1
|
Thakur R, Saini AK, Taliyan R, Chaturvedi N. Neurodegenerative diseases early detection and monitoring system for point-of-care applications. Microchem J 2025; 208:112280. [DOI: 10.1016/j.microc.2024.112280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
2
|
Molla G, Bitew M. Revolutionizing Personalized Medicine: Synergy with Multi-Omics Data Generation, Main Hurdles, and Future Perspectives. Biomedicines 2024; 12:2750. [PMID: 39767657 PMCID: PMC11673561 DOI: 10.3390/biomedicines12122750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 01/11/2025] Open
Abstract
The field of personalized medicine is undergoing a transformative shift through the integration of multi-omics data, which mainly encompasses genomics, transcriptomics, proteomics, and metabolomics. This synergy allows for a comprehensive understanding of individual health by analyzing genetic, molecular, and biochemical profiles. The generation and integration of multi-omics data enable more precise and tailored therapeutic strategies, improving the efficacy of treatments and reducing adverse effects. However, several challenges hinder the full realization of personalized medicine. Key hurdles include the complexity of data integration across different omics layers, the need for advanced computational tools, and the high cost of comprehensive data generation. Additionally, issues related to data privacy, standardization, and the need for robust validation in diverse populations remain significant obstacles. Looking ahead, the future of personalized medicine promises advancements in technology and methodologies that will address these challenges. Emerging innovations in data analytics, machine learning, and high-throughput sequencing are expected to enhance the integration of multi-omics data, making personalized medicine more accessible and effective. Collaborative efforts among researchers, clinicians, and industry stakeholders are crucial to overcoming these hurdles and fully harnessing the potential of multi-omics for individualized healthcare.
Collapse
Affiliation(s)
- Getnet Molla
- College of Veterinary Medicine, Jigjiga University, Jigjiga P.O. Box 1020, Ethiopia
- Bio and Emerging Technology Institute (BETin), Addis Ababa P.O. Box 5954, Ethiopia;
| | - Molalegne Bitew
- Bio and Emerging Technology Institute (BETin), Addis Ababa P.O. Box 5954, Ethiopia;
| |
Collapse
|
3
|
Liu SH, Weber ES, Manz KE, McCarthy KJ, Chen Y, Schüffler PJ, Zhu CW, Tracy M. Assessing the Impact and Cost-Effectiveness of Exposome Interventions on Alzheimer's Disease: A Review of Agent-Based Modeling and Other Data Science Methods for Causal Inference. Genes (Basel) 2024; 15:1457. [PMID: 39596657 PMCID: PMC11593565 DOI: 10.3390/genes15111457] [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: 09/05/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Background: The exposome (e.g., totality of environmental exposures) and its role in Alzheimer's Disease and Alzheimer's Disease and Related Dementias (AD/ADRD) are increasingly critical areas of study. However, little is known about how interventions on the exposome, including personal behavioral modification or policy-level interventions, may impact AD/ADRD disease burden at the population level in real-world settings and the cost-effectiveness of interventions. Methods: We performed a critical review to discuss the challenges in modeling exposome interventions on population-level AD/ADRD burden and the potential of using agent-based modeling (ABM) and other advanced data science methods for causal inference to achieve this. Results: We describe how ABM can be used for empirical causal inference modeling and provide a virtual laboratory for simulating the impacts of personal and policy-level interventions. These hypothetical experiments can provide insight into the optimal timing, targeting, and duration of interventions, identifying optimal combinations of interventions, and can be augmented with economic analyses to evaluate the cost-effectiveness of interventions. We also discuss other data science methods, including structural equation modeling and Mendelian randomization. Lastly, we discuss challenges in modeling the complex exposome, including high dimensional and sparse data, the need to account for dynamic changes over time and over the life course, and the role of exposome burden scores developed using item response theory models and artificial intelligence to address these challenges. Conclusions: This critical review highlights opportunities and challenges in modeling exposome interventions on population-level AD/ADRD disease burden while considering the cost-effectiveness of different interventions, which can be used to aid data-driven policy decisions.
Collapse
Affiliation(s)
- Shelley H. Liu
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ellerie S. Weber
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Katherine E. Manz
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Katharine J. McCarthy
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yitong Chen
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Peter J. Schüffler
- Institute of Pathology, Technical University of Munich, 81675 Munich, Germany
- Munich Data Science Institute, 85748 Garching, Germany
| | - Carolyn W. Zhu
- Department of Geriatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Melissa Tracy
- Department of Epidemiology and Biostatistics, State University of New York at Albany, Albany, NY 12222, USA;
| |
Collapse
|
4
|
Firdaus Z, Li X. Epigenetic Explorations of Neurological Disorders, the Identification Methods, and Therapeutic Avenues. Int J Mol Sci 2024; 25:11658. [PMID: 39519209 PMCID: PMC11546397 DOI: 10.3390/ijms252111658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/26/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Neurodegenerative disorders are major health concerns globally, especially in aging societies. The exploration of brain epigenomes, which consist of multiple forms of DNA methylation and covalent histone modifications, offers new and unanticipated perspective into the mechanisms of aging and neurodegenerative diseases. Initially, chromatin defects in the brain were thought to be static abnormalities from early development associated with rare genetic syndromes. However, it is now evident that mutations and the dysregulation of the epigenetic machinery extend across a broader spectrum, encompassing adult-onset neurodegenerative diseases. Hence, it is crucial to develop methodologies that can enhance epigenetic research. Several approaches have been created to investigate alterations in epigenetics on a spectrum of scales-ranging from low to high-with a particular focus on detecting DNA methylation and histone modifications. This article explores the burgeoning realm of neuroepigenetics, emphasizing its role in enhancing our mechanistic comprehension of neurodegenerative disorders and elucidating the predominant techniques employed for detecting modifications in the epigenome. Additionally, we ponder the potential influence of these advancements on shaping future therapeutic approaches.
Collapse
Affiliation(s)
- Zeba Firdaus
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
5
|
Tripathi A, Pandey VK, Sharma G, Sharma AR, Taufeeq A, Jha AK, Kim JC. Genomic Insights into Dementia: Precision Medicine and the Impact of Gene-Environment Interaction. Aging Dis 2024; 15:2113-2135. [PMID: 38607741 PMCID: PMC11346410 DOI: 10.14336/ad.2024.0322] [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: 01/15/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
The diagnosis, treatment, and management of dementia provide significant challenges due to its chronic cognitive impairment. The complexity of this condition is further highlighted by the impact of gene-environment interactions. A recent strategy combines advanced genomics and precision medicine methods to explore the complex genetic foundations of dementia. Utilizing the most recent research in the field of neurogenetics, the importance of precise genetic data in explaining the variation seen in dementia patients can be investigated. Gene-environment interactions are important because they influence genetic susceptibilities and aid in the development and progression of dementia. Modified to each patient's genetic profile, precision medicine has the potential to detect groups at risk and make previously unheard-of predictions about the course of diseases. Precision medicine techniques have the potential to completely transform treatment and diagnosis methods. Targeted medications that target genetic abnormalities will probably appear, providing the possibility for more efficient and customized medical interventions. Investigating the relationship between genes and the environment may lead to preventive measures that would enable people to change their surroundings and minimize the risk of dementia, leading to the improved lifestyle of affected people. This paper provides a comprehensive overview of the genomic insights into dementia, emphasizing the pivotal role of precision medicine, and gene-environment interactions.
Collapse
Affiliation(s)
- Anjali Tripathi
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Vinay Kumar Pandey
- Division of Research & Innovation (DRI), School of Applied & Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Garima Sharma
- Department of Biomedical Science & Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Republic of Korea
| | - Anam Taufeeq
- Department of Biotechnology, Faculty of Engineering and Technology, Rama University, Kanpur, Uttar Pradesh, India
| | - Abhimanyu Kumar Jha
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Jin-Chul Kim
- Department of Biomedical Science & Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
6
|
Park HH, Armstrong MJ, Gorin FA, Lein PJ. Air Pollution as an Environmental Risk Factor for Alzheimer's Disease and Related Dementias. MEDICAL RESEARCH ARCHIVES 2024; 12:5825. [PMID: 39822906 PMCID: PMC11736697 DOI: 10.18103/mra.v12i10.5825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Alzheimer's disease and related dementias are a leading cause of morbidity in our aging populations. Although influenced by genetic factors, fewer than 5% of Alzheimer's disease and related dementia cases are due solely to genetic causes. There is growing scientific consensus that these dementias arise from complex gene by environment interactions. The 2020 Lancet Commission on dementia prevention, intervention, and care identified 12 modifiable risk factors of dementia, including lifestyle, educational background, comorbidities, and environmental exposures to environmental contaminants. In this review, we summarize the current understanding and data gaps regarding the role(s) of environmental pollutants in the etiology of Alzheimer's disease and related dementias with a focus on air pollution. In addition to summarizing findings from epidemiological and experimental animal studies that link airborne exposures to environmental contaminants to increased risk and/or severity of Alzheimer's disease and related dementias, we discuss currently hypothesized mechanism(s) underlying these associations, including peripheral inflammation, neuroinflammation and epigenetic changes. Key data gaps in this rapidly expanding investigative field and approaches for addressing these gaps are also addressed.
Collapse
Affiliation(s)
- Heui Hye Park
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Matthew J. Armstrong
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Fredric A. Gorin
- Department of Molecular Biosciences, School of Veterinary Medicine, and Department of Neurology, School of Medicine, University of California, Davis, CA 95616, USA
| | - Pamela J. Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| |
Collapse
|
7
|
Tábuas-Pereira M, Bernardes C, Durães J, Lima M, Nogueira AR, Saraiva J, Tábuas T, Coelho M, Paquette K, Westra K, Kun-Rodrigues C, Almeida MR, Baldeiras I, Brás J, Guerreiro R, Santana I. Exploring first-degree family history in a cohort of Portuguese Alzheimer's disease patients: population evidence for X-chromosome linked and recessive inheritance of risk factors. J Neurol 2024; 271:6983-6990. [PMID: 39235525 PMCID: PMC11447147 DOI: 10.1007/s00415-024-12673-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/11/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) heritability is estimated to be around 70-80%. Yet, much of it remains to be explained. Studying transmission patterns may help in understanding other factors contributing to the development of AD. OBJECTIVE In this study, we aimed to search for evidence of autosomal recessive or X- and Y-linked inheritance of risk factors in a large cohort of Portuguese AD patients. METHODS We collected family history from patients with AD and cognitively healthy controls over 75 years of age. We compared the proportions of maternal and paternal history in male and female patients and controls (to search for evidence of X-linked and Y-linked inherited risk factors). We compared the risk of developing AD depending on parents' birthplace (same vs. different), as a proxy of remote consanguinity. We performed linear regressions to study the association of these variables with different endophenotypes. RESULTS We included 3090 participants, 2183 cognitively healthy controls and 907 patients with AD. Men whose mother had dementia have increased odds of developing AD comparing to women whose mother had dementia. In female patients with a CSF biomarker-supported diagnosis of AD, paternal history of dementia is associated with increased CSF phosphorylated Tau levels. People whose parents are from the same town have higher risk of dementia. In multivariate analysis, this proxy is associated with a lower age of onset and higher CSF phosphorylated tau. CONCLUSIONS Our study gives evidence supporting an increased risk of developing AD associated with an X-linked inheritance pattern and remote consanguinity.
Collapse
Affiliation(s)
- Miguel Tábuas-Pereira
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal.
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3004-561, Coimbra, Portugal.
| | - Catarina Bernardes
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3004-561, Coimbra, Portugal
| | - João Durães
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3004-561, Coimbra, Portugal
| | - Marisa Lima
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | | | - Jorge Saraiva
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
| | - Teresa Tábuas
- Instituto Politécnico de Bragança, Bragança, Portugal
| | - Mariana Coelho
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3004-561, Coimbra, Portugal
| | - Kimberly Paquette
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Kaitlyn Westra
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Célia Kun-Rodrigues
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Maria Rosário Almeida
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
| | - Inês Baldeiras
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
| | - José Brás
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Rita Guerreiro
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Isabel Santana
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3004-561, Coimbra, Portugal
| |
Collapse
|
8
|
Kuhn MK, Proctor EA. Microglial Drivers of Alzheimer's Disease Pathology: An Evolution of Diverse Participating States. Proteins 2024. [PMID: 39219300 DOI: 10.1002/prot.26723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 09/04/2024]
Abstract
Microglia, the resident immune-competent cells of the brain, become dysfunctional in Alzheimer's disease (AD), and their aberrant immune responses contribute to the accumulation of pathological proteins and neuronal injury. Genetic studies implicate microglia in the development of AD, prompting interest in developing immunomodulatory therapies to prevent or ameliorate disease. However, microglia take on diverse functional states in disease, playing both protective and detrimental roles in AD, which largely overlap and may shift over the disease course, complicating the identification of effective therapeutic targets. Extensive evidence gathered using transgenic mouse models supports an active role of microglia in pathology progression, though results vary and can be contradictory between different types of models and the degree of pathology at the time of study. Here, we review microglial immune signaling and responses that contribute to the accumulation and spread of pathological proteins or directly affect neuronal health. We additionally explore the use of induced pluripotent stem cell (iPSC)-derived models to study living human microglia and how they have contributed to our knowledge of AD and may begin to fill in the gaps left by mouse models. Ultimately, mouse and iPSC-derived models have their own limitations, and a comprehensive understanding of microglial dysfunction in AD will only be established by an integrated view across models and an appreciation for their complementary viewpoints and limitations.
