101
|
Jiang H, Wang J, Levin BE, Baumel BS, Camargo CJ, Signorile JF, Rundek T. Retinal Microvascular Alterations as the Biomarkers for Alzheimer Disease: Are We There Yet? J Neuroophthalmol 2021; 41:251-260. [PMID: 33136677 PMCID: PMC8079547 DOI: 10.1097/wno.0000000000001140] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Alzheimer disease (AD) is a heterogeneous and multifactorial disorder with an insidious onset and slowly progressive disease course. To date, there are no effective treatments, but biomarkers for early diagnosis and monitoring of disease progression offer a promising first step in developing and testing potential interventions. Cerebral vascular imaging biomarkers to assess the contributions of vascular dysfunction to AD are strongly recommended to be integrated into the current amyloid-β (Aβ) [A], tau [T], and neurodegeneration [(N)]-the "AT(N)" biomarker system for clinical research. However, the methodology is expensive and often requires invasive procedures to document cerebral vascular dysfunction. The retina has been used as a surrogate to study cerebral vascular changes. There is growing interest in the identification of retinal microvascular changes as a safe, easily accessible, low cost, and time-efficient approach to enhancing our understanding of the vascular pathogenesis associated with AD. EVIDENCE ACQUISITION A systemic review of the literature was performed regarding retinal vascular changes in AD and its prodromal stages, focusing on functional and structural changes of large retinal vessels (vessels visible on fundus photographs) and microvasculature (precapillary arterioles, capillary, and postcapillary venules) that are invisible on fundus photographs. RESULTS Static and dynamic retinal microvascular alterations such as retinal arterial wall motion, blood flow rate, and microvascular network density were reported in AD, mild cognitive impairment, and even in the preclinical stages of the disease. The data are somewhat controversial and inconsistent among the articles reviewed and were obtained based on cross-sectional studies that used different patient cohorts, equipment, techniques, and analysis methods. CONCLUSIONS Retinal microvascular alterations exist across the AD spectrum. Further large scale, within-subject longitudinal studies using standardized imaging and analytical methods may advance our knowledge concerning vascular contributions to the pathogenesis of AD.
Collapse
Affiliation(s)
- Hong Jiang
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jianhua Wang
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Bonnie E. Levin
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Bernard S. Baumel
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Christian J. Camargo
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Tania Rundek
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
102
|
Role Played by Receptors for Advanced Glycosylation End Products in Corneal Endothelial Cells after HSV-1 Infection. Int J Mol Sci 2021; 22:ijms22115833. [PMID: 34072468 PMCID: PMC8199122 DOI: 10.3390/ijms22115833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/23/2021] [Accepted: 05/25/2021] [Indexed: 11/16/2022] Open
Abstract
Senescence, sterile inflammation, and infection cause dysfunction of corneal endothelial cells, leading to visual morbidity that may require corneal transplantation. With increasing age, the extracellular matrix is modified by non-enzymatic glycation forming advanced glycation end products (AGEs). The modifications are primarily sensed by the receptors for the AGEs (RAGE) and are manifested as a type I interferon response. Interestingly, in our study, human corneal endothelial cells (HCEn) cells did not respond to the typical RAGE ligands, including the AGEs, high mobility group box 1 (HMGB1), and serum amyloid-A (SAA). Instead, HCEn cells responded exclusively to the CpG DNA, which is possessed by typical corneal pathogen, herpes simplex virus-1 (HSV-1). Upon HSV-1 infection, the surface expression of RAGE was increased, and endocytosed HSV-1 was associated with RAGE and CpG DNA receptor, TLR9. RAGE DNA transfection markedly increased interferon-β secretion by CpG DNA or HSV-1 infection. HSV-1 infection-induced interferon-β secretion was abolished by TLR9 inhibition and partially by RAGE inhibition. Global transcriptional response analysis confirmed that RAGE and TLR9 were both significantly involved in type I interferon responses. We conclude that RAGE is a sensor of HSV-1 infection and provokes a type I interferon response.
Collapse
|
103
|
Pålhaugen L, Sudre CH, Tecelao S, Nakling A, Almdahl IS, Kalheim LF, Cardoso MJ, Johnsen SH, Rongve A, Aarsland D, Bjørnerud A, Selnes P, Fladby T. Brain amyloid and vascular risk are related to distinct white matter hyperintensity patterns. J Cereb Blood Flow Metab 2021; 41:1162-1174. [PMID: 32955960 PMCID: PMC8054718 DOI: 10.1177/0271678x20957604] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
White matter hyperintensities (WMHs) are associated with vascular risk and Alzheimer's disease. In this study, we examined relations between WMH load and distribution, amyloid pathology and vascular risk in 339 controls and cases with either subjective (SCD) or mild cognitive impairment (MCI). Regional deep (DWMH) and periventricular (PWMH) WMH loads were determined using an automated algorithm. We stratified on Aβ1-42 pathology (Aβ+/-) and analyzed group differences, as well as associations with Framingham Risk Score for cardiovascular disease (FRS-CVD) and age. Occipital PWMH (p = 0.001) and occipital DWMH (p = 0.003) loads were increased in SCD-Aβ+ compared with Aβ- controls. In MCI-Aβ+ compared with Aβ- controls, there were differences in global WMH (p = 0.003), as well as occipital DWMH (p = 0.001) and temporal DWMH (p = 0.002) loads. FRS-CVD was associated with frontal PWMHs (p = 0.003) and frontal DWMHs (p = 0.005), after adjusting for age. There were associations between global and all regional WMH loads and age. In summary, posterior WMH loads were increased in SCD-Aβ+ and MCI-Aβ+ cases, whereas frontal WMHs were associated with vascular risk. The differences in WMH topography support the use of regional WMH load as an early-stage marker of etiology.
Collapse
Affiliation(s)
- Lene Pålhaugen
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Carole H Sudre
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK.,Dementia Research Centre, Institute of Neurology, University College London, London, UK.,Department of Medical Physics, University College London, London, UK
| | - Sandra Tecelao
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
| | | | - Ina S Almdahl
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Geriatric Psychiatry, Oslo University Hospital, Oslo, Norway
| | - Lisa F Kalheim
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - M Jorge Cardoso
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK.,Dementia Research Centre, Institute of Neurology, University College London, London, UK.,Department of Medical Physics, University College London, London, UK
| | - Stein H Johnsen
- Department of Neurology, University Hospital of North Norway, Tromsø, Norway.,Department of Clinical Medicine, Brain and Circulation Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Arvid Rongve
- Department of Research and Innovation, Haugesund Hospital, Haugesund, Norway.,Department of Clinical Medicine (K1), University of Bergen, Bergen, Norway
| | - Dag Aarsland
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway.,Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Center for Age-Related Diseases, Stavanger University Hospital, Stavanger, Norway
| | - Atle Bjørnerud
- Department of Diagnostic Physics, Oslo University Hospital, Oslo, Norway.,Department of Physics, University of Oslo, Oslo, Norway
| | - Per Selnes
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tormod Fladby
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
104
|
He JT, Zhao X, Xu L, Mao CY. Vascular Risk Factors and Alzheimer's Disease: Blood-Brain Barrier Disruption, Metabolic Syndromes, and Molecular Links. J Alzheimers Dis 2021; 73:39-58. [PMID: 31815697 DOI: 10.3233/jad-190764] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, marked by cortical and hippocampal deposition of amyloid-β (Aβ) plaques and neurofibrillary tangles and cognitive impairment. Studies indicate a prominent link between cerebrovascular abnormalities and the onset and progression of AD, where blood-brain barrier (BBB) dysfunction and metabolic disorders play key risk factors. Pericyte degeneration, endothelial cell damage, astrocyte depolarization, diminished tight junction integrity, and basement membrane disarray trigger BBB damage. Subsequently, the altered expression of low-density lipoprotein receptor-related protein 1 and receptor for advanced glycation end products at the microvascular endothelial cells dysregulate Aβ transport across the BBB. White matter lesions and microhemorrhages, dyslipidemia, altered brain insulin signaling, and insulin resistance contribute to tau and Aβ pathogenesis, and oxidative stress, mitochondrial damage, inflammation, and hypoperfusion serve as mechanistic links between pathophysiological features of AD and ischemia. Deregulated calcium homeostasis, voltage gated calcium channel functioning, and protein kinase C signaling are also common mechanisms for both AD pathogenesis and cerebrovascular abnormalities. Additionally, APOE polymorphic alleles that characterize impaired cerebrovascular integrity function as primary genetic determinants of AD. Overall, the current review enlightens key vascular risk factors for AD and underscores pathophysiologic relationship between AD and vascular dysfunction.
