1
|
Barak Levitt JA, Ziv M. Dementia Risk in Psoriasis Patients Treated with Biologics: A Propensity Score-matched Population-based Cohort Study. Acta Derm Venereol 2025; 105:adv43243. [PMID: 40364478 DOI: 10.2340/actadv.v105.43243] [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: 02/23/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Translational research and animal models suggest that psoriasis treatments may have neuroprotective effects and reduce dementia risk. This study evaluates the association between biologic therapies for psoriasis and dementia incidence. A retrospective cohort included patients aged 65 or older with psoriasis, divided into 2 groups: those receiving biologic therapy following systemic treatment and those on systemic treatment alone. Patients with prior dementia were excluded. Dementia diagnosis was assessed at least 12 months after biologic initiation. Propensity score matching yielded 1,766 patients (883 per group). Biologic therapy was associated with a 53% reduced dementia risk (hazard ratio 0.47, 95% confidence interval 0.323-0.699), supported by a multivariate Cox model (adjusted hazard ratio 0.52, 95% confidence interval 0.392-0.699). These findings suggest that biologic therapies targeting tumour necrosis factor-alpha, interleukin-17, and interleukin-23 may reduce the risk of dementia, even after adjusting for age and other confounders.
Collapse
Affiliation(s)
- Jen A Barak Levitt
- Department of Dermatology, Emek Medical Center, Afula, Israel; Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa, Israel.
| | - Michael Ziv
- Department of Dermatology, Emek Medical Center, Afula, Israel; Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa, Israel
| |
Collapse
|
2
|
Ding H, Lyu C, Karjadi C, Sunderaraman P, Young CB, Mormino EC, Low S, Devine S, Gifford K, Au R, Lin H. Association of the digital clock drawing test with amyloid and tau PET biomarkers in low age risk adults. Sci Rep 2025; 15:11104. [PMID: 40169870 PMCID: PMC11961701 DOI: 10.1038/s41598-025-95852-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 03/21/2025] [Indexed: 04/03/2025] Open
Abstract
Although brain amyloid and tau deposition measured by PET scans are established as biomarkers of Alzheimer's disease (AD), they can emerge decades before symptoms are detectable on traditional neuropsychological (NP) tests. There is a pressing need for early AD detection tools that are more accessible, cost-effective, and non-invasive. The digital clock drawing test (dCDT), a digital version of the clock drawing test, has emerged as a promising cognitive assessment tool that takes minutes to administer and can reveal clinical symptoms earlier than paper-pencil NP tests. This study explored the association between 53 dCDT measures and amyloid and tau PET biomarkers using data from 87 low age risk participants in the Framingham Heart Study. Our findings revealed a significant association between a dCDT measure related to spatial reasoning function and global amyloid burden (P < 0.05), and 4 dCDT measures correlated with tau accumulation after adjusting for multiple comparisons. Notably, the combination of demographic variables and a composite dCDT score achieved a mean area under the receiver operating characteristics curve of 0.86 in detecting amyloid positivity. These results highlight the potential of dCDT measures as effective predictors of amyloid and tau pathology in preclinical AD.
Collapse
Affiliation(s)
- Huitong Ding
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Chenglin Lyu
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Cody Karjadi
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Preeti Sunderaraman
- The Framingham Heart Study, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Christina B Young
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Spencer Low
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Sherral Devine
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Katherine Gifford
- Department of Neurology, Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rhoda Au
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
- The Framingham Heart Study, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA.
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
- Slone Epidemiology Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
3
|
Dybing KM, Vetter CJ, Dempsey DA, Chaudhuri S, Saykin AJ, Risacher SL. Traumatic Brain Injury and Alzheimer's Disease Biomarkers: A Systematic Review of Findings from Amyloid and Tau Positron Emission Tomography. J Neurotrauma 2025; 42:333-348. [PMID: 39639808 PMCID: PMC11971548 DOI: 10.1089/neu.2024.0055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
Traumatic brain injury (TBI) has been discussed as a risk factor for Alzheimer's disease (AD) due to its association with AD risk and earlier cognitive symptom onset. However, the mechanisms behind this relationship are unclear. Some studies have suggested TBI may increase pathological protein deposition in an AD-like pattern; others have failed to find such associations. This review covers literature that uses positron emission tomography (PET) of β-amyloid (Aβ) and/or tau to examine individuals with a history of TBI who are at increased risk for AD due to age. A comprehensive literature search was conducted on January 9, 2023, and 26 resulting citations met inclusion criteria. Common methodological concerns included small samples, limited clinical detail about participants' TBI, recall bias due to reliance on self-reported TBI, and an inability to establish causation. For both Aβ and tau, results were widespread but inconsistent. The regions that showed the most compelling evidence for increased Aβ deposition were the cingulate gyrus and cuneus/precuneus. Evidence for elevated tau was strongest in the medial temporal lobe, entorhinal cortex, precuneus, and frontal, temporal, parietal, and occipital lobes. However, conflicting findings across most regions in both Aβ- and tau-PET studies indicate the critical need for future work in expanded samples and with greater clinical detail to offer a clearer picture of the relationship between TBI and protein deposition in older individuals at risk for AD.
Collapse
Affiliation(s)
- Kaitlyn M. Dybing
- Address correspondence to: Kaitlyn M. Dybing, BS, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 West 16th Street, Suite 4100, Indianapolis, IN 46202, USA,
| | - Cecelia J. Vetter
- Ruth Lilly Medical Library, Indiana University School of Medicine, Indianapolis, USA
| | | | | | - Andrew J. Saykin
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Shannon L. Risacher
- Address correspondence to: Shannon L. Risacher, PhD, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 West 16th Street, Suite 4100, Indianapolis, IN 46202, USA,
| |
Collapse
|
4
|
Parul, Singh A, Shukla S. Novel techniques for early diagnosis and monitoring of Alzheimer's disease. Expert Rev Neurother 2025; 25:29-42. [PMID: 39435792 DOI: 10.1080/14737175.2024.2415985] [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: 05/30/2024] [Accepted: 10/09/2024] [Indexed: 10/23/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) is the most common neurodegenerative disorder, which is characterized by a progressive loss of cognitive functions. The high prevalence, chronicity, and multimorbidity are very common in AD, which significantly impair the quality of life and functioning of patients. Early detection and accurate diagnosis of Alzheimer's disease (AD) can stop the illness from progressing thereby postponing its symptoms. Therefore, for the early diagnosis and monitoring of AD, more sensitive, noninvasive, straightforward, and affordable screening tools are needed. AREAS COVERED This review summarizes the importance of early detection methods and novel techniques for Alzheimer's disease diagnosis that can be used by healthcare professionals. EXPERT OPINION Early diagnosis assists the patient and caregivers to understand the problem establishing reasonable goals and making future plans together. Early diagnosis techniques not only help in monitoring disease progression but also provide crucial information for the development of novel therapeutic targets. Researchers can plan to potentially alleviate symptoms or slow down the progression of Alzheimer's disease by identifying early molecular changes and targeting altered pathways.
Collapse
Affiliation(s)
- Parul
- Division of Neuroscience and Ageing biology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Animesh Singh
- Division of Neuroscience and Ageing biology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shubha Shukla
- Division of Neuroscience and Ageing biology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
5
|
Hayden MR, Tyagi N. Sodium Thiosulfate: An Innovative Multi-Target Repurposed Treatment Strategy for Late-Onset Alzheimer's Disease. Pharmaceuticals (Basel) 2024; 17:1741. [PMID: 39770582 PMCID: PMC11676759 DOI: 10.3390/ph17121741] [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: 12/06/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Late-onset Alzheimer's disease (LOAD) is a chronic, multifactorial, and progressive neurodegenerative disease that associates with aging and is highly prevalent in our older population (≥65 years of age). This hypothesis generating this narrative review will examine the important role for the use of sodium thiosulfate (STS) as a possible multi-targeting treatment option for LOAD. Sulfur is widely available in our environment and is responsible for forming organosulfur compounds that are known to be associated with a wide range of biological activities in the brain. STS is known to have (i) antioxidant and (ii) anti-inflammatory properties; (iii) chelation properties for calcium and the pro-oxidative cation metals such as iron and copper; (iv) donor properties for hydrogen sulfide production; (v) possible restorative properties for brain endothelial-cell-derived bioavailable nitric oxide. Thus, it becomes apparent that STS has the potential for neuroprotection and neuromodulation and may allow for an attenuation of the progressive nature of neurodegeneration and impaired cognition in LOAD. STS has been successfully used to prevent cisplatin oxidative-stress-induced ototoxicity in the treatment of head and neck and solid cancers, cyanide and arsenic poisoning, and fungal skin diseases. Most recently, intravenous STS has become part of the treatment plan for calciphylaxis globally due to vascular calcification and ischemia-induced skin necrosis and ulceration. Side effects have been minimal with reports of metabolic acidosis and increased anion gap; as with any drug treatment, there is also the possibility of allergic reactions, possible long-term osteoporosis from animal studies to date, and minor side-effects of nausea, headache, and rhinorrhea if infused too rapidly. While STS poorly penetrates the intact blood-brain barrier(s) (BBBs), it could readily penetrate BBBs that are dysfunctional and disrupted to deliver its neuroprotective and neuromodulating effects in addition to its ability to penetrate the blood-cerebrospinal fluid barrier of the choroid plexus. Novel strategies such as the future use of nano-technology may be helpful in allowing an increased entry of STS into the brain.
Collapse
Affiliation(s)
- Melvin R. Hayden
- Department of Internal Medicine, Endocrinology Diabetes and Metabolism, Diabetes and Cardiovascular Disease Center, University of Missouri School of Medicine, One Hospital Drive, Columbia, MO 65211, USA
| | - Neetu Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA;
| |
Collapse
|
6
|
Tenchov R, Sasso JM, Zhou QA. Alzheimer's Disease: Exploring the Landscape of Cognitive Decline. ACS Chem Neurosci 2024; 15:3800-3827. [PMID: 39392435 PMCID: PMC11587518 DOI: 10.1021/acschemneuro.4c00339] [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: 06/03/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired daily functioning. The pathology of AD is marked by the accumulation of amyloid beta plaques and tau protein tangles in the brain, along with neuroinflammation and synaptic dysfunction. Genetic factors, such as mutations in APP, PSEN1, and PSEN2 genes, as well as the APOE ε4 allele, contribute to increased risk of acquiring AD. Currently available treatments provide symptomatic relief but do not halt disease progression. Research efforts are focused on developing disease-modifying therapies that target the underlying pathological mechanisms of AD. Advances in identification and validation of reliable biomarkers for AD hold great promise for enhancing early diagnosis, monitoring disease progression, and assessing treatment response in clinical practice in effort to alleviate the burden of this devastating disease. In this paper, we analyze data from the CAS Content Collection to summarize the research progress in Alzheimer's disease. We examine the publication landscape in effort to provide insights into current knowledge advances and developments. We also review the most discussed and emerging concepts and assess the strategies to combat the disease. We explore the genetic risk factors, pharmacological targets, and comorbid diseases. Finally, we inspect clinical applications of products against AD with their development pipelines and efforts for drug repurposing. The objective of this review is to provide a broad overview of the evolving landscape of current knowledge regarding AD, to outline challenges, and to evaluate growth opportunities to further efforts in combating the disease.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a division of the American Chemical
Society, Columbus Ohio 43210, United States
| | - Janet M. Sasso
- CAS, a division of the American Chemical
Society, Columbus Ohio 43210, United States
| | | |
Collapse
|
7
|
Deza‐Lougovski YI, Weiss LM, Horton HM, Sun A, Borbye‐Lorenzen N, Skogstrand K, Holmgaard S, Andersen‐Ranberg K, Lundmark VP, Börsch‐Supan A, Börsch‐Supan M, Rieckmann A. Circulating apoE4 protein levels from dried blood spots predict cognitive function in a large population-based survey setting. Alzheimers Dement 2024; 20:7613-7623. [PMID: 39234633 PMCID: PMC11567846 DOI: 10.1002/alz.14224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024]
Abstract
INTRODUCTION The apolipoprotein E (APOE) ε4 allele carries risk for cognitive impairment, but whether the level of circulating apoE4 protein in carriers affects cognition is unclear, as is how health and lifestyle impact circulating apoE4 levels. METHODS We assayed apoE4 protein levels in dried blood spots of 12,532 adults aged 50+. Regression analyses tested the likelihood of cognitive impairment between groups and within those with detected apoE4 protein. Predictors of circulating apoE4 were assessed. RESULTS We detected protein binding that indicates the presence of an APOE ε4 allele in 28.4% of this group. This group was more likely to have cognitive impairment, and this risk increases with age. However, higher apoE4 levels were associated with less likelihood of cognitive impairment within this group. Antihypertensive medication predicted apoE4 protein levels. DISCUSSION The apoE4 isoform is associated with a deficient protein and worse cognition. This association is modulated by the level of circulating apoE4 protein in ε4 carriers. HIGHLIGHTS An assay to quantify apoE4 levels from dried blood spot samples was applied. The apoE4 protein was detected as specific binding at ≥30,000 pg/mL in 28.4% of samples. Having the apoE4 protein was associated with worse cognitive performance. Higher apoE4 protein levels in those who have it were associated with better cognition. Cardiovascular factors influenced levels of apoE4 protein.