Collapse
Affiliation(s)
- Madison K Kuhn
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Elizabeth A Proctor
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Engineering Science & Mechanics, The Pennsylvania State University, University Park, Pennsylvania, USA
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
9
|
He Q, Zhang S, Wang J, Ma T, Ma D, Wu L, Zhou M, Zhao L, Chen Y, Liu J, Chen W. The Synergistic Effect Study of Lipopolysaccharide (LPS) and A53T-α-Synuclein: Intranasal LPS Exposure on the A53T-α-Synuclein Transgenic Mouse Model of Parkinson's Disease. Mol Neurobiol 2024; 61:7046-7065. [PMID: 38367134 DOI: 10.1007/s12035-024-04020-y] [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: 10/15/2023] [Accepted: 02/05/2024] [Indexed: 02/19/2024]
Abstract
Aging and interactions between genetic and environmental factors are believed to be involved the chronic development of Parkinson's disease (PD). Among PD patients, abnormally aggregated α-synuclein is a major component of the Lewy body. Generally, the intranasal route is believed to be a gate way to the brain, and it assists environmental neurotoxins in entering the brain and is related to anosmia during early PD. The current study applies the chronic intranasal application of lipopolysaccharides (LPS) in 4-, 8-, 12- and 16-month-old A53T-α-synuclein (A53T-α-Syn) transgenic C57BL/6 mice at 2-day intervals for a 2-month period, for evaluating the behavioral, pathological, and biochemical changes and microglial activation in these animals. According to our results, after intranasal administration of LPS, A53T-α-Syn mice showed severe progressive anosmia, hypokinesia, selective dopaminergic (DAergic) neuronal losses, decreased striatal dopamine (DA) level, and enhanced α-synuclein accumulation within the substantia nigra (SN) in an age-dependent way. In addition, we found obvious NF-кB activation, Nurr1 inhibition, IL-1β, and TNF-α generation within the microglia of the SN. Conversely, the wild-type (WT) mice showed mild, whereas A53T-α-Syn mice had moderate PD-like changes among the old mice. This study demonstrated the synergistic effect of intranasal LPS and α-synuclein burden on PD development. Its underlying mechanism may be associated with Nurr1 inhibition within microglia and the amplification of CNS neuroinflammation. The mice with multiple factors, including aging, neuroinflammation, and α-synuclein mutation, have played a significant role in enhancing our understanding of how inflammation and α-synuclein mutation contribute to the neurodegeneration observed in PD.
Collapse
Affiliation(s)
- Qing He
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuzhen Zhang
- Institute of Neuroscience, Chinese Academy of Sciences (CAS) Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jian Wang
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tengfei Ma
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ding Ma
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Wu
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengxi Zhou
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Zhao
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yajing Chen
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianren Liu
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Wei Chen
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
10
|
Reis J, Spencer PS. An introduction to environmental neurotoxicology: Lessons from a clinical perspective. J Neurol Sci 2024; 463:123108. [PMID: 38991324 DOI: 10.1016/j.jns.2024.123108] [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: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024]
Abstract
In 1992, the Committee on Neurotoxicology and Models for Assessing Risk of the National Academy of Sciences in Washington DC focused with a scientific perspective on the identification of substances with neurotoxic potential, studies of exposed populations, risk assessment, and biologic markers of disease. This Committee recommended: "all physicians should be trained to take a thorough occupational-exposure history and to be aware of other possible sources of toxic exposure". Although convened after several outbreaks of neurotoxic syndromes, clinical neurological considerations were lacking. After defining keys words, namely Environment, Neurotoxicology and Neurotoxicants, we present some demonstrative cases; e.g., the Epidemic Neuropathy in Cuba, Minamata disease, ALS/PDC on Guam, and the ALS hot spot in the French Alps. Always with a clinical and practical approach, we will then review the milieux that contain and convey potential neurotoxicants, the different exposure routes and the clinical presentations. Drawing lessons from clinical cases, we offer some thoughts concerning the future of Environmental Neurotoxicology (ENT). Pointing notably to the diffuse chemical contamination of ecosystems and living beings, including Homo sapiens, we question the real impact of agents with neurotoxic potential on the human brain, considering the effects, for example, of air pollution, endocrine disruptors and nanoparticles. Concern is expressed over the lack of knowledge of the non-monotonic kinetics of many of these chemicals, the major concern being related to mixtures and low-dose exposures, as well as the delayed appearance in clinical expression of prevalent neurodegenerative diseases.
Collapse
Affiliation(s)
- J Reis
- Department of Neurology, University Hospital of Strasbourg, 67000 Strasbourg, France; Association RISE, 67205 Oberhausbergen, France.
| | - P S Spencer
- Department of Neurology, School of Medicine, and Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
11
|
Zhang M, Liang C, Chen X, Cai Y, Cui L. Interplay between microglia and environmental risk factors in Alzheimer's disease. Neural Regen Res 2024; 19:1718-1727. [PMID: 38103237 PMCID: PMC10960290 DOI: 10.4103/1673-5374.389745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/09/2023] [Accepted: 10/24/2023] [Indexed: 12/18/2023] Open
Abstract
Alzheimer's disease, among the most common neurodegenerative disorders, is characterized by progressive cognitive impairment. At present, the Alzheimer's disease main risk remains genetic risks, but major environmental factors are increasingly shown to impact Alzheimer's disease development and progression. Microglia, the most important brain immune cells, play a central role in Alzheimer's disease pathogenesis and are considered environmental and lifestyle "sensors." Factors like environmental pollution and modern lifestyles (e.g., chronic stress, poor dietary habits, sleep, and circadian rhythm disorders) can cause neuroinflammatory responses that lead to cognitive impairment via microglial functioning and phenotypic regulation. However, the specific mechanisms underlying interactions among these factors and microglia in Alzheimer's disease are unclear. Herein, we: discuss the biological effects of air pollution, chronic stress, gut microbiota, sleep patterns, physical exercise, cigarette smoking, and caffeine consumption on microglia; consider how unhealthy lifestyle factors influence individual susceptibility to Alzheimer's disease; and present the neuroprotective effects of a healthy lifestyle. Toward intervening and controlling these environmental risk factors at an early Alzheimer's disease stage, understanding the role of microglia in Alzheimer's disease development, and targeting strategies to target microglia, could be essential to future Alzheimer's disease treatments.
Collapse
Affiliation(s)
- Miaoping Zhang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, China
| | - Chunmei Liang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, China
| | - Xiongjin Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, China
| | - Yujie Cai
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, China
| | - Lili Cui
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, China
| |
Collapse
|
12
|
Duranti E, Villa C. Insights into Dysregulated Neurological Biomarkers in Cancer. Cancers (Basel) 2024; 16:2680. [PMID: 39123408 PMCID: PMC11312413 DOI: 10.3390/cancers16152680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
The link between neurodegenerative diseases (NDs) and cancer has generated greater interest in biomedical research, with decades of global studies investigating neurodegenerative biomarkers in cancer to better understand possible connections. Tau, amyloid-β, α-synuclein, SOD1, TDP-43, and other proteins associated with nervous system diseases have also been identified in various types of solid and malignant tumors, suggesting a potential overlap in pathological processes. In this review, we aim to provide an overview of current evidence on the role of these proteins in cancer, specifically examining their effects on cell proliferation, apoptosis, chemoresistance, and tumor progression. Additionally, we discuss the diagnostic and therapeutic implications of this interconnection, emphasizing the importance of further research to completely comprehend the clinical implications of these proteins in tumors. Finally, we explore the challenges and opportunities in targeting these proteins for the development of new targeted anticancer therapies, providing insight into how to integrate knowledge of NDs in oncology research.
Collapse
Affiliation(s)
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| |
Collapse
|
13
|
Contador I, Buch-Vicente B, del Ser T, Llamas-Velasco S, Villarejo-Galende A, Benito-León J, Bermejo-Pareja F. Charting Alzheimer's Disease and Dementia: Epidemiological Insights, Risk Factors and Prevention Pathways. J Clin Med 2024; 13:4100. [PMID: 39064140 PMCID: PMC11278014 DOI: 10.3390/jcm13144100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD), the most common cause of dementia, is a complex and multifactorial condition without cure at present. The latest treatments, based on anti-amyloid monoclonal antibodies, have only a modest effect in reducing the progression of cognitive decline in AD, whereas the possibility of preventing AD has become a crucial area of research. In fact, recent studies have observed a decrease in dementia incidence in developed regions such as the US and Europe. However, these trends have not been mirrored in non-Western countries (Japan or China), and the contributing factors of this reduction remain unclear. The Lancet Commission has delineated a constrained classification of 12 risk factors across different life stages. Nevertheless, the scientific literature has pointed to over 200 factors-including sociodemographic, medical, psychological, and sociocultural conditions-related to the development of dementia/AD. This narrative review aims to synthesize the risk/protective factors of dementia/AD. Essentially, we found that risk/protective factors vary between individuals and populations, complicating the creation of a unified prevention strategy. Moreover, dementia/AD explanatory mechanisms involve a diverse array of genetic and environmental factors that interact from the early stages of life. In the future, studies across different population-based cohorts are essential to validate risk/protective factors of dementia. This evidence would help develop public health policies to decrease the incidence of dementia.
Collapse
Affiliation(s)
- Israel Contador
- Department of Basic Psychology, Psychobiology, and Methodology of Behavioral Sciences, Faculty of Psychology, University of Salamanca, 37005 Salamanca, Spain
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, 17117 Stockholm, Sweden
| | - Bárbara Buch-Vicente
- Department of Basic Psychology, Psychobiology, and Methodology of Behavioral Sciences, Faculty of Psychology, University of Salamanca, 37005 Salamanca, Spain
| | - Teodoro del Ser
- Alzheimer Centre Reina Sofia—CIEN Foundation, Institute of Health Carlos III, 28031 Madrid, Spain;
| | - Sara Llamas-Velasco
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.L.-V.); (A.V.-G.); (J.B.-L.)
- Department of Neurology, University Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Alberto Villarejo-Galende
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.L.-V.); (A.V.-G.); (J.B.-L.)
- Department of Neurology, University Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Julián Benito-León
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.L.-V.); (A.V.-G.); (J.B.-L.)
- Department of Neurology, University Hospital 12 de Octubre, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
- Department of Medicine, Faculty of Medicine, Complutense University, 28040 Madrid, Spain
| | - Félix Bermejo-Pareja
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
- Department of Medicine, Faculty of Medicine, Complutense University, 28040 Madrid, Spain
| |
Collapse
|
14
|
Kisby GE, Wilson DM, Spencer PS. Introducing the Role of Genotoxicity in Neurodegenerative Diseases and Neuropsychiatric Disorders. Int J Mol Sci 2024; 25:7221. [PMID: 39000326 PMCID: PMC11241460 DOI: 10.3390/ijms25137221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Decades of research have identified genetic and environmental factors involved in age-related neurodegenerative diseases and, to a lesser extent, neuropsychiatric disorders. Genomic instability, i.e., the loss of genome integrity, is a common feature among both neurodegenerative (mayo-trophic lateral sclerosis, Parkinson's disease, Alzheimer's disease) and psychiatric (schizophrenia, autism, bipolar depression) disorders. Genomic instability is associated with the accumulation of persistent DNA damage and the activation of DNA damage response (DDR) pathways, as well as pathologic neuronal cell loss or senescence. Typically, DDR signaling ensures that genomic and proteomic homeostasis are maintained in both dividing cells, including neural progenitors, and post-mitotic neurons. However, dysregulation of these protective responses, in part due to aging or environmental insults, contributes to the progressive development of neurodegenerative and/or psychiatric disorders. In this Special Issue, we introduce and highlight the overlap between neurodegenerative diseases and neuropsychiatric disorders, as well as the emerging clinical, genomic, and molecular evidence for the contributions of DNA damage and aberrant DNA repair. Our goal is to illuminate the importance of this subject to uncover possible treatment and prevention strategies for relevant devastating brain diseases.