Collapse
Affiliation(s)
- Jin-Ting He
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, Jilin Province, China
| | - Xin Zhao
- Department of Paediatrics, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Lei Xu
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, Jilin Province, China
| | - Cui-Ying Mao
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
105
|
Frohlich J, Chaldakov GN, Vinciguerra M. Cardio- and Neurometabolic Adipobiology: Consequences and Implications for Therapy. Int J Mol Sci 2021; 22:ijms22084137. [PMID: 33923652 PMCID: PMC8072708 DOI: 10.3390/ijms22084137] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/08/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
Studies over the past 30 years have revealed that adipose tissue is the major endocrine and paracrine organ of the human body. Arguably, adiopobiology has taken its reasonable place in studying obesity and related cardiometabolic diseases (CMDs), including Alzheimer's disease (AD), which is viewed herein as a neurometabolic disorder. The pathogenesis and therapy of these diseases are multiplex at basic, clinical and translational levels. Our present goal is to describe new developments in cardiometabolic and neurometabolic adipobiology. Accordingly, we focus on adipose- and/or skeletal muscle-derived signaling proteins (adipsin, adiponectin, nerve growth factor, brain-derived neuroptrophic factor, neurotrophin-3, irisin, sirtuins, Klotho, neprilysin, follistatin-like protein-1, meteorin-like (metrnl), as well as growth differentiation factor 11) as examples of metabotrophic factors (MTFs) implicated in the pathogenesis and therapy of obesity and related CMDs. We argue that these pathologies are MTF-deficient diseases. In 1993 the "vascular hypothesis of AD" was published and in the present review we propose the "vasculometabolic hypothesis of AD." We discuss how MTFs could bridge CMDs and neurodegenerative diseases, such as AD. Greater insights on how to manage the MTF network would provide benefits to the quality of human life.
Collapse
Affiliation(s)
- Jan Frohlich
- International Clinical Research Center, St. Anne’s University Hospital, 656 91 Brno, Czech Republic;
| | - George N. Chaldakov
- Department of Anatomy and Cell Biology and Research Institute of the Medical University, 9002 Varna, Bulgaria;
- Department of Translational Stem Cell Biology, Research Institute of the Medical University, 9002 Varna, Bulgaria
| | - Manlio Vinciguerra
- International Clinical Research Center, St. Anne’s University Hospital, 656 91 Brno, Czech Republic;
- Department of Translational Stem Cell Biology, Research Institute of the Medical University, 9002 Varna, Bulgaria
- Correspondence: or
| |
Collapse
|
106
|
Wickstead ES, Irving MA, Getting SJ, McArthur S. Exploiting formyl peptide receptor 2 to promote microglial resolution: a new approach to Alzheimer's disease treatment. FEBS J 2021; 289:1801-1822. [PMID: 33811735 DOI: 10.1111/febs.15861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/09/2021] [Accepted: 03/29/2021] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease and dementia are among the most significant current healthcare challenges given the rapidly growing elderly population, and the almost total lack of effective therapeutic interventions. Alzheimer's disease pathology has long been considered in terms of accumulation of amyloid beta and hyperphosphorylated tau, but the importance of neuroinflammation in driving disease has taken greater precedence over the last 15-20 years. Inflammatory activation of the primary brain immune cells, the microglia, has been implicated in Alzheimer's pathogenesis through genetic, preclinical, imaging and postmortem human studies, and strategies to regulate microglial activity may hold great promise for disease modification. Neuroinflammation is necessary for defence of the brain against pathogen invasion or damage but is normally self-limiting due to the engagement of endogenous pro-resolving circuitry that terminates inflammatory activity, a process that appears to fail in Alzheimer's disease. Here, we discuss the potential for a major regulator and promoter of resolution, the receptor FPR2, to restrain pro-inflammatory microglial activity, and propose that it may serve as a valuable target for therapeutic investigation in Alzheimer's disease.
Collapse
Affiliation(s)
| | - Murray A Irving
- Institute of Dentistry, Barts and the London School of Medicine & Dentistry, Blizard Institute, Queen Mary, University of London, UK
| | - Stephen J Getting
- College of Liberal Arts & Sciences, School of Life Sciences, University of Westminster, London, UK
| | - Simon McArthur
- Institute of Dentistry, Barts and the London School of Medicine & Dentistry, Blizard Institute, Queen Mary, University of London, UK
| |
Collapse
|
107
|
Stefanovski L, Meier JM, Pai RK, Triebkorn P, Lett T, Martin L, Bülau K, Hofmann-Apitius M, Solodkin A, McIntosh AR, Ritter P. Bridging Scales in Alzheimer's Disease: Biological Framework for Brain Simulation With The Virtual Brain. Front Neuroinform 2021; 15:630172. [PMID: 33867964 PMCID: PMC8047422 DOI: 10.3389/fninf.2021.630172] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/08/2021] [Indexed: 12/18/2022] Open
Abstract
Despite the acceleration of knowledge and data accumulation in neuroscience over the last years, the highly prevalent neurodegenerative disease of AD remains a growing problem. Alzheimer's Disease (AD) is the most common cause of dementia and represents the most prevalent neurodegenerative disease. For AD, disease-modifying treatments are presently lacking, and the understanding of disease mechanisms continues to be incomplete. In the present review, we discuss candidate contributing factors leading to AD, and evaluate novel computational brain simulation methods to further disentangle their potential roles. We first present an overview of existing computational models for AD that aim to provide a mechanistic understanding of the disease. Next, we outline the potential to link molecular aspects of neurodegeneration in AD with large-scale brain network modeling using The Virtual Brain (www.thevirtualbrain.org), an open-source, multiscale, whole-brain simulation neuroinformatics platform. Finally, we discuss how this methodological approach may contribute to the understanding, improved diagnostics, and treatment optimization of AD.
Collapse
Affiliation(s)
- Leon Stefanovski
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neurology with Experimental Neurology, Brain Simulation Section, Berlin, Germany
| | - Jil Mona Meier
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neurology with Experimental Neurology, Brain Simulation Section, Berlin, Germany
| | - Roopa Kalsank Pai
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neurology with Experimental Neurology, Brain Simulation Section, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Paul Triebkorn
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neurology with Experimental Neurology, Brain Simulation Section, Berlin, Germany
- Institut de Neurosciences des Systèmes, Aix Marseille Université, Marseille, France
| | - Tristram Lett
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neurology with Experimental Neurology, Brain Simulation Section, Berlin, Germany
| | - Leon Martin
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neurology with Experimental Neurology, Brain Simulation Section, Berlin, Germany
| | - Konstantin Bülau
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neurology with Experimental Neurology, Brain Simulation Section, Berlin, Germany
| | - Martin Hofmann-Apitius
- Fraunhofer Institute for Algorithms and Scientific Computing SCAI, Sankt Augustin, Germany
| | - Ana Solodkin
- Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, United States
| | | | - Petra Ritter
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neurology with Experimental Neurology, Brain Simulation Section, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neuroscience Berlin, Berlin, Germany
- Einstein Center Digital Future, Berlin, Germany
| |
Collapse
|
108
|
Duong MT, Nasrallah IM, Wolk DA, Chang CCY, Chang TY. Cholesterol, Atherosclerosis, and APOE in Vascular Contributions to Cognitive Impairment and Dementia (VCID): Potential Mechanisms and Therapy. Front Aging Neurosci 2021; 13:647990. [PMID: 33841127 PMCID: PMC8026881 DOI: 10.3389/fnagi.2021.647990] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/08/2021] [Indexed: 12/18/2022] Open
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) are a common cause of cognitive decline, yet limited therapies exist. This cerebrovascular disease results in neurodegeneration via acute, chronic, local, and systemic mechanisms. The etiology of VCID is complex, with a significant impact from atherosclerosis. Risk factors including hypercholesterolemia and hypertension promote intracranial atherosclerotic disease and carotid artery stenosis (CAS), which disrupt cerebral blood flow and trigger ischemic strokes and VCID. Apolipoprotein E (APOE) is a cholesterol and phospholipid carrier present in plasma and various tissues. APOE is implicated in dyslipidemia and Alzheimer disease (AD); however, its connection with VCID is less understood. Few experimental models for VCID exist, so much of the present information has been drawn from clinical studies. Here, we review the literature with a focus on the clinical aspects of atherosclerotic cerebrovascular disease and build a working model for the pathogenesis of VCID. We describe potential intermediate steps in this model, linking cholesterol, atherosclerosis, and APOE with VCID. APOE4 is a minor isoform of APOE that promotes lipid dyshomeostasis in astrocytes and microglia, leading to chronic neuroinflammation. APOE4 disturbs lipid homeostasis in macrophages and smooth muscle cells, thus exacerbating systemic inflammation and promoting atherosclerotic plaque formation. Additionally, APOE4 may contribute to stromal activation of endothelial cells and pericytes that disturb the blood-brain barrier (BBB). These and other risk factors together lead to chronic inflammation, atherosclerosis, VCID, and neurodegeneration. Finally, we discuss potential cholesterol metabolism based approaches for future VCID treatment.
Collapse
Affiliation(s)
- Michael Tran Duong
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ilya M Nasrallah
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - David A Wolk
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | | - Ta-Yuan Chang
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
| |
Collapse
|
109
|
Ashraf GM, Ebada MA, Suhail M, Ali A, Uddin MS, Bilgrami AL, Perveen A, Husain A, Tarique M, Hafeez A, Alexiou A, Ahmad A, Kumar R, Banu N, Najda A, Sayed AA, Albadrani GM, Abdel-Daim MM, Peluso I, Barreto GE. Dissecting Sex-Related Cognition between Alzheimer's Disease and Diabetes: From Molecular Mechanisms to Potential Therapeutic Strategies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:4572471. [PMID: 33747345 PMCID: PMC7960032 DOI: 10.1155/2021/4572471] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 01/31/2021] [Accepted: 02/11/2021] [Indexed: 12/16/2022]
Abstract
The brain is a sexually dimorphic organ that implies different functions and structures depending on sex. Current pharmacological approaches against different neurological diseases act distinctly in male and female brains. In all neurodegenerative diseases, including Alzheimer's disease (AD), sex-related outcomes regarding pathogenesis, prevalence, and response to treatments indicate that sex differences are important for precise diagnosis and therapeutic strategy. Pathogenesis of AD includes vascular dementia, and in most cases, this is accompanied by metabolic complications with similar features as those assembled in diabetes. This review discusses how AD-associated dementia and diabetes affect cognition in relation to sex difference, as both diseases share similar pathological mechanisms. We highlight potential protective strategies to mitigate amyloid-beta (Aβ) pathogenesis, emphasizing how these drugs act in the male and female brains.