Collapse
Affiliation(s)
- Yacila I. Deza‐Lougovski
- Institute of Psychology, University of the Bundeswehr MünchenNeubibergGermany
- Max Planck Institute for Social Law and Social PolicyMunichGermany
| | - Luzia M. Weiss
- Max Planck Institute for Social Law and Social PolicyMunichGermany
| | - Hannah M. Horton
- Max Planck Institute for Social Law and Social PolicyMunichGermany
- Munich Research Institute for the Economics of Aging and SHARE Analyses (MEA)MunichGermany
| | - Aijing Sun
- Max Planck Institute for Social Law and Social PolicyMunichGermany
- Munich Research Institute for the Economics of Aging and SHARE Analyses (MEA)MunichGermany
| | - Nis Borbye‐Lorenzen
- Department for Congenital DisordersCenter for Neonatal Screening, Statens Serum InstitutCopenhagenDenmark
| | - Kristin Skogstrand
- Department for Congenital DisordersCenter for Neonatal Screening, Statens Serum InstitutCopenhagenDenmark
| | - Solveig Holmgaard
- Department for Congenital DisordersCenter for Neonatal Screening, Statens Serum InstitutCopenhagenDenmark
| | - Karen Andersen‐Ranberg
- Department of Public Health Epidemiology, Biostatistics and BiodemographyUniversity of Southern DenmarkOdenseDenmark
- Department of Clinical Research Geriatric Research UnitUniversity of Southern DenmarkOdenseDenmark
| | - Vania Panes Lundmark
- Department of Integrative Medical BiologyUmeå UniversityUmeåSweden
- Umeå Center for Functional Brain ImagingUmeå UniversityUmeåSweden
| | - Axel Börsch‐Supan
- Max Planck Institute for Social Law and Social PolicyMunichGermany
- Munich Research Institute for the Economics of Aging and SHARE Analyses (MEA)MunichGermany
| | - Martina Börsch‐Supan
- Munich Research Institute for the Economics of Aging and SHARE Analyses (MEA)MunichGermany
- Survey of Health, Ageing and Retirement in Europe (SHARE Biomarker Project)MunichGermany
| | - Anna Rieckmann
- Institute of Psychology, University of the Bundeswehr MünchenNeubibergGermany
- Max Planck Institute for Social Law and Social PolicyMunichGermany
| |
Collapse
|
8
|
Villareal JAB, Bathe T, Hery GP, Phillips JL, Tsering W, Prokop S. Deterioration of neuroimmune homeostasis in Alzheimer's Disease patients who survive a COVID-19 infection. J Neuroinflammation 2024; 21:202. [PMID: 39154174 PMCID: PMC11330027 DOI: 10.1186/s12974-024-03196-3] [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: 05/21/2024] [Accepted: 08/06/2024] [Indexed: 08/19/2024] Open
Abstract
Growing evidence has implicated systemic infection as a significant risk factor for the development and advancement of Alzheimer's disease (AD). With the emergence of SARS-CoV-2 (COVID-19) and the resultant pandemic, many individuals from the same aging population vulnerable to AD suffered a severe systemic infection with potentially unidentified long-term consequences for survivors. To study the impact of COVID-19 survival on the brain's intrinsic immune system in a population also suffering from AD, we profiled post-mortem brain tissue from patients in the UF Neuromedicine Human Brain and Tissue Bank with a diagnosis of AD who survived a COVID-19 infection (COVID-AD) and contrasted our findings with AD patients who did not experience a COVID-19 infection, including a group of brain donors who passed away before arrival of SARS-CoV-2 in the United States. We assessed disease-relevant protein pathology and microglial and astrocytic markers by quantitative immunohistochemistry and supplemented these data with whole tissue gene expression analysis performed on the NanoString nCounter® platform. COVID-AD patients showed slightly elevated Aβ burden in the entorhinal, fusiform, and inferior temporal cortices compared to non-COVID-AD patients, while tau pathology burden did not differ between groups. Analysis of microglia revealed a significant loss of microglial homeostasis as well as exacerbated microgliosis in COVID-AD patients compared to non-COVID-AD patients in a brain region-dependent manner. Furthermore, COVID-AD patients showed reduced cortical astrocyte numbers, independent of functional subtype. Transcriptomic analysis supported these histological findings and, in addition, identified a dysregulation of oligodendrocyte and myelination pathways in the hippocampus of COVID-AD patients. In summary, our data demonstrate a profound impact of COVID-19 infection on neuroimmune and glial pathways in AD patients persisting for months post-infection, highlighting the importance of peripheral to central neuroimmune crosstalk in neurodegenerative diseases.
Collapse
Affiliation(s)
- Jonathan A B Villareal
- Department of Pathology, Immunology & Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
| | - Tim Bathe
- Department of Pathology, Immunology & Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Gabriela P Hery
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32608, USA
| | - Jennifer L Phillips
- Department of Pathology, Immunology & Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
| | - Wangchen Tsering
- Department of Pathology, Immunology & Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Stefan Prokop
- Department of Pathology, Immunology & Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA.
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA.
- Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32608, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
9
|
Wahl D, Clayton ZS. Peripheral vascular dysfunction and the aging brain. Aging (Albany NY) 2024; 16:9280-9302. [PMID: 38805248 PMCID: PMC11164523 DOI: 10.18632/aging.205877] [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: 01/04/2024] [Accepted: 03/28/2024] [Indexed: 05/29/2024]
Abstract
Aging is the greatest non-modifiable risk factor for most diseases, including cardiovascular diseases (CVD), which remain the leading cause of mortality worldwide. Robust evidence indicates that CVD are a strong determinant for reduced brain health and all-cause dementia with advancing age. CVD are also closely linked with peripheral and cerebral vascular dysfunction, common contributors to the development and progression of all types of dementia, that are largely driven by excessive levels of oxidative stress (e.g., reactive oxygen species [ROS]). Emerging evidence suggests that several fundamental aging mechanisms (e.g., "hallmarks" of aging), including chronic low-grade inflammation, mitochondrial dysfunction, cellular senescence and deregulated nutrient sensing contribute to excessive ROS production and are common to both peripheral and cerebral vascular dysfunction. Therefore, targeting these mechanisms to reduce ROS-related oxidative stress and improve peripheral and/or cerebral vascular function may be a promising strategy to reduce dementia risk with aging. Investigating how certain lifestyle strategies (e.g., aerobic exercise and diet modulation) and/or select pharmacological agents (natural and synthetic) intersect with aging "hallmarks" to promote peripheral and/or cerebral vascular health represent a viable option for reducing dementia risk with aging. Therefore, the primary purpose of this review is to explore mechanistic links among peripheral vascular dysfunction, cerebral vascular dysfunction, and reduced brain health with aging. Such insight and assessments of non-invasive measures of peripheral and cerebral vascular health with aging might provide a new approach for assessing dementia risk in older adults.
Collapse
Affiliation(s)
- Devin Wahl
- Department of Health and Exercise Science and Center for Healthy Aging, Colorado State University, Fort Collins, CO 80523, USA
| | - Zachary S. Clayton
- University of Colorado Anschutz Medical Campus, Department of Medicine, Division of Geriatric Medicine, Aurora, CO 80045, USA
| |
Collapse
|
10
|
De Sousa Rodrigues ME, Bolen ML, Blackmer-Raynolds L, Schwartz N, Chang J, Tansey MG, Sampson TR. Diet-induced metabolic and immune impairments are sex-specifically modulated by soluble TNF signaling in the 5xFAD mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.28.582516. [PMID: 38464096 PMCID: PMC10925304 DOI: 10.1101/2024.02.28.582516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Emerging evidence indicates that high-fat, high carbohydrate diet (HFHC) impacts central pathological features of Alzheimer's disease (AD) across both human incidences and animal models. However, the mechanisms underlying this association are poorly understood. Here, we identify compartment-specific metabolic and inflammatory dysregulations that are induced by HFHC diet in the 5xFAD mouse model of AD pathology. We observe that both male and female 5xFAD mice display exacerbated adiposity, cholesterolemia, and dysregulated insulin signaling. Independent of biological sex, HFHC diet also resulted in altered inflammatory cytokine profiles across the gastrointestinal, circulating, and central nervous systems (CNS) compartments demonstrating region-specific impacts of metabolic inflammation. In male mice, we note that HFHC triggered increases in amyloid beta, an observation not seen in female mice. Interestingly, inhibiting the inflammatory cytokine, soluble tumor necrosis factor (TNF) with the brain-permeant soluble TNF inhibitor XPro1595 was able to restore aspects of HFHC-induced metabolic inflammation, but only in male mice. Targeted transcriptomics of CNS regions revealed that inhibition of soluble TNF was sufficient to alter expression of hippocampal and cortical genes associated with beneficial immune and metabolic responses. Collectively, these results suggest that HFHC diet impairs metabolic and inflammatory pathways in an AD-relevant genotype and that soluble TNF has sex-dependent roles in modulating these pathways across anatomical compartments. Modulation of energy homeostasis and inflammation may provide new therapeutic avenues for AD.
Collapse
Affiliation(s)
| | - MacKenzie L. Bolen
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, The University of Florida College of Medicine, Gainesville, Florida, USA
| | | | - Noah Schwartz
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia USA
| | - Jianjun Chang
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia USA
| | - Malú Gámez Tansey
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, The University of Florida College of Medicine, Gainesville, Florida, USA
| | | |
Collapse
|
11
|
Cichońska D, Mazuś M, Kusiak A. Recent Aspects of Periodontitis and Alzheimer's Disease-A Narrative Review. Int J Mol Sci 2024; 25:2612. [PMID: 38473858 DOI: 10.3390/ijms25052612] [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: 01/16/2024] [Revised: 02/13/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Periodontitis is an inflammatory condition affecting the supporting structures of the teeth. Periodontal conditions may increase the susceptibility of individuals to various systemic illnesses, including Alzheimer's disease. Alzheimer's disease is a neurodegenerative condition characterized by a gradual onset and progressive deterioration, making it the primary cause of dementia, although the exact cause of the disease remains elusive. Both Alzheimer's disease and periodontitis share risk factors and clinical studies comparing the associations and occurrence of periodontitis among individuals with Alzheimer's disease have suggested a potential correlation between these conditions. Brains of individuals with Alzheimer's disease have substantiated the existence of microorganisms related to periodontitis, especially Porphyromonas gingivalis, which produces neurotoxic gingipains and may present the capability to breach the blood-brain barrier. Treponema denticola may induce tau hyperphosphorylation and lead to neuronal apoptosis. Lipopolysaccharides-components of bacterial cell membranes and mediators of inflammation-also have an impact on brain function. Further research could unveil therapeutic approaches targeting periodontal pathogens to potentially alleviate AD progression.
Collapse
Affiliation(s)
- Dominika Cichońska
- Department of Periodontology and Oral Mucosa Diseases, Medical University of Gdańsk, Orzeszkowej 18 St. 18, 80-208 Gdańsk, Poland
| | - Magda Mazuś
- Student Research Group of the Department of Periodontology and Oral Mucosa Diseases, Medical University of Gdańsk, Orzeszkowej 18 St. 18, 80-208 Gdańsk, Poland
| | - Aida Kusiak
- Department of Periodontology and Oral Mucosa Diseases, Medical University of Gdańsk, Orzeszkowej 18 St. 18, 80-208 Gdańsk, Poland
| |
Collapse
|
12
|
Zuliani G, Zuin M, Romagnoli T, Polastri M, Cervellati C, Brombo G. Acetyl-cholinesterase-inhibitors reconsidered. A narrative review of post-marketing studies on Alzheimer's disease. Aging Clin Exp Res 2024; 36:23. [PMID: 38321321 PMCID: PMC10847178 DOI: 10.1007/s40520-023-02675-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 12/18/2023] [Indexed: 02/08/2024]
Abstract
The real efficacy of Acetyl-cholinesterase-inhibitors (AChEI) has been questioned. In this narrative review we evaluated their effect on cognitive decline, measured by Mini Mental State Examination (MMSE), and on total mortality rates in patients with Alzheimer's disease (AD) recruited into post-marketing open/non-randomized/retrospective studies. In AD patients treated with AChEI, the mean MMSE loss ranged from 0.2 to 1.37 points/years, compared with 1.07-3.4 points/years in non-treated patients. Six studies also reported data about survival; a reduction in total mortality relative risk between 27% and 42% was observed, over a period of 2-8 years. The type of studies and the use of MMSE to assess cognitive decline, may have introduced several biases. However, the clinical effects of AChEI seem to be of the same order of magnitude as the drugs currently used in most common chronic disorders, as regards progression of the disease and total mortality. In the absence of long-term randomized trials on "standard" unselected AD outpatients, open/retrospective studies and health databases represent the best available evidence on the possible effect of AChEI in the real-word setting. Our data support the clinical benefit of AChEI in older patients affected by AD.
Collapse
Affiliation(s)
- Giovanni Zuliani
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy
| | - Marco Zuin
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy
| | - Tommaso Romagnoli
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy
| | - Michele Polastri
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy
| | - Carlo Cervellati
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy.
| | - Gloria Brombo
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy
| |
Collapse
|
13
|
Kreutzer AG, Parrocha CMT, Haerianardakani S, Guaglianone G, Nguyen JT, Diab MN, Yong W, Perez-Rosendahl M, Head E, Nowick JS. Antibodies Raised Against an Aβ Oligomer Mimic Recognize Pathological Features in Alzheimer's Disease and Associated Amyloid-Disease Brain Tissue. ACS CENTRAL SCIENCE 2024; 10:104-121. [PMID: 38292607 PMCID: PMC10823522 DOI: 10.1021/acscentsci.3c00592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 11/27/2023] [Accepted: 11/27/2023] [Indexed: 02/01/2024]
Abstract
Antibodies that target the β-amyloid peptide (Aβ) and its associated assemblies are important tools in Alzheimer's disease research and have emerged as promising Alzheimer's disease therapies. This paper reports the creation and characterization of a triangular Aβ trimer mimic composed of Aβ17-36 β-hairpins and the generation and study of polyclonal antibodies raised against the Aβ trimer mimic. The Aβ trimer mimic is covalently stabilized by three disulfide bonds at the corners of the triangular trimer to create a homogeneous oligomer. Structural, biophysical, and cell-based studies demonstrate that the Aβ trimer mimic shares characteristics with oligomers of full-length Aβ. X-ray crystallography elucidates the structure of the trimer and reveals that four copies of the trimer assemble to form a dodecamer. SDS-PAGE, size exclusion chromatography, and dynamic light scattering reveal that the trimer also forms higher-order assemblies in solution. Cell-based toxicity assays show that the trimer elicits LDH release, decreases ATP levels, and activates caspase-3/7 mediated apoptosis. Immunostaining studies on brain slices from people who lived with Alzheimer's disease and people who lived with Down syndrome reveal that the polyclonal antibodies raised against the Aβ trimer mimic recognize pathological features including different types of Aβ plaques and cerebral amyloid angiopathy.
Collapse
Affiliation(s)
- Adam G. Kreutzer
- Department
of Chemistry, University of California Irvine, Irvine, California 92697, United States
| | - Chelsea Marie T. Parrocha
- Department
of Pharmaceutical Sciences, University of
California Irvine, Irvine, California 92697, United States
| | - Sepehr Haerianardakani
- Department
of Chemistry, University of California Irvine, Irvine, California 92697, United States
| | - Gretchen Guaglianone
- Department
of Chemistry, University of California Irvine, Irvine, California 92697, United States
| | - Jennifer T. Nguyen
- Department
of Pharmaceutical Sciences, University of
California Irvine, Irvine, California 92697, United States
| | - Michelle N. Diab
- Department
of Chemistry, University of California Irvine, Irvine, California 92697, United States
| | - William Yong
- Department
of Pathology and Laboratory Medicine, University
of California Irvine, Irvine, California 92697, United States
| | - Mari Perez-Rosendahl
- Department
of Pathology and Laboratory Medicine, University
of California Irvine, Irvine, California 92697, United States
| | - Elizabeth Head
- Department
of Pathology and Laboratory Medicine, University
of California Irvine, Irvine, California 92697, United States
| | - James S. Nowick
- Department
of Chemistry, University of California Irvine, Irvine, California 92697, United States
- Department
of Pharmaceutical Sciences, University of
California Irvine, Irvine, California 92697, United States
| |
Collapse
|
14
|
Shigemizu D, Fukunaga K, Yamakawa A, Suganuma M, Fujita K, Kimura T, Watanabe K, Mushiroda T, Sakurai T, Niida S, Ozaki K. The HLA-DRB1*09:01-DQB1*03:03 haplotype is associated with the risk for late-onset Alzheimer's disease in APOE
ε
4-negative Japanese adults. NPJ AGING 2024; 10:3. [PMID: 38167405 PMCID: PMC10761915 DOI: 10.1038/s41514-023-00131-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/22/2023] [Indexed: 01/05/2024]
Abstract
Late-onset Alzheimer's disease (LOAD) is the most common cause of dementia among those older than 65 years. The onset of LOAD is influenced by neuroinflammation. The human leukocyte antigen (HLA) system is involved in regulating inflammatory responses. Numerous HLA alleles and their haplotypes have shown varying associations with LOAD in diverse populations, yet their impact on the Japanese population remains to be elucidated. Here, we conducted a comprehensive investigation into the associations between LOAD and HLA alleles within the Japanese population. Using whole-genome sequencing (WGS) data from 303 LOAD patients and 1717 cognitively normal (CN) controls, we identified four-digit HLA class I alleles (A, B, and C) and class II alleles (DRB1, DQB1, and DPB1). We found a significant association between the HLA-DRB1*09:01-DQB1*03:03 haplotype and LOAD risk in APOEε 4-negative samples (odds ratio = 1.81, 95% confidence interval = 1.38-2.38, P = 2.03× 10 − 5 ). These alleles not only showed distinctive frequencies specific to East Asians but demonstrated a high degree of linkage disequilibrium in APOEε 4-negative samples (r2 = 0.88). Because HLA class II molecules interact with T-cell receptors (TCRs), we explored potential disparities in the diversities of TCR α chain (TRA) and β chain (TRB) repertoires between APOEε 4-negative LOAD and CN samples. Lower diversity of TRA repertoires was associated with LOAD in APOEε 4-negative samples, irrespective of the HLA DRB1*09:01-DQB1*03:03 haplotype. Our study enhances the understanding of the etiology of LOAD in the Japanese population and provides new insights into the underlying mechanisms of its pathogenesis.