Collapse
Affiliation(s)
- Glen E. Kisby
- Department of Biomedical Sciences, College of Osteopathic Medicine of Pacific Northwest, Western University of Health Sciences, Lebanon, OR 97355, USA
| | - David M. Wilson
- Biomedical Research Institute, BIOMED, Hasselt University, 3500 Hasselt, Belgium;
| | - Peter S. Spencer
- Department of Neurology, School of Medicine, Oregon Institute of Occupational Health Sciences, Oregon Health & Sciences University (OHSU), Portland, OR 97239, USA
| |
Collapse
|
15
|
Neale ZE, Fonda JR, Miller MW, Wolf EJ, Zhang R, Sherva R, Harrington KM, Merritt V, Panizzon MS, Hauger RL, Gaziano JM, Logue MW. Subjective cognitive concerns, APOE ε4, PTSD symptoms, and risk for dementia among older veterans. Alzheimers Res Ther 2024; 16:143. [PMID: 38951900 PMCID: PMC11218206 DOI: 10.1186/s13195-024-01512-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/20/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND Posttraumatic stress disorder (PTSD) and traumatic brain injury (TBI) are associated with self-reported problems with cognition as well as risk for Alzheimer's disease and related dementias (ADRD). Overlapping symptom profiles observed in cognitive disorders, psychiatric disorders, and environmental exposures (e.g., head injury) can complicate the detection of early signs of ADRD. The interplay between PTSD, head injury, subjective (self-reported) cognitive concerns and genetic risk for ADRD is also not well understood, particularly in diverse ancestry groups. METHODS Using data from the U.S. Department of Veterans Affairs (VA) Million Veteran Program (MVP), we examined the relationship between dementia risk factors (APOE ε4, PTSD, TBI) and subjective cognitive concerns (SCC) measured in individuals of European (n = 140,921), African (n = 15,788), and Hispanic (n = 8,064) ancestry (EA, AA, and HA, respectively). We then used data from the VA electronic medical record to perform a retrospective survival analysis evaluating PTSD, TBI, APOE ε4, and SCC and their associations with risk of conversion to ADRD in Veterans aged 65 and older. RESULTS PTSD symptoms (B = 0.50-0.52, p < 1E-250) and probable TBI (B = 0.05-0.19, p = 1.51E-07 - 0.002) were positively associated with SCC across all three ancestry groups. APOE ε4 was associated with greater SCC in EA Veterans aged 65 and older (B = 0.037, p = 1.88E-12). Results of Cox models indicated that PTSD symptoms (hazard ratio [HR] = 1.13-1.21), APOE ε4 (HR = 1.73-2.05) and SCC (HR = 1.18-1.37) were positively associated with risk for ADRD across all three ancestry groups. In the EA group, probable TBI also contributed to increased risk of ADRD (HR = 1.18). CONCLUSIONS The findings underscore the value of SCC as an indicator of ADRD risk in Veterans 65 and older when considered in conjunction with other influential genetic, clinical, and demographic risk factors.
Collapse
Affiliation(s)
- Zoe E Neale
- National Center for PTSD, Behavioral Sciences Division, VA Boston Healthcare System, 150 South Huntington Ave (116B-2), Boston, MA, 02130, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Jennifer R Fonda
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02130, USA
- Translational Research Center for TBI and Stress Disorders (TRACTS) and Geriatric Research, Educational and Clinical Center (GRECC), VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Mark W Miller
- National Center for PTSD, Behavioral Sciences Division, VA Boston Healthcare System, 150 South Huntington Ave (116B-2), Boston, MA, 02130, USA
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02130, USA
| | - Erika J Wolf
- National Center for PTSD, Behavioral Sciences Division, VA Boston Healthcare System, 150 South Huntington Ave (116B-2), Boston, MA, 02130, USA
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02130, USA
| | - Rui Zhang
- National Center for PTSD, Behavioral Sciences Division, VA Boston Healthcare System, 150 South Huntington Ave (116B-2), Boston, MA, 02130, USA
| | - Richard Sherva
- Biomedical Genetics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02130, USA
| | - Kelly M Harrington
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02130, USA
- Million Veteran Program (MVP) Coordinating Center, VA Boston Healthcare System, Boston, MA, USA
| | - Victoria Merritt
- Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, La Jolla, San Diego, CA, USA
| | - Matthew S Panizzon
- Department of Psychiatry, University of California, La Jolla, San Diego, CA, USA
- Center for Behavior Genetics of Aging, University of California, La Jolla, San Diego, CA, USA
| | - Richard L Hauger
- Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, La Jolla, San Diego, CA, USA
- Center for Behavior Genetics of Aging, University of California, La Jolla, San Diego, CA, USA
| | - J Michael Gaziano
- Million Veteran Program (MVP) Coordinating Center, VA Boston Healthcare System, Boston, MA, USA
- Division of Aging, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark W Logue
- National Center for PTSD, Behavioral Sciences Division, VA Boston Healthcare System, 150 South Huntington Ave (116B-2), Boston, MA, 02130, USA.
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02130, USA.
- Biomedical Genetics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02130, USA.
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA.
| |
Collapse
|
16
|
Chmielarz M, Sobieszczańska B, Środa-Pomianek K. Metabolic Endotoxemia: From the Gut to Neurodegeneration. Int J Mol Sci 2024; 25:7006. [PMID: 39000116 PMCID: PMC11241432 DOI: 10.3390/ijms25137006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/22/2024] [Accepted: 06/23/2024] [Indexed: 07/16/2024] Open
Abstract
Metabolic endotoxemia is a severe health problem for residents in developed countries who follow a Western diet, disrupting intestinal microbiota and the whole organism's homeostasis. Although the effect of endotoxin on the human immune system is well known, its long-term impact on the human body, lasting many months or even years, is unknown. This is due to the difficulty of conducting in vitro and in vivo studies on the prolonged effect of endotoxin on the central nervous system. In this article, based on the available literature, we traced the path of endotoxin from the intestines to the blood through the intestinal epithelium and factors promoting the development of metabolic endotoxemia. The presence of endotoxin in the bloodstream and the inflammation it induces may contribute to lowering the blood-brain barrier, potentially allowing its penetration into the central nervous system; although, the theory is still controversial. Microglia, guarding the central nervous system, are the first line of defense and respond to endotoxin with activation, which may contribute to the development of neurodegenerative diseases. We traced the pro-inflammatory role of endotoxin in neurodegenerative diseases and its impact on the epigenetic regulation of microglial phenotypes.
Collapse
Affiliation(s)
- Mateusz Chmielarz
- Department of Microbiology, Wroclaw University of Medicine, Chalubinskiego 4 Street, 50-368 Wroclaw, Poland
| | - Beata Sobieszczańska
- Department of Microbiology, Wroclaw University of Medicine, Chalubinskiego 4 Street, 50-368 Wroclaw, Poland
| | - Kamila Środa-Pomianek
- Department of Biophysics and Neuroscience, Wroclaw University of Medicine, Chalubinskiego 3a, 50-368 Wroclaw, Poland
| |
Collapse
|
17
|
Dhapola R, Sharma P, Kumari S, Bhatti JS, HariKrishnaReddy D. Environmental Toxins and Alzheimer's Disease: a Comprehensive Analysis of Pathogenic Mechanisms and Therapeutic Modulation. Mol Neurobiol 2024; 61:3657-3677. [PMID: 38006469 DOI: 10.1007/s12035-023-03805-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/13/2023] [Indexed: 11/27/2023]
Abstract
Alzheimer's disease is a leading cause of mortality worldwide. Inorganic and organic hazards, susceptibility to harmful metals, pesticides, agrochemicals, and air pollution are major environmental concerns. As merely 5% of AD cases are directly inherited indicating that these environmental factors play a major role in disease development. Long-term exposure to environmental toxins is believed to progress neuropathology, which leads to the development of AD. Numerous in-vitro and in-vivo studies have suggested the harmful impact of environmental toxins at cellular and molecular level. Common mechanisms involved in the toxicity of these environmental pollutants include oxidative stress, neuroinflammation, mitochondrial dysfunction, abnormal tau, and APP processing. Increased expression of GSK-3β, BACE-1, TNF-α, and pro-apoptotic molecules like caspases is observed upon exposure to these environmental toxins. In addition, the expression of neurotrophins like BDNF and GAP-43 have been found to be reduced as a result of toxicity. Further, modulation of signaling pathways involving PARP-1, PGC-1α, and MAPK/ERK induced by toxins have been reported to contribute in AD pathogenesis. These pathways are a promising target for developing novel AD therapeutics. Drugs like epigallocatechin-gallate, neflamapimod, salsalate, dexmedetomidine, and atabecestat are in different phases of clinical trials targeting the pathways for possible treatment of AD. This review aims to culminate the correlation between environmental toxicants and AD development. We emphasized upon the signaling pathways involved in the progression of the disease and the therapeutics under clinical trial targeting the altered pathways for possible treatment of AD.
Collapse
Affiliation(s)
- Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, Central University of Punjab, Bathinda, 151 401, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, Central University of Punjab, Bathinda, 151 401, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, Central University of Punjab, Bathinda, 151 401, India
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151 401, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, Central University of Punjab, Bathinda, 151 401, India.
| |
Collapse
|
18
|
Brown RE. Measuring the replicability of our own research. J Neurosci Methods 2024; 406:110111. [PMID: 38521128 DOI: 10.1016/j.jneumeth.2024.110111] [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: 01/21/2024] [Revised: 03/08/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024]
Abstract
In the study of transgenic mouse models of neurodevelopmental and neurodegenerative disorders, we use batteries of tests to measure deficits in behaviour and from the results of these tests, we make inferences about the mental states of the mice that we interpret as deficits in "learning", "memory", "anxiety", "depression", etc. This paper discusses the problems of determining whether a particular transgenic mouse is a valid mouse model of disease X, the problem of background strains, and the question of whether our behavioural tests are measuring what we say they are. The problem of the reliability of results is then discussed: are they replicable between labs and can we replicate our results in our own lab? This involves the study of intra- and inter- experimenter reliability. The variables that influence replicability and the importance of conducting a complete behavioural phenotype: sensory, motor, cognitive and social emotional behaviour are discussed. Then the thorny question of failure to replicate is examined: Is it a curse or a blessing? Finally, the role of failure in research and what it tells us about our research paradigms is examined.
Collapse
Affiliation(s)
- Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
19
|
Dong Z, Jiang W, Li H, DeWan AT, Zhao H. LDER-GE estimates phenotypic variance component of gene-environment interactions in human complex traits accurately with GE interaction summary statistics and full LD information. Brief Bioinform 2024; 25:bbae335. [PMID: 38980374 PMCID: PMC11232466 DOI: 10.1093/bib/bbae335] [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: 04/08/2024] [Revised: 06/05/2024] [Accepted: 06/26/2024] [Indexed: 07/10/2024] Open
Abstract
Gene-environment (GE) interactions are essential in understanding human complex traits. Identifying these interactions is necessary for deciphering the biological basis of such traits. In this study, we review state-of-art methods for estimating the proportion of phenotypic variance explained by genome-wide GE interactions and introduce a novel statistical method Linkage-Disequilibrium Eigenvalue Regression for Gene-Environment interactions (LDER-GE). LDER-GE improves the accuracy of estimating the phenotypic variance component explained by genome-wide GE interactions using large-scale biobank association summary statistics. LDER-GE leverages the complete Linkage Disequilibrium (LD) matrix, as opposed to only the diagonal squared LD matrix utilized by LDSC (Linkage Disequilibrium Score)-based methods. Our extensive simulation studies demonstrate that LDER-GE performs better than LDSC-based approaches by enhancing statistical efficiency by ~23%. This improvement is equivalent to a sample size increase of around 51%. Additionally, LDER-GE effectively controls type-I error rate and produces unbiased results. We conducted an analysis using UK Biobank data, comprising 307 259 unrelated European-Ancestry subjects and 966 766 variants, across 217 environmental covariate-phenotype (E-Y) pairs. LDER-GE identified 34 significant E-Y pairs while LDSC-based method only identified 23 significant E-Y pairs with 22 overlapped with LDER-GE. Furthermore, we employed LDER-GE to estimate the aggregated variance component attributed to multiple GE interactions, leading to an increase in the explained phenotypic variance with GE interactions compared to considering main genetic effects only. Our results suggest the importance of impacts of GE interactions on human complex traits.
