1
|
Agostinho D, Simões M, Castelo-Branco M. Predicting conversion from mild cognitive impairment to Alzheimer's disease: a multimodal approach. Brain Commun 2024; 6:fcae208. [PMID: 38961871 PMCID: PMC11220508 DOI: 10.1093/braincomms/fcae208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/09/2024] [Accepted: 06/12/2024] [Indexed: 07/05/2024] Open
Abstract
Successively predicting whether mild cognitive impairment patients will progress to Alzheimer's disease is of significant clinical relevance. This ability may provide information that can be leveraged by emerging intervention approaches and thus mitigate some of the negative effects of the disease. Neuroimaging biomarkers have gained some attention in recent years and may be useful in predicting the conversion of mild cognitive impairment to Alzheimer's disease. We implemented a novel multi-modal approach that allowed us to evaluate the potential of different imaging modalities, both alone and in different degrees of combinations, in predicting the conversion to Alzheimer's disease of mild cognitive impairment patients. We applied this approach to the imaging data from the Alzheimer's Disease Neuroimaging Initiative that is a multi-modal imaging dataset comprised of MRI, Fluorodeoxyglucose PET, Florbetapir PET and diffusion tensor imaging. We included a total of 480 mild cognitive impairment patients that were split into two groups: converted and stable. Imaging data were segmented into atlas-based regions of interest, from which relevant features were extracted for the different imaging modalities and used to construct machine-learning models to classify mild cognitive impairment patients into converted or stable, using each of the different imaging modalities independently. The models were then combined, using a simple weight fusion ensemble strategy, to evaluate the complementarity of different imaging modalities and their contribution to the prediction accuracy of the models. The single-modality findings revealed that the model, utilizing features extracted from Florbetapir PET, demonstrated the highest performance with a balanced accuracy of 83.51%. Concerning multi-modality models, not all combinations enhanced mild cognitive impairment conversion prediction. Notably, the combination of MRI with Fluorodeoxyglucose PET emerged as the most promising, exhibiting an overall improvement in predictive capabilities, achieving a balanced accuracy of 78.43%. This indicates synergy and complementarity between the two imaging modalities in predicting mild cognitive impairment conversion. These findings suggest that β-amyloid accumulation provides robust predictive capabilities, while the combination of multiple imaging modalities has the potential to surpass certain single-modality approaches. Exploring modality-specific biomarkers, we identified the brainstem as a sensitive biomarker for both MRI and Fluorodeoxyglucose PET modalities, implicating its involvement in early Alzheimer's pathology. Notably, the corpus callosum and adjacent cortical regions emerged as potential biomarkers, warranting further study into their role in the early stages of Alzheimer's disease.
Collapse
Affiliation(s)
- Daniel Agostinho
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), ICNAS, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Faculty of Science and Technology, Centre for Informatics and Systems of the University of Coimbra (CISUC), 3030-790 Coimbra, Portugal
- Intelligent Systems Associate Laboratory (LASI), 4800-058 Guimarães, Portugal
| | - Marco Simões
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), ICNAS, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Faculty of Science and Technology, Centre for Informatics and Systems of the University of Coimbra (CISUC), 3030-790 Coimbra, Portugal
- Intelligent Systems Associate Laboratory (LASI), 4800-058 Guimarães, Portugal
| | - Miguel Castelo-Branco
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), ICNAS, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Intelligent Systems Associate Laboratory (LASI), 4800-058 Guimarães, Portugal
| |
Collapse
|
2
|
Noche JA, Radhakrishnan H, Ubele MF, Boaz K, Mefford JL, Jones ED, van Rooyen HY, Perpich JA, McCarty K, Meacham B, Smiley J, Bembenek Bailey SA, Puskás LG, Powell DK, Sordo L, Phelan MJ, Norris CM, Head E, Stark CEL. Age-Related Brain Atrophy and the Positive Effects of Behavioral Enrichment in Middle-Aged Beagles. J Neurosci 2024; 44:e2366232024. [PMID: 38561226 PMCID: PMC11097262 DOI: 10.1523/jneurosci.2366-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/08/2024] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Aging dogs serve as a valuable preclinical model for Alzheimer's disease (AD) due to their natural age-related development of β-amyloid (Aβ) plaques, human-like metabolism, and large brains that are ideal for studying structural brain aging trajectories from serial neuroimaging. Here we examined the effects of chronic treatment with the calcineurin inhibitor (CNI) tacrolimus or the nuclear factor of activated T cells (NFAT)-inhibiting compound Q134R on age-related canine brain atrophy from a longitudinal study in middle-aged beagles (36 females, 7 males) undergoing behavioral enrichment. Annual MRI was analyzed using modern, automated techniques for region-of-interest-based and voxel-based volumetric assessments. We found that the frontal lobe showed accelerated atrophy with age, while the caudate nucleus remained relatively stable. Remarkably, the hippocampus increased in volume in all dogs. None of these changes were influenced by tacrolimus or Q134R treatment. Our results suggest that behavioral enrichment can prevent atrophy and increase the volume of the hippocampus but does not prevent aging-associated prefrontal cortex atrophy.
Collapse
Affiliation(s)
| | - Hamsanandini Radhakrishnan
- University of California, Irvine, California 92697
- University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | | | - Kathy Boaz
- University of Kentucky, Lexington, Kentucky 40506
| | | | - Erin D Jones
- University of Kentucky, Lexington, Kentucky 40506
| | | | | | | | | | | | | | | | | | - Lorena Sordo
- University of California, Irvine, California 92697
| | | | | | | | | |
Collapse
|
3
|
Al-Kuraishy HM, Jabir MS, Al-Gareeb AI, Albuhadily AK, Albukhaty S, Sulaiman GM, Batiha GES. Evaluation and targeting of amyloid precursor protein (APP)/amyloid beta (Aβ) axis in amyloidogenic and non-amyloidogenic pathways: A time outside the tunnel. Ageing Res Rev 2023; 92:102119. [PMID: 37931848 DOI: 10.1016/j.arr.2023.102119] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023]
Abstract
In Alzheimer disease (AD), amyloid precursor protein (APP) and production of amyloid beta (Aβ) which is generated by amyloidogenic pathway is implicated in neurotoxicity and neuronal cell deaths. However, physiological Aβ level is essential to improves neuronal survival, attenuates neuronal apoptosis and has neuroprotective effect. In addition, physiological APP level has neurotrophic effect on the central nervous system (CNS). APP has a critical role in the brain growth and development via activation of long-term potentiation (LTP) and acceleration of neurite outgrowth. Moreover, APP is cleaved by α secretase to form a neuroprotective soluble APP alpha (sAPPα) in non-amyloidogenic pathway. Consequently, this mini-review purposes to highlight the possible beneficial role of APP and Aβ. In addition, this mini-review discussed the modulation of APP processing and Aβ production.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Majid S Jabir
- Department of Applied science, University of Technology, Iraq.