Collapse
Affiliation(s)
- Daichi Shigemizu
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan.
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan.
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8551, Japan.
| | - Koya Fukunaga
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan
| | - Akiko Yamakawa
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| | - Mutsumi Suganuma
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| | - Kosuke Fujita
- Department of Prevention and Care Science, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
- Japan Society for the Promotion of Science, Tokyo, 102-0083, Japan
| | - Tetsuaki Kimura
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| | - Ken Watanabe
- NCGG Biobank, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| | - Taisei Mushiroda
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan
| | - Takashi Sakurai
- Department of Prevention and Care Science, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| | - Shumpei Niida
- Core Facility Administration, Research Institute, National Center for Geriatrics and Gerontology, Aichi, 474-8511, Japan
| | - Kouichi Ozaki
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan.
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan.
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8551, Japan.
| |
Collapse
|
15
|
Tu R, Xia J. Stroke and Vascular Cognitive Impairment: The Role of Intestinal Microbiota Metabolite TMAO. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:102-121. [PMID: 36740795 DOI: 10.2174/1871527322666230203140805] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/18/2022] [Accepted: 12/12/2022] [Indexed: 02/07/2023]
Abstract
The gut microbiome interacts with the brain bidirectionally through the microbiome-gutbrain axis, which plays a key role in regulating various nervous system pathophysiological processes. Trimethylamine N-oxide (TMAO) is produced by choline metabolism through intestinal microorganisms, which can cross the blood-brain barrier to act on the central nervous system. Previous studies have shown that elevated plasma TMAO concentrations increase the risk of major adverse cardiovascular events, but there are few studies on TMAO in cerebrovascular disease and vascular cognitive impairment. This review summarized a decade of research on the impact of TMAO on stroke and related cognitive impairment, with particular attention to the effects on vascular cognitive disorders. We demonstrated that TMAO has a marked impact on the occurrence, development, and prognosis of stroke by regulating cholesterol metabolism, foam cell formation, platelet hyperresponsiveness and thrombosis, and promoting inflammation and oxidative stress. TMAO can also influence the cognitive impairment caused by Alzheimer's disease and Parkinson's disease via inducing abnormal aggregation of key proteins, affecting inflammation and thrombosis. However, although clinical studies have confirmed the association between the microbiome-gut-brain axis and vascular cognitive impairment (cerebral small vessel disease and post-stroke cognitive impairment), the molecular mechanism of TMAO has not been clarified, and TMAO precursors seem to play the opposite role in the process of poststroke cognitive impairment. In addition, several studies have also reported the possible neuroprotective effects of TMAO. Existing therapies for these diseases targeted to regulate intestinal flora and its metabolites have shown good efficacy. TMAO is probably a new target for early prediction and treatment of stroke and vascular cognitive impairment.
Collapse
Affiliation(s)
- Ruxin Tu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jian Xia
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
- Human Clinical Research Center for Cerebrovascular Disease, Changsha, China
| |
Collapse
|
16
|
Adiga D, Eswaran S, Sriharikrishnaa S, Khan NG, Prasada Kabekkodu S, Kumar D. Epigenetics of Alzheimer’s Disease: Past, Present and Future. ENZYMATIC TARGETS FOR DRUG DISCOVERY AGAINST ALZHEIMER'S DISEASE 2023:27-72. [DOI: 10.2174/9789815136142123010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Alzheimer’s disease (AD) exemplifies a looming epidemic lacking effective
treatment and manifests with the accumulation of neurofibrillary tangles, amyloid-β
plaques, neuroinflammation, behavioral changes, and acute cognitive impairments. It is
a complex, multifactorial disorder that arises from the intricate interaction between
environment and genetic factors, restrained via epigenetic machinery. Though the
research progress has improved the understanding of clinical manifestations and
disease advancement, the causal mechanism of detrimental consequences remains
undefined. Despite the substantial improvement in recent diagnostic modalities, it is
challenging to distinguish AD from other forms of dementia. Accurate diagnosis is a
major glitch in AD as it banks on the symptoms and clinical criteria. Several studies are
underway in exploring novel and reliable biomarkers for AD. In this direction,
epigenetic alterations have transpired as key modulators in AD pathogenesis with the
impeding inferences for the management of this neurological disorder. The present
chapter aims to discuss the significance of epigenetic modifications reported in the
pathophysiology of AD such as DNA methylation, hydroxy-methylation, methylation
of mtDNA, histone modifications, and noncoding RNAs. Additionally, the chapter also
describes the possible therapeutic avenues that target epigenetic modifications in AD.
Collapse
Affiliation(s)
- Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy
of Higher Education (MAHE), Manipal – 576104, Karnataka, India
| | - Sangavi Eswaran
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy
of Higher Education (MAHE), Manipal – 576104, Karnataka, India
| | - S. Sriharikrishnaa
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy
of Higher Education (MAHE), Manipal – 576104, Karnataka, India
| | - Nadeem G. Khan
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy
of Higher Education (MAHE), Manipal – 576104, Karnataka, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy
of Higher Education (MAHE), Manipal – 576104, Karnataka, India
| | - Dileep Kumar
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth
(Deemed to be University), Erandwane, Pune – 411038, Maharashtra, India
| |
Collapse
|
17
|
Dybing KM, Vetter CJ, Dempsey DA, Chaudhuri S, Saykin AJ, Risacher SL. Traumatic brain injury and Alzheimer's Disease biomarkers: A systematic review of findings from amyloid and tau positron emission tomography (PET). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.30.23298528. [PMID: 38077068 PMCID: PMC10705648 DOI: 10.1101/2023.11.30.23298528] [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: 12/21/2023]
Abstract
Traumatic brain injury (TBI) has been discussed as a risk factor for Alzheimer's disease (AD) due to its association with dementia risk and earlier cognitive symptom onset. However, the mechanisms behind this relationship are unclear. Some studies have suggested TBI may increase pathological protein deposition in an AD-like pattern; others have failed to find such associations. This review covers literature that uses positron emission tomography (PET) of amyloid-β and/or tau to examine subjects with history of TBI who are at risk for AD due to advanced age. A comprehensive literature search was conducted on January 9, 2023, and 24 resulting citations met inclusion criteria. Common methodological concerns included small samples, limited clinical detail about subjects' TBI, recall bias due to reliance on self-reported TBI, and an inability to establish causation. For both amyloid and tau, results were widespread but inconsistent. The regions which showed the most compelling evidence for increased amyloid deposition were the cingulate gyrus, cuneus/precuneus, and parietal lobe. Evidence for increased tau was strongest in the medial temporal lobe, entorhinal cortex, precuneus, and frontal, temporal, parietal, and occipital lobes. However, conflicting findings across most regions of interest in both amyloid- and tau-PET studies indicate the critical need for future work in expanded samples and with greater clinical detail to offer a clearer picture of the relationship between TBI and protein deposition in older subjects at risk for AD.
Collapse
Affiliation(s)
- Kaitlyn M. Dybing
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Cecelia J. Vetter
- Ruth Lilly Medical Library, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Desarae A. Dempsey
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Soumilee Chaudhuri
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Shannon L. Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| |
Collapse
|
18
|
Griffin P, Apostolova L, Dickerson BC, Rabinovici G, Salloway S, Raghuram S, Brandt K, Hall S, Masdeu J, Carrillo MC, Hammers D. Developments in understanding early onset Alzheimer's disease. Alzheimers Dement 2023; 19 Suppl 9:S126-S131. [PMID: 37390354 PMCID: PMC10756918 DOI: 10.1002/alz.13353] [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: 02/13/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 07/02/2023]
Abstract
On September 25 and 26, 2021, the Alzheimer's Association hosted the first meeting focused on people with early-onset Alzheimer's disease (EOAD)-sometimes referred to as younger onset Alzheimer's disease (AD). Though a diagnosis of AD can be devastating at any age, those with a younger onset-defined as symptoms developing prior to 65 years of age-face unique challenges. EOAD occurs when people are in the prime of their lives, often with multiple responsibilities including careers, community activities, and raising children and caring for older family members. These challenges warrant special consideration and study, yet people with EOAD are often excluded from AD research because of their atypical age of onset. To help fill this gap, we designed and launched the Longitudinal Early-Onset Alzheimer's Disease Study (LEADS) to enroll and follow 500 people with EOAD from > 15 sites in the United States, which the National Institute on Aging funded in 2018. The September 2021 meeting was designed to inform people with EOAD and their family members and caregivers about the latest research on the biology of EOAD, treatments in the pipeline, practical considerations about legal and financial arrangements for families, and the support networks available to them. More than 217 registrants attended.
Collapse
Affiliation(s)
- Percy Griffin
- Alzheimer’s Association, Medical & Scientific Relations, Chicago, Illinois, USA
| | - Liana Apostolova
- Departments of Neurology, Radiology, Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Bradford C. Dickerson
- Frontotemporal Disorders Unit & Alzheimer’s Disease Research Center, Departments of Neurology and Psychiatry, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts, USA
| | - Gil Rabinovici
- Departments of Neurology, Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Stephen Salloway
- Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Srilatha Raghuram
- Alzheimer’s Association, Medical & Scientific Relations, Chicago, Illinois, USA
| | - Katie Brandt
- Frontotemporal Disorders Unit & Alzheimer’s Disease Research Center, Departments of Neurology and Psychiatry, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen Hall
- Alzheimer’s Association, Medical & Scientific Relations, Chicago, Illinois, USA
| | - Joseph Masdeu
- Department of Neurology, Houston Methodist, Houston, Texas, USA
| | - Maria C. Carrillo
- Alzheimer’s Association, Medical & Scientific Relations, Chicago, Illinois, USA
| | - Dustin Hammers
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
19
|
Alatrany AS, Khan W, Hussain AJ, Mustafina J, Al-Jumeily D. Transfer Learning for Classification of Alzheimer's Disease Based on Genome Wide Data. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:2700-2711. [PMID: 37018274 DOI: 10.1109/tcbb.2022.3233869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Alzheimer's disease (AD) is a type of brain disorder that is regarded as a degenerative disease because the corresponding symptoms aggravate with the time progression. Single nucleotide polymorphisms (SNPs) have been identified as relevant biomarkers for this condition. This study aims to identify SNPs biomarkers associated with the AD in order to perform a reliable classification of AD. In contrast to existing related works, we utilize deep transfer learning with varying experimental analysis for reliable classification of AD. For this purpose, the convolutional neural networks (CNN) are firstly trained over the genome-wide association studies (GWAS) dataset requested from the AD neuroimaging initiative. We then employ the deep transfer learning for further training of our CNN (as base model) over a different AD GWAS dataset, to extract the final set of features. The extracted features are then fed into Support Vector Machine for classification of AD. Detailed experiments are performed using multiple datasets and varying experimental configurations. The statistical outcomes indicate an accuracy of 89% which is a significant improvement when benchmarked with existing related works.
Collapse
|
20
|
Tureson KN, Beam CR, Medina LD, Segal-Gidan F, D'Orazio LM, Chui H, Torres M, Varma R, Ringman JM. Use of the Spanish English Neuropsychological Assessment Scale in older adult Latines and those at risk for autosomal dominant Alzheimer's disease. J Clin Exp Neuropsychol 2023; 45:553-569. [PMID: 37990912 PMCID: PMC10926998 DOI: 10.1080/13803395.2023.2284971] [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: 05/07/2023] [Accepted: 11/12/2023] [Indexed: 11/23/2023]
Abstract
OBJECTIVE The Spanish English Neuropsychological Assessment Scale (SENAS) is a cognitive battery with English and Spanish versions for use with persons for whom either language is predominant. Few studies have examined its utility outside the normative sample. The current study examined SENAS performance in samples of older adult Latines and Latines with or at risk for autosomal dominant Alzheimer's disease (ADAD) mutations. METHOD The SENAS was administered to 202 older adults from the Los Angeles Latino Eye Study (LALES) and 29 adults with (carriers) or without (non-carriers) mutations causing ADAD. We examined associations between SENAS, age, education, and language (LALES) and between SENAS, estimated years from familial age of dementia diagnosis, education, language, and acculturation (ADAD). Partial correlations were used to examine differences in correlational strength between estimated years from familial age of dementia diagnosis and SENAS scores among ADAD carriers compared to chronological age and SENAS in the LALES sample. Exploratory t-tests were performed to examine SENAS performance differences between ADAD carriers and non-carriers. RESULTS In an older adult sample (LALES), increased age correlated with worse verbal delayed recall; English fluency and higher education correlated with better naming and visuospatial subtest performance. Among ADAD carriers, verbal and nonverbal delayed recall and object naming subtest performance worsened as they approached their familial age of dementia diagnosis. English fluency and higher U.S.-acculturation were related to better SENAS performance among carriers and non-carriers. Tests of verbal delayed recall and object naming best distinguished ADAD carriers from their familial non-carrier counterparts. CONCLUSIONS Verbal delayed recall and object naming measures appear to be most sensitive to age-related changes in older adult samples and mutation-related changes in distinguishing ADAD carriers from non-carriers. Future research should examine the sensitivity of SENAS in other samples, such as larger samples of symptomatic ADAD carriers and other AD subtypes.