Collapse
Affiliation(s)
- Zihan Dong
- Department of Biostatistics, Yale School of Public Health, 60 College Street, New Haven, CT 06510, United States
- Center for Perinatal, Pediatric and Environmental Epidemiology, 60 College Street, Yale School of Public Health, New Haven, CT 06510, United States
| | - Wei Jiang
- Department of Biostatistics, Yale School of Public Health, 60 College Street, New Haven, CT 06510, United States
| | - Hongyu Li
- Department of Biostatistics, Yale School of Public Health, 60 College Street, New Haven, CT 06510, United States
| | - Andrew T DeWan
- Center for Perinatal, Pediatric and Environmental Epidemiology, 60 College Street, Yale School of Public Health, New Haven, CT 06510, United States
- Department of Chronic Disease Epidemiology, Yale School of Public Health, 60 College Street, New Haven, CT 06510, United States
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, 60 College Street, New Haven, CT 06510, United States
| |
Collapse
|
20
|
Ma H, Shi Z, Kim M, Liu B, Smith PJ, Liu Y, Wu G. Disentangling sex-dependent effects of APOE on diverse trajectories of cognitive decline in Alzheimer's disease. Neuroimage 2024; 292:120609. [PMID: 38614371 PMCID: PMC11069285 DOI: 10.1016/j.neuroimage.2024.120609] [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: 09/05/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/15/2024] Open
Abstract
Current diagnostic systems for Alzheimer's disease (AD) rely upon clinical signs and symptoms, despite the fact that the multiplicity of clinical symptoms renders various neuropsychological assessments inadequate to reflect the underlying pathophysiological mechanisms. Since putative neuroimaging biomarkers play a crucial role in understanding the etiology of AD, we sought to stratify the diverse relationships between AD biomarkers and cognitive decline in the aging population and uncover risk factors contributing to the diversities in AD. To do so, we capitalized on a large amount of neuroimaging data from the ADNI study to examine the inflection points along the dynamic relationship between cognitive decline trajectories and whole-brain neuroimaging biomarkers, using a state-of-the-art statistical model of change point detection. Our findings indicated that the temporal relationship between AD biomarkers and cognitive decline may differ depending on the synergistic effect of genetic risk and biological sex. Specifically, tauopathy-PET biomarkers exhibit a more dynamic and age-dependent association with Mini-Mental State Examination scores (p<0.05), with inflection points at 72, 78, and 83 years old, compared with amyloid-PET and neurodegeneration (cortical thickness from MRI) biomarkers. In the landscape of health disparities in AD, our analysis indicated that biological sex moderates the rate of cognitive decline associated with APOE4 genotype. Meanwhile, we found that higher education levels may moderate the effect of APOE4, acting as a marker of cognitive reserve.
Collapse
Affiliation(s)
- Haixu Ma
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Zhuoyu Shi
- Department of Psychiatry, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Minjeong Kim
- Department of Computer Science, University of North Carolina at Greensboro, NC 27412, USA
| | - Bin Liu
- Department of Statistics and Data Science, School of Management at Fudan University, Shanghai, 200433, PR China
| | - Patrick J Smith
- Department of Psychiatry, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Yufeng Liu
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, NC 27599, USA; Department of Genetics, Department of Biostatistics, University of North Carolina at Chapel Hill, NC 27599, USA.
| | - Guorong Wu
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, NC 27599, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, NC 27599, USA; Department of Computer Science, University of North Carolina at Chapel Hill, NC 27599, USA; UNC Neuroscience Center, University of North Carolina at Chapel Hill, NC 27599, USA.
| |
Collapse
|
21
|
Yao XI, Tong X, Shen C, Song Y, Sun S, Chen K, Shen H. Green space, genetic susceptibility, and risk of osteoporosis:a cohort study from the UK Biobank. CHEMOSPHERE 2024; 353:141632. [PMID: 38442776 DOI: 10.1016/j.chemosphere.2024.141632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
OBJECTIVE This study aimed to investigate the effect of residential exposure to green space on the incident osteoporosis and further explore the modification effect of genetic susceptibility. METHODS Participants from the UK Biobank were followed from 2006 to 2010 (baseline) to December 31st, 2022. Using land use coverage, we evaluated exposure to residential surrounding green space, natural environment, and domestic gardens. We used the Cox regression to examine the association between the residential environment and incident osteoporosis. The interactive effects between polygenic risk score (PRS) of osteoporosis and residential environments on incident osteoporosis were investigated. RESULTS This study included 292,662 participants. Over a median follow-up period of 13.65 years, we documented 9177 incidents of osteoporosis. Per interquartile (IQR) increase in greenness and natural environment at a 300 m buffer was associated with a 4% lower risk of incident osteoporosis [HR = 0.96 (95% CI: 0.93, 0.99)] and [HR = 0.96 (95% CI: 0.93, 0.98)], respectively. We did not identify any interactive effects between genetic risk and residential environment on incident osteoporosis. CONCLUSIONS This study found that public greenness and natural environments could reduce the risk of incident osteoporosis regardless of genetic predisposition. Developing sustainable and publicly accessible natural environments might benefit populations' bone health.
Collapse
Affiliation(s)
- Xiaoxin I Yao
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, PR China; Department of Clinical Research, The Eighth Affiliated Hospital, Sun Yat-sen University, PR China
| | - Xinning Tong
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, PR China
| | - Chen Shen
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK; National Institute for Health Research Health Protection Research Unit in Chemical and Radiation Threats and Hazards, Imperial College London, UK
| | - Yichang Song
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, PR China
| | - Shengzhi Sun
- School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China.
| | - Keng Chen
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, PR China.
| | - Huiyong Shen
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, PR China; Department of Clinical Research, The Eighth Affiliated Hospital, Sun Yat-sen University, PR China.
| |
Collapse
|
22
|
Soni N, Hohsfield LA, Tran KM, Kawauchi S, Walker A, Javonillo D, Phan J, Matheos D, Da Cunha C, Uyar A, Milinkeviciute G, Gomez‐Arboledas A, Tran K, Kaczorowski CC, Wood MA, Tenner AJ, LaFerla FM, Carter GW, Mortazavi A, Swarup V, MacGregor GR, Green KN. Genetic diversity promotes resilience in a mouse model of Alzheimer's disease. Alzheimers Dement 2024; 20:2794-2816. [PMID: 38426371 PMCID: PMC11032575 DOI: 10.1002/alz.13753] [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: 11/17/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 03/02/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a neurodegenerative disorder with multifactorial etiology, including genetic factors that play a significant role in disease risk and resilience. However, the role of genetic diversity in preclinical AD studies has received limited attention. METHODS We crossed five Collaborative Cross strains with 5xFAD C57BL/6J female mice to generate F1 mice with and without the 5xFAD transgene. Amyloid plaque pathology, microglial and astrocytic responses, neurofilament light chain levels, and gene expression were assessed at various ages. RESULTS Genetic diversity significantly impacts AD-related pathology. Hybrid strains showed resistance to amyloid plaque formation and neuronal damage. Transcriptome diversity was maintained across ages and sexes, with observable strain-specific variations in AD-related phenotypes. Comparative gene expression analysis indicated correlations between mouse strains and human AD. DISCUSSION Increasing genetic diversity promotes resilience to AD-related pathogenesis, relative to an inbred C57BL/6J background, reinforcing the importance of genetic diversity in uncovering resilience in the development of AD. HIGHLIGHTS Genetic diversity's impact on AD in mice was explored. Diverse F1 mouse strains were used for AD study, via the Collaborative Cross. Strain-specific variations in AD pathology, glia, and transcription were found. Strains resilient to plaque formation and plasma neurofilament light chain (NfL) increases were identified. Correlations with human AD transcriptomics were observed.
Collapse
Affiliation(s)
- Neelakshi Soni
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Lindsay A. Hohsfield
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Kristine M. Tran
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Shimako Kawauchi
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Transgenic Mouse Facility, ULAROffice of ResearchUniversity of CaliforniaIrvineCaliforniaUSA
| | - Amber Walker
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Transgenic Mouse Facility, ULAROffice of ResearchUniversity of CaliforniaIrvineCaliforniaUSA
| | - Dominic Javonillo
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Jimmy Phan
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Dina Matheos
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Celia Da Cunha
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Asli Uyar
- The Jackson LaboratoryBar HarborMaineUSA
| | - Giedre Milinkeviciute
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Angela Gomez‐Arboledas
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Katelynn Tran
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | | | - Marcelo A. Wood
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Andrea J. Tenner
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Pathology and Laboratory MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Frank M. LaFerla
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | | | - Ali Mortazavi
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Developmental and Cellular BiologyUniversity of CaliforniaIrvineCaliforniaUSA
- Center for Complex Biological SystemsUniversity of CaliforniaIrvineCaliforniaUSA
| | - Vivek Swarup
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Grant R. MacGregor
- Transgenic Mouse Facility, ULAROffice of ResearchUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Developmental and Cellular BiologyUniversity of CaliforniaIrvineCaliforniaUSA
| | - Kim N. Green
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| |
Collapse
|
23
|
Liu X, Liu Y, Liu J, Zhang H, Shan C, Guo Y, Gong X, Cui M, Li X, Tang M. Correlation between the gut microbiome and neurodegenerative diseases: a review of metagenomics evidence. Neural Regen Res 2024; 19:833-845. [PMID: 37843219 PMCID: PMC10664138 DOI: 10.4103/1673-5374.382223] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/19/2023] [Accepted: 06/17/2023] [Indexed: 10/17/2023] Open
Abstract
A growing body of evidence suggests that the gut microbiota contributes to the development of neurodegenerative diseases via the microbiota-gut-brain axis. As a contributing factor, microbiota dysbiosis always occurs in pathological changes of neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. High-throughput sequencing technology has helped to reveal that the bidirectional communication between the central nervous system and the enteric nervous system is facilitated by the microbiota's diverse microorganisms, and for both neuroimmune and neuroendocrine systems. Here, we summarize the bioinformatics analysis and wet-biology validation for the gut metagenomics in neurodegenerative diseases, with an emphasis on multi-omics studies and the gut virome. The pathogen-associated signaling biomarkers for identifying brain disorders and potential therapeutic targets are also elucidated. Finally, we discuss the role of diet, prebiotics, probiotics, postbiotics and exercise interventions in remodeling the microbiome and reducing the symptoms of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaoyan Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Yi Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
- Institute of Animal Husbandry, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu Province, China
| | - Junlin Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Hantao Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Chaofan Shan
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Yinglu Guo
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Xun Gong
- Department of Rheumatology & Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Mengmeng Cui
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Xiubin Li
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Min Tang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| |
Collapse
|
24
|
Sakowski SA, Koubek EJ, Chen KS, Goutman SA, Feldman EL. Role of the Exposome in Neurodegenerative Disease: Recent Insights and Future Directions. Ann Neurol 2024; 95:635-652. [PMID: 38411261 PMCID: PMC11023772 DOI: 10.1002/ana.26897] [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: 11/03/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/28/2024]
Abstract
Neurodegenerative diseases are increasing in prevalence and place a significant burden on society. The causes are multifactorial and complex, and increasing evidence suggests a dynamic interplay between genes and the environment, emphasizing the importance of identifying and understanding the role of lifelong exposures, known as the exposome, on the nervous system. This review provides an overview of recent advances toward defining neurodegenerative disease exposomes, focusing on Parkinson's disease, amyotrophic lateral sclerosis, and Alzheimer's disease. We present the current state of the field based on emerging data, elaborate on key themes and potential mechanisms, and conclude with limitations and future directions. ANN NEUROL 2024;95:635-652.
Collapse
Affiliation(s)
- Stacey A. Sakowski
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - Emily J. Koubek
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kevin S. Chen
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Stephen A. Goutman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
25
|
Teixeira L. The nervous system and associated disorders. BRITISH JOURNAL OF NURSING (MARK ALLEN PUBLISHING) 2024; 33:194-199. [PMID: 38386518 DOI: 10.12968/bjon.2024.33.4.194] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Disorders of the nervous system, encompassing the brain, spinal cord and peripheral nerves, have emerged as a significant public health issue, with profound implications for individuals worldwide. These conditions result in significant morbidity and mortality. Many patients with neurological disorders often have comorbidities, further complicating their clinical presentation. Therefore, nurses must possess a comprehensive understanding of the nervous system and its associated disorders to formulate detailed care plans that address the unique needs of each patient. This article aims to explore the underlying pathophysiology of some of the most prevalent neurological disorders and how this informs effective patient assessment and diagnostic strategies. A further article will build on this to consider patient assessment and formulating a care plan in more detail.