Collapse
Affiliation(s)
- Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmoud Ahmed Ebada
- Faculty of Medicine, Zagazig University, Zagazig, El-Sharkia, Egypt
- National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt
| | - Mohd Suhail
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ashraf Ali
- Department of Sciences of Agriculture, Food, Natural Resources, and Engineering (DAFNE), University of Foggia, Via Napoli 25, 71122 Foggia, Italy
| | - Md. Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Anwar L. Bilgrami
- Department of Entomology, Rutgers University, New Brunswick, NJ 018901, USA
- Deanship of Scientific Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Amjad Husain
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
- Centre for Science and Society, IISER Bhopal, India
- Innovation and Incubation Centre for Entrepreneurship, IISER Bhopal, India
| | - Mohd Tarique
- Department of Child Health, University of Missouri, Columbia, MO 65201, USA
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Saharanpur, India
| | - Athanasios Alexiou
- Novel Global Community Educational Foundation, New South Wales, Australia
- AFNP Med Austria, Wien, Austria
| | - Ausaf Ahmad
- Amity Institute of Biotechnology, Amity University Uttar Pradesh Lucknow Campus, Uttar Pradesh, India
| | - Rajnish Kumar
- Amity Institute of Biotechnology, Amity University Uttar Pradesh Lucknow Campus, Uttar Pradesh, India
| | - Naheed Banu
- Department of Physical Therapy, College of Medical Rehabilitation, Qassim University, Buraidah, Qassim, Saudi Arabia
| | - Agnieszka Najda
- Laboratory of Quality of Vegetables and Medicinal Plants, Department of Vegetable Crops and Medicinal Plants, University of Life Sciences in Lublin, 15 Akademicka Street, 20-950 Lublin, Poland
| | - Amany A. Sayed
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Ghadeer M. Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia
| | - Mohamed M. Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Ilaria Peluso
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), 00142 Rome, Italy
| | - George E. Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
- Health Research Institute, University of Limerick, Limerick, Ireland
| |
Collapse
|
110
|
Huang Y, Ma J, Jiang B, Yang N, Fu F, Chen X, Liu C, Miao X, Mao H, Zheng R, Wang J, Ding K, Zhang X. Effect of nutritional risk on cognitive function in patients with chronic obstructive pulmonary disease. J Int Med Res 2021; 49:300060521990127. [PMID: 33535842 PMCID: PMC7869158 DOI: 10.1177/0300060521990127] [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] [Indexed: 12/31/2022] Open
Abstract
Objective We aimed to clarify the cognitive function of patients with chronic obstructive pulmonary disease (COPD) and different nutritional status. Methods Among 95 patients with COPD in this retrospective study, we administered the Nutritional Risk Screening 2002 (NRS 2002) and Mini-Mental State Examination (MMSE). We recorded patients’ clinical characteristics, comorbidities, and laboratory measurements. According to NRS 2002 scores, patients were divided into two groups: no nutritional risk with NRS 2002 < 3 (n = 54) and nutritional risk, with NRS 2002 ≥ 3 (n = 41). Results We found a negative correlation between NRS 2002 and MMSE scores in participants with COPD (r = −0.313). Patients with nutritional risk were more likely to be cognitively impaired than those with no nutritional risk. Multivariate logistic regression analysis indicated that malnutrition was an independent risk factor for cognitive impairment, after adjusting for confounders (odds ratio [OR] = 4.120, 95% confidence interval [CI]: 1.072–15.837). We found a similar association between NRS 2002 and MMSE scores at 90-day follow-up using a Pearson’s correlation test (r = −0.493) and logistic regression analysis (OR = 7.333, 95% CI: 1.114–48.264). Conclusions Patients with COPD at nutritional risk are more likely to have cognitive impairment.
Collapse
Affiliation(s)
- Yiben Huang
- Department of Respiratory Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiedong Ma
- Department of Respiratory Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Bingqian Jiang
- Department of Respiratory Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Naiping Yang
- Department of Respiratory Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Fangyi Fu
- Department of Respiratory Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xianjing Chen
- Department of Respiratory Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chunyan Liu
- Department of Respiratory Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaqi Miao
- Department of Respiratory Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Huanhuan Mao
- Department of Respiratory Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rongrong Zheng
- Department of Respiratory Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jianing Wang
- Department of Respiratory Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Keke Ding
- Department of Respiratory Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaodiao Zhang
- Department of Respiratory Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
111
|
Bone WP, Siewert KM, Jha A, Klarin D, Damrauer SM, Chang KM, Tsao PS, Assimes TL, Ritchie MD, Voight BF. Multi-trait association studies discover pleiotropic loci between Alzheimer's disease and cardiometabolic traits. Alzheimers Res Ther 2021; 13:34. [PMID: 33541420 PMCID: PMC7860582 DOI: 10.1186/s13195-021-00773-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Identification of genetic risk factors that are shared between Alzheimer's disease (AD) and other traits, i.e., pleiotropy, can help improve our understanding of the etiology of AD and potentially detect new therapeutic targets. Previous epidemiological correlations observed between cardiometabolic traits and AD led us to assess the pleiotropy between these traits. METHODS We performed a set of bivariate genome-wide association studies coupled with colocalization analysis to identify loci that are shared between AD and eleven cardiometabolic traits. For each of these loci, we performed colocalization with Genotype-Tissue Expression (GTEx) project expression quantitative trait loci (eQTL) to identify candidate causal genes. RESULTS We identified three previously unreported pleiotropic trait associations at known AD loci as well as four novel pleiotropic loci. One associated locus was tagged by a low-frequency coding variant in the gene DOCK4 and is potentially implicated in its alternative splicing. Colocalization with GTEx eQTL data identified additional candidate genes for the loci we detected, including ACE, the target of the hypertensive drug class of ACE inhibitors. We found that the allele associated with decreased ACE expression in brain tissue was also associated with increased risk of AD, providing human genetic evidence of a potential increase in AD risk from use of an established anti-hypertensive therapeutic. CONCLUSION Our results support a complex genetic relationship between AD and these cardiometabolic traits, and the candidate causal genes identified suggest that blood pressure and immune response play a role in the pleiotropy between these traits.
Collapse
Affiliation(s)
- William P Bone
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Katherine M Siewert
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Anupama Jha
- Department of Computer and Information Science, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Derek Klarin
- Boston VA Healthcare System, Boston, MA, 02130, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Scott M Damrauer
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, PA, 19104, Philadelphia, USA
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kyong-Mi Chang
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Philip S Tsao
- VA Palo Alto Health Care System, Palo Alto, CA, 94550, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Themistocles L Assimes
- VA Palo Alto Health Care System, Palo Alto, CA, 94550, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Marylyn D Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center for Precision Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Benjamin F Voight
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, 19104, USA.
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
112
|
Hsiao SH, Hwang TJ, Lin FJ, Sheu JJ, Wu CH. The Association Between the Use of Cholinesterase Inhibitors and Cardiovascular Events Among Older Patients With Alzheimer Disease. Mayo Clin Proc 2021; 96:350-362. [PMID: 33549256 DOI: 10.1016/j.mayocp.2020.05.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/19/2020] [Accepted: 05/26/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To evaluate the association between the use of cholinesterase inhibitors (ChEIs) and incident cardiovascular events (CVEs) among older patients with Alzheimer disease (AD). PATIENTS AND METHODS This retrospective cohort study was conducted with a new-user design and active-comparator design. The data source was the 2005-2014 Full Population file from the Health and Welfare Database in Taiwan. Patients were included if they were aged 50 years or older and had been diagnosed with AD between January 1, 2006, and December 31, 2010. The association between ChEI use and the risk of CVEs was investigated in patients with AD. Among the ChEI users, the risk of CVEs was further compared between patients with different cumulative doses and different ChEI treatment strategies. The propensity score method, which included matching and inverse probability of treatment weighting, was used to balance the potential confounders. A Cox proportional hazards model with competing risks was used to estimate the hazard ratio of CVEs. RESULTS The study included 6070 patients with AD. After covariate adjustment, ChEI users had a significantly lower risk of CVEs than nonusers (hazard ratio, 0.57; 95% CI, 0.51 to 0.62). Among ChEI users, patients with a high cumulative dose had a significantly lower risk of CVEs than those with a low cumulative dose (hazard ratio, 0.82; 95% CI, 0.70 to 0.96). CONCLUSION The use of ChEIs was associated with a decreased risk of incident CVEs among patients with AD. The cardioprotective effect of ChEIs showed a dose-response relationship.