Collapse
Affiliation(s)
- Kayla N Tureson
- Alzheimer's Disease Research Center, Department of Neurology, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Christopher R Beam
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
- USC Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Luis D Medina
- Department of Psychology, University of Houston, Houston, TX, USA
| | - Freddi Segal-Gidan
- Alzheimer's Disease Research Center, Department of Neurology, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | - Lina M D'Orazio
- Alzheimer's Disease Research Center, Department of Neurology, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | - Helena Chui
- Alzheimer's Disease Research Center, Department of Neurology, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | - Mina Torres
- CHA Hollywood Presbyterian Medical Center, Southern California Eye Institute, Los Angeles, CA, USA
| | - Rohit Varma
- CHA Hollywood Presbyterian Medical Center, Southern California Eye Institute, Los Angeles, CA, USA
| | - John M Ringman
- Alzheimer's Disease Research Center, Department of Neurology, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
21
|
Damestani NL, Jacoby J, Yadav SM, Lovely AE, Michael A, Terpstra M, Eshghi M, Rashid B, Cruchaga C, Salat DH, Juttukonda MR. Associations between age, sex, APOE genotype, and regional vascular physiology in typically aging adults. Neuroimage 2023; 275:120167. [PMID: 37187365 PMCID: PMC10339339 DOI: 10.1016/j.neuroimage.2023.120167] [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: 01/05/2023] [Revised: 05/02/2023] [Accepted: 05/11/2023] [Indexed: 05/17/2023] Open
Abstract
Altered blood flow in the human brain is characteristic of typical aging. However, numerous factors contribute to inter-individual variation in patterns of blood flow throughout the lifespan. To better understand the mechanisms behind such variation, we studied how sex and APOE genotype, a primary genetic risk factor for Alzheimer's disease (AD), influence associations between age and brain perfusion measures. We conducted a cross-sectional study of 562 participants from the Human Connectome Project - Aging (36 to >90 years of age). We found widespread associations between age and vascular parameters, where increasing age was associated with regional decreases in cerebral blood flow (CBF) and increases in arterial transit time (ATT). When grouped by sex and APOE genotype, interactions between group and age demonstrated that females had relatively greater CBF and lower ATT compared to males. Females carrying the APOEε4 allele showed the strongest association between CBF decline and ATT incline with age. This demonstrates that sex and genetic risk for AD modulate age-associated patterns of cerebral perfusion measures.
Collapse
Affiliation(s)
- Nikou L Damestani
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Harvard Medical School, Boston, MA, USA.
| | - John Jacoby
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Shrikanth M Yadav
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Allison E Lovely
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Aurea Michael
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Melissa Terpstra
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, USA
| | | | - Barnaly Rashid
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO, USA
| | - David H Salat
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Harvard Medical School, Boston, MA, USA; Neuroimaging Research for Veterans Center, VA Boston Healthcare System, Boston MA, USA
| | - Meher R Juttukonda
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Shigemizu D, Akiyama S, Suganuma M, Furutani M, Yamakawa A, Nakano Y, Ozaki K, Niida S. Classification and deep-learning-based prediction of Alzheimer disease subtypes by using genomic data. Transl Psychiatry 2023; 13:232. [PMID: 37386009 DOI: 10.1038/s41398-023-02531-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/01/2023] Open
Abstract
Late-onset Alzheimer's disease (LOAD) is the most common multifactorial neurodegenerative disease among elderly people. LOAD is heterogeneous, and the symptoms vary among patients. Genome-wide association studies (GWAS) have identified genetic risk factors for LOAD but not for LOAD subtypes. Here, we examined the genetic architecture of LOAD based on Japanese GWAS data from 1947 patients and 2192 cognitively normal controls in a discovery cohort and 847 patients and 2298 controls in an independent validation cohort. Two distinct groups of LOAD patients were identified. One was characterized by major risk genes for developing LOAD (APOC1 and APOC1P1) and immune-related genes (RELB and CBLC). The other was characterized by genes associated with kidney disorders (AXDND1, FBP1, and MIR2278). Subsequent analysis of albumin and hemoglobin values from routine blood test results suggested that impaired kidney function could lead to LOAD pathogenesis. We developed a prediction model for LOAD subtypes using a deep neural network, which achieved an accuracy of 0.694 (2870/4137) in the discovery cohort and 0.687 (2162/3145) in the validation cohort. These findings provide new insights into the pathogenic mechanisms of LOAD.
Collapse
Affiliation(s)
- Daichi Shigemizu
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan.
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan.
| | - Shintaro Akiyama
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| | - Mutsumi Suganuma
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| | - Motoki Furutani
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan
| | - Akiko Yamakawa
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan
| | - Kouichi Ozaki
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan
| | - Shumpei Niida
- Core Facility Administration, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| |
Collapse
|
23
|
Dohoney RA, Joseph JA, Baysah C, Thomas AG, Siwakoti A, Ball TD, Kumar S. "Common-Precursor" Protein Mimetic Approach to Rescue Aβ Aggregation-Mediated Alzheimer's Phenotypes. ACS Chem Biol 2023. [PMID: 37367833 DOI: 10.1021/acschembio.3c00120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Abberent protein-protein interactions (aPPIs) are associated with an array of pathological conditions, which make them important therapeutic targets. The aPPIs are mediated via specific chemical interactions that spread over a large and hydrophobic surface. Therefore, ligands that can complement the surface topography and chemical fingerprints could manipulate aPPIs. Oligopyridylamides (OPs) are synthetic protein mimetics that have been shown to manipulate aPPIs. However, the previous OP library used to disrupt these aPPIs was moderate in number (∼30 OPs) with very limited chemical diversity. The onus is on the laborious and time-consuming synthetic pathways with multiple chromatography steps. We have developed a novel chromatography-free technique to synthesize a highly diverse chemical library of OPs using a "common-precursor" approach. We significantly expanded the chemical diversity of OPs using a chromatography-free high-yielding method. To validate our novel approach, we have synthesized an OP with identical chemical diversity to a pre-existing OP-based potent inhibitor of Aβ aggregation, a process central to Alzheimer's disease (AD). The newly synthesized OP ligand (RD242) was very potent in inhibiting Aβ aggregation and rescuing AD phenotypes in an in vivo model. Moreover, RD242 was very effective in rescuing AD phenotypes in a post-disease onset AD model. We envision that our "common-precursor" synthetic approach will have tremendous potential as it is expandable for other oligoamide scaffolds to enhance affinity for disease-relevant targets.
Collapse
Affiliation(s)
- Ryan A Dohoney
- The Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
| | - Johnson A Joseph
- The Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
| | - Charles Baysah
- The Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
| | - Alexandra G Thomas
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
- The Department of Biological Sciences, University of Denver, Denver, Colorado 80210, United States
| | - Apshara Siwakoti
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
- The Department of Biological Sciences, University of Denver, Denver, Colorado 80210, United States
| | - Tyler D Ball
- The Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
| | - Sunil Kumar
- The Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
| |
Collapse
|
24
|
Han ZZ, Fleet A, Larrieu D. Can accelerated ageing models inform us on age-related tauopathies? Aging Cell 2023; 22:e13830. [PMID: 37013265 PMCID: PMC10186612 DOI: 10.1111/acel.13830] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 04/05/2023] Open
Abstract
Ageing is the greatest risk factor of late-onset neurodegenerative diseases. In the realm of sporadic tauopathies, modelling the process of biological ageing in experimental animals forms the foundation of searching for the molecular origin of pathogenic tau and developing potential therapeutic interventions. Although prior research into transgenic tau models offers valuable lessons for studying how tau mutations and overexpression can drive tau pathologies, the underlying mechanisms by which ageing leads to abnormal tau accumulation remains poorly understood. Mutations associated with human progeroid syndromes have been proposed to be able to mimic an aged environment in animal models. Here, we summarise recent attempts in modelling ageing in relation to tauopathies using animal models that carry mutations associated with human progeroid syndromes, or genetic elements unrelated to human progeroid syndromes, or have exceptional natural lifespans, or a remarkable resistance to ageing-related disorders.
Collapse
Affiliation(s)
- Zhuang Zhuang Han
- Department of PharmacologyUniversity of CambridgeTennis Ct RdCambridgeCB2 1PDUK
| | - Alex Fleet
- Department of PharmacologyUniversity of CambridgeTennis Ct RdCambridgeCB2 1PDUK
| | - Delphine Larrieu
- Department of PharmacologyUniversity of CambridgeTennis Ct RdCambridgeCB2 1PDUK
| |
Collapse
|
25
|
Kommaddi RP, Verma A, Muniz-Terrera G, Tiwari V, Chithanathan K, Diwakar L, Gowaikar R, Karunakaran S, Malo PK, Graff-Radford NR, Day GS, Laske C, Vöglein J, Nübling G, Ikeuchi T, Kasuga K, Ravindranath V. Sex difference in evolution of cognitive decline: studies on mouse model and the Dominantly Inherited Alzheimer Network cohort. Transl Psychiatry 2023; 13:123. [PMID: 37045867 PMCID: PMC10097702 DOI: 10.1038/s41398-023-02411-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/15/2023] [Accepted: 03/24/2023] [Indexed: 04/14/2023] Open
Abstract
Women carry a higher burden of Alzheimer's disease (AD) compared to men, which is not accounted entirely by differences in lifespan. To identify the mechanisms underlying this effect, we investigated sex-specific differences in the progression of familial AD in humans and in APPswe/PS1ΔE9 mice. Activity dependent protein translation and associative learning and memory deficits were examined in APPswe/PS1ΔE9 mice and wild-type mice. As a human comparator group, progression of cognitive dysfunction was assessed in mutation carriers and non-carriers from DIAN (Dominantly Inherited Alzheimer Network) cohort. Female APPswe/PS1ΔE9 mice did not show recall deficits after contextual fear conditioning until 8 months of age. Further, activity dependent protein translation and Akt1-mTOR signaling at the synapse were impaired in male but not in female mice until 8 months of age. Ovariectomized APPswe/PS1ΔE9 mice displayed recall deficits at 4 months of age and these were sustained until 8 months of age. Moreover, activity dependent protein translation was also impaired in 4 months old ovariectomized APPswe/PS1ΔE9 mice compared with sham female APPswe/PS1ΔE9 mice. Progression of memory impairment differed between men and women in the DIAN cohort as analyzed using linear mixed effects model, wherein men showed steeper cognitive decline irrespective of the age of entry in the study, while women showed significantly greater performance and slower decline in immediate recall (LOGIMEM) and delayed recall (MEMUNITS) than men. However, when the performance of men and women in several cognitive tasks (such as Wechsler's logical memory) are compared with the estimated year from expected symptom onset (EYO) we found no significant differences between men and women. We conclude that in familial AD patients and mouse models, females are protected, and the onset of disease is delayed as long as estrogen levels are intact.
Collapse
Affiliation(s)
- Reddy Peera Kommaddi
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India.
| | - Aditi Verma
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, India
| | - Graciela Muniz-Terrera
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland, UK
- The Department of Social Medicine, Ohio University, Athens, OH, 45701, USA
| | - Vivek Tiwari
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India
| | | | - Latha Diwakar
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India
| | - Ruturaj Gowaikar
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, India
| | - Smitha Karunakaran
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, India
| | - Palash Kumar Malo
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India
| | - Neill R Graff-Radford
- Department of Neurology, Mayo Clinic Florida, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA
| | - Gregory S Day
- Department of Neurology, Mayo Clinic Florida, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA
| | - Christoph Laske
- German Center for Neurodegenerative Diseases, Munich, Germany
- Section for Dementia Research, Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Jonathan Vöglein
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Georg Nübling
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Center for Bioresources, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8585, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Center for Bioresources, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8585, Japan
| | - Vijayalakshmi Ravindranath
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
26
|
Beason-Held LL, Kerley CI, Chaganti S, Moghekar A, Thambisetty M, Ferrucci L, Resnick SM, Landman BA. Health Conditions Associated with Alzheimer's Disease and Vascular Dementia. Ann Neurol 2023; 93:805-818. [PMID: 36571386 PMCID: PMC11973975 DOI: 10.1002/ana.26584] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 12/15/2022] [Accepted: 12/17/2022] [Indexed: 12/27/2022]
Abstract
OBJECTIVE We examined medical records to determine health conditions associated with dementia at varied intervals prior to dementia diagnosis in participants from the Baltimore Longitudinal Study of Aging (BLSA). METHODS Data were available for 347 Alzheimer's disease (AD), 76 vascular dementia (VaD), and 811 control participants without dementia. Logistic regressions were performed associating International Classification of Diseases, 9th Revision (ICD-9) health codes with dementia status across all time points, at 5 and 1 year(s) prior to dementia diagnosis, and at the year of diagnosis, controlling for age, sex, and follow-up length of the medical record. RESULTS In AD, the earliest and most consistent associations across all time points included depression, erectile dysfunction, gait abnormalities, hearing loss, and nervous and musculoskeletal symptoms. Cardiomegaly, urinary incontinence, non-epithelial skin cancer, and pneumonia were not significant until 1 year before dementia diagnosis. In VaD, the earliest and most consistent associations across all time points included abnormal electrocardiogram (EKG), cardiac dysrhythmias, cerebrovascular disease, non-epithelial skin cancer, depression, and hearing loss. Atrial fibrillation, occlusion of cerebral arteries, essential tremor, and abnormal reflexes were not significant until 1 year before dementia diagnosis. INTERPRETATION These findings suggest that some health conditions are associated with future dementia beginning at least 5 years before dementia diagnosis and are consistently seen over time, while others only reach significance closer to the date of diagnosis. These results also show that there are both shared and distinctive health conditions associated with AD and VaD. These results reinforce the need for medical intervention and treatment to lessen the impact of health comorbidities in the aging population. ANN NEUROL 2023;93:805-818.
Collapse
Affiliation(s)
- Lori L Beason-Held
- National Institute on Aging Intramural Research Program, Baltimore, Maryland, USA
| | - Cailey I Kerley
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Shikha Chaganti
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Madhav Thambisetty
- National Institute on Aging Intramural Research Program, Baltimore, Maryland, USA
| | - Luigi Ferrucci
- National Institute on Aging Intramural Research Program, Baltimore, Maryland, USA
| | - Susan M Resnick
- National Institute on Aging Intramural Research Program, Baltimore, Maryland, USA
| | - Bennett A Landman
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
27
|
Mathew A, Balaji E V, Pai SRK, Kishore A, Pai V, Pemmireddy R, K S C. Current Drug Targets in Alzheimer's Associated Memory Impairment: A Comprehensive Review. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:255-275. [PMID: 35366787 DOI: 10.2174/1871527321666220401124719] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/17/2021] [Accepted: 01/19/2022] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia among geriatrics. It is a progressive, degenerative neurologic disorder that causes memory and cognition loss. The accumulation of amyloid fibrils and neurofibrillary tangles in the brain of AD patients is a distinguishing feature of the disease. Therefore, most of the current therapeutic goals are targeting inhibition of beta-amyloid synthesis and aggregation as well as tau phosphorylation and aggregation. There is also a loss of the cholinergic neurons in the basal forebrain, and first-generation therapeutic agents were primarily focused on compensating for this loss of neurons. However, cholinesterase inhibitors can only alleviate cognitive symptoms of AD and cannot reduce the progression of the disease. Understanding the molecular and cellular changes associated with AD pathology has advanced significantly in recent decades. The etiology of AD is complex, with a substantial portion of sporadic AD emerging from unknown reasons and a lesser proportion of early-onset familial AD (FAD) caused by a mutation in several genes, such as the amyloid precursor protein (APP), presenilin 1 (PS1), and presenilin 2 (PS2) genes. Hence, efforts are being made to discover novel strategies for these targets for AD therapy. A new generation of AChE and BChE inhibitors is currently being explored and evaluated in human clinical trials for AD symptomatic treatment. Other approaches for slowing the progression of AD include serotonergic modulation, H3 receptor antagonism, phosphodiesterase, COX-2, and MAO-B inhibition. The present review provides an insight into the possible therapeutic strategies and their molecular mechanisms, enlightening the perception of classical and future treatment approaches.