Collapse
Affiliation(s)
- Luis Teixeira
- Lecturer in Adult Nursing Complex Care, Kings College London
| |
Collapse
|
26
|
Abondio P, Bruno F, Passarino G, Montesanto A, Luiselli D. Pangenomics: A new era in the field of neurodegenerative diseases. Ageing Res Rev 2024; 94:102180. [PMID: 38163518 DOI: 10.1016/j.arr.2023.102180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/14/2023] [Accepted: 12/28/2023] [Indexed: 01/03/2024]
Abstract
A pangenome is composed of all the genetic variability of a group of individuals, and its application to the study of neurodegenerative diseases may provide valuable insights into the underlying aspects of genetic heterogenetiy for these complex ailments, including gene expression, epigenetics, and translation mechanisms. Furthermore, a reference pangenome allows for the identification of previously undetected structural commonalities and differences among individuals, which may help in the diagnosis of a disease, support the prediction of what will happen over time (prognosis) and aid in developing novel treatments in the perspective of personalized medicine. Therefore, in the present review, the application of the pangenome concept to the study of neurodegenerative diseases will be discussed and analyzed for its potential to enable an improvement in diagnosis and prognosis for these illnesses, leading to the development of tailored treatments for individual patients from the knowledge of the genomic composition of a whole population.
Collapse
Affiliation(s)
- Paolo Abondio
- Laboratory of Ancient DNA, Department of Cultural Heritage, University of Bologna, Via degli Ariani 1, 48121 Ravenna, Italy.
| | - Francesco Bruno
- Academy of Cognitive Behavioral Sciences of Calabria (ASCoC), Lamezia Terme, Italy; Regional Neurogenetic Centre (CRN), Department of Primary Care, Azienda Sanitaria Provinciale Di Catanzaro, Viale A. Perugini, 88046 Lamezia Terme, CZ, Italy; Association for Neurogenetic Research (ARN), Lamezia Terme, CZ, Italy
| | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende 87036, Italy
| | - Alberto Montesanto
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende 87036, Italy
| | - Donata Luiselli
- Laboratory of Ancient DNA, Department of Cultural Heritage, University of Bologna, Via degli Ariani 1, 48121 Ravenna, Italy
| |
Collapse
|
27
|
Saul MC, Litkowski EM, Hadad N, Dunn AR, Boas SM, Wilcox JAL, Robbins JE, Wu Y, Philip VM, Merrihew GE, Park J, De Jager PL, Bridges DE, Menon V, Bennett DA, Hohman TJ, MacCoss MJ, Kaczorowski CC. Hippocampus Glutathione S Reductase Potentially Confers Genetic Resilience to Cognitive Decline in the AD-BXD Mouse Population. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.09.574219. [PMID: 38260300 PMCID: PMC10802440 DOI: 10.1101/2024.01.09.574219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Alzheimer's disease (AD) is a prevalent and costly age-related dementia. Heritable factors account for 58-79% of variation in late-onset AD, but substantial variation remains in age-of- onset, disease severity, and whether those with high-risk genotypes acquire AD. To emulate the diversity of human populations, we utilized the AD-BXD mouse panel. This genetically diverse resource combines AD genotypes with multiple BXD strains to discover new genetic drivers of AD resilience. Comparing AD-BXD carriers to noncarrier littermates, we computed a novel quantitative metric for resilience to cognitive decline in the AD-BXDs. Our quantitative AD resilience trait was heritable and genetic mapping identified a locus on chr8 associated with resilience to AD mutations that resulted in amyloid brain pathology. Using a hippocampus proteomics dataset, we nominated the mitochondrial glutathione S reductase protein (GR or GSHR) as a resilience factor, finding that the DBA/2J genotype was associated with substantially higher GR abundance. By mapping protein QTLs (pQTLs), we identified synaptic organization and mitochondrial proteins coregulated in trans with a cis-pQTL for GR. We found four coexpression modules correlated with the quantitative resilience score in aged 5XFAD mice using paracliques, which were related to cell structure, protein folding, and postsynaptic densities. Finally, we found significant positive associations between human GSR transcript abundance in the brain and better outcomes on AD-related cognitive and pathology traits in the Religious Orders Study/Memory and Aging project (ROSMAP). Taken together, these data support a framework for resilience in which neuronal antioxidant pathway activity provides for stability of synapses within the hippocampus.
Collapse
|
28
|
Mishra V, Yadav D, Solanki KS, Koul B, Song M. A Review on the Protective Effects of Probiotics against Alzheimer's Disease. BIOLOGY 2023; 13:8. [PMID: 38248439 PMCID: PMC10813289 DOI: 10.3390/biology13010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/01/2023] [Accepted: 12/13/2023] [Indexed: 01/23/2024]
Abstract
This review summarizes the protective effects of probiotics against Alzheimer's disease (AD), one of the most common neurodegenerative disorders affecting older adults. This disease is characterized by the deposition of tau and amyloid β peptide (Aβ) in different parts of the brain. Symptoms observed in patients with AD include struggles with writing, speech, memory, and knowledge. The gut microbiota reportedly plays an important role in brain functioning due to its bidirectional communication with the gut via the gut-brain axis. The emotional and cognitive centers in the brain are linked to the functions of the peripheral intestinal system via this gut-brain axis. Dysbiosis has been linked to neurodegenerative disorders, indicating the significance of gut homeostasis for proper brain function. Probiotics play an important role in protecting against the symptoms of AD as they restore gut-brain homeostasis to a great extent. This review summarizes the characteristics, status of gut-brain axis, and significance of gut microbiota in AD. Review and research articles related to the role of probiotics in the treatment of AD were searched in the PubMed database. Recent studies conducted using animal models were given preference. Recent clinical trials were searched for separately. Several studies conducted on animal and human models clearly explain the benefits of probiotics in improving cognition and memory in experimental subjects. Based on these studies, novel therapeutic approaches can be designed for the treatment of patients with AD.
Collapse
Affiliation(s)
- Vibhuti Mishra
- School of Studies in Biochemistry, Jiwaji University, Gwalior 474003, India;
| | - Dhananjay Yadav
- Department of Life Science, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Kavita Singh Solanki
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA;
| | - Bhupendra Koul
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, India;
| | - Minseok Song
- Department of Life Science, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
29
|
Yu C, Ryan J, Orchard SG, Robb C, Woods RL, Wolfe R, Renton AE, Goate AM, Brodtmann A, Shah RC, Chong TTJ, Sheets K, Kyndt C, Sood A, Storey E, Murray AM, McNeil JJ, Lacaze P. Validation of newly derived polygenic risk scores for dementia in a prospective study of older individuals. Alzheimers Dement 2023; 19:5333-5342. [PMID: 37177856 PMCID: PMC10640662 DOI: 10.1002/alz.13113] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/27/2023] [Accepted: 04/03/2023] [Indexed: 05/15/2023]
Abstract
INTRODUCTION Recent genome-wide association studies identified new dementia-associated variants. We assessed the performance of updated polygenic risk scores (PRSs) using these variants in an independent cohort. METHODS We used Cox models and area under the curve (AUC) to validate new PRSs (PRS-83SNP, PRS-SBayesR, and PRS-CS) compared with an older PRS-23SNP in 12,031 initially-healthy participants ≥70 years of age. Dementia was rigorously adjudicated according to Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (DSM-IV) criteria. RESULTS PRS-83SNP, PRS-SBayesR, and PRS-CS were associated with incident dementia, with fully adjusted (including apolipoprotein E [APOE] ε4) hazard ratios per standard deviation (SD) of 1.35 (1.23-1.47), 1.37 (1.25-1.50), and 1.42 (1.30-1.56), respectively. The AUC of a model containing conventional/non-genetic factors and APOE was 74.7%. This was improved to 75.7% (p = 0.007), 76% (p = 0.004), and 76.1% (p = 0.003) with addition of PRS-83SNP, PRS-SBayesR, and PRS-CS, respectively. The PRS-23SNP did not improve AUC (74.7%, p = 0.95). CONCLUSION New PRSs for dementia significantly improve risk-prediction performance, but still account for less risk than APOE genotype overall.
Collapse
Affiliation(s)
- Chenglong Yu
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Joanne Ryan
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Suzanne G. Orchard
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Catherine Robb
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Robyn L. Woods
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Rory Wolfe
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Alan E. Renton
- Department Genetics and Genomic Sciences and Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Departments of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alison M. Goate
- Department Genetics and Genomic Sciences and Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Departments of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Amy Brodtmann
- Cognitive Health Initiative, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia
| | - Raj C. Shah
- Department of Family & Preventive Medicine and the Rush Alzheimer’s Disease Center, Chicago, Illinois, USA
| | - Trevor T.-J. Chong
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
- Department of Clinical Neurosciences, St. Vincent’s Hospital, Melbourne, Victoria, Australia
| | - Kerry Sheets
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Geriatrics, Hennepin Healthcare, Minneapolis, Minnesota, USA
| | - Christopher Kyndt
- Department of Neurology, Melbourne Health, Parkville, Victoria, Australia
- Department of Neuroscience, Eastern Health, Box Hill, Victoria, Australia
| | - Ajay Sood
- Department of Neurology and the Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | - Elsdon Storey
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Anne M. Murray
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Geriatrics, Hennepin Healthcare, Minneapolis, Minnesota, USA
- Berman Center for Outcomes and Clinical Research, Hennepin Healthcare Research Institute, Hennepin Healthcare, and University of Minnesota, Minneapolis, Minnesota, USA
| | - John J. McNeil
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Paul Lacaze
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
30
|
Pandics T, Major D, Fazekas-Pongor V, Szarvas Z, Peterfi A, Mukli P, Gulej R, Ungvari A, Fekete M, Tompa A, Tarantini S, Yabluchanskiy A, Conley S, Csiszar A, Tabak AG, Benyo Z, Adany R, Ungvari Z. Exposome and unhealthy aging: environmental drivers from air pollution to occupational exposures. GeroScience 2023; 45:3381-3408. [PMID: 37688657 PMCID: PMC10643494 DOI: 10.1007/s11357-023-00913-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/14/2023] [Indexed: 09/11/2023] Open
Abstract
The aging population worldwide is facing a significant increase in age-related non-communicable diseases, including cardiovascular and brain pathologies. This comprehensive review paper delves into the impact of the exposome, which encompasses the totality of environmental exposures, on unhealthy aging. It explores how environmental factors contribute to the acceleration of aging processes, increase biological age, and facilitate the development and progression of a wide range of age-associated diseases. The impact of environmental factors on cognitive health and the development of chronic age-related diseases affecting the cardiovascular system and central nervous system is discussed, with a specific focus on Alzheimer's disease, Parkinson's disease, stroke, small vessel disease, and vascular cognitive impairment (VCI). Aging is a major risk factor for these diseases. Their pathogenesis involves cellular and molecular mechanisms of aging such as increased oxidative stress, impaired mitochondrial function, DNA damage, and inflammation and is influenced by environmental factors. Environmental toxicants, including ambient particulate matter, pesticides, heavy metals, and organic solvents, have been identified as significant contributors to cardiovascular and brain aging disorders. These toxicants can inflict both macro- and microvascular damage and many of them can also cross the blood-brain barrier, inducing neurotoxic effects, neuroinflammation, and neuronal dysfunction. In conclusion, environmental factors play a critical role in modulating cardiovascular and brain aging. A deeper understanding of how environmental toxicants exacerbate aging processes and contribute to the pathogenesis of neurodegenerative diseases, VCI, and dementia is crucial for the development of preventive strategies and interventions to promote cardiovascular, cerebrovascular, and brain health. By mitigating exposure to harmful environmental factors and promoting healthy aging, we can strive to reduce the burden of age-related cardiovascular and brain pathologies in the aging population.
Collapse
Affiliation(s)
- Tamas Pandics
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Department of Public Health Laboratory, National Public Health Centre, Budapest, Hungary
- Department of Public Health Siences, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - David Major
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Vince Fazekas-Pongor
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Zsofia Szarvas
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Peterfi
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Ungvari
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Monika Fekete
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Anna Tompa
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Shannon Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Adam G Tabak
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- UCL Brain Sciences, University College London, London, UK
- Department of Internal Medicine and Oncology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Zoltan Benyo
- Department of Translational Medicine, Semmelweis University, Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University (ELKH-SE) Cerebrovascular and Neurocognitive Disorders Research Group, Budapest, H-1052, Hungary
| | - Roza Adany
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- ELKH-DE Public Health Research Group, Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary
- Epidemiology and Surveillance Centre, Semmelweis University, 1085, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA.