| | - Ali I Al-Gareeb
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali K Albuhadily
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Salim Albukhaty
- Department of Chemistry, College of Science, University of Misan, Maysan 62001, Iraq
| | | | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira 22511, Egypt
| |
Collapse
|
4
|
Rao RV, Subramaniam KG, Gregory J, Bredesen AL, Coward C, Okada S, Kelly L, Bredesen DE. Rationale for a Multi-Factorial Approach for the Reversal of Cognitive Decline in Alzheimer's Disease and MCI: A Review. Int J Mol Sci 2023; 24:ijms24021659. [PMID: 36675177 PMCID: PMC9865291 DOI: 10.3390/ijms24021659] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/03/2023] [Accepted: 01/09/2023] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's disease (AD) is a multifactorial, progressive, neurodegenerative disease typically characterized by memory loss, personality changes, and a decline in overall cognitive function. Usually manifesting in individuals over the age of 60, this is the most prevalent type of dementia and remains the fifth leading cause of death among Americans aged 65 and older. While the development of effective treatment and prevention for AD is a major healthcare goal, unfortunately, therapeutic approaches to date have yet to find a treatment plan that produces long-term cognitive improvement. Drugs that may be able to slow down the progression rate of AD are being introduced to the market; however, there has been no previous solution for preventing or reversing the disease-associated cognitive decline. Recent studies have identified several factors that contribute to the progression and severity of the disease: diet, lifestyle, stress, sleep, nutrient deficiencies, mental health, socialization, and toxins. Thus, increasing evidence supports dietary and other lifestyle changes as potentially effective ways to prevent, slow, or reverse AD progression. Studies also have demonstrated that a personalized, multi-therapeutic approach is needed to improve metabolic abnormalities and AD-associated cognitive decline. These studies suggest the effects of abnormalities, such as insulin resistance, chronic inflammation, hypovitaminosis D, hormonal deficiencies, and hyperhomocysteinemia, in the AD process. Therefore a personalized, multi-therapeutic program based on an individual's genetics and biochemistry may be preferable over a single-drug/mono-therapeutic approach. This article reviews these multi-therapeutic strategies that identify and attenuate all the risk factors specific to each affected individual. This article systematically reviews studies that have incorporated multiple strategies that target numerous factors simultaneously to reverse or treat cognitive decline. We included high-quality clinical trials and observational studies that focused on the cognitive effects of programs comprising lifestyle, physical, and mental activity, as well as nutritional aspects. Articles from PubMed Central, Scopus, and Google Scholar databases were collected, and abstracts were reviewed for relevance to the subject matter. Epidemiological, pathological, toxicological, genetic, and biochemical studies have all concluded that AD represents a complex network insufficiency. The research studies explored in this manuscript confirm the need for a multifactorial approach to target the various risk factors of AD. A single-drug approach may delay the progression of memory loss but, to date, has not prevented or reversed it. Diet, physical activity, sleep, stress, and environment all contribute to the progression of the disease, and, therefore, a multi-factorial optimization of network support and function offers a rational therapeutic strategy. Thus, a multi-therapeutic program that simultaneously targets multiple factors underlying the AD network may be more effective than a mono-therapeutic approach.
Collapse
Affiliation(s)
- Rammohan V. Rao
- Apollo Health, Burlingame, CA 94011, USA
- Correspondence: (R.V.R.); (D.E.B.)
| | | | | | | | | | - Sho Okada
- Apollo Health, Burlingame, CA 94011, USA
| | | | - Dale E. Bredesen
- Apollo Health, Burlingame, CA 94011, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90024, USA
- Correspondence: (R.V.R.); (D.E.B.)
| |
Collapse
|
5
|
Sandison H, Callan NG, Rao RV, Phipps J, Bradley R. Observed Improvement in Cognition During a Personalized Lifestyle Intervention in People with Cognitive Decline. J Alzheimers Dis 2023; 94:993-1004. [PMID: 37355891 PMCID: PMC10473097 DOI: 10.3233/jad-230004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a chronic condition marked by progressive objective cognitive impairment (OCI). No monotherapy has substantially altered disease progression, suggesting the disease is multifactorial and may require a multimodal therapeutic approach. OBJECTIVE We sought to determine if cognitive function in a sample with OCI would change in response to a multimodal, individualized care plan based on potential contributors to cognitive decline (e.g., nutritional status, infection, etc.). METHODS Participants (n = 34) were recruited from the San Diego, CA area. The multimodal intervention included lifestyle changes (i.e., movement, diet, and stress management), nutraceutical support, and medications. It was delivered pragmatically over four clinical visits, and outcome measures were gathered at four study visits, occurring at baseline, one, three, and six months (primary endpoint). Study participants received weekly phone calls for nutrition support throughout study participation. Outcome measures included the Cambridge Brain Sciences (CBS) battery, and the Montreal Cognitive Assessment (MoCA). RESULTS At 6 months, mean MoCA scores improved from 19.6±3.1 to 21.7±6.2 (p = 0.013). Significant improvement was observed in mean scores of the CBS memory domain [25.2 (SD 23.3) to 35.8 (SD 26.9); p < 0.01] and CBS overall composite cognition score [24.5 (SD 16.1) to 29.7 (SD 20.5); p = 0.02]. All CBS domains improved. CONCLUSION Multiple measures of cognitive function improved after six months of intervention. Our results support the feasibility and impact of a multimodal, individualized treatment approach to OCI, warranting further research.
Collapse
Affiliation(s)
| | - Nini G.L. Callan
- Helfgott Research Institute, National University of Natural Medicine, Portland, OR, USA
| | | | - John Phipps
- Helfgott Research Institute, National University of Natural Medicine, Portland, OR, USA
| | - Ryan Bradley
- Helfgott Research Institute, National University of Natural Medicine, Portland, OR, USA
- Herbert Wertheim School of Public Health and Human Longevity Sciences, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
6
|
Sidenkova A, Calabrese V, Tomasello M, Fritsch T. Subjective cognitive decline and cerebral-cognitive reserve in late age. TRANSLATIONAL MEDICINE OF AGING 2023; 7:137-147. [DOI: 10.1016/j.tma.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2024] Open
|
7
|
Perus L, Busto GU, Mangin JF, Le Bars E, Gabelle A. Effects of preventive interventions on neuroimaging biomarkers in subjects at-risk to develop Alzheimer's disease: A systematic review. Front Aging Neurosci 2022; 14:1014559. [PMID: 36506466 PMCID: PMC9730537 DOI: 10.3389/fnagi.2022.1014559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/01/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer's Disease (AD) is a multifactorial and complex neurodegenerative disorder. Some modifiable risk factors have been associated with an increased risk of appearance of the disease and/or cognitive decline. Preventive clinical trials aiming at reducing one or combined risk factors have been implemented and their potential effects assessed on cognitive trajectories and on AD biomarkers. However, the effect of interventions on surrogate markers, in particular imaging biomarkers, remains poorly understood. We conducted a review of the literature and analyzed 43 interventional studies that included physical exercise, nutrition, cognitive training or multidomain interventions, and assessed various brain imaging biomarkers, to determine the effects of preventive interventions on imaging biomarkers for subjects at-risk to develop AD. Deciphering the global and regional brain effect of each and combined interventions will help to better understand the interplay relationship between multimodal interventions, cognition, surrogate brain markers, and to better design primary and secondary outcomes for future preventive clinical trials. Those studies were pondered using generally-admitted quality criteria to reveal that interventions may affect the brain of patients with cognitive impairment rather than those without cognitive impairment thus indicating that particular care should be taken when selecting individuals for interventions. Additionally, a majority of the studies concurred on the effect of the interventions and particularly onto the frontal brain areas.