Collapse
Affiliation(s)
- Shih-Han Hsiao
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei City, Taiwan
| | - Tzung-Jeng Hwang
- Department of Psychiatry, College of Medicine and National Taiwan University Hospital, National Taiwan University, Taipei City
| | - Fang-Ju Lin
- Graduate Institute of Clinical Pharmacy, College of Medicine, National Taiwan University, Taipei City; School of Pharmacy, College of Medicine, National Taiwan University, Taipei City; Department of Pharmacy, National Taiwan University Hospital, Taipei City
| | - Jau-Jiuan Sheu
- Department of Neurology, Taipei Medical University Hospital, Taipei City, Taiwan; Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan
| | - Chung-Hsuen Wu
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei City, Taiwan.
| |
Collapse
|
113
|
Clark AL, Weigand AJ, Bangen KJ, Merritt VC, Bondi MW, Delano-Wood L. Repetitive mTBI is associated with age-related reductions in cerebral blood flow but not cortical thickness. J Cereb Blood Flow Metab 2021; 41:431-444. [PMID: 32248731 PMCID: PMC8369996 DOI: 10.1177/0271678x19897443] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mild traumatic brain injury (mTBI) is a risk factor for Alzheimer's disease (AD), and evidence suggests cerebrovascular dysregulation initiates deleterious neurodegenerative cascades. We examined whether mTBI history alters cerebral blood flow (CBF) and cortical thickness in regions vulnerable to early AD-related changes. Seventy-four young to middle-aged Veterans (mean age = 34, range = 23-48) underwent brain scans. Participants were divided into: (1) Veteran Controls (n = 27), (2) 1-2 mTBIs (n = 26), and (2) 3+ mTBIs (n = 21) groups. Resting CBF was measured using MP-PCASL. T1 structural scans were processed with FreeSurfer. CBF and cortical thickness estimates were extracted from nine AD-vulnerable regions. Regression analyses examined whether mTBI moderated the association between age, CBF, and cortical thickness. Regressions adjusting for sex and posttraumatic stress revealed mTBI moderated the association between age and CBF of the precuneus as well as superior and inferior parietal cortices (p's < .05); increasing age was associated with lower CBF in the 3+ mTBIs group, but not in the VCs or 1-2 mTBIs groups. mTBI did not moderate associations between age and cortical thickness (p's >.05). Repetitive mTBI is associated with cerebrovascular dysfunction in AD-vulnerable regions and may accelerate pathological aging trajectories.
Collapse
Affiliation(s)
- Alexandra L Clark
- VA San Diego Healthcare System (VASDHS), San Diego, CA, USA.,School of Medicine, Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Alexandra J Weigand
- San Diego State University/University of California, San Diego (SDSU/UCSD) Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
| | - Katherine J Bangen
- VA San Diego Healthcare System (VASDHS), San Diego, CA, USA.,School of Medicine, Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Victoria C Merritt
- VA San Diego Healthcare System (VASDHS), San Diego, CA, USA.,School of Medicine, Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Mark W Bondi
- VA San Diego Healthcare System (VASDHS), San Diego, CA, USA.,School of Medicine, Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Lisa Delano-Wood
- VA San Diego Healthcare System (VASDHS), San Diego, CA, USA.,School of Medicine, Department of Psychiatry, University of California San Diego, San Diego, CA, USA.,Center of Excellence for Stress and Mental Health, VASDHS, San Diego, CA, USA
| |
Collapse
|
114
|
Zenuni H, Grillo P, Sancesario GM, Bernardini S, Mercuri NB, Schirinzi T. How Comorbidity Reflects on Cerebrospinal Fluid Biomarkers of Neurodegeneration in Aging. J Alzheimers Dis Rep 2021; 5:87-92. [PMID: 33681720 PMCID: PMC7902985 DOI: 10.3233/adr-200280] [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] [Accepted: 12/29/2020] [Indexed: 12/23/2022] Open
Abstract
Systemic comorbidity precipitates the risk for dementia. To comprehend the underlying mechanisms into a therapeutic perspective, we analyzed how comorbidity affects neurodegeneration-related cerebrospinal fluid (CSF) biomarkers of 55 cognitively intact subjects. The Charson Comorbidity Index (CCI) was correlated with CSF amyloid-β42 (Aβ42), amyloid-β40, total-tau, 181-phosphorylated-tau (p-tau), the Aβ42/p-tau ratio, neurogranin, and lactate. The age-related brain lesions at imaging were also considered. CCI had a raw association with Aβ42/p-tau and p-tau, and a stronger, age-independent correlation with lactate. These preliminary findings suggested that, in normal subjects, systemic comorbidity might increase CNS oxidative stress and, together with aging, contribute to develop an Alzheimer's disease-like biochemical profile.
Collapse
Affiliation(s)
- Henri Zenuni
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Piergiorgio Grillo
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | | | - Sergio Bernardini
- Department of Experimental Medicine, University of Roma Tor Vergata, Rome, Italy
- Department of Clinical Biochemistry, Tor Vergata University Hospital, Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
- IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Tommaso Schirinzi
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| |
Collapse
|
115
|
Dake MD, De Marco M, Blackburn DJ, Wilkinson ID, Remes A, Liu Y, Pikkarainen M, Hallikainen M, Soininen H, Venneri A. Obesity and Brain Vulnerability in Normal and Abnormal Aging: A Multimodal MRI Study. J Alzheimers Dis Rep 2021; 5:65-77. [PMID: 33681718 PMCID: PMC7903016 DOI: 10.3233/adr-200267] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background: How the relationship between obesity and MRI-defined neural properties varies across distinct stages of cognitive impairment due to Alzheimer’s disease is unclear. Objective: We used multimodal neuroimaging to clarify this relationship. Methods: Scans were acquired from 47 patients clinically diagnosed with mild Alzheimer’s disease dementia, 68 patients with mild cognitive impairment, and 57 cognitively healthy individuals. Voxel-wise associations were run between maps of gray matter volume, white matter integrity, and cerebral blood flow, and global/visceral obesity. Results: Negative associations were found in cognitively healthy individuals between obesity and white matter integrity and cerebral blood flow of temporo-parietal regions. In mild cognitive impairment, negative associations emerged in frontal, temporal, and brainstem regions. In mild dementia, a positive association was found between obesity and gray matter volume around the right temporoparietal junction. Conclusion: Obesity might contribute toward neural tissue vulnerability in cognitively healthy individuals and mild cognitive impairment, while a healthy weight in mild Alzheimer’s disease dementia could help preserve brain structure in the presence of age and disease-related weight loss.
Collapse
Affiliation(s)
- Manmohi D Dake
- Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Matteo De Marco
- Department of Neuroscience, University of Sheffield, Sheffield, UK
| | | | - Iain D Wilkinson
- Academic Unit of Radiology, University of Sheffield, Sheffield, UK
| | - Anne Remes
- Department of Neurology, University of Eastern Finland, Kuopio, Finland
| | - Yawu Liu
- Department of Neurology, University of Eastern Finland, Kuopio, Finland
| | - Maria Pikkarainen
- Department of Neurology, University of Eastern Finland, Kuopio, Finland
| | - Merja Hallikainen
- Department of Neurology, University of Eastern Finland, Kuopio, Finland
| | - Hilkka Soininen
- Department of Neurology, University of Eastern Finland, Kuopio, Finland
| | - Annalena Venneri
- Department of Neuroscience, University of Sheffield, Sheffield, UK
| |
Collapse
|
116
|
de Bem AF, Krolow R, Farias HR, de Rezende VL, Gelain DP, Moreira JCF, Duarte JMDN, de Oliveira J. Animal Models of Metabolic Disorders in the Study of Neurodegenerative Diseases: An Overview. Front Neurosci 2021; 14:604150. [PMID: 33536868 PMCID: PMC7848140 DOI: 10.3389/fnins.2020.604150] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/24/2020] [Indexed: 12/21/2022] Open
Abstract
The incidence of metabolic disorders, as well as of neurodegenerative diseases—mainly the sporadic forms of Alzheimer’s and Parkinson’s disease—are increasing worldwide. Notably, obesity, diabetes, and hypercholesterolemia have been indicated as early risk factors for sporadic forms of Alzheimer’s and Parkinson’s disease. These conditions share a range of molecular and cellular features, including protein aggregation, oxidative stress, neuroinflammation, and blood-brain barrier dysfunction, all of which contribute to neuronal death and cognitive impairment. Rodent models of obesity, diabetes, and hypercholesterolemia exhibit all the hallmarks of these degenerative diseases, and represent an interesting approach to the study of the phenotypic features and pathogenic mechanisms of neurodegenerative disorders. We review the main pathological aspects of Alzheimer’s and Parkinson’s disease as summarized in rodent models of obesity, diabetes, and hypercholesterolemia.
Collapse
Affiliation(s)
- Andreza Fabro de Bem
- Department of Physiological Sciences, Institute of Biology, University of Brasilia, Brazilia, Brazil
| | - Rachel Krolow
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Hémelin Resende Farias
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Victória Linden de Rezende
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Daniel Pens Gelain
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - José Cláudio Fonseca Moreira
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - João Miguel das Neves Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Jade de Oliveira
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
117
|
Daniels RD, Clouston SAP, Hall CB, Anderson KR, Bennett DA, Bromet EJ, Calvert GM, Carreón T, DeKosky ST, Diminich ED, Finch CE, Gandy S, Kreisl WC, Kritikos M, Kubale TL, Mielke MM, Peskind ER, Raskind MA, Richards M, Sano M, Santiago-Colón A, Sloan RP, Spiro A, Vasdev N, Luft BJ, Reissman DB. A Workshop on Cognitive Aging and Impairment in the 9/11-Exposed Population. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:E681. [PMID: 33466931 PMCID: PMC7830144 DOI: 10.3390/ijerph18020681] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 12/11/2022]
Abstract
The terrorist attacks on 11 September 2001 potentially exposed more than 400,000 responders, workers, and residents to psychological and physical stressors, and numerous hazardous pollutants. In 2011, the World Trade Center Health Program (WTCHP) was mandated to monitor and treat persons with 9/11-related adverse health conditions and conduct research on physical and mental health conditions related to the attacks. Emerging evidence suggests that persons exposed to 9/11 may be at increased risk of developing mild cognitive impairment. To investigate further, the WTCHP convened a scientific workshop that examined the natural history of cognitive aging and impairment, biomarkers in the pathway of neurodegenerative diseases, the neuropathological changes associated with hazardous exposures, and the evidence of cognitive decline and impairment in the 9/11-exposed population. Invited participants included scientists actively involved in health-effects research of 9/11-exposed persons and other at-risk populations. Attendees shared relevant research results from their respective programs and discussed several options for enhancements to research and surveillance activities, including the development of a multi-institutional collaborative research network. The goal of this report is to outline the meeting's agenda and provide an overview of the presentation materials and group discussion.