Collapse
Affiliation(s)
- Anna Mathew
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Vignesh Balaji E
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Sreedhara Ranganath K Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Anoop Kishore
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Vasudev Pai
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Ramadevi Pemmireddy
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Chandrashekar K S
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| |
Collapse
|
28
|
Agarwal A, Mishra B, Gupta A, Srivastava MVP, Basheer A, Sharma J, Vishnu VY. Importance of high-quality evidence regarding the use of Bacopa monnieri in dementia. Front Aging Neurosci 2023; 15:1134775. [PMID: 36936504 PMCID: PMC10014812 DOI: 10.3389/fnagi.2023.1134775] [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: 12/30/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
Background Bacopa monnieri (BM), a commonly used herb, has shown neuroprotective effects in animal and in vitro studies; but human studies on patients with Alzheimer's Disease (AD) have been inconclusive. Further high-quality trials are required to conclusively state the utility of BM in AD and other neurodegenerative dementias. Methods In the present study, we did a narrative review of the current challenges in designing clinical trials of BM in dementia and their evidence-based recommendations. Results Many facets of the BM trials need improvement, especially effect size and sample size estimation. Current assessment and outcomes measures need a more holistic approach and newer scales for diagnosing and monitoring prodromal AD. The stringent guidelines in CONSORT and STROBE are often considered difficult to implement for clinical trials in ayurvedic medications like BM. However, adherence to these guidelines will undoubtedly improve the quality of evidence and go a long way in assessing whether BM is efficacious in treating AD/prodromal AD patients and other neurodegenerative dementias. Conclusion Future studies on BM should implement more randomized controlled trials (RCTs) with an appropriate sample size of accurately diagnosed AD/prodromal AD patients, administering a recommended dosage of BM and for a pre-specified time calculated to achieve adequate power for the study. Researchers should also develop and validate more sensitive cognitive scales, especially for prodromal AD. BM should be evaluated in accordance with the same rigorous standards as conventional drugs to generate the best quality evidence.
Collapse
Affiliation(s)
- Ayush Agarwal
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Biswamohan Mishra
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Anu Gupta
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | | | - Aneesh Basheer
- DM Wayanad Institute of Medical Sciences (DM WIMS), Wayanad, India
| | - Jyoti Sharma
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Venugopalan Y. Vishnu
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
- *Correspondence: Venugopalan Y. Vishnu,
| |
Collapse
|
29
|
Akhtar A, Gupta SM, Dwivedi S, Kumar D, Shaikh MF, Negi A. Preclinical Models for Alzheimer's Disease: Past, Present, and Future Approaches. ACS OMEGA 2022; 7:47504-47517. [PMID: 36591205 PMCID: PMC9798399 DOI: 10.1021/acsomega.2c05609] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/22/2022] [Indexed: 05/13/2023]
Abstract
A robust preclinical disease model is a primary requirement to understand the underlying mechanisms, signaling pathways, and drug screening for human diseases. Although various preclinical models are available for several diseases, clinical models for Alzheimer's disease (AD) remain underdeveloped and inaccurate. The pathophysiology of AD mainly includes the presence of amyloid plaques and neurofibrillary tangles (NFT). Furthermore, neuroinflammation and free radical generation also contribute to AD. Currently, there is a wide gap in scientific approaches to preventing AD progression. Most of the available drugs are limited to symptomatic relief and improve deteriorating cognitive functions. To mimic the pathogenesis of human AD, animal models like 3XTg-AD and 5XFAD are the primarily used mice models in AD therapeutics. Animal models for AD include intracerebroventricular-streptozotocin (ICV-STZ), amyloid beta-induced, colchicine-induced, etc., focusing on parameters such as cognitive decline and dementia. Unfortunately, the translational rate of the potential drug candidates in clinical trials is poor due to limitations in imitating human AD pathology in animal models. Therefore, the available preclinical models possess a gap in AD modeling. This paper presents an outline that critically assesses the applicability and limitations of the current approaches in disease modeling for AD. Also, we attempted to provide key suggestions for the best-fit model to evaluate potential therapies, which might improve therapy translation from preclinical studies to patients with AD.
Collapse
Affiliation(s)
- Ansab Akhtar
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Shraddha M. Gupta
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Shubham Dwivedi
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Devendra Kumar
- Faculty
of Pharmacy, DIT University, Uttarakhand, Dehradun 248009, India
| | - Mohd. Farooq Shaikh
- Neuropharmacology
Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 47500, Malaysia
| | - Arvind Negi
- Department
of Bioproducts and Biosystems, Aalto University, FI-00076 Espoo, Finland
- E-mail:
| |
Collapse
|
30
|
Miller A, Desai A, Roley LT, Goodwin RL, Nathaniel AI, Nathaniel TI. The role of ethnicity, biological sex, and psychotropic agents in early and late onset Alzheimer's disease. Front Aging Neurosci 2022; 14:1052330. [PMID: 36620767 PMCID: PMC9815502 DOI: 10.3389/fnagi.2022.1052330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Objective This study investigates differences in pharmacological and demographic factors among male and female patients with Late-onset Alzheimer's disease (LOAD) and Early-onset Alzheimer's disease (EOAD). Method Data are from 10,126 AD patients, 9,290 were diagnosed with LOAD, while 836 were diagnosed with EOAD. Data were collected from the Prisma Health Upstate Alzheimer's patients' registry between 2016 and 2021. The logistic regression analysis was used to assess the association between pharmacological and demographic factors in males and females with LOAD and EOAD. Results In the adjusted analysis for males, patients that were administered memantine [odd ratio (OR) = 1.588, 95% CI, 1.175-2.145, p = 0.003], and buspirone [OR = 1.971, 95% CI, 1.221-3.183, p = 0.006] were more likely to be associated with EOAD, while increasing age [OR = 0.816, 95% CI, 0.799-0.834, p < 0.001] was associated with LOAD. Female patients with a history of alcohol (ETOH) use were more likely to be associated with EOAD while increasing age [OR = 0.845, 95% CI, 0.834-0.857, p < 0.001], treatment with memantine [OR = 0.774, 95% CI, 0.627-0.956, p = 0.017], African Americans [OR = 0.621, 95% CI, 0.462-0.835, p = 0.002] and tobacco use [OR = 0.529, 95% CI, 0.424-0.660, p < 0.001] were associated with LOAD. Conclusion Our findings identified specific demographic and pharmacological factors associated with males and females with LOAD and EOAD. These findings suggest the need to develop strategies to eliminate disparity in the care of LOAD or EOAD patients.
Collapse
Affiliation(s)
- Alyssa Miller
- Department of Biology, North Greenville University, Tigerville, SC, United States
| | - Ashna Desai
- Department of Biology, University of South Carolina, Columbia, SC, United States
| | | | - Richard L. Goodwin
- School of Medicine Greenville, University of South Carolina, Greenville, SC, United States
| | | | - Thomas I. Nathaniel
- School of Medicine Greenville, University of South Carolina, Greenville, SC, United States,*Correspondence: Thomas I. Nathaniel,
| |
Collapse
|
31
|
Longitudinal Analysis of the Microbiome and Metabolome in the 5xfAD Mouse Model of Alzheimer's Disease. mBio 2022; 13:e0179422. [PMID: 36468884 PMCID: PMC9765021 DOI: 10.1128/mbio.01794-22] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Recent reports implicate gut microbiome dysbiosis in the onset and progression of Alzheimer's disease (AD), yet studies involving model animals overwhelmingly omit the microbial perspective. Here, we evaluate longitudinal microbiomes and metabolomes from a popular transgenic mouse model for familial AD (5xfAD). Cecal and fecal samples from 5xfAD and wild-type B6J (WT) mice from 4 to 18 months of age were subjected to shotgun Illumina sequencing. Metabolomics was performed on plasma and feces from a subset of the same animals. Significant genotype, sex, age, and cage-specific differences were observed in the microbiome, with the variance explained by genotype at 4 and 18 months of age rising from 0.9 to 9% and 0.3 to 8% for the cecal and fecal samples, respectively. Bacteria at significantly higher abundances in AD mice include multiple Alistipes spp., two Ligilactobacillus spp., and Lactobacillus sp. P38, while multiple species of Turicibacter, Lactobacillus johnsonii, and Romboutsia ilealis were less abundant. Turicibacter is similarly depleted in people with AD, and members of this genus both consume and induce the production of gut-derived serotonin. Contradicting previous findings in humans, serotonin is significantly more concentrated in the blood of older 5xfAD animals compared to their WT littermates. 5xfAD animals exhibited significantly lower plasma concentrations of carnosine and the lysophospholipid lysoPC a C18:1. Correlations between the microbiome and metabolome were also explored. Taken together, these findings strengthen the link between Turicibacter abundance and AD, provide a basis for further microbiome studies of murine models for AD, and suggest that greater control over animal model microbiomes is needed in AD research. IMPORTANCE Microorganisms residing within the gastrointestinal tract are implicated in the onset and progression of Alzheimer's disease (AD) through the mediation of inflammation, exchange of small-molecules across the blood-brain barrier, and stimulation of the vagus nerve. Unfortunately, most animal models for AD are housed under conditions that do not reflect real-world human microbial exposure and do not sufficiently account for (or meaningfully consider) variations in the microbiome. An improved understanding of AD model animal microbiomes will increase model efficacy and the translatability of research findings into humans. Here, we present the characterization of the microbiome and metabolome of the 5xfAD mouse model, which is one of the most common animal models for familial AD. The manuscript highlights the importance of considering the microbiome in study design and aims to lay the groundwork for future studies involving mouse models for AD.
Collapse
|
32
|
Pan X, Zhang D, Heo JH, Park C, Li G, Dengler-Crish CM, Li Y, Gu Y, Young HN, Lavender DL, Shi L. Antihypertensive Use and the Risk of Alzheimer's Disease and Related Dementias among Older Adults in the USA. Drugs Aging 2022; 39:875-886. [PMID: 36251143 PMCID: PMC9851394 DOI: 10.1007/s40266-022-00981-8] [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] [Accepted: 09/30/2022] [Indexed: 01/21/2023]
Abstract
BACKGROUND Epidemiological evidence on different classes of antihypertensives and risks of Alzheimer's disease and related dementias (ADRD) is inconclusive and limited. This study examined the association between antihypertensive use (including therapy type and antihypertensive class) and ADRD diagnoses among older adults with hypertension. METHODS A retrospective, cross-sectional study was conducted, involving 539 individuals aged ≥ 65 years who used antihypertensives and had ADRD diagnosis selected from 2013 to 2018 Medical Expenditure Panel Survey (MEPS) data. The predictors were therapy type (monotherapy or polytherapy) and class of antihypertensives defined using Multum Lexicon therapeutic classification (with calcium channel blockers [CCBs] as the reference group). Weighted logistic regression was used to assess the relationships of therapy type and class of antihypertensives use with ADRD diagnosis, adjusting for sociodemographic characteristics and health status. RESULTS We found no significant difference between monotherapy and polytherapy on the odds of ADRD diagnosis. As to monotherapy, those who used angiotensin-converting enzyme inhibitors (ACEIs) had significantly lower odds of developing AD compared to those who used CCBs (OR 0.36, 95 % CI 0.13-0.99). CONCLUSIONS Findings of the study suggest the need for evidence-based drug therapy to manage hypertension in later adulthood and warrant further investigation into the mechanism underlying the protective effect of antihypertensives, particularly ACEIs, against the development of AD among older adults with hypertension.
Collapse
Affiliation(s)
- Xi Pan
- Department of Sociology, Texas State University, 601 University Drive, San Marcos, Texas, 78666, USA.
| | - Donglan Zhang
- Division of Health Services Research, Department of Foundations of Medicine, New York University Long Island School of Medicine, 101 Mineola Boulevard, Suite 3-047, Mineola, NY, 11501, USA
| | - Ji Haeng Heo
- Health Outcomes Division, College of Pharmacy, The University of Texas at Austin, College of Pharmacy-Health Outcomes and Pharmacy Practice, 2409 University Avenue, STOP A1930, Austin, TX, 78712-1120, USA
| | - Chanhyun Park
- Health Outcomes Division, College of Pharmacy, The University of Texas at Austin, College of Pharmacy-Health Outcomes and Pharmacy Practice, 2409 University Avenue, STOP A1930, Austin, TX, 78712-1120, USA
| | - Gang Li
- Huazhong University of Science and Technology, Wuhan, China
| | - Christine M Dengler-Crish
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, 4209 St, OH-44, Rootstown, OH, 44272, USA
| | - Yan Li
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1077, New York, NY, 10029, USA
| | - Yian Gu
- Department of Neurology, Columbia University Irving Medical Center, 710 W 168th St, New York, NY, 10033, USA
| | - Henry N Young
- Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia, 250 W. Green Street, Athens, GA, 30602, USA
| | - Devin L Lavender
- Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia, 250 W. Green Street, Athens, GA, 30602, USA
| | - Lu Shi
- Department of Public Health Sciences, Clemson University, 507 Edwards Hall, Clemson, SC, 29634, USA
| |
Collapse
|
33
|
An Alzheimer’s Disease Patient-Derived Olfactory Stem Cell Model Identifies Gene Expression Changes Associated with Cognition. Cells 2022; 11:cells11203258. [PMID: 36291125 PMCID: PMC9601087 DOI: 10.3390/cells11203258] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/25/2022] Open
Abstract
An early symptom of Alzheimer’s disease (AD) is an impaired sense of smell, for which the molecular basis remains elusive. Here, we generated human olfactory neurosphere-derived (ONS) cells from people with AD and mild cognitive impairment (MCI), and performed global RNA sequencing to determine gene expression changes. ONS cells expressed markers of neuroglial differentiation, providing a unique cellular model to explore changes of early AD-associated pathways. Our transcriptomics data from ONS cells revealed differentially expressed genes (DEGs) associated with cognitive processes in AD cells compared to MCI, or matched healthy controls (HC). A-Kinase Anchoring Protein 6 (AKAP6) was the most significantly altered gene in AD compared to both MCI and HC, and has been linked to cognitive function. The greatest change in gene expression of all DEGs occurred between AD and MCI. Gene pathway analysis revealed defects in multiple cellular processes with aging, intellectual deficiency and alternative splicing being the most significantly dysregulated in AD ONS cells. Our results demonstrate that ONS cells can provide a cellular model for AD that recapitulates disease-associated differences. We have revealed potential novel genes, including AKAP6 that may have a role in AD, particularly MCI to AD transition, and should be further examined.