Collapse
Affiliation(s)
- Lisa Perus
- INM, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
- Department of Neurology, Memory Resources and Research Center, Gui de Chauliac Hospital, Montpellier, France
- Institut d'Imagerie Fonctionnelle Humaine, I2FH, Department of Neuroradiology, Gui de Chauliac Hospital and University of Montpellier, Montpellier, France
- CATI, US52-UAR2031, CEA, ICM, SU, CNRS, INSERM, APHP, Ile de France, France
| | - Germain U. Busto
- INM, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
- Department of Neurology, Memory Resources and Research Center, Gui de Chauliac Hospital, Montpellier, France
| | - Jean-François Mangin
- CATI, US52-UAR2031, CEA, ICM, SU, CNRS, INSERM, APHP, Ile de France, France
- Université Paris-Saclay, CEA, CNRS, Neurospin, UMR9027 Baobab, Gif-sur-Yvette, France
| | - Emmanuelle Le Bars
- Institut d'Imagerie Fonctionnelle Humaine, I2FH, Department of Neuroradiology, Gui de Chauliac Hospital and University of Montpellier, Montpellier, France
| | - Audrey Gabelle
- INM, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
- Department of Neurology, Memory Resources and Research Center, Gui de Chauliac Hospital, Montpellier, France
| |
Collapse
|
8
|
Treyer V, Meyer RS, Buchmann A, Crameri GAG, Studer S, Saake A, Gruber E, Unschuld PG, Nitsch RM, Hock C, Gietl AF. Physical activity is associated with lower cerebral beta-amyloid and cognitive function benefits from lifetime experience-a study in exceptional aging. PLoS One 2021; 16:e0247225. [PMID: 33606797 PMCID: PMC7895362 DOI: 10.1371/journal.pone.0247225] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 02/03/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Exceptional agers (85+ years) are characterized by preserved cognition presumably due to high cognitive reserve. In the current study, we examined whether personality, risk and protective factors for dementia as well as quality of life are associated with core features of Alzheimer's disease (amyloid-deposition and hippocampal volume) as well as cognition in exceptional aging. METHODS We studied 49 exceptional agers (average 87.8 years, range 84-94 years), with preserved activities of daily living and absence of dementia. All participants received a detailed clinical and neuropsychological examination. We used established questionnaires to measure lifetime experience, personality, recent physical and cognitive activity as well as quality of life. Cerebral amyloid-deposition was estimated by 18-[F]-Flutemetamol-PET and manual hippocampal volumetry was performed on 3D T1 MRI images. RESULTS In this sample of exceptional agers with preserved activities of daily living, we found intact cognitive performance in the subjects with the highest amyloid-load in the brain, but a lower quality of life with respect to autonomy as well as higher neuroticism. Higher self-reported physical activity in the last twelve months went with a lower amyloid load. Higher self-reported leisure-time/ not work-related activity went with better executive functioning at older age. CONCLUSION Even in exceptional aging, high amyloid load may subtly influence personality and quality of life. Our findings support a close relationship between high physical activity and low amyloid-deposition and underscore the importance of extracurricular activities for executive functions. As executive functions are known to be a central resource for everyday functioning in fostering extracurricular activities may be effective in delaying the onset of dementia.
Collapse
Affiliation(s)
- Valerie Treyer
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Department of Nuclear Medicine, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Rafael S. Meyer
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Andreas Buchmann
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | | | - Sandro Studer
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Antje Saake
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Esmeralda Gruber
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Paul G. Unschuld
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Hospital for Psychogeriatric Medicine, Psychiatric University Hospital Zurich, Zurich, Switzerland
| | - Roger M. Nitsch
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Neurimmune, Schlieren-Zurich, Switzerland
| | - Christoph Hock
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Neurimmune, Schlieren-Zurich, Switzerland
| | - Anton F. Gietl
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| |
Collapse
|
9
|
Mobile Application for Monitoring and Preventing Cognitive Decline Through Lifestyle Intervention. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1338:89-96. [DOI: 10.1007/978-3-030-78775-2_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
10
|
Wang Z, Meng L, Liu H, Shen L, Ji HF. AlzRiskMR database: an online database for the impact of exposure factors on Alzheimer's disease. Brief Bioinform 2020; 22:5909008. [PMID: 32951050 DOI: 10.1093/bib/bbaa213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/22/2020] [Accepted: 08/12/2020] [Indexed: 11/12/2022] Open
Abstract
In view of great difficulties in the pathogenesis analysis of Alzheimer's disease (AD) presently, profiling the modifiable risk factors is crucial for early detection and intervention of AD. However, the causal associations among them have yet to be identified, and the effective integration and application of these data also remain considerable challenges due to the lack of efficient collection and analysis procedures. To address this issue, we performed comprehensive analyses by two-sample Mendelian randomization (2SMR) and established the AlzRiskMR database (https://github.com/SDBMC/RiskFactors2AD). Four 2SMR analysis methods, including inverse variance weighted (IVW), MR-Egger, weighted median, and weighted mode, were used for the complementary calculation to test the reliability of the results. The database currently comprises 1870 sets of data of Genome-Wide Association Studies (GWAS) from the MR-Base and NHGRI-EBI GWAS Catalog database. AlzRiskMR database not only estimates causal associations between modifiable risk factors and AD but also offers a useful and timely resource for early intervention of AD development incidence.
Collapse
Affiliation(s)
- Zhe Wang
- Shandong Provincial Research Center for Bioinformatic Engineering and Technique, Institute of Biomedical Research, School of Life Sciences, Shandong University of Technology, Zibo, Shandong, China
| | - Lei Meng
- Shandong Provincial Research Center for Bioinformatic Engineering and Technique, Institute of Biomedical Research, School of Life Sciences, Shandong University of Technology, Zibo, Shandong, China
| | - Hong Liu
- Shandong Provincial Research Center for Bioinformatic Engineering and Technique, Institute of Biomedical Research, School of Life Sciences, Shandong University of Technology, Zibo, Shandong, China
| | - Liang Shen
- Shandong Provincial Research Center for Bioinformatic Engineering and Technique, Institute of Biomedical Research, School of Life Sciences, Shandong University of Technology, Zibo, Shandong, China
| | - Hong-Fang Ji
- Shandong Provincial Research Center for Bioinformatic Engineering and Technique, Institute of Biomedical Research, School of Life Sciences, Shandong University of Technology, Zibo, Shandong, China
| |
Collapse
|
11
|
Barbaresko J, Lellmann AW, Schmidt A, Lehmann A, Amini AM, Egert S, Schlesinger S, Nöthlings U. Dietary Factors and Neurodegenerative Disorders: An Umbrella Review of Meta-Analyses of Prospective Studies. Adv Nutr 2020; 11:1161-1173. [PMID: 32427314 PMCID: PMC7490166 DOI: 10.1093/advances/nmaa053] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/13/2020] [Accepted: 04/17/2020] [Indexed: 12/16/2022] Open
Abstract
Diet has been hypothesized to be associated with neurodegenerative disorders. The aim was to conduct an umbrella review to summarize and evaluate the current evidence of prospective associations between any dietary factors and the incidence of neurodegenerative disorders. We conducted a systematic search in PubMed, Embase, and the Cochrane library up to November 2019 to identify systematic reviews with meta-analyses of prospective studies investigating the association between dietary factors (dietary patterns, foods and beverages, nutrients, and phytochemicals) and neurodegenerative disorders (cognitive decline, cognitive impairment, Alzheimer disease, all-cause dementia, and Parkinson disease). Summary risk ratios (SRRs) and 95% CIs were recalculated using a random effects model. We evaluated the risk of bias of identified meta-analyses and the quality of evidence for all associations. In total, 20 meta-analyses including 98 SRRs were identified. All original meta-analyses were rated as being at high risk of bias. Methodological concerns related mainly to the inappropriate synthesis, assessment, and discussion of the risk of bias of primary studies. For the recalculated meta-analyses, quality of evidence was moderate for inverse associations between higher adherence to the Mediterranean diet (SRR: 0.63; 95% CI: 0.48, 0.82; n = 4 primary studies) and higher fish intake (SRR: 0.72; 95% CI: 0.59, 0.89; n = 6) and Alzheimer disease, as well as for tea consumption and all-cause dementia (SRR: 0.74; 95% CI: 0.63, 0.88; n = 2) and Parkinson disease (SRR per 2 cups/d: 0.69; 95% CI: 0.54, 0.87; n = 5). This umbrella review provides a comprehensive overview of the available evidence on dietary factors and neurodegenerative disorders. The results indicate that the Mediterranean diet, fish, and tea could be inversely associated with neurodegenerative disorders. However, the quality of evidence was generally low, suggesting that further studies are likely to change the overall estimates. Thus, more well-conducted research, also investigating other dietary factors in association with neurodegenerative disorders, is warranted.
Collapse
Affiliation(s)
- Janett Barbaresko
- German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany,Department of Nutrition and Food Sciences, Nutritional Epidemiology, University of Bonn, Bonn, Germany,Address correspondence to JB (e-mail: )
| | - Arno Werner Lellmann
- Department of Nutrition and Food Sciences, Nutritional Epidemiology, University of Bonn, Bonn, Germany,German Nutrition Society, Bonn, Germany
| | | | | | | | - Sarah Egert
- German Nutrition Society, Bonn, Germany,Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Sabrina Schlesinger
- German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ute Nöthlings
- Department of Nutrition and Food Sciences, Nutritional Epidemiology, University of Bonn, Bonn, Germany
| |
Collapse
|
12
|
De Plano LM, Carnazza S, Franco D, Rizzo MG, Conoci S, Petralia S, Nicoletti A, Zappia M, Campolo M, Esposito E, Cuzzocrea S, Guglielmino SPP. Innovative IgG Biomarkers Based on Phage Display Microbial Amyloid Mimotope for State and Stage Diagnosis in Alzheimer's Disease. ACS Chem Neurosci 2020; 11:1013-1026. [PMID: 32176482 PMCID: PMC7997372 DOI: 10.1021/acschemneuro.9b00549] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
![]()
An
innovative approach to identify new conformational antigens
of Aβ1–42 recognized by IgG autoantibodies
as biomarkers of state and stage in Alzheimer’s disease (AD)
patients is described. In particular, through the use of bioinformatics
modeling, conformational similarities between several Aβ1–42 forms and other amyloid-like proteins with F1 capsular
antigen (Caf1) of Yersinia pestis were first found.
pVIII M13 phage display libraries were then screened against YPF19,
anti-Caf1 monoclonal antibody, and IgGs of AD patients, in alternate
biopanning cycles of a so-called “double binding” selection.
From the selected phage clones, one, termed 12III1, was found to be
able to prevent in vitro Aβ1–42-induced cytotoxicity in SH-SY5Y cells, as well as to promote disaggregation
of preformed fibrils, to a greater extent with respect to wild-type
phage (pC89). IgG levels detected by 12III1 provided a significant
level of discrimination between diseased and nondemented subjects,
as well as a good correlation with the state progression of the disease.
These results give significant impact in AD state and stage diagnosis,
paving the way for the development not only for an innovative blood
diagnostic assay for AD precise diagnosis, progressive clinical assessment,
and screening but also for new effective treatments.
Collapse
Affiliation(s)
- Laura M. De Plano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Santina Carnazza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Domenico Franco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Maria Giovanna Rizzo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Sabrina Conoci
- STmicroelectronics, Stradale Primosole, 50, 95121 Catania, Italy
- Distretto Tecnologico Micro e Nano Sistemi Sicilia, Strada VII-Zona Industriale, 95121 Catania, Italy
| | | | - Alessandra Nicoletti
- Neurology Clinic, Department “G.F. Ingrassia”, Section of Neurosciences, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy
| | - Mario Zappia
- Neurology Clinic, Department “G.F. Ingrassia”, Section of Neurosciences, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy
| | - Michela Campolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Salvatore P. P. Guglielmino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
13
|
Ghaffari M, Sanadgol N, Abdollahi M. A Systematic Review of Current Progresses in the Nucleic Acid-Based Therapies for Neurodegeneration with Implications for Alzheimer's Disease. Mini Rev Med Chem 2020; 20:1499-1517. [PMID: 32400332 DOI: 10.2174/1389557520666200513122357] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/31/2020] [Accepted: 04/20/2020] [Indexed: 12/30/2022]
Abstract
Recently, manipulation of gene expression and switching genes on or off highlight the potential of nucleic acid-based therapies (NA-BTs). Alzheimer's Disease (AD) is a common devastating neurodegenerative disease (NDs) responsible for 60-80% of all cases of dementia and predicted as a main public health concern among aged populations. The aim of this study was to outline the current research in the field of NA-BTs for the treatment of AD disabilities, including strategies to suppress the memory and learning defects, to promote recovery processes, and to reinforce social relationships in these patients. This review was performed via evaluating PubMed reported studies from January 2010 to November 2019. Also, reference lists were checked to find additional studies. All intermediation or complementarity of animal models, case-control and cohort studies, and controlled trials (CTs) on specific NA-BTs to AD were acceptable, although in vitro studies were excluded due to the considerable diversities and heterogeneities. After removing the duplicates according to preferred reporting items for systematic reviews and meta-analyses (PRISMA) instruction, we merged remaining titles across search databases. There are 48 ongoing studies related to the application of nucleic acids in the treatment and diagnosis of AD where more consideration is given to DNA targeting strategies (18 targets for vectors and aptamers), antisense oligonucleotides (10 targets), micro-RNAs mimics (7 targets), antagomiRs (6 targets), small interferences-RNAs (5 targets), as well as mRNAs (2 targets) respectively. All of these targets are grouped into 4 categories according to their role in molecular pathways where amyloid-β (18 targets), neural survival (11 targets), memory and cognition (8 targets), and tau (3 targets) are more targeted pathways, respectively. With recent successes in the systemic delivery of nucleic acids via intravenous injection; it is worth investing in the production of new-generation medicines. There are still several challenges for NA-BTs including, their delivery to the effective modulators, mass production at low cost, sustaining efficacy and minimizing off-target effects. Regarding miRNA-based therapies, given the obvious involvement of miRNAs in numerous facets of brain disease, and the many sophisticated techniques for delivery to the brain, miRNA-based therapies will make new hope for the treatment of neurological diseases such as AD.
Collapse
Affiliation(s)
- Maryam Ghaffari
- Department of Biology, Faculty of Sciences, University of Zabol, Zabol, Iran
| | - Nima Sanadgol
- Department of Biology, Faculty of Sciences, University of Zabol, Zabol, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Department of Toxicology and Pharmacology, School of Pharmacy, Tehran University of Medical Sciences, Iran
| |
Collapse
|
14
|
Lue LF, Kuo YM, Sabbagh M. Advance in Plasma AD Core Biomarker Development: Current Findings from Immunomagnetic Reduction-Based SQUID Technology. Neurol Ther 2019; 8:95-111. [PMID: 31833027 PMCID: PMC6908530 DOI: 10.1007/s40120-019-00167-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Indexed: 11/28/2022] Open
Abstract
New super-sensitive biomarker assay platforms for measuring Alzheimer's disease (AD) core pathological markers in plasma have recently been developed and tested. Research findings from these technologies offer promising evidence for identifying the earliest stages of AD and correlating them with brain pathological progression. Here, we review findings using immunomagnetic reduction, one of these ultrasensitive technologies. The principles, technology and assays developed, along with selected published findings will be discussed. The major findings from this technology were significant increases of amyloid beta (Aβ) 42 and total tau (t-tau) levels in subjects clinically diagnosed with early AD when compared with cognitively normal control (NC) subjects. The composite marker of the product of Aβ42 and t-tau discriminated subjects with early AD from NC subjects with high accuracy. The potential of this technology for the purpose of early or preclinical disease stage detection has yet to be explored in subjects who have also been assessed with brain imaging and cerebrospinal fluid AD core biomarker measurements.
Collapse
Affiliation(s)
- Lih-Fen Lue
- Civin Neuropathology Laboratory, Banner Sun Health Research Institute, 10515 West Santa Fe Drive, Sun City, AZ, 85351, USA.
- School of Life Sciences, Arizona State University, 427 East Tyler Mall, Tempe, AZ, 85281, USA.
| | - Yu-Min Kuo
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University Medical School, 1 Dasyue Road, Tainan, Taiwan
| | - Marwan Sabbagh
- Cleveland Clinic Lou Ruvo Center for Brain Health, 888 W Bonneville Ave, Las Vegas, NV, 89106, USA
| |
Collapse
|
15
|
Abstract
Background Alzheimer’s disease (AD) imposes a heavy burden on society and every family. Therefore, diagnosing AD in advance and discovering new drug targets are crucial, while these could be achieved by identifying AD-related proteins. The time-consuming and money-costing biological experiment makes researchers turn to develop more advanced algorithms to identify AD-related proteins. Results Firstly, we proposed a hypothesis “similar diseases share similar related proteins”. Therefore, five similarity calculation methods are introduced to find out others diseases which are similar to AD. Then, these diseases’ related proteins could be obtained by public data set. Finally, these proteins are features of each disease and could be used to map their similarity to AD. We developed a novel method ‘LRRGD’ which combines Logistic Regression (LR) and Gradient Descent (GD) and borrows the idea of Random Forest (RF). LR is introduced to regress features to similarities. Borrowing the idea of RF, hundreds of LR models have been built by randomly selecting 40 features (proteins) each time. Here, GD is introduced to find out the optimal result. To avoid the drawback of local optimal solution, a good initial value is selected by some known AD-related proteins. Finally, 376 proteins are found to be related to AD. Conclusion Three hundred eight of three hundred seventy-six proteins are the novel proteins. Three case studies are done to prove our method’s effectiveness. These 308 proteins could give researchers a basis to do biological experiments to help treatment and diagnostic AD.