Collapse
Affiliation(s)
- Robert D. Daniels
- World Trade Center Health Program, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Washington, DC 20201, USA; (K.R.A.); (G.M.C.); (T.C.); (T.L.K.); (A.S.-C.); (D.B.R.)
| | - Sean A. P. Clouston
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (S.A.P.C.); (E.J.B.); (E.D.D.); (M.K.); (B.J.L.)
| | - Charles B. Hall
- Department of Epidemiology & Population Health (Biostatistics), Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Kristi R. Anderson
- World Trade Center Health Program, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Washington, DC 20201, USA; (K.R.A.); (G.M.C.); (T.C.); (T.L.K.); (A.S.-C.); (D.B.R.)
| | - David A. Bennett
- Department of Neurological Sciences, Rush Medical College, Rush University, Chicago, IL 60612, USA;
| | - Evelyn J. Bromet
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (S.A.P.C.); (E.J.B.); (E.D.D.); (M.K.); (B.J.L.)
| | - Geoffrey M. Calvert
- World Trade Center Health Program, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Washington, DC 20201, USA; (K.R.A.); (G.M.C.); (T.C.); (T.L.K.); (A.S.-C.); (D.B.R.)
| | - Tania Carreón
- World Trade Center Health Program, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Washington, DC 20201, USA; (K.R.A.); (G.M.C.); (T.C.); (T.L.K.); (A.S.-C.); (D.B.R.)
| | - Steven T. DeKosky
- McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA;
| | - Erica D. Diminich
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (S.A.P.C.); (E.J.B.); (E.D.D.); (M.K.); (B.J.L.)
| | - Caleb E. Finch
- USC Leonard Davis School of Gerontology, Los Angeles, CA 90089, USA;
| | - Sam Gandy
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (S.G.); (M.S.)
| | - William C. Kreisl
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, New York, NY 10032, USA;
| | - Minos Kritikos
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (S.A.P.C.); (E.J.B.); (E.D.D.); (M.K.); (B.J.L.)
| | - Travis L. Kubale
- World Trade Center Health Program, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Washington, DC 20201, USA; (K.R.A.); (G.M.C.); (T.C.); (T.L.K.); (A.S.-C.); (D.B.R.)
| | - Michelle M. Mielke
- Division of Epidemiology and Department of Neurology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA;
| | - Elaine R. Peskind
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA;
| | - Murray A. Raskind
- Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA;
| | - Marcus Richards
- Faculty of Population Health Sciences, University College London, London WC1E 6BT, UK;
| | - Mary Sano
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (S.G.); (M.S.)
| | - Albeliz Santiago-Colón
- World Trade Center Health Program, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Washington, DC 20201, USA; (K.R.A.); (G.M.C.); (T.C.); (T.L.K.); (A.S.-C.); (D.B.R.)
| | - Richard P. Sloan
- Division of Behavioral Medicine, Columbia University, New York, NY 10027, USA;
| | - Avron Spiro
- Boston University Schools of Public Health and Medicine and Veterans Affairs Boston Healthcare System, Boston, MA 02130, USA;
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH) & Department of Psychiatry, University of Toronto, Toronto, ON M5S, Canada;
| | - Benjamin J. Luft
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (S.A.P.C.); (E.J.B.); (E.D.D.); (M.K.); (B.J.L.)
| | - Dori B. Reissman
- World Trade Center Health Program, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Washington, DC 20201, USA; (K.R.A.); (G.M.C.); (T.C.); (T.L.K.); (A.S.-C.); (D.B.R.)
| |
Collapse
|
118
|
Paul KC, Debes F, Eliasen E, Weihe P, Petersen MS. Incidence, gender influence, and neuropsychological predictors of all cause dementia in the Faroe Islands-the Faroese Septuagenarian cohort. Aging Clin Exp Res 2021; 33:105-114. [PMID: 32207093 PMCID: PMC7508821 DOI: 10.1007/s40520-020-01520-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/18/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Using the Faroese Septuagenarian cohort, we aimed to describe the incidence of dementia and assess the validity of neurocognitive tests to predict subsequent dementia diagnosis. METHODS In this population-based cohort, 713 Faroese septuagenarians aged 70-74 years without dementia, underwent clinical and neuropsychological examinations. After 10-years of follow-up, information was collected on all participants referred for cognitive evaluations and diagnosed with dementia. Incidence rates were calculated and presented with 95% confidence intervals (CIs), assuming a Poisson distribution. We then performed discriminant analysis to determine the best set of neuropsychological tests to identify those who would develop dementia. RESULTS Over the 10-years, 65 participants (9.1%) were diagnosed with dementia, with a 10-year incidence rate of 1063 cases per 100,000 person years (95% CI 825, 1343). Women had a greater incidence than men (incidence rate ratio (IRR) = 1.58; 95% CI 0.93, 2.71). After stepwise selection, gender and six neuropsychological measures were selected to discriminate between those who would and would not develop dementia. Overall, the model was able to correctly identify 82% of those who would not develop dementia (specificity) and 71% of those who would (sensitivity). CONCLUSIONS These results indicate that among a greater number of tests covering a broad range of cognitive abilities, tests reflecting verbal and visual learning and recall, visuospatial function, attention, and encoding into and retrieval from long-term memory may be helpful in identifying patients in the pre-symptomatic phase of dementia. Thus, helping care-givers identify patients at a higher risk of developing dementia and adjusting management of care accordingly.
Collapse
Affiliation(s)
- Kimberly C Paul
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Fróði Debes
- Department of Occupational Medicine and Public Health, The Faroese Hospital System, Tórshavn, Faroe Islands
| | - Eina Eliasen
- Department of Occupational Medicine and Public Health, The Faroese Hospital System, Tórshavn, Faroe Islands
| | - Pál Weihe
- Department of Occupational Medicine and Public Health, The Faroese Hospital System, Tórshavn, Faroe Islands
- Faculty of Health Sciences, Centre of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
| | - Maria Skaalum Petersen
- Department of Occupational Medicine and Public Health, The Faroese Hospital System, Tórshavn, Faroe Islands.
- Faculty of Health Sciences, Centre of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands.
| |
Collapse
|
119
|
Sharma MJ, Callahan BL. Cerebrovascular and Neurodegenerative Pathologies in Long-Term Stable Mild Cognitive Impairment. J Alzheimers Dis 2021; 79:1269-1283. [PMID: 33427736 DOI: 10.3233/jad-200829] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Mild cognitive impairment (MCI) is considered by some to be a prodromal phase of a progressive disease (i.e., neurodegeneration) resulting in dementia; however, a substantial portion of individuals (ranging from 5-30%) remain cognitively stable over the long term (sMCI). The etiology of sMCI is unclear but may be linked to cerebrovascular disease (CVD), as evidence from longitudinal studies suggest a significant proportion of individuals with vasculopathy remain stable over time. OBJECTIVE To quantify the presence of neurodegenerative and vascular pathologies in individuals with long-term (>5-year) sMCI, in a preliminary test of the hypothesis that CVD may be a contributor to non-degenerative cognitive impairment. We expect frequent vasculopathy at autopsy in sMCI relative to neurodegenerative disease, and relative to individuals who convert to dementia. METHODS In this retrospective study, using data from the National Alzheimer's Coordinating Center, individuals with sMCI (n = 28) were compared to those with MCI who declined over a 5 to 9-year period (dMCI; n = 139) on measures of neurodegenerative pathology (i.e., Aβ plaques, neurofibrillary tangles, TDP-43, and cerebral amyloid angiopathy) and CVD (infarcts, lacunes, microinfarcts, hemorrhages, and microbleeds). RESULTS Alzheimer's disease pathology (Aβ plaques, neurofibrillary tangles, and cerebral amyloid angiopathy) was significantly higher in the dMCI group than the sMCI group. Microinfarcts were the only vasculopathy associated with group membership; these were more frequent in sMCI. CONCLUSION The most frequent neuropathology in this sample of long-term sMCI was microinfarcts, tentatively suggesting that silent small vessel disease may characterize non-worsening cognitive impairment.