Collapse
|
34
|
Juhl AL, Grenzer IM, Teegen B, Wiltfang J, Fitzner D, Hansen N. Biomarkers of neurodegeneration in neural autoantibody-associated psychiatric syndromes: A retrospective cohort study. J Transl Autoimmun 2022; 5:100169. [PMID: 36238527 PMCID: PMC9550648 DOI: 10.1016/j.jtauto.2022.100169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/01/2022] [Accepted: 10/03/2022] [Indexed: 10/25/2022] Open
Abstract
Background Autoantibody-associated psychiatric syndromes are a novel disease entity that is not fully understood. Several lines of evidence suggest that neurodegenerative processes are involved here. We are investigating whether autoantibody-positive psychiatric syndromes differ from those that are autoantibody-negative in cerebrospinal fluid (CSF) neurodegeneration markers. Methods We retrospectively analyzed data from 167 psychiatric patients at the University Medical Center Göttingen from 2017 to 2020. We divided this patient cohort into two, namely antibody-positive and antibody-negative. We compared various clinical features, neurodegeneration markers, and their autoantibody status in CSF and serum. We then compared both cohorts' neurodegeneration markers to a representative Alzheimer cohort. We subdivided the patients into their diverse psychiatric syndromes according to the manual to assess and document psychopathology in psychiatry (the AMDP), and compared the neurodegeneration markers. Results Antibody-associated psychiatric syndromes do not appear to reveal significantly greater neurodegeneration than their antibody-negative psychiatric syndromes. 71% of antibody-positive patients fulfilled the criteria for a possible and 22% for a definitive autoimmune encephalitis. Our autoantibody-positive patient cohort's relative risk to develop an possible autoimmune encephalitis was 9%. We also noted that phosphorylated tau protein 181 (ptau 181) did not significantly differ between antibody-associated psychiatric syndromes and our Alzheimer cohort. The psycho-organic syndrome usually exhibits the most prominent neurodegeneration markers, both in antibody-positive and antibody-negative psychiatric patients. Discussion We did not find hints for neurodegenerative processes in our antibody-positive versus AD cohort considering total tau or amyloid markers. However, our findings indicate that the neurodegeneration marker ptau181 does not differ significantly between antibody-positive and Alzheimer cohorts, further suggesting axonal neurodegeneration in antibody-positive patients as AD patients have an elevated ptau181. The evidence we uncovered thus suggests that axonal neurodegeneration might affect patients suffering from autoantibody-associated psychiatric syndromes.
Collapse
Affiliation(s)
- Aaron Levin Juhl
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany,Translational Psychoneuroscience, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
| | - Insa Maria Grenzer
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany,Translational Psychoneuroscience, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
| | - Bianca Teegen
- Clinical Immunological Laboratory Prof. Stöcker, Groß Grönau, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany,German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany,Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Dirk Fitzner
- Department of Neurology, University Medical Center Göttingen, Robert-Koch Straße 40, 37075, Göttingen, Germany
| | - Niels Hansen
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany,Translational Psychoneuroscience, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany,Corresponding author. University Medical Center of Göttingen, Department of Psychiatry and Psychotherapy, Translational Psychoneuroscience, Von-Siebold Str. 5, 37075, Göttingen.
| |
Collapse
|
35
|
Pizzini FB, Conti E, Bianchetti A, Splendiani A, Fusco D, Caranci F, Bozzao A, Landi F, Gandolfo N, Farina L, Miele V, Trabucchi M, Frisoni GB, Bastianello S. Radiological assessment of dementia: the Italian inter-society consensus for a practical and clinically oriented guide to image acquisition, evaluation, and reporting. LA RADIOLOGIA MEDICA 2022; 127:998-1022. [PMID: 36070064 PMCID: PMC9508052 DOI: 10.1007/s11547-022-01534-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/25/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Radiological evaluation of dementia is expected to increase more and more in routine practice due to both the primary role of neuroimaging in the diagnostic pathway and the increasing incidence of the disease. Despite this, radiologists often do not follow a disease-oriented approach to image interpretation, for several reasons, leading to reports of limited value to clinicians. In our work, through an intersocietal consensus on the main mandatory knowledge about dementia, we proposed a disease-oriented protocol to optimize and standardize the acquisition/evaluation/interpretation and reporting of radiological images. Our main purpose is to provide a practical guideline for the radiologist to help increase the effectiveness of interdisciplinary dialogue and diagnostic accuracy in daily practice. RESULTS We defined key clinical and imaging features of the dementias (A), recommended MRI protocol (B), proposed a disease-oriented imaging evaluation and interpretation (C) and report (D) with a glimpse to future avenues (E). The proposed radiological practice is to systematically evaluate and score atrophy, white matter changes, microbleeds, small vessel disease, consider the use of quantitative measures using commercial software tools critically, and adopt a structured disease-oriented report. In the expanding field of cognitive disorders, the only effective assessment approach is the standardized disease-oriented one, which includes a multidisciplinary integration of the clinical picture, MRI, CSF and blood biomarkers and nuclear medicine.
Collapse
Affiliation(s)
- Francesca B. Pizzini
- Radiology, Department of Diagnostic and Public Health, University of Verona, Piazzale L.A. Scuro, 10, 37100 Verona, Italy
| | - Enrico Conti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Angelo Bianchetti
- Department of Medicine and Rehabilitation, Clinical Institute S. Anna-Gruppo San Donato, Brescia, Italy
- Italian Society of Gerontology and Geriatrics (SIGG), Florence, Italy
- Italian Association of Psychogeriatrics (AIP), Brescia, Italy
| | - Alessandra Splendiani
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Domenico Fusco
- Foundation Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Ferdinando Caranci
- Department of Medicine of Precision, School of Medicine, “Luigi Vanvitelli” University of Campania, 80147 Naples, Italy
| | - Alessandro Bozzao
- NESMOS, Department of Neuroradiology, S. Andrea Hospital, University Sapienza, Rome, Italy
| | - Francesco Landi
- Foundation Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Nicoletta Gandolfo
- Diagnostic Imaging Department, Villa Scassi Hospital-ASL 3, Corso Scassi 1, Genoa, Italy
| | - Lisa Farina
- Neuroradiology Department, IRCCS Mondino Foundation, Pavia, Italy
| | - Vittorio Miele
- Dipartimento Di Radiodiagnostica Emergenza-Urgenza, Azienda Universitaria Careggi, Florence, Italy
| | - Marco Trabucchi
- Italian Society of Gerontology and Geriatrics (SIGG), Florence, Italy
- Italian Association of Psychogeriatrics (AIP), Brescia, Italy
- University of “Tor Vergata”, Rome, Italy
| | - Giovanni B. Frisoni
- Centre de La Mémoire, Geneva University and University Hospitals, 1205 Geneva, Switzerland
| | - Stefano Bastianello
- Neuroradiology Department, IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
36
|
Janahi M, Aksman L, Schott JM, Mokrab Y, Altmann A. Nomograms of human hippocampal volume shifted by polygenic scores. eLife 2022; 11:e78232. [PMID: 35938915 PMCID: PMC9391046 DOI: 10.7554/elife.78232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/06/2022] [Indexed: 11/25/2022] Open
Abstract
Nomograms are important clinical tools applied widely in both developing and aging populations. They are generally constructed as normative models identifying cases as outliers to a distribution of healthy controls. Currently used normative models do not account for genetic heterogeneity. Hippocampal volume (HV) is a key endophenotype for many brain disorders. Here, we examine the impact of genetic adjustment on HV nomograms and the translational ability to detect dementia patients. Using imaging data from 35,686 healthy subjects aged 44-82 from the UK Biobank (UKB), we built HV nomograms using Gaussian process regression (GPR), which - compared to a previous method - extended the application age by 20 years, including dementia critical age ranges. Using HV polygenic scores (HV-PGS), we built genetically adjusted nomograms from participants stratified into the top and bottom 30% of HV-PGS. This shifted the nomograms in the expected directions by ~100 mm3 (2.3% of the average HV), which equates to 3 years of normal aging for a person aged ~65. Clinical impact of genetically adjusted nomograms was investigated by comparing 818 subjects from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database diagnosed as either cognitively normal (CN), having mild cognitive impairment (MCI) or Alzheimer's disease (AD) patients. While no significant change in the survival analysis was found for MCI-to-AD conversion, an average of 68% relative decrease was found in intra-diagnostic-group variance, highlighting the importance of genetic adjustment in untangling phenotypic heterogeneity.
Collapse
Affiliation(s)
- Mohammed Janahi
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Biomedical Engineering, University College LondonLondonUnited Kingdom
- Medical and Population Genomics Lab, Human Genetics Department, Research Branch, Sidra MedicineDohaQatar
| | - Leon Aksman
- Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Jonathan M Schott
- Dementia Research Centre (DRC), Queen Square Institute of Neurology, University College LondonLondonUnited Kingdom
| | - Younes Mokrab
- Medical and Population Genomics Lab, Human Genetics Department, Research Branch, Sidra MedicineDohaQatar
- Department of Genetic Medicine, Weill Cornell Medicine-QatarDohaQatar
| | - Andre Altmann
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Biomedical Engineering, University College LondonLondonUnited Kingdom
| |
Collapse
|
37
|
Kim KW, Choi J, Chin J, Lee BH, Na DL. Eye-Tracking Metrics for Figure-Copying Processes in Early- vs. Late-Onset Alzheimer's Disease. Front Neurol 2022; 13:844341. [PMID: 35651346 PMCID: PMC9149280 DOI: 10.3389/fneur.2022.844341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/12/2022] [Indexed: 12/04/2022] Open
Abstract
Visuospatial dysfunction is a common symptom in patients with Alzheimer's disease (AD). To more focus on copying processes rather than on finally completed figures, we conceptually split the copying processes into three stages: visuoperceptual function, visuoconstructional function, and working memory function. We constructed perceptual and working spaces to investigate the different stages of copying, and then, we compared the number and duration of fixations and saccades and the number of switches across the two spaces. We used eye-tracking glasses to assess eye-tracking metrics in patients with early-onset AD (EOAD), patients with late-onset AD (LOAD), and normal control (NC) participants while they copied the simplified Rey–Osterrieth complex figure test (RCFT). Regarding eye metrics on the perceptual space, the number and duration of fixations were greater in both groups of patients with AD than in the NC participants group (number: EOAD vs. NC: p < 0.001, LOAD vs. NC: p = 0. 003/ duration: EOAD vs. NC: p < 0.001, LOAD vs. NC: p < 0.001). On the working space, the number and duration of fixations were greater in the patients with EOAD than in the patients with LOAD and NC participants (number: EOAD vs. LOAD: p = 0. 007, EOAD vs. NC: p = 0. 001/duration: EOAD vs. LOAD: p = 0. 008, EOAD vs. NC: p = 0. 002). The number of saccades and switching was higher in patients with EOAD than in NC participants (p < 0.001). The eye-tracking metrics from the simplified RCFT correlated with the neuropsychological test scores. Patients with EOAD and LOAD achieved the same level of performance at the simplified and original RCFT scores. However, patients with EOAD than LOAD showed a greater number and duration of fixations on the working space and more frequent switching between the perceptual and working spaces, which may reflect more cognitive efforts to achieve the same level of performance.
Collapse
Affiliation(s)
- Ko Woon Kim
- Department of Neurology, Jeonbuk National University Medical School and Hospital, Jeonju, South Korea.,Research Institute of Clinical Medicine of Jeonbuk National University, Jeonju, South Korea.,Biomedical Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Jongdoo Choi
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Neuroscience Center, Samsung Medical Center, Seoul, South Korea
| | - Juhee Chin
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Neuroscience Center, Samsung Medical Center, Seoul, South Korea
| | - Byung Hwa Lee
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Neuroscience Center, Samsung Medical Center, Seoul, South Korea
| | - Duk L Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, South Korea.,Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, South Korea
| |
Collapse
|
38
|
Şimşek H, Uçar A. Is Ketogenic Diet Therapy a Remedy for Alzheimer’s Disease or Mild Cognitive Impairments?: A Narrative Review of Randomized Controlled Trials. ADVANCES IN GERONTOLOGY 2022. [DOI: 10.1134/s2079057022020175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
39
|
Ghasemi-Tarie R, Kiasalari Z, Fakour M, Khorasani M, Keshtkar S, Baluchnejadmojarad T, Roghani M. Nobiletin prevents amyloid β 1-40-induced cognitive impairment via inhibition of neuroinflammation and oxidative/nitrosative stress. Metab Brain Dis 2022; 37:1337-1349. [PMID: 35294678 DOI: 10.1007/s11011-022-00949-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 03/01/2022] [Indexed: 01/17/2023]
Abstract
Alzheimer's disease (AD) is presented as an age-related neurodegenerative disease with multiple cognitive deficits and amyloid β (Aβ) accumulation is the most important involved factor in its development. Nobiletin is a bioflavonoid isolated from citrus fruits peels with anti-inflammatory and anti-oxidative activity as well as anti-dementia property that has shown potency to ameliorate intracellular and extracellular Ab. The aim of the present study was to assess protective effect of nobiletin against Aβ1-40-induced cognitive impairment as a consistent model of AD. After bilateral intrahippocampal (CA1 subfield) injection of Aβ1-40, rats were treated with nobiletin (10 mg/kg/day; p.o.) from stereotaxic surgery day (day 0) till day + 7. Cognition function was evaluated in a battery of behavioral tasks at week 3 with final assessment of hippocampal oxidative stress and inflammation besides Nissl staining and 3-nitrotyrosine (3-NT) immunohistochemistry. Analysis of behavioral data showed notable and significant improvement of alternation in Y maze test, discrimination ratio in novel object recognition task, and step through latency in passive avoidance test in nobiletin-treated Aβ group. Additionally, nobiletin treatment was associated with lower hippocampal levels of MDA and ROS and partial reversal of SOD activity and also improvement of Nrf2 with no significant effect on GSH and catalase. Furthermore, nobiletin attenuated hippocampal neuroinflammation in Aβ group as shown by lower tissue levels of TLR4, NF-kB, and TNFa. Histochemical findings showed that nobiletin prevents CA1 neuronal loss in Nissl staining in addition to its alleviation of 3-nitrotyrosine (3-NT) immunoreactivity as a marker of nitrosative stress. Collectively, these findings indicated neuroprotective and anti-dementia potential of nobiletin that is partly attributed to its anti-oxidative, anti-nitrosative, and anti-inflammatory property associated with proper modulation of TLR4/NF-kB/Nrf2 pathways.