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
31
|
David GS, Viana JMS, das Graças Dias KO. A simulation-based assessment of the efficiency of QTL mapping under environment and genotype x environment interaction effects. PLoS One 2023; 18:e0295245. [PMID: 38033088 PMCID: PMC10688852 DOI: 10.1371/journal.pone.0295245] [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: 07/20/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023] Open
Abstract
The objective of this simulation-based study was to assess how genes, environments, and genotype x environment (GxE) interaction affect the quantitative trait loci (QTL) mapping efficiency. The simulation software performed 50 samplings of 300 recombinant inbred lines (RILs) from a F2, which were assessed in six environments. The RILs were genotyped for 977 single nucleotide polymorphisms (SNP) and phenotyped for grain yield. The average SNP density was 2 cM. We defined six QTLs and 190 minor genes. The trait heritability ranged from 30 to 80%. We fitted the single QTL model and the multiple QTL model on multiple phenotypes. The environment and complex GxE interaction effects led to a low correlation between the QTL heritability and power. The single- and across-environment analyses allowed all QTLs be declared, with an average power of 28 to 100%. In the across-environment analysis, five QTLs showed average power in the range 46 to 82%. Both models provided a good control of the false positive rate (6%, on average) and a precise localization of the QTLs (bias of 2 cM, on average). The QTL power in each environment has a high positive correlation with the range between QTL genotypes for the sum of the additive, environment, and GxE interaction effects (0.76 to 0.96). The uncertainty about the magnitude and sign of the environment and GxE interaction effects makes QTL mapping in multi-environment trials unpredictable. Unfortunately, this uncertainty has no solution because the geneticist has no control over the magnitude and sign of the environment and GxE interaction effects. However, the single- and across-environment analyses are efficient even under a low correlation between QTL heritability and power.
Collapse
Affiliation(s)
- Grace Sunshine David
- Department of Crop Science, University of Calabar, Calabar, Cross River State, Nigeria
| | | | | |
Collapse
|
32
|
Yuan S, Li W, Ling Y, Huang X, Feng A, Tan S, He N, Li L, Li S, Xu A, Lyu J. Associations of screen-based sedentary activities with all cause dementia, Alzheimer's disease, vascular dementia: a longitudinal study based on 462,524 participants from the UK Biobank. BMC Public Health 2023; 23:2141. [PMID: 37919716 PMCID: PMC10621115 DOI: 10.1186/s12889-023-17050-3] [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: 04/20/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Current drug treatments for dementia aren't effective. Studying gene-environment interactions can help develop personalized early intervention strategies for Alzheimer's disease (AD). However, no studies have examined the relationship between screen-based sedentary activities and genetic susceptibility to AD risk, and further understanding of the causal relationship is also crucial. METHODS This study included 462,524 participants from the UK Biobank with a follow-up of 13.6 years. Participants' screen-based sedentary activities time was categorized into three groups based on recorded time: ≥ 4 h/day, 2-3 h/day, and ≤ 1 h/day. Cox proportional risk models were used to analyze the association between computer use/TV viewing groups and the risk of all-cause dementia, AD and vascular dementia (VD). Generalized linear model (GLM) were used to examine the relationship between screen-based sedentary activities and brain structure. Bidirectional Mendelian randomization (MR) was used to validate the causal relationship between TV viewing and AD. RESULTS Compared to TV viewing ≤ 1 h/day, 1)TV viewing 2-3 h/day was correlated with a higher risk of all-cause dementia (HR = 1.09, 95% CI:1.01-1.18, P < 0.05), and TV viewing ≥ 4 h/day was associated with a higher risk of all-cause dementia (HR = 1.29, 95% CI: 1.19-1.40, P < 0.001), AD (HR = 1.25, 95% CI:1.1-1.42, P < 0.001), and VD (HR = 1.24, 95% CI: 1.04-1.49, P < 0.05); 2) TV viewing ≥ 4 h/day was correlated with a higher AD risk at intermediate (HR = 1.34, 95% CI: 1.03-1.75, P < 0.001) and high AD genetic risk score (AD-GRS) (HR = 2.18, 95% CI: 1.65-2.87, P < 0.001);3) TV viewing 2-3 h/day [β = (-94.8), 95% CI: (-37.9) -(-151.7), P < 0.01] and TV viewing ≥ 4 h/day [β = (-92.94), 95% CI: (-17.42) -(-168.46), P < 0.05] were correlated with a less hippocampus volume. In addition, a causal effect of TV viewing times was observed on AD analyzed using MR Egger (OR = 5.618, 95%CI:1.502-21.013, P < 0.05). CONCLUSIONS There was a causal effect between TV viewing time and AD analyzed using bidirectional MR, and more TV viewing time exposure was correlated with a higher AD risk. Therefore, it is recommended that people with intermediate and high AD-GRS should control their TV viewing time to be less than 4 h/ day or even less than 1 h/day.
Collapse
Affiliation(s)
- Shiqi Yuan
- Department of Neurology, Guangdong Province, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, 510630, China
| | - Wanyue Li
- Department of Rehabilitation, Guangdong Province, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, 510630, China
| | - Yitong Ling
- Department of Neurology, Guangdong Province, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, 510630, China
| | - Xiaxuan Huang
- Department of Neurology, Guangdong Province, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, 510630, China
| | - Aozi Feng
- Department of Clinical Research, Guangdong Province, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, 510630, China
| | - Shanyuan Tan
- Department of Neurology, Guangdong Province, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, 510630, China
| | - Ningxia He
- Department of Clinical Research, Guangdong Province, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, 510630, China
| | - Li Li
- Department of Clinical Research, Guangdong Province, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, 510630, China
| | - Shuna Li
- Department of Clinical Research, Guangdong Province, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, 510630, China
| | - Anding Xu
- Department of Neurology, Guangdong Province, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, 510630, China.
| | - Jun Lyu
- Department of Clinical Research, Guangdong Province, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, 510630, China.
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
33
|
Hannan AJ. Gene-environment interactions modulating brain disorders: Neurobiological mechanisms and therapeutic applications. Neurobiol Dis 2023; 188:106325. [PMID: 37838008 DOI: 10.1016/j.nbd.2023.106325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2023] Open
Affiliation(s)
- Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
34
|
Oliva CA, Lira M, Jara C, Catenaccio A, Mariqueo TA, Lindsay CB, Bozinovic F, Cavieres G, Inestrosa NC, Tapia-Rojas C, Rivera DS. Long-term social isolation stress exacerbates sex-specific neurodegeneration markers in a natural model of Alzheimer's disease. Front Aging Neurosci 2023; 15:1250342. [PMID: 37810621 PMCID: PMC10557460 DOI: 10.3389/fnagi.2023.1250342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/30/2023] [Indexed: 10/10/2023] Open
Abstract
Social interactions have a significant impact on health in humans and animal models. Social isolation initiates a cascade of stress-related physiological disorders and stands as a significant risk factor for a wide spectrum of morbidity and mortality. Indeed, social isolation stress (SIS) is indicative of cognitive decline and risk to neurodegenerative conditions, including Alzheimer's disease (AD). This study aimed to evaluate the impact of chronic, long-term SIS on the propensity to develop hallmarks of AD in young degus (Octodon degus), a long-lived animal model that mimics sporadic AD naturally. We examined inflammatory factors, bioenergetic status, reactive oxygen species (ROS), oxidative stress, antioxidants, abnormal proteins, tau protein, and amyloid-β (Aβ) levels in the hippocampus of female and male degus that were socially isolated from post-natal and post-weaning until adulthood. Additionally, we explored the effect of re-socialization following chronic isolation on these protein profiles. Our results showed that SIS promotes a pro-inflammatory scenario more severe in males, a response that was partially mitigated by a period of re-socialization. In addition, ATP levels, ROS, and markers of oxidative stress are severely affected in female degus, where a period of re-socialization fails to restore them as it does in males. In females, these effects might be linked to antioxidant enzymes like catalase, which experience a decline across all SIS treatments without recovery during re-socialization. Although in males, a previous enzyme in antioxidant pathway diminishes in all treatments, catalase rebounds during re-socialization. Notably, males have less mature neurons after chronic isolation, whereas phosphorylated tau and all detectable forms of Aβ increased in both sexes, persisting even post re-socialization. Collectively, these findings suggest that long-term SIS may render males more susceptible to inflammatory states, while females are predisposed to oxidative states. In both scenarios, the accumulation of tau and Aβ proteins increase the individual susceptibility to early-onset neurodegenerative conditions such as AD.
Collapse
Affiliation(s)
- Carolina A. Oliva
- Centro para la Transversalización de Género en I+D+i+e, Vicerrectoría de Investigación y Doctorados, Universidad Autónoma de Chile, Santiago, Chile
| | - Matías Lira
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
| | - Claudia Jara
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Alejandra Catenaccio
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
| | - Trinidad A. Mariqueo
- Centro de Investigaciones Médicas, Laboratorio de Neurofarmacología, Escuela de Medicina, Universidad de Talca, Talca, Chile
| | - Carolina B. Lindsay
- Laboratory of Neurosystems, Department of Neuroscience and Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Francisco Bozinovic
- Center of Applied Ecology and Sustainability (CAPES), Departamento de Ecología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Grisel Cavieres
- Center of Applied Ecology and Sustainability (CAPES), Departamento de Ecología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Zoología, Facultad de Ciencias Naturales y Oceanográficas, Universidad de Concepción, Concepción, Chile
| | - Nibaldo C. Inestrosa
- Center of Aging and Regeneration UC (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Cheril Tapia-Rojas
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
| | - Daniela S. Rivera
- GEMA Center for Genomics, Ecology and Environment, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
| |
Collapse
|
35
|
Tan Z, Lin Y, Zhou M, Guo W, Qiu J, Ding L, Wu Z, Xu P, Chen X. Correlation of SV2C rs1423099 single nucleotide polymorphism with sporadic Parkinson's disease in Han population in Southern China. Neurosci Lett 2023; 813:137426. [PMID: 37544580 DOI: 10.1016/j.neulet.2023.137426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/18/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND The synaptic vesicle glycoprotein 2 (SV2) has been implicated in synaptic function throughout the brain. Accumulating evidence investigated that SV2C contributed to dopamine release and the disrupted expression of SV2C was considered to be a unique feature of PD that may facilitate dopaminergic neuron dysfunction. OBJECTIVE This study aimed to examine the relationship between the SV2C rs1423099 single nucleotide polymorphism and sporadic Parkinson's disease (PD) in the Chinese Han population. MATERIALS AND METHODS This study enrolled 351 patients with sporadic PD and 240 normal controls in Chinese Han population. Peripheral blood DNA was extracted by DNA extraction kits and the rs1423099 genotype was analyzed by Agena MassARRAY DNA mass spectrometry. The differences in genotype and allele distribution frequencies between PD patients and control groups were compared using chi-squared tests or Fisher's exact tests. RESULTS No statistical difference was revealed in age and sex distribution between the cases and control groups, and the distribution of genotype and allele frequencies was consistent with the Hardy-Weinberg equilibrium test. In SV2C rs1423099 dominant model, the frequency of the CC/CT genotype was significantly higher in the PD group compared to the control group (OR = 4.065,95% CI: 2.801-10.870, p = 0.002). Nevertheless, in the recessive model, CC or CT/TT genotypes have no statistical difference in the two groups (p = 0.09). Additionally, in allelic analysis, the C allele was investigated to increase the risk of PD (OR = 1.346, 95% CI: 1.036-1.745, p = 0.026); Furthermore, subgroup analysis suggested that those carrying the C allele in the male subgroup were at a higher risk to afflicted with PD (OR = 1.637, 95% CI: 1.147-2.336, p = 0.006). CONCLUSION SV2C rs1423099 single nucleotide polymorphism was associated with sporadic Parkinson's disease in the Chinese Han population, particularly in males.
Collapse
Affiliation(s)
- Zixin Tan
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Yuwan Lin
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Miaomiao Zhou
- Department of Neurology, Shanghai General Hospital, Shanghai 200940, China
| | - Wenyuan Guo
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Jiewen Qiu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Liuyan Ding
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Zhuohua Wu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| | - Pingyi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| | - Xiang Chen
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| |
Collapse
|
36
|
Cozachenco D, Zimmer ER, Lourenco MV. Emerging concepts towards a translational framework in Alzheimer's disease. Neurosci Biobehav Rev 2023; 152:105246. [PMID: 37236385 DOI: 10.1016/j.neubiorev.2023.105246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/20/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023]
Abstract
Over the past decades, significant efforts have been made to understand the precise mechanisms underlying the pathogenesis of Alzheimer's disease (AD), the most common cause of dementia. However, clinical trials targeting AD pathological hallmarks have consistently failed. Refinement of AD conceptualization, modeling, and assessment is key to developing successful therapies. Here, we review critical findings and discuss emerging ideas to integrate molecular mechanisms and clinical approaches in AD. We further propose a refined workflow for animal studies incorporating multimodal biomarkers used in clinical studies - delineating critical paths for drug discovery and translation. Addressing unresolved questions with the proposed conceptual and experimental framework may accelerate the development of effective disease-modifying strategies for AD.
Collapse
Affiliation(s)
- Danielle Cozachenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Eduardo R Zimmer
- Department of Pharmacology, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Graduate Program in Biological Sciences: Biochemistry (PPGBioq), UFRGS, Porto Alegre, RS, Brazil; Pharmacology and Therapeutics (PPGFT), UFRGS, Porto Alegre, RS, Brazil; McGill Centre for Studies in Aging, McGill University, Montreal, Canada; Brain Institute of Rio Grande do Sul, PUCRS, Porto Alegre, Brazil.