Collapse
Affiliation(s)
- Manu J Sharma
- Department of Psychology, University of Calgary, Calgary (AB), Canada
- Hotchkiss Brain Institute, Calgary (AB), Canada
| | - Brandy L Callahan
- Department of Psychology, University of Calgary, Calgary (AB), Canada
- Hotchkiss Brain Institute, Calgary (AB), Canada
| |
Collapse
|
120
|
Leszek J, Mikhaylenko EV, Belousov DM, Koutsouraki E, Szczechowiak K, Kobusiak-Prokopowicz M, Mysiak A, Diniz BS, Somasundaram SG, Kirkland CE, Aliev G. The Links between Cardiovascular Diseases and Alzheimer's Disease. Curr Neuropharmacol 2021; 19:152-169. [PMID: 32727331 PMCID: PMC8033981 DOI: 10.2174/1570159x18666200729093724] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/02/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
The root cause of non-inherited Alzheimer's disease (AD) remains unknown despite hundreds of research studies performed to attempt to solve this problem. Since proper prophylaxis remains the best strategy, many scientists have studied the risk factors that may affect AD development. There is robust evidence supporting the hypothesis that cardiovascular diseases (CVD) may contribute to AD progression, as the diseases often coexist. Therefore, a lack of well-defined diagnostic criteria makes studying the relationship between AD and CVD complicated. Additionally, inflammation accompanies the pathogenesis of AD and CVD, and is not only a consequence but also implicated as a significant contributor to the course of the diseases. Of note, АроЕε4 is found to be one of the major risk factors affecting both the cardiovascular and nervous systems. According to genome wide association and epidemiological studies, numerous common risk factors have been associated with the development of AD-related pathology. Furthermore, the risk of developing AD and CVDs appears to be increased by a wide range of conditions and lifestyle factors: hypertension, dyslipidemia, hypercholesterolemia, hyperhomocysteinemia, gut/oral microbiota, physical activity, and diet. This review summarizes the literature and provides possible mechanistic links between CVDs and AD.
Collapse
Affiliation(s)
- Jerzy Leszek
- Address correspondence to these authors at the Department of Psychiatry, Wrocław Medical University, Ul. Pasteura 10, 50-367, Wroclaw, Poland;, E-mail: and GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX 78229, USA; Tel: +1-210-442-8625 or +1-440-263-7461; E-mails: ,
| | | | | | | | | | | | | | | | | | | | - Gjumrakch Aliev
- Address correspondence to these authors at the Department of Psychiatry, Wrocław Medical University, Ul. Pasteura 10, 50-367, Wroclaw, Poland;, E-mail: and GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX 78229, USA; Tel: +1-210-442-8625 or +1-440-263-7461; E-mails: ,
| |
Collapse
|
121
|
Ostrovska SS, Liholetov EО, Pavlova VV, Derkach АK, Shevchenko IF, Adegova LY. RELATIONSHIP BETWEEN ALZHEIMER’S DISEASE, CEREBROVASCULAR AND CARDIOVASCULAR DISEASES (literature review). BULLETIN OF PROBLEMS BIOLOGY AND MEDICINE 2021. [DOI: 10.29254/2077-4214-2021-1-159-302-307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- S. S. Ostrovska
- Dnipro Medical Institute of Traditional and Alternative Medicine (Dnipro)
| | - E. О. Liholetov
- Dnipro Medical Institute of Traditional and Alternative Medicine (Dnipro)
| | - V. V. Pavlova
- Dnipro Medical Institute of Traditional and Alternative Medicine (Dnipro)
| | - А. K. Derkach
- Dnipro Medical Institute of Traditional and Alternative Medicine (Dnipro)
| | - I. F. Shevchenko
- Dnipro Medical Institute of Traditional and Alternative Medicine (Dnipro)
| | - L. Y. Adegova
- Dnipro Medical Institute of Traditional and Alternative Medicine (Dnipro)
| |
Collapse
|
122
|
Camarda C, Torelli P, Pipia C, Sottile G, Cilluffo G, Camarda R. APOE Genotypes and Brain Imaging Classes in Normal Cognition, Mild Cognitive Impairment, and Alzheimer's Disease: A Longitudinal Study. Curr Alzheimer Res 2020; 17:766-780. [PMID: 33167837 DOI: 10.2174/1567205017666201109093314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 08/20/2020] [Accepted: 10/10/2020] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To evaluate in 419 stroke-free cognitively normal subjects (CN) aged 45-82 years covering during a long prospective study (11.54 ± 1.47 years) the preclinical to dementia spectrum: 1) the distribution of small vessel disease (V) and brain atrophy (A) aggregated as following: V-/A-, V-/A+, V+/A-, V+/A+; 2) the relationship of these imaging classes with individual apolipoprotein E (APOE) genotypes; 3) the risk of progression to Alzheimer Disease (AD) of the individual APOE genotypes. METHODS Participants underwent one baseline (t0), and 4 clinical and neuropsychological assessments (t1,t2,t3, and t4). Brain MRI was performed in all subjects at t0, t2, t3 and t4.. White matter hyperintensities were assessed through two visual rating scales. Lacunes were also rated. Subcortical and global brain atrophy were determined through the bicaudate ratio and the lateral ventricle to brain ratio, respectively. APOE genotypes were determined at t0 in all subjects. Cox proportional hazard model was used to evaluate the risk of progression to AD. RESULTS The imaging class of mixed type was very common in AD, and in non amnestic mild cognitive impaired APOE ε4 non carriers. In these subjects, frontal and parieto-occipital regions were most affected by small vessel disease. CONCLUSION Our findings suggest that the APOE ε3 allele is probably linked to the brain vascular pathology.
Collapse
Affiliation(s)
- Cecilia Camarda
- Department of Biomedicine, Neurosciences, and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Paola Torelli
- Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy
| | | | - Gianluca Sottile
- Department of Economics, Business, and Statistics, University of Palermo, Palermo, Italy,Institute for Research and Biomedical Innovation (IRIB), National Research Council, Palermo, Italy
| | - Giovanna Cilluffo
- Institute for Research and Biomedical Innovation (IRIB), National Research Council, Palermo, Italy
| | - Rosolino Camarda
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| |
Collapse
|
123
|
Rosli NHM, Yahya HM, Ibrahim FW, Shahar S, Ismail IS, Azam AA, Rajab NF. Serum Metabolomics Profiling of Commercially Mixed Functional Foods—Effects in Beta-Amyloid Induced Rats Measured Using 1H NMR Spectroscopy. Nutrients 2020; 12:nu12123812. [PMID: 33322743 PMCID: PMC7764480 DOI: 10.3390/nu12123812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/04/2020] [Accepted: 12/09/2020] [Indexed: 11/16/2022] Open
Abstract
Functional foods such as pomegranate, dates and honey were shown by various previous studies to individually have a neuroprotective effect, especially in neurodegenerative disease such as Alzheimer’s disease (AD). In this novel and original study, an 1H NMR spectroscopy tool was used to identify the metabolic neuroprotective mechanism of commercially mixed functional foods (MFF) consisting of pomegranate, dates and honey, in rats injected with amyloid-beta 1-42 (Aβ-42). Forty-five male albino Wistar rats were randomly divided into five groups: NC (0.9% normal saline treatment + phosphate buffer solution (PBS) solution injection), Abeta (0.9% normal saline treatment + 0.2 µg/µL Aβ-42 injection), MFF (4 mL/kg MFF treatment + PBS solution injection), Abeta–MFF (4 mL/kg MFF treatment + 0.2 µg/µL Aβ-42 injection) and Abeta–NAC (150 mg/kg N-acetylcysteine + 0.2 µg/µL Aβ-42 injection). Based on the results, the MFF and NAC treatment improved the spatial memory and learning using Y-maze. In the metabolic analysis, a total of 12 metabolites were identified, for which levels changed significantly among the treatment groups. Systematic metabolic pathway analysis found that the MFF and NAC treatments provided a neuroprotective effect in Aβ-42 injected rats by improving the acid amino and energy metabolisms. Overall, this finding showed that MFF might serve as a potential neuroprotective functional food for the prevention of AD.
Collapse
Affiliation(s)
- Nur Hasnieza Mohd Rosli
- Biomedical Science Program, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Hanis Mastura Yahya
- Centre for Healthy Aging and Wellness (H-Care), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia; (H.M.Y.); (S.S.)
| | - Farah Wahida Ibrahim
- Centre for Toxicology and Health Risk Studies (CORE), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Suzana Shahar
- Centre for Healthy Aging and Wellness (H-Care), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia; (H.M.Y.); (S.S.)
| | - Intan Safinar Ismail
- Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (I.S.I.); (A.A.A.)
| | - Amalina Ahmad Azam
- Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (I.S.I.); (A.A.A.)
| | - Nor Fadilah Rajab
- Centre for Healthy Aging and Wellness (H-Care), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia; (H.M.Y.); (S.S.)