Collapse
Affiliation(s)
| | - Zahra Kiasalari
- Neurophysiology Research Center, Department of Physiology, Shahed University, Tehran, Iran
| | - Marzieh Fakour
- Department of Physiology, School of Medicine, Shahed University, Tehran, Iran.
| | - Maryam Khorasani
- Department of Physiology, School of Medicine, Shahed University, Tehran, Iran
| | - Sedigheh Keshtkar
- Department of Physiology, School of Medicine, Shahed University, Tehran, Iran
| | | | - Mehrdad Roghani
- Neurophysiology Research Center, Department of Physiology, Shahed University, Tehran, Iran.
| |
Collapse
|
40
|
Basheer A, Agarwal A, Misra B, Gupta A, Srivastava P, Kirubakaran R, Vishnu V. Bacopa monnieri in the treatment of dementia due to Alzheimer's disease: A systematic review of randomised controlled trials. Interact J Med Res 2022; 11:e38542. [PMID: 35612544 PMCID: PMC9379783 DOI: 10.2196/38542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/21/2022] [Accepted: 05/25/2022] [Indexed: 11/14/2022] Open
Abstract
Background Bacopa monnieri, a herb that has been used for many centuries in India, has shown neuroprotective effects in animal and in vitro studies; human studies on patients with Alzheimer disease have been inconclusive. Objective The primary objective of this review was to determine the clinical efficacy and safety of B. monnieri in persons with mild, moderate, or severe dementia, or mild cognitive impairment, due to Alzheimer disease. Methods We searched PubMed, Embase, Cochrane Library, clinical trial registries (World Health Organization, Australia-New Zealand, United States, and South Africa), the metaRegister of Controlled Trials, and CINAHL. We intended to include all randomized and quasi-randomized controlled trials that compared B. monnieri, its extract or active ingredients (at any dosage), with a placebo or a cholinesterase inhibitor among adults with dementia due to Alzheimer disease and in those with mild cognitive impairment due to Alzheimer disease. Results Our comprehensive search yielded 5 eligible studies. A total of 3 studies used B. monnieri in combination with herbal extracts while the remaining 2 used B. monnieri extracts only. Two studies compared B. monnieri with donepezil while the others used a placebo as the control. There was considerable variation in the B. monnieri dose used (ranging between 125 mg to 500 mg twice daily) and heterogeneity in treatment duration, follow-up, and outcomes. The major outcomes were Mini-Mental State Examination scores reported in 3 trials, Cognitive subscale scores of the Alzheimer’s Disease Assessment Scale in 1 study, and a battery of cognitive tests in 2 studies. Using the Cochrane risk-of-bias tool, overall, we judged all 5 studies to be at high risk of bias. While all studies reported a statistically significant difference between B. monnieri and the comparator in at least one outcome, we rated the overall quality of evidence for the Alzheimer’s Disease Assessment Scale-Cognitive Subscale, Postgraduate Institute Memory Scale, Mini-Mental State Examination, and Wechsler Memory Scale to be very low due to downgrading by 2 levels for high risk of bias and 1 more level for impreciseness due to small sample sizes and wide CIs. Conclusions There was no difference between B. monnieri and the placebo or donepezil in the treatment of Alzheimer disease based on very low certainty evidence. No major safety issues were reported in the included trials. Future randomized controlled trials should aim to recruit more participants and report clinically meaningful outcomes. Trial Registration PROSPERO CRD42020169421; https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=169421
Collapse
Affiliation(s)
- Aneesh Basheer
- DM Wayanad Institute of Medical Sciences (DM WIMS), Wayanad, IN
| | - Ayush Agarwal
- All India Institute of Medical Sciences, New Delhi, RN 704, 7th floor, CN CenterAnsari Nagar, New Delhi, IN
| | - Biswamohan Misra
- All India Institute of Medical Sciences, New Delhi, RN 704, 7th floor, CN CenterAnsari Nagar, New Delhi, IN
| | - Anu Gupta
- All India Institute of Medical Sciences, New Delhi, RN 704, 7th floor, CN CenterAnsari Nagar, New Delhi, IN
| | - Padma Srivastava
- All India Institute of Medical Sciences, New Delhi, RN 704, 7th floor, CN CenterAnsari Nagar, New Delhi, IN
| | | | - Venugopalan Vishnu
- All India Institute of Medical Sciences, New Delhi, RN 704, 7th floor, CN CenterAnsari Nagar, New Delhi, IN
| |
Collapse
|
41
|
Brezovakova V, Sykova E, Jadhav S. Astrocytes Derived from Familial and Sporadic Alzheimer's Disease iPSCs Show Altered Calcium Signaling and Respond Differently to Misfolded Protein Tau. Cells 2022; 11:cells11091429. [PMID: 35563735 PMCID: PMC9101114 DOI: 10.3390/cells11091429] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 12/22/2022] Open
Abstract
Astrocytes regulate important functions in the brain, and their dysregulation has been linked to the etiology of neurodegenerative diseases, such as Alzheimer’s disease (AD). The role of astroglia in human AD remains enigmatic, owing to the limitations of animal models, which, while recreating some pathological aspects of the disease, do not fully mirror its course. In addition, the recognition of major structural and functional differences between human and mouse astrocytes has also prompted research into human glial cells. In the current study, astrocytes were generated using human iPSCs from patients with sporadic Alzheimer’s disease (sAD), familial Alzheimer’s disease (fAD) and non-demented controls (NDC). All clones gained astrocyte-specific morphological and proteomic characteristics upon in vitro differentiation, without considerable inter-clonal variances. In comparison to NDC, AD astrocytes displayed aberrant calcium dynamics in response to glutamate. When exposed to monomeric and aggregated tau, AD astrocytes demonstrated hypertrophy and elevated GFAP expression, differential expression of select signaling and receptor proteins, and the enhanced production of metalloproteinases (MMPs). Moreover, astrocytic secretomes were able to degrade tau in both monomeric and pathologically aggregated forms, which was mediated by MMP-2 and -9. The capacity to neutralize tau varied considerably between clones, with fAD astrocytes having the lowest degradability relative to sAD and healthy astrocytes. Importantly, when compared to aggregated tau alone, astrocytic secretome pretreatment of tau differentially reduced its detrimental effects on neurons. Our results show crucial differences in sporadic and familial AD astrocytes and suggests that these cells may play distinctive roles in the pathogenesis of early and late onset Alzheimer’s disease.
Collapse
|
42
|
Pomilio AB, Vitale AA, Lazarowski AJ. Uncommon Noninvasive Biomarkers for the Evaluation and Monitoring of the Etiopathogenesis of Alzheimer's Disease. Curr Pharm Des 2022; 28:1152-1169. [DOI: 10.2174/1381612828666220413101929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/25/2022] [Indexed: 11/22/2022]
Abstract
Background:
Alzheimer´s disease (AD) is the most widespread dementia in the world, followed by vascular dementia. Since AD is a heterogeneous disease that shows several varied phenotypes, it is not easy to make an accurate diagnosis, so it arises when the symptoms are clear and the disease is already very advanced. Therefore, it is important to find out biomarkers for AD early diagnosis that facilitate treatment or slow down the disease. Classic biomarkers are obtained from cerebrospinal fluid and plasma, along with brain imaging by positron emission tomography. Attempts have been made to discover uncommon biomarkers from other body fluids, which are addressed in this update.
Objective:
This update aims to describe recent biomarkers from minimally invasive body fluids for the patients, such as saliva, urine, eye fluid or tears.
Methods:
Biomarkers were determined in patients versus controls by single tandem mass spectrometry, and immunoassays. Metabolites were identified by nuclear magnetic resonance, and microRNAs with genome-wide high-throughput real-time polymerase chain reaction-based platforms.
Results:
Biomarkers from urine, saliva, and eye fluid were described, including peptides/proteins, metabolites, and some microRNAs. The association with AD neuroinflammation and neurodegeneration was analyzed, highlighting the contribution of matrix metalloproteinases, the immune system and microglia, as well as the vascular system.
Conclusion:
Unusual biomarkers have been developed, which distinguish each stage and progression of the disease, and are suitable for the early AD diagnosis. An outstanding relationship of biomarkers with neuroinflammation and neurodegeneration was assessed, clearing up concerns of the etiopathogenesis of AD.
Collapse
Affiliation(s)
- Alicia B. Pomilio
- Departamento de Bioquímica Clínica, Área Hematología, Hospital de Clínicas “José de San Martín”, Universidad de Buenos Aires, Av. Córdoba 2351, C1120AAF Buenos Aires, Argentina
| | - Arturo A. Vitale
- Departamento de Bioquímica Clínica, Área Hematología, Hospital de Clínicas “José de San Martín”, Universidad de Buenos Aires, Av. Córdoba 2351, C1120AAF Buenos Aires, Argentina
| | - Alberto J. Lazarowski
- Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Córdoba 2351, C1120AAF Buenos Aires, Argentina
| |
Collapse
|
43
|
Pomilio AB, Vitale AA, Lazarowski AJ. Neuroproteomics Chip-Based Mass Spectrometry and Other Techniques for Alzheimer´S Disease Biomarkers – Update. Curr Pharm Des 2022; 28:1124-1151. [DOI: 10.2174/1381612828666220413094918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/25/2022] [Indexed: 11/22/2022]
Abstract
Background:
Alzheimer's disease (AD) is a progressive neurodegenerative disease of growing interest given that there is cognitive damage and symptom onset acceleration. Therefore, it is important to find AD biomarkers for early diagnosis, disease progression, and discrimination of AD and other diseases.
Objective:
To update the relevance of mass spectrometry for the identification of peptides and proteins involved in AD useful as discriminating biomarkers.
Methods:
Proteomics and peptidomics technologies that show the highest possible specificity and selectivity for AD biomarkers are analyzed, together with the biological fluids used. In addition to positron emission tomography and magnetic resonance imaging, MALDI-TOF mass spectrometry is widely used to identify proteins and peptides involved in AD. The use of protein chips in SELDI technology and electroblotting chips for peptides makes feasible small amounts (L) of samples for analysis.
Results:
Suitable biomarkers are related to AD pathology, such as intracellular neurofibrillary tangles; extraneuronal senile plaques; neuronal and axonal degeneration; inflammation and oxidative stress. Recently, peptides were added to the candidate list, which are not amyloid-b or tau fragments, but are related to coagulation, brain plasticity, and complement/neuroinflammation systems involving the neurovascular unit.
Conclusion:
The progress made in the application of mass spectrometry and recent chip techniques is promising for discriminating between AD, mild cognitive impairment, and matched healthy controls. The application of this technique to blood samples from patients with AD has shown to be less invasive and fast enough to determine the diagnosis, stage of the disease, prognosis, and follow-up of the therapeutic response.
Collapse
Affiliation(s)
- Alicia B. Pomilio
- Departamento de Bioquímica Clínica, Área Hematología, Hospital de Clínicas “José de San Martín”, Universidad de Buenos Aires, Av. Córdoba 2351, C1120AAF Buenos Aires, Argentina
| | - Arturo A. Vitale
- Departamento de Bioquímica Clínica, Área Hematología, Hospital de Clínicas “José de San Martín”, Universidad de Buenos Aires, Av. Córdoba 2351, C1120AAF Buenos Aires, Argentina
| | - Alberto J. Lazarowski
- Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Córdoba 2351, C1120AAF Buenos Aires, Argentina
| |
Collapse
|
44
|
Aguilar-Pineda JA, Paco-Coralla SG, Febres-Molina C, Gamero-Begazo PL, Shrivastava P, Vera-López KJ, Davila-Del-Carpio G, López-C P, Gómez B, Lino Cardenas CL. In Silico Analysis of the Antagonist Effect of Enoxaparin on the ApoE4–Amyloid-Beta (Aβ) Complex at Different pH Conditions. Biomolecules 2022; 12:biom12040499. [PMID: 35454088 PMCID: PMC9027285 DOI: 10.3390/biom12040499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 11/16/2022] Open
Abstract
Apolipoprotein E4 (ApoE4) is thought to increase the risk of developing Alzheimer’s disease. Several studies have shown that ApoE4-Amyloid β (Aβ) interactions can increment amyloid depositions in the brain and that this can be augmented at low pH values. On the other hand, experimental studies in transgenic mouse models have shown that treatment with enoxaparin significantly reduces cortical Aβ levels, as well as decreases the number of activated astrocytes around Aβ plaques. However, the interactions between enoxaparin and the ApoE4-Aβ proteins have been poorly explored. In this work, we combine molecular dynamics simulations, molecular docking, and binding free energy calculations to elucidate the molecular properties of the ApoE4-Aβ interactions and the competitive binding affinity of the enoxaparin on the ApoE4 binding sites. In addition, we investigated the effect of the environmental pH levels on those interactions. Our results showed that under different pH conditions, the closed form of the ApoE4 protein, in which the C-terminal domain folds into the protein, remains stabilized by a network of hydrogen bonds. This closed conformation allowed the generation of six different ApoE4-Aβ interaction sites, which were energetically favorable. Systems at pH5 and 6 showed the highest energetic affinity. The enoxaparin molecule was found to have a strong energetic affinity for ApoE4-interacting sites and thus can neutralize or disrupt ApoE4-Aβ complex formation.
Collapse
Affiliation(s)
- Jorge Alberto Aguilar-Pineda
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de Investigación, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru; (S.G.P.-C.); (P.S.); (K.J.V.-L.); (G.D.-D.-C.)
- Centro de Investigación en Ingeniería Molecular—CIIM, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru; (P.L.G.-B.); (B.G.)
- Correspondence: (J.A.A.-P.); (C.L.L.C.)
| | - Silvana G. Paco-Coralla
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de Investigación, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru; (S.G.P.-C.); (P.S.); (K.J.V.-L.); (G.D.-D.-C.)
| | - Camilo Febres-Molina
- Doctorado en Fisicoquímica Molecular, Facultad de Ciencias Exactas, Universidad Andres Bello, Santiago 8370134, Chile;
| | - Pamela L. Gamero-Begazo
- Centro de Investigación en Ingeniería Molecular—CIIM, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru; (P.L.G.-B.); (B.G.)
| | - Pallavi Shrivastava
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de Investigación, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru; (S.G.P.-C.); (P.S.); (K.J.V.-L.); (G.D.-D.-C.)
| | - Karin J. Vera-López
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de Investigación, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru; (S.G.P.-C.); (P.S.); (K.J.V.-L.); (G.D.-D.-C.)
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru
| | - Gonzalo Davila-Del-Carpio
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de Investigación, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru; (S.G.P.-C.); (P.S.); (K.J.V.-L.); (G.D.-D.-C.)
- Vicerrectorado de Investigación, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru;
| | - Patricia López-C
- Vicerrectorado de Investigación, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru;
| | - Badhin Gómez
- Centro de Investigación en Ingeniería Molecular—CIIM, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru; (P.L.G.-B.); (B.G.)