Collapse
Affiliation(s)
- Tianyi Zhao
- School of Life Science and Technology, Department of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yang Hu
- School of Life Science and Technology, Department of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Tianyi Zang
- School of Life Science and Technology, Department of Computer Science and Technology, Harbin Institute of Technology, Harbin, China.
| | - Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
16
|
Levy B, Hess C, Hogan J, Hogan M, Ellison JM, Greenspan S, Elber A, Falcon K, Driscoll DF, Hashmi AZ. Machine Learning Enhances the Efficiency of Cognitive Screenings for Primary Care. J Geriatr Psychiatry Neurol 2019; 32:137-144. [PMID: 30879363 DOI: 10.1177/0891988719834349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Incorporation of cognitive screening into the busy primary care will require the development of highly efficient screening tools. We report the convergence validity of a very brief, self-administered, computerized assessment protocol against one of the most extensively used, clinician-administered instruments-the Montreal Cognitive Assessment (MoCA). METHOD Two hundred six participants (mean age = 67.44, standard deviation [SD] = 11.63) completed the MoCA and the computerized test. Three machine learning algorithms (ie, Support Vector Machine, Random Forest, and Gradient Boosting Trees) were trained to classify participants according to the clinical cutoff score of the MoCA (ie, < 26) from participant performance on 25 features of the computerized test. Analysis employed Synthetic Minority Oversampling TEchnic to correct the sample for class imbalance. RESULTS Gradient Boosting Trees achieved the highest performance (accuracy = 0.81, specificity = 0.88, sensitivity = 0.74, F1 score = 0.79, and area under the curve = 0.81). A subsequent K-means clustering of the prediction features yielded 3 categories that corresponded to the unimpaired (mean = 26.98, SD = 2.35), mildly impaired (mean = 23.58, SD = 3.19), and moderately impaired (mean = 17.24, SD = 4.23) ranges of MoCA score ( F = 222.36, P < .00). In addition, compared to the MoCA, the computerized test correlated more strongly with age in unimpaired participants (ie, MoCA ≥26, n = 165), suggesting greater sensitivity to age-related changes in cognitive functioning. CONCLUSION Future studies should examine ways to improve the sensitivity of the computerized test by expanding the cognitive domains it measures without compromising its efficiency.
Collapse
Affiliation(s)
- Boaz Levy
- 1 Department of Counseling and School Psychology, University of Massachusetts, Boston, MA, USA
| | - Courtney Hess
- 1 Department of Counseling and School Psychology, University of Massachusetts, Boston, MA, USA
| | - Jacqueline Hogan
- 1 Department of Counseling and School Psychology, University of Massachusetts, Boston, MA, USA
| | | | - James M Ellison
- 3 Christiana Care Health System, Department of Psychiatry and Human Behavior, Sidney Kimmel Medical College, Thomas Jefferson University, DE, USA
| | - Sarah Greenspan
- 1 Department of Counseling and School Psychology, University of Massachusetts, Boston, MA, USA
| | - Allison Elber
- 1 Department of Counseling and School Psychology, University of Massachusetts, Boston, MA, USA
| | - Kathryn Falcon
- 1 Department of Counseling and School Psychology, University of Massachusetts, Boston, MA, USA
| | | | | |
Collapse
|
17
|
Ishola IO, Osele MO, Chijioke MC, Adeyemi OO. Isorhamnetin enhanced cortico-hippocampal learning and memory capability in mice with scopolamine-induced amnesia: Role of antioxidant defense, cholinergic and BDNF signaling. Brain Res 2019; 1712:188-196. [PMID: 30772273 DOI: 10.1016/j.brainres.2019.02.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/10/2019] [Accepted: 02/14/2019] [Indexed: 12/22/2022]
Abstract
Isorhamnetin (IRN), a 3'-O-methylated metabolite of quercetin has antioxidant, anti-inflammatory and neuroprotective properties. In this study, we investigated the learning and memory enhancing effects of IRN on spatial and non-spatial learning and memory deficits induced by scopolamine (3 mg/kg, i.p; muscarinic antagonist) using the novel object recognition test (NORT) and Morris water maze (MWM) task. IRN (1, 5 or 50 mg/kg, p.o.) or vehicle was administered to male albino for 3 consecutive days, scopolamine was given 1 h after last administration on day 3. Five minutes post scopolamine administration the behavioural test of cognitive function was carried out. One hour after probe test (MWM task) on day 7, the brains were isolated to assay for oxidative stress, cholinesterase activity and brain derived neurotrophic factor (BDNF) levels in the prefrontal cortex (PFC) and hippocampus (HIPPO). IRN treatment significantly improved scopolamine-induced learning and memory impairment in behavioural tests. IRN reduced malondialdehyde and nitrite generation induced by scopolamine through increase in glutathione (GSH) level, superoxide dismutase (SOD) and catalase (CAT) activities in the prefrontal cortex and hippocampus. In addition, IRN attenuates scopolamine induced cholinesterase activity and BDNF level in the prefrontal cortex and hippocampus of mice. Findings from this study showed that IRN possesses cognition and memory enhancing properties possibly through enhancement of antioxidant defense system, cholinergic signaling and synaptic plasticity.
Collapse
Affiliation(s)
- Ismail O Ishola
- Department of Pharmacology, Therapeutics and Toxicology, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, PMB 12003, Surulere, Lagos State, Nigeria
| | - Mmesomachukwu O Osele
- Department of Pharmacology, Therapeutics and Toxicology, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, PMB 12003, Surulere, Lagos State, Nigeria
| | - Micah C Chijioke
- Department of Pharmacology, Therapeutics and Toxicology, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, PMB 12003, Surulere, Lagos State, Nigeria
| | - Olufunmilayo O Adeyemi
- Department of Pharmacology, Therapeutics and Toxicology, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, PMB 12003, Surulere, Lagos State, Nigeria
| |
Collapse
|
18
|
Morley JE, Farr SA, Nguyen AD, Xu F. Editorial: What is the Physiological Function of Amyloid-Beta Protein? J Nutr Health Aging 2019; 23:225-226. [PMID: 30820508 DOI: 10.1007/s12603-019-1162-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- J E Morley
- John E. Morley, MB, BCh, Division of Geriatric Medicine, Saint Louis University School of Medicine, 1402 S. Grand Blvd., M238, St. Louis, MO 63104,
| | | | | | | |
Collapse
|
19
|
Hampel H, O'Bryant SE, Molinuevo JL, Zetterberg H, Masters CL, Lista S, Kiddle SJ, Batrla R, Blennow K. Blood-based biomarkers for Alzheimer disease: mapping the road to the clinic. Nat Rev Neurol 2018; 14:639-652. [PMID: 30297701 PMCID: PMC6211654 DOI: 10.1038/s41582-018-0079-7] [Citation(s) in RCA: 452] [Impact Index Per Article: 64.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Biomarker discovery and development for clinical research, diagnostics and therapy monitoring in clinical trials have advanced rapidly in key areas of medicine - most notably, oncology and cardiovascular diseases - allowing rapid early detection and supporting the evolution of biomarker-guided, precision-medicine-based targeted therapies. In Alzheimer disease (AD), breakthroughs in biomarker identification and validation include cerebrospinal fluid and PET markers of amyloid-β and tau proteins, which are highly accurate in detecting the presence of AD-associated pathophysiological and neuropathological changes. However, the high cost, insufficient accessibility and/or invasiveness of these assays limit their use as viable first-line tools for detecting patterns of pathophysiology. Therefore, a multistage, tiered approach is needed, prioritizing development of an initial screen to exclude from these tests the high numbers of people with cognitive deficits who do not demonstrate evidence of underlying AD pathophysiology. This Review summarizes the efforts of an international working group that aimed to survey the current landscape of blood-based AD biomarkers and outlines operational steps for an effective academic-industry co-development pathway from identification and assay development to validation for clinical use.
Collapse
Affiliation(s)
- Harald Hampel
- AXA Research Fund and Sorbonne University Chair, Paris, France.
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France.
- Brain & Spine Institute (ICM), INSERM U 1127, Paris, France.
- Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, AP-HP, Pitié-Salpêtrière Hospital, Paris, France.
| | - Sid E O'Bryant
- University of North Texas Health Science Center, Fort Worth, TX, USA
| | - José L Molinuevo
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Melbourne, Australia
| | - Simone Lista
- AXA Research Fund and Sorbonne University Chair, Paris, France
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- Brain & Spine Institute (ICM), INSERM U 1127, Paris, France
- Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Steven J Kiddle
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| | | | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
| |
Collapse
|
20
|
Agrawal M, Saraf S, Saraf S, Antimisiaris SG, Chougule MB, Shoyele SA, Alexander A. Nose-to-brain drug delivery: An update on clinical challenges and progress towards approval of anti-Alzheimer drugs. J Control Release 2018; 281:139-177. [DOI: 10.1016/j.jconrel.2018.05.011] [Citation(s) in RCA: 366] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/10/2018] [Accepted: 05/10/2018] [Indexed: 01/02/2023]
|
21
|
Ye F, Luo YJ, Xiao J, Yu NW, Yi G. Impact of Insulin Sensitizers on the Incidence of Dementia: A Meta-Analysis. Dement Geriatr Cogn Disord 2018; 41:251-60. [PMID: 27250528 DOI: 10.1159/000445941] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/02/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Abundant evidence from epidemiological and clinical studies has proven that diabetes mellitus (DM) is correlated with an increased incidence of dementia and Alzheimer's disease (AD). Insulin resistance is considered to play an important role in the associations between DM and dementia. However, whether insulin sensitizer drugs are effective in preventing dementia still remains unclear. METHODS Electronic searches of PubMed, EMBASE, Google Scholar, and the ISI Web of Science were conducted to identify studies that reported about the associations between insulin sensitizers and the incidence of dementia. The included studies were reviewed, and a meta-analysis was performed using STATA to determine the combined relative risk (RR) for the incidence of dementia when using insulin sensitizers. Subgroup analysis and meta regression were also conducted. RESULTS In total, nine comparisons out of six studies were qualified for inclusion, and data from 544,093 participants were evaluated. The results of the meta-analysis revealed a combined RR of 0.78 (95% CI 0.64-0.95, p = 0.015) for the incidence of dementia when using insulin sensitizers. The incidence rate of dementia was reduced with either metformin (RR 0.79, 95% CI 0.62-1.01, p = 0.064) or thiazolidinediones (RR 0.75, 95% CI 0.56-1.00, p = 0.050), both with a marginal trend toward significance. CONCLUSIONS The results indicate that insulin sensitizer drugs might provide protection against incident dementia. Controlled studies with large samples should be conducted to further confirm these conclusions and provide information for clinical strategies.
Collapse
Affiliation(s)
- Fang Ye
- Department of Neurology, Sichuan Provincial Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | | | | | | | | |
Collapse
|
22
|
Levy B, Tsoy E, Gable S. Developing Cognitive Markers of Alzheimer's Disease for Primary Care: Implications for Behavioral and Global Prevention. J Alzheimers Dis 2018; 54:1259-1272. [PMID: 27567831 DOI: 10.3233/jad-160309] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
A comprehensive approach to the prevention of Alzheimer's disease (AD) warrants a synergy across multiple domains and procedures. Whereas the study of biological markers has mobilized major activity in the field, the development of cognitive markers is largely ignored, despite the unique advantages they may offer. Cognitive markers essentially assess the core clinical feature that biological markers intend to predict. In this respect, cognitive markers expand the foundation of preclinical diagnostics and disease staging in a manner that integrates both physiological and psychological factors. In addition, the cost-effective implementation of cognitive markers makes them remarkably conducive to community-wide screenings, and thereby a vital component of any global blueprint for prevention. Specifically, in the primary care setting, cognitive markers may provide effective gate keeping for more invasive, labor intensive, and expensive procedures. From this perspective, cognitive markers may provide the first step for identifying preclinical treatment recipients in general public. Moreover, the detection of preclinical decline via cognitive markers can increase awareness of AD risk and the motivation for making protective lifestyle changes. The behavioral approach might be expedient for prevention in light of the compelling evidence of lifestyle amelioration of AD risk. In an integrative view, incorporating cognitive markers to primary care may facilitate a synergetic development in preventive interventions that carries epidemiological significance. This paper addresses the theoretical, methodological, and pragmatic aspects of this prospect.
Collapse
|
23
|
Baldacci F, Lista S, O'Bryant SE, Ceravolo R, Toschi N, Hampel H. Blood-Based Biomarker Screening with Agnostic Biological Definitions for an Accurate Diagnosis Within the Dimensional Spectrum of Neurodegenerative Diseases. Methods Mol Biol 2018; 1750:139-155. [PMID: 29512070 DOI: 10.1007/978-1-4939-7704-8_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The discovery, development, and validation of novel candidate biomarkers in Alzheimer's disease (AD) and other neurodegenerative diseases (NDs) are increasingly gaining momentum. As a result, evolving diagnostic research criteria of NDs are beginning to integrate biofluid and neuroimaging indicators of pathophysiological mechanisms. More than 10% of people aged over 65 suffer from NDs. There is an urgent need for a refined two-stage diagnostic model to first initiate an early, sensitive, and noninvasive process in primary care settings. Individuals that meet detection criteria will then be channeled to more specific, costly (positron-emission tomography), and invasive (cerebrospinal fluid) assessment methods for confirmatory biological characterization and diagnosis.A reliable and sensitive blood test for AD and other NDs is not yet established; however, it would provide the golden screening gate for an efficient primary care management. A limitation to the development of a large-scale blood-screening biomarker-based test is the traditional application of clinically descriptive criteria for the categorization of single late-stage ND constructs. These are genetically and biologically heterogeneous, reflected in multiple pathophysiological mechanisms and subsequent pathologies throughout a dimensional continuum. Evidence suggests that a shared, "open-source" integrated multilevel categorization of NDs that clusters individuals based on descriptive clinical phenotypes and pathophysiological biomarker signatures will provide the next incremental step toward an improved diagnostic process of NDs. This intermediate objective toward unbiased biomarker-guided early detection of individuals at risk for NDs is currently carried out by the international pilot Alzheimer Precision Medicine Initiative Cohort Program (APMI-CP).