- Correspondence: ; Tel.: +60-3-9289-7002
| |
Collapse
|
124
|
Gao H, Song R, Li Y, Zhang W, Wan Z, Wang Y, Zhang H, Han S. Effects of Oat Fiber Intervention on Cognitive Behavior in LDLR -/- Mice Modeling Atherosclerosis by Targeting the Microbiome-Gut-Brain Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:14480-14491. [PMID: 33237770 DOI: 10.1021/acs.jafc.0c05677] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
It is known that cardiovascular disease can result in cognitive impairment. However, whether oat fiber improves cognitive behavior through a cardiovascular-related mechanism remains unclear. The present work was aimed to elucidate the potential of oat fiber on cognitive behavior by targeting the neuroinflammation signal and microbiome-gut-brain axis in a mouse model of atherosclerosis. Male low-density lipoprotein receptor knock-out (LDLR-/-) mice were treated with a high fat/cholesterol diet without or with 0.8% oat fiber for 14 weeks. Behavioral tests indicated that LDLR-/- mice exhibited a significant cognitive impairment; however, oat fiber can improve cognitive behavior by reducing latency to the platform and increasing the number of crossing and swimming distance in the target quadrant. Oat fiber can inhibit Aβ plaque processing in both the cortex and hippocampus via decreasing the relative protein expression of GFAP and IBα1. Notably, oat fiber inhibited the nod-like receptor family pyrin domain-containing 3 inflammasome activation and blocked the toll-like receptor 4 signal pathway in both the cortex and hippocampus, accompanied by a reduction of circulating serum lipopolysaccharide. In addition, oat fiber raised the expressions of short-chain fatty acid (SCFA) receptors and tight junction proteins (zonula occludens-1 and occludin) and improved intestinal microbiota diversity via increasing the contents of gut metabolites SCFAs. In summary, the present study provided experimental evidence that dietary oat fiber retarded the progression of cognitive impairment in a mouse model of atherosclerosis. Mechanistically, the neuroprotective potential was related to oat fiber and its metabolites SCFAs on the diversity and abundance of gut microbiota that produced anti-inflammatory metabolites, leading to repressed neuroinflammation and reduced gut permeability through the microbiome-gut-brain axis.
Collapse
Affiliation(s)
- Hui Gao
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, 199 Renai Road, Suzhou, 215123 Jiangsu, P.R. China
| | - Ruijuan Song
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, 199 Renai Road, Suzhou, 215123 Jiangsu, P.R. China
| | - Yuezhen Li
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, 199 Renai Road, Suzhou, 215123 Jiangsu, P.R. China
| | - Weiguo Zhang
- Independent Scientist, Irving, Texas 75039, United States
| | - Zhongxiao Wan
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, 199 Renai Road, Suzhou, 215123 Jiangsu, P.R. China
| | - Ying Wang
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, 199 Renai Road, Suzhou, 215123 Jiangsu, P.R. China
| | - Hong Zhang
- Department of Food and Nutrition, School of Public Health, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009 Jiangsu, P.R. China
| | - Shufen Han
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, 199 Renai Road, Suzhou, 215123 Jiangsu, P.R. China
| |
Collapse
|
125
|
Breijyeh Z, Karaman R. Comprehensive Review on Alzheimer's Disease: Causes and Treatment. Molecules 2020; 25:E5789. [PMID: 33302541 PMCID: PMC7764106 DOI: 10.3390/molecules25245789] [Citation(s) in RCA: 947] [Impact Index Per Article: 236.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/03/2020] [Accepted: 12/06/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is a disorder that causes degeneration of the cells in the brain and it is the main cause of dementia, which is characterized by a decline in thinking and independence in personal daily activities. AD is considered a multifactorial disease: two main hypotheses were proposed as a cause for AD, cholinergic and amyloid hypotheses. Additionally, several risk factors such as increasing age, genetic factors, head injuries, vascular diseases, infections, and environmental factors play a role in the disease. Currently, there are only two classes of approved drugs to treat AD, including inhibitors to cholinesterase enzyme and antagonists to N-methyl d-aspartate (NMDA), which are effective only in treating the symptoms of AD, but do not cure or prevent the disease. Nowadays, the research is focusing on understanding AD pathology by targeting several mechanisms, such as abnormal tau protein metabolism, β-amyloid, inflammatory response, and cholinergic and free radical damage, aiming to develop successful treatments that are capable of stopping or modifying the course of AD. This review discusses currently available drugs and future theories for the development of new therapies for AD, such as disease-modifying therapeutics (DMT), chaperones, and natural compounds.
Collapse
Affiliation(s)
| | - Rafik Karaman
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem 20002, Palestine;
| |
Collapse
|
126
|
Salisbury DL, Yu F. A Comparison of Cardiopulmonary Exercise Testing and Field Walking Tests in Community-Dwelling Older Adults With Mild-to-Moderate Alzheimer's Dementia. J Aging Phys Act 2020; 28:911-919. [PMID: 32498039 DOI: 10.1123/japa.2019-0387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 03/13/2020] [Accepted: 03/30/2020] [Indexed: 11/18/2022]
Abstract
The purpose of this study was to investigate the relationships among peak exercise parameters on 6-min walk test, shuttle walk test, and laboratory-based cardiopulmonary exercise testing in persons with Alzheimer's dementia. This study is a cross-sectional analysis of the baseline data of 90 participants (age 77.1 [6.6] years, 43% female) from the FIT-AD trial. Cardiopulmonary exercise testing produced significantly higher peak heart rate (118.6 [17.5] vs. 106 [22.8] vs. 106 [18.8] beats/min), rating of perceived exertion (16 [2.1] vs. 12 [2.3] vs. 11 [2.1]), and systolic blood pressure (182 [23.7] vs. 156 [18.9] vs. 150 [16.9] mmHg) compared with the shuttle walk test and 6-min walk test, respectively. Peak walking distance on shuttle walk test (241.3 [127.3] m) and 6-min walk test (365.0 [107.9] m) significantly correlated with peak oxygen consumption (17.1 [4.3] ml·kg-1·min-1) on cardiopulmonary exercise testing (r = .449, p ≤ .001 and r = .435, p ≤ .001), respectively, which is considerably lower than what is seen in older adults and persons with cardiopulmonary diseases.
Collapse
|
127
|
Lao PJ, Gutierrez J, Keator D, Rizvi B, Banerjee A, Igwe KC, Laing KK, Sathishkumar M, Moni F, Andrews H, Krinsky-McHale S, Head E, Lee JH, Lai F, Yassa MA, Rosas HD, Silverman W, Lott IT, Schupf N, Brickman AM. Alzheimer-Related Cerebrovascular Disease in Down Syndrome. Ann Neurol 2020; 88:1165-1177. [PMID: 32944999 PMCID: PMC7729262 DOI: 10.1002/ana.25905] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/15/2020] [Accepted: 09/15/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Adults with Down syndrome (DS) develop Alzheimer disease (AD) pathology by their 5th decade. Compared with the general population, traditional vascular risks in adults with DS are rare, allowing examination of cerebrovascular disease in this population and insight into its role in AD without the confound of vascular risk factors. We examined in vivo magnetic resonance imaging (MRI)-based biomarkers of cerebrovascular pathology in adults with DS, and determined their cross-sectional relationship with age, beta-amyloid pathology, and mild cognitive impairment or clinical AD diagnostic status. METHODS Participants from the Biomarkers of Alzheimer's Disease in Down Syndrome study (n = 138, 50 ± 7 years, 39% women) with MRI data and a subset (n = 90) with amyloid positron emission tomography (PET) were included. We derived MRI-based biomarkers of cerebrovascular pathology, including white matter hyperintensities (WMH), infarcts, cerebral microbleeds, and enlarged perivascular spaces (PVS), as well as PET-based biomarkers of amyloid burden. Participants were characterized as cognitively stable (CS), mild cognitive impairment-DS (MCI-DS), possible AD dementia, or definite AD dementia based on in-depth assessments of cognition, function, and health status. RESULTS There were detectable WMH, enlarged PVS, infarcts, and microbleeds as early as the 5th decade of life. There was a monotonic increase in WMH volume, enlarged PVS, and presence of infarcts across diagnostic groups (CS < MCI-DS < possible AD dementia < definite AD dementia). Higher amyloid burden was associated with a higher likelihood of an infarct. INTERPRETATION The findings highlight the prevalence of cerebrovascular disease in adults with DS and add to a growing body of evidence that implicates cerebrovascular disease as a core feature of AD and not simply a comorbidity. ANN NEUROL 2020;88:1165-1177.
Collapse
Affiliation(s)
- Patrick J Lao
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
| | - José Gutierrez
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY
| | - David Keator
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA
| | - Batool Rizvi
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Arit Banerjee
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Kay C Igwe
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Krystal K Laing
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Mithra Sathishkumar
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA
| | - Fahmida Moni
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Howard Andrews
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Psychology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY
| | - Sharon Krinsky-McHale
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Psychology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY
| | - Elizabeth Head
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA
| | - Joseph H Lee
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY
| | - Florence Lai
- Department of Neurology, Massachusetts General Hospital, Harvard University, Boston, MA
| | - Michael A Yassa
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA
| | - H Diana Rosas
- Department of Neurology, Massachusetts General Hospital, Harvard University, Boston, MA
- Department of Radiology, Athinoula Martinos Center, Massachusetts General Hospital, Harvard University, Charlestown, MA
| | - Wayne Silverman
- Department of Pediatrics, University of California, Irvine, Irvine, CA
| | - Ira T Lott
- Department of Pediatrics, University of California, Irvine, Irvine, CA
| | - Nicole Schupf
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Adam M Brickman
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY
| |
Collapse
|
128
|
Shabir O, Moll TA, Matuszyk MM, Eyre B, Dake MD, Berwick J, Francis SE. Preclinical models of disease and multimorbidity with focus upon cardiovascular disease and dementia. Mech Ageing Dev 2020; 192:111361. [DOI: 10.1016/j.mad.2020.111361] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/28/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022]
|
129
|
Howe MD, McCullough LD, Urayama A. The Role of Basement Membranes in Cerebral Amyloid Angiopathy. Front Physiol 2020; 11:601320. [PMID: 33329053 PMCID: PMC7732667 DOI: 10.3389/fphys.2020.601320] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/28/2020] [Indexed: 12/25/2022] Open
Abstract
Dementia is a neuropsychiatric syndrome characterized by cognitive decline in multiple domains, often leading to functional impairment in activities of daily living, disability, and death. The most common causes of age-related progressive dementia include Alzheimer's disease (AD) and vascular cognitive impairment (VCI), however, mixed disease pathologies commonly occur, as epitomized by a type of small vessel pathology called cerebral amyloid angiopathy (CAA). In CAA patients, the small vessels of the brain become hardened and vulnerable to rupture, leading to impaired neurovascular coupling, multiple microhemorrhage, microinfarction, neurological emergencies, and cognitive decline across multiple functional domains. While the pathogenesis of CAA is not well understood, it has long been thought to be initiated in thickened basement membrane (BM) segments, which contain abnormal protein deposits and amyloid-β (Aβ). Recent advances in our understanding of CAA pathogenesis link BM remodeling to functional impairment of perivascular transport pathways that are key to removing Aβ from the brain. Dysregulation of this process may drive CAA pathogenesis and provides an important link between vascular risk factors and disease phenotype. The present review summarizes how the structure and composition of the BM allows for perivascular transport pathways to operate in the healthy brain, and then outlines multiple mechanisms by which specific dementia risk factors may promote dysfunction of perivascular transport pathways and increase Aβ deposition during CAA pathogenesis. A better understanding of how BM remodeling alters perivascular transport could lead to novel diagnostic and therapeutic strategies for CAA patients.