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru
| | - Christian L. Lino Cardenas
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Boston, MA 02114, USA
- Correspondence: (J.A.A.-P.); (C.L.L.C.)
| |
Collapse
|
45
|
Ribarič S. Physical Exercise, a Potential Non-Pharmacological Intervention for Attenuating Neuroinflammation and Cognitive Decline in Alzheimer's Disease Patients. Int J Mol Sci 2022; 23:ijms23063245. [PMID: 35328666 PMCID: PMC8952567 DOI: 10.3390/ijms23063245] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/14/2022] Open
Abstract
This narrative review summarises the evidence for considering physical exercise (PE) as a non-pharmacological intervention for delaying cognitive decline in patients with Alzheimer’s disease (AD) not only by improving cardiovascular fitness but also by attenuating neuroinflammation. Ageing is the most important risk factor for AD. A hallmark of the ageing process is a systemic low-grade chronic inflammation that also contributes to neuroinflammation. Neuroinflammation is associated with AD, Parkinson’s disease, late-onset epilepsy, amyotrophic lateral sclerosis and anxiety disorders. Pharmacological treatment of AD is currently limited to mitigating the symptoms and attenuating progression of the disease. AD animal model studies and human studies on patients with a clinical diagnosis of different stages of AD have concluded that PE attenuates cognitive decline not only by improving cardiovascular fitness but possibly also by attenuating neuroinflammation. Therefore, low-grade chronic inflammation and neuroinflammation should be considered potential modifiable risk factors for AD that can be attenuated by PE. This opens the possibility for personalised attenuation of neuroinflammation that could also have important health benefits for patients with other inflammation associated brain disorders (i.e., Parkinson’s disease, late-onset epilepsy, amyotrophic lateral sclerosis and anxiety disorders). In summary, life-long, regular, structured PE should be considered as a supplemental intervention for attenuating the progression of AD in human. Further studies in human are necessary to develop optimal, personalised protocols, adapted to the progression of AD and the individual’s mental and physical limitations, to take full advantage of the beneficial effects of PE that include improved cardiovascular fitness, attenuated systemic inflammation and neuroinflammation, stimulated brain Aβ peptides brain catabolism and brain clearance.
Collapse
Affiliation(s)
- Samo Ribarič
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
46
|
Zhang L, Cao J, Yang H, Pham P, Khan U, Brown B, Wang Y, Zieneldien T, Cao C. Commercial and Instant Coffees Effectively Lower Aβ1-40 and Aβ1-42 in N2a/APPswe Cells. Front Nutr 2022; 9:850523. [PMID: 35369094 PMCID: PMC8965317 DOI: 10.3389/fnut.2022.850523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background Alzheimer's disease (AD) is a multifactorial neurological disease with neurofibrillary tangles and neuritic plaques as histopathological markers. Due to this, although AD is the leading cause of dementia worldwide, clinical AD dementia cannot be certainly diagnosed until neuropathological post-mortem evaluation. Coffee has been reported to have neurologically protective factors, particularly against AD, but coffee brand and type have not been taken into consideration in previous studies. We examined the discrepancies among popular commercial and instant coffees in limiting the development and progression through Aβ1-40 and Aβ1-42 production, and hypothesized that coffee consumption, regardless of brand or type, is beneficial for stalling the progression and development of Aβ-related AD. Methods Coffee samples from four commercial coffee brands and four instant coffees were purchased or prepared following given instructions and filtered for the study. 5, 2.5, and 1.25% concentrations of each coffee were used to treat N2a/APPswe cell lines. MTT assay was used to assess cell viability for coffee concentrations, as well as pure caffeine concentrations. Sandwich ELISA assay was used to determine Aβ concentration for Aβ1-40 and Aβ1-42 peptides of coffee-treated cells. Results Caffeine concentrations were significantly varied among all coffees (DC vs. MDC, PC, SB, NIN, MIN p < 0.05). There was no correlation between caffeine concentration and cell toxicity among brands and types of coffee, with no toxicity at 0.5 mg/ml caffeine and lower. Most coffees were toxic to N2a/APPswe cells at 5% (p < 0.05), but not at 2.5%. Most coffees at a 2.5% concentration reduced Aβ1-40 and Aβ1-42 production, with comparable results between commercial and instant coffees. Conclusion All coffees tested have beneficial health effects for AD through lowering Aβ1-40 and Aβ1-42 production, with Dunkin' Donuts® medium roast coffee demonstrating the most consistent and optimal cell survival rates and Aβ concentration. On the other hand, Starbucks® coffee exhibited the highest cell toxicity rates among the tested coffees.
Collapse
Affiliation(s)
- Lifang Zhang
- Department of Neurological Rehabilitation, The Affiliated Brain Hospital of Guangzhou Medical University, Guanzhou, China
| | - Jessica Cao
- Department of Kinesiology, Wiess School of Natural Sciences, Rice University, Houston, TX, United States
| | - Haiqiang Yang
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Phillip Pham
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Umer Khan
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Breanna Brown
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Yanhong Wang
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Tarek Zieneldien
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Chuanhai Cao
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
- Department of Neurology, College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
47
|
Goyzueta-Mamani LD, Barazorda-Ccahuana HL, Chávez-Fumagalli MA, F. Alvarez KL, Aguilar-Pineda JA, Vera-Lopez KJ, Lino Cardenas CL. In Silico Analysis of Metabolites from Peruvian Native Plants as Potential Therapeutics against Alzheimer's Disease. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030918. [PMID: 35164183 PMCID: PMC8838509 DOI: 10.3390/molecules27030918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 12/19/2022]
Abstract
Background: Despite research on the molecular bases of Alzheimer’s disease (AD), effective therapies against its progression are still needed. Recent studies have shown direct links between AD progression and neurovascular dysfunction, highlighting it as a potential target for new therapeutics development. In this work, we screened and evaluated the inhibitory effect of natural compounds from native Peruvian plants against tau protein, amyloid beta, and angiotensin II type 1 receptor (AT1R) pathologic AD markers. Methods: We applied in silico analysis, such as virtual screening, molecular docking, molecular dynamics simulation (MD), and MM/GBSA estimation, to identify metabolites from Peruvian plants with inhibitory properties, and compared them to nicotinamide, telmisartan, and grapeseed extract drugs in clinical trials. Results: Our results demonstrated the increased bioactivity of three plants’ metabolites against tau protein, amyloid beta, and AT1R. The MD simulations indicated the stability of the AT1R:floribundic acid, amyloid beta:rutin, and tau:brassicasterol systems. A polypharmaceutical potential was observed for rutin due to its high affinity to AT1R, amyloid beta, and tau. The metabolite floribundic acid showed bioactivity against the AT1R and tau, and the metabolite brassicasterol showed bioactivity against the amyloid beta and tau. Conclusions: This study has identified molecules from native Peruvian plants that have the potential to bind three pathologic markers of AD.
Collapse
Affiliation(s)
- Luis Daniel Goyzueta-Mamani
- Laboratory of Genomics and Neurovascular Diseases, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru; (M.A.C.-F.); (K.L.F.A.); (J.A.A.-P.); (K.J.V.-L.)
- Correspondence: (L.D.G.-M.); (C.L.L.C.)
| | - Haruna Luz Barazorda-Ccahuana
- Vicerrectorado de Investigación, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru;
| | - Miguel Angel Chávez-Fumagalli
- Laboratory of Genomics and Neurovascular Diseases, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru; (M.A.C.-F.); (K.L.F.A.); (J.A.A.-P.); (K.J.V.-L.)
| | - Karla Lucia F. Alvarez
- Laboratory of Genomics and Neurovascular Diseases, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru; (M.A.C.-F.); (K.L.F.A.); (J.A.A.-P.); (K.J.V.-L.)
| | - Jorge Alberto Aguilar-Pineda
- Laboratory of Genomics and Neurovascular Diseases, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru; (M.A.C.-F.); (K.L.F.A.); (J.A.A.-P.); (K.J.V.-L.)
| | - Karin Jannet Vera-Lopez
- Laboratory of Genomics and Neurovascular Diseases, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru; (M.A.C.-F.); (K.L.F.A.); (J.A.A.-P.); (K.J.V.-L.)
| | - Christian Lacks Lino Cardenas
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
- Correspondence: (L.D.G.-M.); (C.L.L.C.)
| |
Collapse
|
48
|
Khullar S, Wang D. Predicting gene regulatory networks from multi-omics to link genetic risk variants and neuroimmunology to Alzheimer's disease phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34189529 DOI: 10.1101/2021.06.21.449165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Genome-wide association studies have found many genetic risk variants associated with Alzheimer's disease (AD). However, how these risk variants affect deeper phenotypes such as disease progression and immune response remains elusive. Also, our understanding of cellular and molecular mechanisms from disease risk variants to various phenotypes is still limited. To address these problems, we performed an integrative multi-omics analysis of genotype, transcriptomics, and epigenomics for revealing gene regulatory mechanisms from disease variants to AD phenotypes. METHOD First, given the population gene expression data of a cohort, we construct and cluster its gene co-expression network to identify gene co-expression modules for various AD phenotypes. Next, we predict transcription factors (TFs) regulating co-expressed genes and AD risk SNPs that interrupt TF binding sites on regulatory elements. Finally, we construct a gene regulatory network (GRN) linking SNPs, interrupted TFs, and regulatory elements to target genes and gene modules for each phenotype in the cohort. This network thus provides systematic insights into gene regulatory mechanisms from risk variants to AD phenotypes. RESULTS Our analysis predicted GRNs in three major AD-relevant regions: Hippocampus, Dorsolateral Prefrontal Cortex (DLPFC), Lateral Temporal Lobe (LTL). Comparative analyses revealed cross-region-conserved and region-specific GRNs, in which many immunological genes are present. For instance, SNPs rs13404184 and rs61068452 disrupt SPI1 binding and regulation of AD gene INPP5D in the Hippocampus and LTL. However, SNP rs117863556 interrupts bindings of REST to regulate GAB2 in DLPFC only. Driven by emerging neuroinflammation in AD, we used Covid-19 as a proxy to identify possible regulatory mechanisms for neuroimmunology in AD. To this end, we looked at the GRN subnetworks relating to genes from shared AD-Covid pathways. From those subnetworks, our machine learning analysis prioritized the AD-Covid genes for predicting Covid-19 severity. Decision Curve Analysis also validated our AD-Covid genes outperform known Covid-19 genes for classifying severe Covid-19 patients. This suggests AD-Covid genes along with linked SNPs can be potential novel biomarkers for neuroimmunology in AD. Finally, our results are open-source available as a comprehensive functional genomic map for AD, providing a deeper mechanistic understanding of the interplay among multi-omics, brain regions, gene functions like neuroimmunology, and phenotypes.
Collapse
|
49
|
Xu X, Du L, Jiang J, Yang M, Wang Z, Wang Y, Tang T, Fu X, Hao J. Microglial TREM2 Mitigates Inflammatory Responses and Neuronal Apoptosis in Angiotensin II-Induced Hypertension in Middle-Aged Mice. Front Aging Neurosci 2021; 13:716917. [PMID: 34489683 PMCID: PMC8417947 DOI: 10.3389/fnagi.2021.716917] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/16/2021] [Indexed: 11/15/2022] Open
Abstract
Growing evidence suggests that hypertension and aging are prominent risk factors for the development of late-onset Alzheimer's disease (LOAD) by inducement of neuroinflammation. Recent study showed that neuroinflammation via activated microglia induces reactive astrocytes, termed A1 astrocytes, that highly upregulate numerous classical complement cascade genes that are destructive to neurons in neurodegeneration diseases. Moreover, triggering receptor expressed on myeloid cells 2 (TREM2) is considered as one of the strongest single-allele genetic risk factors and plays important roles in neuroinflammation for LOAD. However, the mechanisms of microglia in the regulation of A1 astrocytic activation are still not clear. We introduced angiotensin II-induced hypertension in middle-aged mice and found that hypertension-upregulated TREM2 expression and A1 astrocytic activation were involved in neuroinflammation in the animal models used in this study. The in vitro results revealed that overexpression of microglial TREM2 not only mitigated microglial inflammatory response but also had salutary effects on reverse A1 astrocytic activation and neuronal toxicity.
Collapse
Affiliation(s)
- Xiaotian Xu
- Department of Neurology, The Affiliated Hospital, Yangzhou University, Yangzhou, China
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Lin Du
- Department of Cardiology, The Affiliated Hospital, Yangzhou University, Yangzhou, China
| | - Jianxiong Jiang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ming Yang
- Department of Neurology, The Affiliated Hospital, Yangzhou University, Yangzhou, China
| | - Zhaoxia Wang
- Department of Neurology, The Affiliated Hospital, Yangzhou University, Yangzhou, China
| | - Yingge Wang
- Department of Neurology, The Affiliated Hospital, Yangzhou University, Yangzhou, China
| | - Tieyu Tang
- Department of Neurology, The Affiliated Hospital, Yangzhou University, Yangzhou, China
| | - Xuetao Fu
- Department of Neurology, The Affiliated Hospital, Yangzhou University, Yangzhou, China
| | - Jiukuan Hao
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| |
Collapse
|
50
|
Kim YJ, Soto M, Branigan GL, Rodgers K, Brinton RD. Association between menopausal hormone therapy and risk of neurodegenerative diseases: Implications for precision hormone therapy. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12174. [PMID: 34027024 PMCID: PMC8118114 DOI: 10.1002/trc2.12174] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/18/2021] [Accepted: 03/12/2021] [Indexed: 01/03/2023]
Abstract
INTRODUCTION The impact of menopausal hormone therapy (HT) on age-associated Alzheimer's and neurodegenerative diseases (NDDs) remains unresolved. To determine the effect of HT, formulation, type, and duration on risk of NDDs, a retrospective analysis was performed using a 10-year Humana claims dataset. METHODS Study population included women aged 45 years or older with or without claim records of HT medications. Patients diagnosed with NDDs including Alzheimer's disease (AD), Parkinson's disease (PD), dementia, multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS) were identified. Relative risk (RR) ratios and 95% confidence intervals (CI) for combined NDDs, or AD, PD, dementia, MS, and ALS were determined. Cumulative hazard ratios were determined to investigate the association between HT and NDDs at different age groups. RESULTS In 379,352 women with or without claim records of HT, use of HT was associated with significantly reduced risk for combined NDDs (RR 0.42, 95% CI 0.40-0.43, P < 0.001). Average follow-up time was 5.1 [2.3] years. Formulations containing natural steroids 17β-estradiol and/or progesterone were associated with greater reduction in NDD risk. Oral- HT users showed significantly reduced RRs (0.42, 0.41-0.44, P < 0.001) for combined NDDs compared to non-HT users. The RRs for transdermal-HT users were significantly decreased for all-cause dementia (0.73, 0.60-0.88, P = 0.001) and MS (0.55, 0.36-0.84, P = 0.005). Greatest reduction in risk of NDD, AD, and dementia emerged in patients aged 65 years or older. Further, the protective effect of long-term therapy (>1 year) on combined NDDs, AD, PD, and dementia was greater compared to short-term therapy (≤1 year). DISCUSSION HT was associated with reduced risk of all NDDs including AD and dementia, with greater duration of therapy and natural steroid formulations associated with greater efficacy. These findings advance precision HT to prevent NDDs including AD.
Collapse
Affiliation(s)
- Yu Jin Kim
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
| | - Maira Soto
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of PharmacologyUniversity of ArizonaTucsonArizonaUSA
| | - Gregory L Branigan
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of PharmacologyUniversity of ArizonaTucsonArizonaUSA
- College of MedicineMD‐PhD Training ProgramUniversity of ArizonaTucsonArizonaUSA
| | - Kathleen Rodgers
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of PharmacologyUniversity of ArizonaTucsonArizonaUSA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of PharmacologyUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of NeurologyUniversity of ArizonaTucsonArizonaUSA
| |
Collapse
|