Collapse
Affiliation(s)
- Filippo Baldacci
- AXA Research Fund & UPMC Chair, F-75013, Paris, France.,Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l'hôpital, F-75013, Paris, France.,Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, F-75013, Paris, France.,Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l'hôpital, F-75013, Paris, France.,Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Simone Lista
- AXA Research Fund & UPMC Chair, F-75013, Paris, France. .,Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l'hôpital, F-75013, Paris, France. .,Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, F-75013, Paris, France. .,Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l'hôpital, F-75013, Paris, France.
| | - Sid E O'Bryant
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy.,Department of Radiology"Athinoula A. Martinos", Center for Biomedical Imaging, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Harald Hampel
- AXA Research Fund & UPMC Chair, F-75013, Paris, France.,Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l'hôpital, F-75013, Paris, France.,Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, F-75013, Paris, France.,Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l'hôpital, F-75013, Paris, France
| | | |
Collapse
|
24
|
Abstract
Alzheimer's disease (AD) affects more than 5 million Americans, with substantial consequences for individuals with AD, families, and society in terms of morbidity, mortality, and healthcare costs. With disease-modifying treatment trials unsuccessful at the present time and only medications to treat symptoms available, an emerging approach is prevention. Advances in diagnostic criteria, biomarker development, and greater understanding of the biophysiological basis of AD make these initiatives feasible. Ongoing pharmacological trials using anti-amyloid therapies are underway in sporadic and genetic forms of AD, although a large number of modifiable risk factors for AD have been identified in observational studies, many of which do not appear to exert effects through amyloid or tau. This suggests that prevention studies focusing on risk reduction and lifestyle modification may offer additional benefits. Rather than relying solely on large-sample, long-duration, randomized clinical trial designs, a precision medicine approach using N-of-1 trials may provide more-rapid information on whether personalized prevention plans can improve person-centered outcomes. Because there appear to be multiple pathways to developing AD, there may also be multiple ways to prevent or delay the onset of AD. Even if these precision approaches alone are not successful in preventing AD, they may greatly improve the likelihood of amyloid- or tau-specific therapies to reach their endpoints by reducing comorbidities. Keeping this in mind, dementia may be a disorder that develops over a lifetime, with individualized ways to build a better brain as we age.
Collapse
Affiliation(s)
- James E Galvin
- Comprehensive Center for Brain Health, Department of Integrated Medical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida
| |
Collapse
|
25
|
Du K, Liu M, Pan Y, Zhong X, Wei M. Association of Serum Manganese Levels with Alzheimer's Disease and Mild Cognitive Impairment: A Systematic Review and Meta-Analysis. Nutrients 2017; 9:nu9030231. [PMID: 28273828 PMCID: PMC5372894 DOI: 10.3390/nu9030231] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 02/28/2017] [Indexed: 01/20/2023] Open
Abstract
Manganese (Mn) is one of the most studied environmental heavy metals linked to Alzheimer’s disease (AD). However, it remains unclear whether serum Mn levels are associated with AD and mild cognition impairment (MCI, a prodromal stage of AD). We conducted a meta-analysis to analyze the serum Mn levels in patients with AD and MCI. A systematic database search of PubMed, Web of Science, and the China National Knowledge Infrastructure (CNKI) identified 17 studies, including 836 cases and 1254 health controls (HC). Random-effects meta-analysis showed that patients with AD had significantly reduced serum Mn levels compared with HC subjects (SMD = −0.39; 95% CI (−0.71, −0.08); p = 0.015). MCI individuals had a tendency toward reduced serum Mn levels compared with HC subjects (SMD = −0.31; 95% CI (−0.70, 0.08); p = 0.117). A significant decrease in serum Mn levels was found in patients with cognitive impairment (including both AD patients and MCI patients) (SMD = −0.37, 95% CI (−0.60; −0.13); p = 0.002). Finally, no significant differences were observed between AD and MCI patients in serum levels (SMD = 0.24; 95% CI (−0.23, 0.72); p = 0.310). Our findings show that the serum Mn levels are lower in AD patients, and Mn deficiency may be a risk factor for AD.
Collapse
Affiliation(s)
- Ke Du
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang 110122, China.
| | - Mingyan Liu
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang 110122, China.
| | - Yanzhu Pan
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang 110122, China.
| | - Xin Zhong
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang 110122, China.
| | - Minjie Wei
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang 110122, China.
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang 110122, China.
| |
Collapse
|
26
|
Touchon J, Rosenbaum J, Aisen P, Andrieu S, Carrillo MC, Ceccaldi M, Dartiques JF, Feldman H, Gabelle A, Isaac M, Fitten LJ, Sperling RA, Vellas B, Tariot P, Weiner M. Editorial: Collaborative Efforts to Prevent Alzheimer's Disease. J Nutr Health Aging 2017; 21:1072-1074. [PMID: 29188862 DOI: 10.1007/s12603-017-0961-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- J Touchon
- Jacques Touchon, University Hospital of Montpellier, 34025 Montpellier Cedex 5, France,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Schindler SM, Klegeris A. Elucidating the link between the modifiable risk factors of Alzheimer's disease and neuroinflammation. Neurodegener Dis Manag 2016; 6:375-84. [DOI: 10.2217/nmt-2016-0028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Increased worldwide longevity through medical interventions, although beneficial, has allowed the age-related Alzheimer's disease (AD) to become an epidemic of the 21st century. AD pathology involves adverse activation of microglia, the immune cells of the brain and resulting chronic neuroinflammation. Certain diets, physical inactivity and Type 2 diabetes mellitus have been identified as the risk factors for developing AD, which may increase the risk of AD by neuroimmune mechanisms primarily through the overactivation of microglia. Thus, modifying these risk factors may represent an alternative therapeutic strategy for lowering the incidence of AD. We highlight the link between select modifiable risk factors and neuroimmune mechanisms, and demonstrate that by controlling microglial activation and neuroinflammation the prevalence of AD may be decreased.
Collapse
Affiliation(s)
- Stephanie M Schindler
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia, V1V 1V7, Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia, V1V 1V7, Canada
| |
Collapse
|
28
|
Molino S, Dossena M, Buonocore D, Ferrari F, Venturini L, Ricevuti G, Verri M. Polyphenols in dementia: From molecular basis to clinical trials. Life Sci 2016; 161:69-77. [PMID: 27493077 DOI: 10.1016/j.lfs.2016.07.021] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/25/2016] [Accepted: 07/30/2016] [Indexed: 10/21/2022]
Abstract
Dementia is common in the elderly, but there are currently no effective therapies available to prevent or treat this syndrome. In the last decade, polyphenols (particularly curcumin, resveratrol and tea catechins) have been under very close scrutiny as potential therapeutic agents for neurodegenerative diseases, diabetes, inflammatory diseases and aging. Data were collected from Web of Science (ISI Web of Knowledge), Pubmed and Medline (from 2000 to 2015), by searching for the keywords "dementia" AND "curcumin", "resveratrol", "EGCG", "tea catechins". The same keywords were used to investigate the current state of clinical trials recorded in the NIH clinicaltrials.gov registry. Starting from the intrinsic properties of the compounds, we explain their specific action in patients with AD and the most common types of dementia. The pharmacological actions of curcumin, resveratrol and tea catechins have mainly been attributed to their antioxidant activity, interaction with cell signaling pathways, anti-inflammatory effect, chelation of metal ions, and neuroprotection. Evidence from in vitro and in vivo studies on polyphenols have demonstrated that they may play an integral role in preventing and treating diseases associated with neurodegeneration. Furthermore, we critically analyze the clinical trials that we found, which investigate the real pharmacological actions and the possible side effects of these compounds. This review highlights the potential role of polyphenols in the prevention/treatment of dementia and describes the current limitations of research in this field.
Collapse
Affiliation(s)
- Silvia Molino
- Dipartimento di Biologia e Biotecnologie "Lazzaro Spallanzani", Università degli Studi di Pavia, Via Ferrata, 9-27100, Pavia (PV), Italy
| | - Maurizia Dossena
- Dipartimento di Biologia e Biotecnologie "Lazzaro Spallanzani", Università degli Studi di Pavia, Via Ferrata, 9-27100, Pavia (PV), Italy
| | - Daniela Buonocore
- Dipartimento di Biologia e Biotecnologie "Lazzaro Spallanzani", Università degli Studi di Pavia, Via Ferrata, 9-27100, Pavia (PV), Italy
| | - Federica Ferrari
- Dipartimento di Biologia e Biotecnologie "Lazzaro Spallanzani", Università degli Studi di Pavia, Via Ferrata, 9-27100, Pavia (PV), Italy
| | - Letizia Venturini
- Dipartimento di Medicina Interna e Terapia Medica, Divisione di Geriatria - ASP - IDR S. Margherita - Università degli Studi di Pavia, Via Emilia, 12-27100, Pavia, (PV), Italy
| | - Giovanni Ricevuti
- Dipartimento di Medicina Interna e Terapia Medica, Divisione di Geriatria - ASP - IDR S. Margherita - Università degli Studi di Pavia, Via Emilia, 12-27100, Pavia, (PV), Italy
| | - Manuela Verri
- Dipartimento di Biologia e Biotecnologie "Lazzaro Spallanzani", Università degli Studi di Pavia, Via Ferrata, 9-27100, Pavia (PV), Italy.