Collapse
Affiliation(s)
| | | | - Akihiko Urayama
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
130
|
How Repair-or-Dispose Decisions Under Stress Can Initiate Disease Progression. iScience 2020; 23:101701. [PMID: 33235980 PMCID: PMC7670198 DOI: 10.1016/j.isci.2020.101701] [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: 02/07/2020] [Revised: 07/17/2020] [Accepted: 10/15/2020] [Indexed: 11/20/2022] Open
Abstract
Glia, the helper cells of the brain, are essential in maintaining neural resilience across time and varying challenges: By reacting to changes in neuronal health glia carefully balance repair or disposal of injured neurons. Malfunction of these interactions is implicated in many neurodegenerative diseases. We present a reductionist model that mimics repair-or-dispose decisions to generate a hypothesis for the cause of disease onset. The model assumes four tissue states: healthy and challenged tissue, primed tissue at risk of acute damage propagation, and chronic neurodegeneration. We discuss analogies to progression stages observed in the most common neurodegenerative conditions and to experimental observations of cellular signaling pathways of glia-neuron crosstalk. The model suggests that the onset of neurodegeneration can result as a compromise between two conflicting goals: short-term resilience to stressors versus long-term prevention of tissue damage.
Collapse
|
131
|
Tóth ME, Dukay B, Hoyk Z, Sántha M. Cerebrovascular Changes and Neurodegeneration Related to Hyperlipidemia: Characteristics of the Human ApoB-100 Transgenic Mice. Curr Pharm Des 2020; 26:1486-1494. [PMID: 32067608 PMCID: PMC7403644 DOI: 10.2174/1381612826666200218101818] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 11/27/2019] [Indexed: 01/07/2023]
Abstract
Serum lipid levels are closely related to the structure and function of blood vessels. Chronic hyperlipidemia may lead to damage in both the cardio- and the cerebrovascular systems. Vascular dysfunctions, including impairments of the blood-brain barrier, are known to be associated with neurodegenerative diseases. A growing number of evidence suggests that cardiovascular risk factors, such as hyperlipidemia, may increase the likelihood of developing dementia. Due to differences in lipoprotein metabolism, wild-type mice are protected against diet-induced hypercholesterolemia, and their serum lipid profile is different from that observed in humans. Therefore, several transgenic mouse models have been established to study the role of different apolipoproteins and their receptors in lipid metabolism, as well as the complications related to pathological lipoprotein levels. This mini-review focused on a transgenic mouse model overexpressing an apolipoprotein, the human ApoB-100. We discussed literature data and current advancements on the understanding of ApoB-100 induced cardio- and cerebrovascular lesions in order to demonstrate the involvement of this type of apolipoprotein in a wide range of pathologies, and a link between hyperlipidemia and neurodegeneration.
Collapse
Affiliation(s)
- Melinda E Tóth
- Institute of Biochemistry, Biological Research Centre, Sezeged, Hungary
| | - Brigitta Dukay
- Institute of Biochemistry, Biological Research Centre, Sezeged, Hungary.,Doctoral School in Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Zsófia Hoyk
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Temesvári krt. 62., Hungary
| | - Miklós Sántha
- Institute of Biochemistry, Biological Research Centre, Sezeged, Hungary
| |
Collapse
|
132
|
Kulshreshtha A, Goetz M, Alonso A, Shah AJ, Bremner JD, Goldberg J, Vaccarino V. Association Between Cardiovascular Health and Cognitive Performance: A Twins Study. J Alzheimers Dis 2020; 71:957-968. [PMID: 31476151 PMCID: PMC6918828 DOI: 10.3233/jad-190217] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND/OBJECTIVE The 2020 Strategic Impact Goal introduced by the American Heart Association (AHA) aims at improving cardiovascular health (CVH) of all Americans by 20%. AHA defined ideal CVH across seven established modifiable risk factors for cardiovascular diseases. Prior studies have indicated that ideal CVH also benefits brain health and cognitive aging, but it is possible that this association is explained by familial factors. METHODS We examined 272 male monozygotic and dizygotic twin pairs (total 544 subjects) free of overt cardiovascular disease and dementia from the Vietnam Era Twin Registry. Memory and learning were measured by Trail Making tests and Wechsler Memory Scale (Immediate and Delayed Memory tests and Visual Reproductive Test). Each of the seven CVH components (smoking, body mass index, physical activity, diet, total cholesterol, blood pressure, and blood glucose) was scored per established criterion. RESULTS The mean age of the twins was 55 years, 96% were whites, and 61% monozygotic. When considering twins as individuals, for every unit increase in CVH score (indicating better cardiovascular health), twins demonstrated faster cognitive processing speed (Trail B: - 5.6 s, 95% CI - 10.3, - 0.9; p = 0.03) and better story recall, both immediate (0.35, 95% CI 0.06, 0.62; p = 0.02) and delayed (0.39, 95% CI 0.08, 0.70; p = 0.01). CONCLUSIONS Better CVH is associated with better cognitive health in several domains. As suggested by within-pair analysis, this association is largely explained by familial factors, implying that early life exposures are shared determinants of both brain health and cardiovascular health.
Collapse
Affiliation(s)
- Ambar Kulshreshtha
- Department of Family and Preventive Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Margarethe Goetz
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Amit J Shah
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.,Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - J Douglas Bremner
- Department of Psychiatry and Behavioral Sciences and the Department of Radiology, Emory University School of Medicine, and the Atlanta VA Medical Center, Decatur, GA, USA
| | - Jack Goldberg
- Vietnam Era Twin Registry, Seattle, WA, USA.,University of Washington School of Public Health, Seattle, WA, USA
| | - Viola Vaccarino
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.,Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
133
|
White Matter Hyperintensities Contribute to Language Deficits in Primary Progressive Aphasia. Cogn Behav Neurol 2020; 33:179-191. [PMID: 32889950 DOI: 10.1097/wnn.0000000000000237] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To determine the contribution of white matter hyperintensities (WMH) to language deficits while accounting for cortical atrophy in individuals with primary progressive aphasia (PPA). METHOD Forty-three individuals with PPA completed neuropsychological assessments of nonverbal semantics, naming, and sentence repetition plus T2-weighted and fluid-attenuated inversion recovery scans. Using three visual scales, we rated WMH and cerebral ventricle size for both scan types. We used Spearman correlations to evaluate associations between the scales and scans. To test whether visual ratings-particularly of WMH-are associated with language, we compared a base model (including gray matter component scores obtained via principal component analysis, age, and days between assessment and MRI as independent variables) with full models (ie, the base model plus visual ratings) for each language variable. RESULTS Visual ratings were significantly associated within and between scans and were significantly correlated with age but not with other vascular risk factors. Only the T2 scan ratings were associated with language abilities. Specifically, controlling for other variables, poorer naming was significantly related to larger ventricles (P = 0.033) and greater global (P = 0.033) and periventricular (P = 0.049) WMH. High global WMH (P = 0.034) were also correlated with worse sentence repetition skills. CONCLUSION Visual ratings of global brain health were associated with language deficits in PPA independent of cortical atrophy and age. While WMH are not unique to PPA, measuring WMH in conjunction with cortical atrophy may elucidate more accurate brain structure-behavior relationships in PPA than cortical atrophy measures alone.
Collapse
|
134
|
Friberg L, Andersson T, Rosenqvist M. Less dementia and stroke in low-risk patients with atrial fibrillation taking oral anticoagulation. Eur Heart J 2020; 40:2327-2335. [PMID: 31095295 PMCID: PMC6642728 DOI: 10.1093/eurheartj/ehz304] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/30/2019] [Accepted: 05/03/2019] [Indexed: 12/04/2022] Open
Affiliation(s)
- Leif Friberg
- Department of Clinical Sciences, Karolinska Institutet at Danderyd University Hospital, Storskogsvägen 5, Stockholm, Sweden
| | - