditional Chinese Medicine, Shanghai, China
| | - Chenyi Zhao
- Department of Physiology, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yongkang Wu
- Department of Physiology, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhiyuan Lu
- Department of Physiology, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yining Gu
- School of Rehabilitation Science, University of Traditional Chinese Medicine, Shanghai, China
| | - Zongtao Ba
- School of Rehabilitation Science, University of Traditional Chinese Medicine, Shanghai, China
| | - Xingyu Wang
- Department of Physiology, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian Wang
- School of Rehabilitation Science, University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Xu
- Department of Physiology, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|