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
37
|
Chen Z, Wu B, Li G, Zhou L, Zhang L, Liu J. MAPT rs17649553 T allele is associated with better verbal memory and higher small-world properties in Parkinson's disease. Neurobiol Aging 2023; 129:219-231. [PMID: 37413784 DOI: 10.1016/j.neurobiolaging.2023.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 07/08/2023]
Abstract
Currently, over 90 genetic loci have been found to be associated with Parkinson's disease (PD) in genome-wide association studies, nevertheless, the effects of these genetic variants on the clinical features and brain structure of PD patients are largely unknown. This study investigated the effects of microtubule-associated protein tau (MAPT) rs17649553 (C>T), a genetic variant associated with reduced PD risk, on the clinical manifestations and brain networks of PD patients. We found MAPT rs17649553 T allele was associated with better verbal memory in PD patients. In addition, MAPT rs17649553 significantly shaped the topology of gray matter covariance network and white matter network. Both the network metrics in gray matter covariance network and white matter network were correlated with verbal memory, however, the mediation analysis showed that it was the small-world properties in white matter network that mediated the effects of MAPT rs17649553 on verbal memory. These results suggest that MAPT rs17649553 T allele is associated with higher small-world properties in structural network and better verbal memory in PD.
Collapse
Affiliation(s)
- Zhichun Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bin Wu
- Department of Neurology, Xuchang Central Hospital Affiliated with Henan University of Science and Technology, Henan, China
| | - Guanglu Li
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Liche Zhou
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lina Zhang
- Department of Biostatistics, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
38
|
Gabbert C, König IR, Lüth T, Kasten M, Grünewald A, Klein C, Trinh J. Lifestyle factors and clinical severity of Parkinson's disease. Sci Rep 2023; 13:9537. [PMID: 37308498 DOI: 10.1038/s41598-023-31531-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 03/14/2023] [Indexed: 06/14/2023] Open
Abstract
Genetic factors, environmental factors, and gene-environment interactions have been found to modify PD risk, age at onset (AAO), and disease progression. The objective of this study was to explore the association of coffee drinking, aspirin intake, and smoking, with motor and non-motor symptoms in a cohort of 35,959 American patients with PD from the Fox Insight Study using generalized linear models. Coffee drinkers had fewer problems swallowing but dosage and duration of coffee intake were not associated with motor or non-motor symptoms. Aspirin intake correlated with more tremor (p = 0.0026), problems getting up (p = 0.0185), light-headedness (p = 0.0043), and problems remembering (p = 1 × 10-5). Smoking was directly associated with symptoms: smokers had more problems with drooling (p = 0.0106), swallowing (p = 0.0002), and freezing (p < 1 × 10-5). Additionally, smokers had more possibly mood-related symptoms: unexplained pains (p < 1 × 10-5), problems remembering (p = 0.0001), and feeling sad (p < 1 × 10-5). Confirmatory and longitudinal studies are warranted to investigate the clinical correlation over time.
Collapse
Affiliation(s)
- Carolin Gabbert
- Institute of Neurogenetics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Inke R König
- Institute of Medical Biometry and Statistics, University of Lübeck, Lübeck, Germany
| | - Theresa Lüth
- Institute of Neurogenetics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Meike Kasten
- Institute of Neurogenetics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
| | - Anne Grünewald
- Institute of Neurogenetics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-Sur-Alzette, Luxembourg
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Joanne Trinh
- Institute of Neurogenetics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany.
| |
Collapse
|
39
|
Virolainen SJ, VonHandorf A, Viel KCMF, Weirauch MT, Kottyan LC. Gene-environment interactions and their impact on human health. Genes Immun 2023; 24:1-11. [PMID: 36585519 PMCID: PMC9801363 DOI: 10.1038/s41435-022-00192-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/31/2022]
Abstract
The molecular processes underlying human health and disease are highly complex. Often, genetic and environmental factors contribute to a given disease or phenotype in a non-additive manner, yielding a gene-environment (G × E) interaction. In this work, we broadly review current knowledge on the impact of gene-environment interactions on human health. We first explain the independent impact of genetic variation and the environment. We next detail well-established G × E interactions that impact human health involving environmental toxicants, pollution, viruses, and sex chromosome composition. We conclude with possibilities and challenges for studying G × E interactions.
Collapse
Affiliation(s)
- Samuel J Virolainen
- Division of Human Genetics, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
- Immunology Graduate Program, University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH, 45229, USA
| | - Andrew VonHandorf
- Division of Human Genetics, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Kenyatta C M F Viel
- Division of Human Genetics, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Matthew T Weirauch
- Division of Human Genetics, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA.
- Immunology Graduate Program, University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH, 45229, USA.
- Divisions of Biomedical Informatics and Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH, 45229, USA.
| | - Leah C Kottyan
- Division of Human Genetics, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA.
- Immunology Graduate Program, University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH, 45229, USA.
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., MLC 15012, Cincinnati, OH, 45229, USA.
| |
Collapse
|
40
|
Raval M, Mishra S, Tiwari AK. Epigenetic regulons in Alzheimer's disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 198:185-247. [DOI: 10.1016/bs.pmbts.2023.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
|
41
|
Guo M, Liu W, Luo H, Shao Q, Li Y, Gu Y, Guan Y, Ma W, Chen M, Yang H, Ji X, Liu J. Hypoxic stress accelerates the propagation of pathological alpha-synuclein and degeneration of dopaminergic neurons. CNS Neurosci Ther 2022; 29:544-558. [PMID: 36514210 PMCID: PMC9873519 DOI: 10.1111/cns.14055] [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: 10/02/2022] [Revised: 11/12/2022] [Accepted: 11/27/2022] [Indexed: 12/15/2022] Open
Abstract
AIMS The etiology of Parkinson's disease (PD) is complex and the mechanism is unclear. It has become a top priority to find common factors that induce and affect PD pathology. We explored the key role of hypoxia in promoting the pathological propagation of α-synuclein (α-syn) and the progression of PD. METHODS We performed PD modeling by conducting intracranial stereotaxic surgery in the unilateral striatum of mice. We then measured protein aggregation in vitro. The rotarod and pole tests were employed next to measure the damage of the phenotype. Pathological deposition and autophagy were also observed by immunofluorescence staining and protein levels measured by western blotting. RESULTS We demonstrated that short-term hypoxia activated phosphorylated (p)-α-syn in mice. We confirmed that p-α-syn was more readily formed aggregates than α-syn in vitro. Furthermore, we found that hypoxia promoted the activation and propagation of endogenous α-syn, contributing to the earlier degeneration of dopaminergic neurons in the substantia nigra and the deposition of p-α-syn in our animal model. Finally, autophagy inhibition contributed to the above pathologies. CONCLUSION Hypoxia was shown to accelerate the pathological progression and damage phenotype in PD model mice. The results provided a promising research target for determining common interventions for PD in the future.
Collapse
Affiliation(s)
- Mengyuan Guo
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Weijin Liu
- Department of Neurobiology, School of Basic Medical SciencesCapital Medical UniversityBeijingChina,School of Rehabilitation MedicineCapital Medical UniversityBeijingChina
| | - Hanjiang Luo
- Neuroscience LaboratoryAffiliated Hospital of Guilin Medical UniversityGuangxiChina
| | - Qianqian Shao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Yuning Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Yakun Gu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Yuying Guan
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Wei Ma
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Min Chen
- Neuroscience LaboratoryAffiliated Hospital of Guilin Medical UniversityGuangxiChina
| | - Hui Yang
- Department of Neurobiology, School of Basic Medical SciencesCapital Medical UniversityBeijingChina
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina,Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Jia Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| |
Collapse
|
42
|
Rikos D, Siokas V, Mentis AFA, Aloizou AM, Liampas I, Tsouris Z, Peristeri E, Stamati P, Hadjigeorgiou GM, Dardiotis E. TREM2 R47H variant and risk for Alzheimer's disease: assessment in a Greek population and updated meta-analysis. Int J Neurosci 2022:1-9. [PMID: 36408688 DOI: 10.1080/00207454.2022.2150844] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 02/06/2022] [Accepted: 11/10/2022] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Rare coding variants in TREM2 and their association with the susceptibility towards Alzheimer's disease (AD) were recently studied in various ethnic groups with contradictory results. The T allele of the rs75932628 (p.R47H variant) has shown a positive risk association with AD in several studies; however, neither a study in Greece nor an updated meta-analysis have been conducted. OBJECTIVE To assess the association between TREM2 rs75932628 and late-onset (sporadic) AD in a Greek population, and perform a meta-analysis of current data. MATERIALS AND METHODS The rs75932628 was genotyped in a total of 327 patients with AD and 700 cognitively healthy controls. A systematic search and meta-analyses of studies presenting data regarding rs75932628 in AD cases and controls were also performed. RESULTS Three patients vs. none of the controls were found to carry the heterozygous risk allele of the rs75932628, yielding a significant association (p = 0.032), in the Greek sample. In the meta-analysis, the overall odds ratio (OR) under a fixed-effects model was 2.98 (Confidence Interval (CI):2.52-3.53) showing a significant association of the rs75932628-T allele with AD in the overall dataset, based on data from 27 studies (26200 AD cases and 142084controls). Caucasian population-only studies (n = 16) revealed a similar OR of 2.93 (CI:2.45-3.51), whereas Asian population-only studies (n = 5) had a non-significant OR of 0.84 (CI:0.19-3.74). CONCLUSION The rs75932628 was associated with AD in the Greek sample. Our meta-analysis, covering a total population of over 168,000 people, also showed a significant association of the allele with AD in Caucasian populations.
Collapse
Affiliation(s)
- Dimitrios Rikos
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Larissa, Greece
- Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Vasileios Siokas
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Larissa, Greece
- Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Alexios-Fotios A Mentis
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Larissa, Greece
- Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
- Public Health Laboratories, Hellenic Pasteur Institute, Athens, Greece
| | - Athina-Maria Aloizou
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Larissa, Greece
- Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Ioannis Liampas
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Larissa, Greece
- Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Zisis Tsouris
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Larissa, Greece
- Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Eleni Peristeri
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Larissa, Greece
- Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Polyxeni Stamati
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Larissa, Greece
- Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Georgios M Hadjigeorgiou
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Larissa, Greece
- Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
- Department of Neurology, Medical School, University of Cyprus, Nicosia, Cyprus
| | - Efthimios Dardiotis
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Larissa, Greece
- Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| |
Collapse
|
43
|
Abd El Hamid MM, Omar YM, Shaheen M, Mabrouk MS. Discovering epistasis interactions in Alzheimer's disease using deep learning model. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
44
|
Reagan AM, Christensen KE, Graham LC, Bedwell AA, Eldridge K, Speedy R, Figueiredo LL, Persohn SC, Bottiglieri T, Nho K, Sasner M, Territo PR, Rozen R, Howell GR. The 677C > T variant in methylenetetrahydrofolate reductase causes morphological and functional cerebrovascular deficits in mice. J Cereb Blood Flow Metab 2022; 42:2333-2350. [PMID: 36050860 PMCID: PMC9670012 DOI: 10.1177/0271678x221122644] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 06/30/2022] [Accepted: 07/12/2022] [Indexed: 02/03/2023]
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) particularly Alzheimer's disease and related dementias (ADRDs) are increasing; however, mechanisms driving cerebrovascular decline are poorly understood. Methylenetetrahydrofolate reductase (MTHFR) is a critical enzyme in the folate and methionine cycles. Variants in MTHFR, notably 677 C > T, are associated with dementias, but no mouse model existed to identify mechanisms by which MTHFR677C > T increases risk. Therefore, MODEL-AD created a novel knock-in (KI) strain carrying the Mthfr677C > T allele on the C57BL/6J background (Mthfr677C > T) to characterize morphology and function perturbed by the variant. Consistent with human clinical data, Mthfr677C > T mice have reduced enzyme activity in the liver and elevated plasma homocysteine levels. MTHFR enzyme activity is also reduced in the Mthfr677C > T brain. Mice showed reduced tissue perfusion in numerous brain regions by PET/CT as well as significantly reduced vascular density, pericyte number and increased GFAP-expressing astrocytes in frontal cortex. Electron microscopy revealed cerebrovascular damage including endothelial and pericyte apoptosis, reduced luminal size, and increased astrocyte and microglial presence in the microenvironment. Collectively, these data support a mechanism by which variations in MTHFR perturb cerebrovascular health laying the foundation to incorporate our new Mthfr677C > T mouse model in studies examining genetic susceptibility for cerebrovascular dysfunction in ADRDs.
Collapse
Affiliation(s)
| | - Karen E Christensen
- Departments of Human Genetics and Pediatrics, McGill University,
Research Institute of the Health Center, Montreal, QC, Canada
| | | | - Amanda A Bedwell
- Department of Medicine, Division of Clinical Pharmacology,
Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kierra Eldridge
- Department of Medicine, Division of Clinical Pharmacology,
Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rachael Speedy
- Department of Medicine, Division of Clinical Pharmacology,
Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lucas L Figueiredo
- Department of Medicine, Division of Clinical Pharmacology,
Indiana University School of Medicine, Indianapolis, IN, USA
| | - Scott C Persohn
- Department of Medicine, Division of Clinical Pharmacology,
Indiana University School of Medicine, Indianapolis, IN, USA
| | - Teodoro Bottiglieri
- Center of Metabolomics, Institute of Metabolic Disease, Baylor
Scott & White Research Institute, Dallas, TX, USA
| | - Kwangsik Nho
- Center for Neuroimaging, Indiana Alzheimer’s Disease Research
Center, Department of Radiology and Imaging Sciences, Indiana University School
of Medicine, Indianapolis, IN, USA
| | | | - Paul R Territo
- Department of Medicine, Division of Clinical Pharmacology,
Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rima Rozen
- Departments of Human Genetics and Pediatrics, McGill University,
Research Institute of the Health Center, Montreal, QC, Canada
| | - Gareth R Howell
- The Jackson Laboratory, Bar Harbor, ME, USA
- Graduate School of Biomedical Sciences, Tufts University School
of Medicine, Boston, MA, USA
- Graduate School of Biomedical Sciences and Engineering,
University of Maine, Orono, ME, USA
| |
Collapse
|
45
|
Aborageh M, Krawitz P, Fröhlich H. Genetics in parkinson's disease: From better disease understanding to machine learning based precision medicine. FRONTIERS IN MOLECULAR MEDICINE 2022; 2:933383. [PMID: 39086979 PMCID: PMC11285583 DOI: 10.3389/fmmed.2022.933383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 08/30/2022] [Indexed: 08/02/2024]
Abstract
Parkinson's Disease (PD) is a neurodegenerative disorder with highly heterogeneous phenotypes. Accordingly, it has been challenging to robustly identify genetic factors associated with disease risk, prognosis and therapy response via genome-wide association studies (GWAS). In this review we first provide an overview of existing statistical methods to detect associations between genetic variants and the disease phenotypes in existing PD GWAS. Secondly, we discuss the potential of machine learning approaches to better quantify disease phenotypes and to move beyond disease understanding towards a better-personalized treatment of the disease.
Collapse
Affiliation(s)
- Mohamed Aborageh
- Bonn-Aachen International Center for Information Technology (B-IT), Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Peter Krawitz
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Bonn, Germany
| | - Holger Fröhlich
- Bonn-Aachen International Center for Information Technology (B-IT), Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, Germany
| |
Collapse
|
46
|
Ouellette AR, Hadad N, Deighan A, Robinson L, O'Connell K, Freund A, Churchill GA, Kaczorowski CC. Life-long dietary restrictions have negligible or damaging effects on late-life cognitive performance: A key role for genetics in outcomes. Neurobiol Aging 2022; 118:108-116. [PMID: 35914473 PMCID: PMC9583241 DOI: 10.1016/j.neurobiolaging.2022.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/22/2022] [Accepted: 07/07/2022] [Indexed: 11/21/2022]
Abstract
Several studies report that caloric restriction (CR) or intermittent fasting (IF) can improve cognition, while others report limited or no cognitive benefits. Here, we compare the effects of 20% CR, 40% CR, 1-day IF, and 2-day IF feeding paradigms to ad libitum controls on Y-maze working memory (WM) and contextual fear memory (CFM) in a large population of Diversity Outbred mice that model the genetic diversity of humans. While CR and IF interventions improve lifespan, we observed no enhancement of working memory or CFM in mice on these feeding paradigms, and report 40% CR to be damaging to recall of CFM. Using Quantitative Trait Loci mapping, we identified the gene Slc16a7 to be associated with CFM outcomes in aged mice on lifespan promoting feeding paradigms. Limited utility of dieting and fasting on memory in mice that recapitulate genetic diversity in the human population highlights the need for anti-aging therapeutics that promote cognitive function, with the neuronal monocarboxylate transporter MCT2 encoded by Slc16a7 highlighted as novel target.
Collapse
Affiliation(s)
- Andrew R Ouellette
- The University of Maine, Graduate School of Biomedical Science and Engineering, Orono ME, USA; The Jackson Laboratory, Bar Harbor ME, USA
| | | | | | | | | | - Adam Freund
- Calico Life Sciences LLC, San Francisco CA, USA
| | | | - Catherine C Kaczorowski
- The University of Maine, Graduate School of Biomedical Science and Engineering, Orono ME, USA; The Jackson Laboratory, Bar Harbor ME, USA.
| |
Collapse
|
47
|
Migliore L, Coppedè F. Gene-environment interactions in Alzheimer disease: the emerging role of epigenetics. Nat Rev Neurol 2022; 18:643-660. [PMID: 36180553 DOI: 10.1038/s41582-022-00714-w] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2022] [Indexed: 12/15/2022]
Abstract
With the exception of a few monogenic forms, Alzheimer disease (AD) has a complex aetiology that is likely to involve multiple susceptibility genes and environmental factors. The role of environmental factors is difficult to determine and, until a few years ago, the molecular mechanisms underlying gene-environment (G × E) interactions in AD were largely unknown. Here, we review evidence that has emerged over the past two decades to explain how environmental factors, such as diet, lifestyle, alcohol, smoking and pollutants, might interact with the human genome. In particular, we discuss how various environmental AD risk factors can induce epigenetic modifications of key AD-related genes and pathways and consider how epigenetic mechanisms could contribute to the effects of oxidative stress on AD onset. Studies on early-life exposures are helping to uncover critical time windows of sensitivity to epigenetic influences from environmental factors, thereby laying the foundations for future primary preventative approaches. We conclude that epigenetic modifications need to be considered when assessing G × E interactions in AD.
Collapse
Affiliation(s)
- Lucia Migliore
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy. .,Department of Laboratory Medicine, Pisa University Hospital, Pisa, Italy.
| | - Fabio Coppedè
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| |
Collapse
|
48
|
A Mouse Model of Neurodegeneration Induced by Blade Penetrating Stab Wound to the Hippocampus. BIOLOGY 2022; 11:biology11091365. [PMID: 36138848 PMCID: PMC9495452 DOI: 10.3390/biology11091365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 08/28/2022] [Accepted: 09/14/2022] [Indexed: 12/02/2022]
Abstract
Simple Summary To date, various animal models of traumatic brain injury (TBI) have been developed to investigate the cellular and molecular networks underlying the pathogenesis of neurodegeneration, and to examine the safeness and therapeutic efficacy of drugs. However, the commonly used animal TBI models usually have the weaknesses of low reproducibility and high mortality rates. In the present study, we created a mouse model of cognitive deficits induced by a blade penetrating stab wound to the hippocampus (HBSI). This model will contribute a better understanding of neurodegeneration and accelerate the translation of preclinical research to clinical applications. Abstract Traumatic brain injury (TBI) is closely associated with the later development of neurodegenerative and psychiatric diseases which are still incurable. Although various animal TBI models have been generated, they usually have weaknesses in standardization, survivability and/or reproducibility. In the present study, we investigated whether applying a blade penetrating stab wound to the hippocampus would create an animal model of cognitive deficits. Open-field, Morris water maze and Barnes maze tests were used to evaluate the animal behaviors. The immunofluorescence staining of NeuN, GFAP, IBA1, and TUNEL was conducted to analyze the changes in neurons, astrocytes, and microglia, as well as cell death. Mice with a hippocampal blade stab injury (HBSI) displayed the activation of microglia and astrocytes, inflammation, neuronal apoptosis, and deficits in spatial learning and memory. These findings suggest that HBSI is an easy approach to generate a reliable in vivo model of TBI to capture hemorrhage, neuroinflammation, reactive gliosis, and neural death, as well as cognitive deficits observed in human patients.
Collapse
|
49
|
Body mass index, genetic susceptibility, and Alzheimer's disease: a longitudinal study based on 475,813 participants from the UK Biobank. J Transl Med 2022; 20:417. [PMID: 36085169 PMCID: PMC9463868 DOI: 10.1186/s12967-022-03621-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/01/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The association between body mass index (BMI) and Alzheimer's disease (AD) remains controversial. Genetic and environmental factors are now considered contributors to AD risk. However, little is known about the potential interaction between genetic risk and BMI on AD risk. OBJECTIVE To study the causal relationship between BMI and AD, and the potential interaction between AD genetic risk and BMI on AD risk. METHODS AND RESULTS Using the UK Biobank database, 475,813 participants were selected for an average follow-up time of more than 10 years. MAIN FINDINGS 1) there was a nonlinear relationship between BMI and AD risk in participants aged 60 years or older (p for non-linear < 0.001), but not in participants aged 37-59 years (p for non-linear = 0.717) using restricted cubic splines; 2) for participants aged 60 years and older, compared with the BMI (23-30 kg/m2) group, the BMI (< 23 kg/m2) group was associated with a higher AD risk (HR = 1.585; 95% CI 1.304-1.928, p < 0.001) and the BMI (> 30 kg/m2) group was associated with a lower AD risk (HR = 0.741; 95% CI 0.618-0.888, p < 0.01) analyzed using the Cox proportional risk model; 3) participants with a combination of high AD genetic risk score (AD-GRS) and BMI (< 23 kg/m2) were associated with the highest AD risk (HR = 3.034; 95% CI 2.057-4.477, p < 0.001). In addition, compared with the BMI (< 23 kg/m2), the higher BMI was associated with a lower risk of AD in participants with the same intermediate or high AD-GRS; 4) there was a reverse causality between BMI and AD when analyzed using bidirectional Mendelian randomization (MR). CONCLUSION There was a reverse causality between BMI and AD analyzed using MR. For participants aged 60 years and older, the higher BMI was associated with a lower risk of AD in participants with the same intermediate or high AD genetic risk. BMI (23-30 kg/m2) may be a potential intervention for AD.
Collapse
|
50
|
Lin ZH, Liu Y, Xue NJ, Zheng R, Yan YQ, Wang ZX, Li YL, Ying CZ, Song Z, Tian J, Pu JL, Zhang BR. Quercetin Protects against MPP +/MPTP-Induced Dopaminergic Neuron Death in Parkinson's Disease by Inhibiting Ferroptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7769355. [PMID: 36105483 PMCID: PMC9467739 DOI: 10.1155/2022/7769355] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 12/17/2022]
Abstract
Ferroptosis, a novel form of regulated cell death, is caused by accumulation of lipid peroxides and excessive iron deposition. This process has been linked to the death of dopaminergic neurons in substantia nigra compacta (SNc) of Parkinson's disease (PD) patients. Quercetin (QCT), a natural flavonoid, has multiple pharmacological activities. However, it has not been established whether QCT can protect against dopaminergic neuron death by inhibiting ferroptosis. In this study, we investigated the potential antiferroptotic effects of QCT in cellular models established using specific ferroptosis inducers (Erastin and RSL-3) and MPP+. The effects were also explored using MPTP-induced PD mouse models. The cell counting kit-8 (CCK-8) assay was performed to assess cell viability. Variations in mitochondrial morphology were evaluated by transmission electron microscopy (TEM) while the mitochondrial membrane potential, mass, and ROS were measured by fluorescent probes. Lipid peroxidation levels were assayed through measurement of lipid ROS, MDA, GSH, and SOD levels. The effects of QCT on MPTP-induced behavioral disorders were examined by rotarod and open field tests. In vitro and in vivo, QCT significantly inhibited ferroptosis by activating the nuclear factor erythroid 2-related factor 2 (Nrf2) protein. Additionally, QCT ameliorated motor behavioral impairments and protected against the loss of dopaminergic neurons in MPTP-induced PD models. Interestingly, Nrf2 knockdown alleviated the protective effects of QCT against ferroptosis. In conclusion, these results demonstrate that ferroptosis is involved in MPP+/MPTP-induced PD, and QCT inhibits ferroptosis by activating the Nrf2 protein. Therefore, QCT is a potential agent for preventing the loss of dopaminergic neurons by targeting ferroptosis.
Collapse
Affiliation(s)
- Zhi-Hao Lin
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Yi Liu
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Nai-Jia Xue
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Ran Zheng
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Yi-Qun Yan
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Zhong-Xuan Wang
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Yao-Lin Li
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Chang-Zhou Ying
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Zhe Song
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Jun Tian
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Jia-Li Pu
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Bao-Rong Zhang
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| |
Collapse
|