| |
Collapse
|
29
|
Boespflug EL, McNamara RK, Eliassen JC, Schidler MD, Krikorian R. Fish Oil Supplementation Increases Event-Related Posterior Cingulate Activation in Older Adults with Subjective Memory Impairment. J Nutr Health Aging 2016; 20:161-9. [PMID: 26812512 DOI: 10.1007/s12603-015-0609-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To determine the effects of long-chain omega-3 (LCn-3) fatty acids found in fish oil, including eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), on cortical blood oxygen level-dependent (BOLD) activity during a working memory task in older adults with subjective memory impairment. DESIGN Randomized, double-blind, placebo-controlled study. SETTING Academic medical center. PARTICIPANTS Healthy older adults (62-80 years) with subjective memory impairment, but not meeting criteria for mild cognitive impairment or dementia. INTERVENTION Fish oil (EPA+DHA: 2.4 g/d, n=11) or placebo (corn oil, n=10) for 24 weeks. MEASUREMENTS Cortical BOLD response patterns during performance of a sequential letter n-back working memory task were determined at baseline and week 24 by functional magnetic resonance imaging (fMRI). RESULTS At 24 weeks erythrocyte membrane EPA+DHA composition increased significantly from baseline in participants receiving fish oil (+31%, p ≤ 0.0001) but not placebo (-17%, p=0.06). Multivariate modeling of fMRI data identified a significant interaction among treatment, visit, and memory loading in the right cingulate (BA 23/24), and in the right sensorimotor area (BA 3/4). In the fish oil group, BOLD increases at 24 weeks were observed in the right posterior cingulate and left superior frontal regions during memory loading. A region-of-interest analysis indicated that the baseline to endpoint change in posterior cingulate cortex BOLD activity signal was significantly greater in the fish oil group compared with the placebo group during the 1-back (p=0.0003) and 2-back (p=0.0005) conditions. Among all participants, the change in erythrocyte EPA+DHA during the intervention was associated with performance in the 2-back working memory task (p = 0.01), and with cingulate BOLD signal during the 1-back (p = 0.005) with a trend during the 2-back (p = 0.09). Further, cingulate BOLD activity was related to performance in the 2-back condition. CONCLUSION Dietary fish oil supplementation increases red blood cell omega-3 content, working memory performance, and BOLD signal in the posterior cingulate cortex during greater working memory load in older adults with subjective memory impairment suggesting enhanced neuronal response to working memory challenge.
Collapse
Affiliation(s)
- E L Boespflug
- Robert Krikorian, PhD, Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati Academic Health Center, PO Box 670559, Cincinnati, OH 45267-0559, PH: 513-558-2455, FX: 513-558-0877, EM:
| | | | | | | | | |
Collapse
|
30
|
Rolandi E, Frisoni GB, Cavedo E. Efficacy of lifestyle interventions on clinical and neuroimaging outcomes in elderly. Ageing Res Rev 2016; 25:1-12. [PMID: 26589097 DOI: 10.1016/j.arr.2015.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 10/30/2015] [Accepted: 11/09/2015] [Indexed: 11/19/2022]
Abstract
The prevalence of Alzheimer's disease (AD) is constantly growing worldwide in absence of any effective treatment. Methodology and technique advancements facilitated the early diagnosis of AD leading to a shift toward preclinical AD stages investigation in order to delay the disease onset in individuals at risk for AD. Recent evidence demonstrating the aging related multifactorial nature of AD supported the hypothesis that modifiable environmental factors can accelerate or delay the disease onset. In particular, healthy dietary habits, constant physical and cognitive activities are associated with reduced brain atrophy, amyloid load and incidence of AD cases. Due to these promising results, an emerging field of studies is currently investigating the efficacy of interventions addressing different lifestyle habits in cognitive intact elderly individuals as a potential preventive strategy against AD onset. We provide a critical overview of the current evidence on nonpharmacologic treatments in elderly individuals, discussing their efficacy on clinical and neuroimaging outcomes and identifying current methodological issues. Future perspectives, relevant for the scientific community and the worldwide public health institutes will be further discussed.
Collapse
Affiliation(s)
- Elena Rolandi
- Laboratory of Alzheimer's Neuroimaging and Epidemiology, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| | - Giovanni Battista Frisoni
- Laboratory of Alzheimer's Neuroimaging and Epidemiology, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Memory Clinic and Laboratory of Neuroimaging of Ageing - LANVIE, University Hospitals and University of Geneva, Geneva, Switzerland.
| | - Enrica Cavedo
- Laboratory of Alzheimer's Neuroimaging and Epidemiology, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Paris 06, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A) & Institut du Cerveau et de la Moelle épinière (ICM), UMR S 1127, Département de Neurologie, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France and the CATI multicenter neuroimaging platform (cati-neuroimaging.com), France.
| |
Collapse
|
31
|
Brouillette MJ, Fellows LK, Palladini L, Finch L, Thomas R, Mayo NE. Quantifying cognition at the bedside: a novel approach combining cognitive symptoms and signs in HIV. BMC Neurol 2015; 15:224. [PMID: 26563353 PMCID: PMC4643497 DOI: 10.1186/s12883-015-0483-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 11/01/2015] [Indexed: 11/24/2022] Open
Abstract
Background Up to half of all people with HIV infection have some degree of cognitive impairment. This impairment is typically mild, but nonetheless often disabling. Although early detection of cognitive impairment offers the greatest hope of effective intervention, there are important barriers to this goal in most clinical settings. These include uncertainty about how self-reported cognitive symptoms relate to objective impairments, and the paucity of bedside measurement tools suitable for mild deficits. Clinicians need guidance in interpreting cognitive symptoms in this population, and a brief cognitive measurement tool targeted to mild impairment. We addressed these two problems together here. The objective of this study was to determine the extent to which performance on cognitive tests and self-reported cognitive symptoms form a unidimensional construct. Methods Two hundred three HIV+ individuals completed the Montreal Cognitive Assessment, computerized cognitive tasks and a questionnaire eliciting cognitive symptoms. Rasch measurement theory was applied to determine whether patient-reported and performance items could be combined to measure cognition as a unidimensional latent construct. Results Performance-based items and cognitive symptoms are arranged hierarchically along the same continuum of cognitive ability, forming a measure with thresholds covering a broad spectrum of ability that has good internal reliability. The cognitive symptoms that fit the measurement model relate to important aspects of everyday life, providing evidence that the identified construct is meaningful. Conclusions This finding lays the foundation for a rapid measure of cognitive ability in people with HIV infection that is feasible for routine clinical use, and shows that some cognitive symptoms are systematically related to performance in this population.
Collapse
Affiliation(s)
- Marie-Josée Brouillette
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC, Canada. .,Chronic Viral Illness Service, McGill University Health Center, Montreal, QC, Canada. .,Research Institute of the McGill University Health Centre, Montreal, QC, Canada. .,McGill University Health Centre, Glen site, 1001 Decarie, D02.4110, Montreal, QC, H4A 3J1, Canada.
| | - Lesley K Fellows
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC, Canada. .,Montreal Neurological Institute, McGill University, Montreal, QC, Canada.
| | - Lisa Palladini
- School of Physical and Occupational Therapy, Faculty of Medicine, McGill University, Montreal, QC, Canada.
| | - Lois Finch
- Division of Clinical Epidemiology, McGill University Health Center, Montreal, QC, Canada.
| | | | - Nancy E Mayo
- School of Physical and Occupational Therapy, Faculty of Medicine, McGill University, Montreal, QC, Canada. .,Division of Clinical Epidemiology, McGill University Health Center, Montreal, QC, Canada.
| |
Collapse
|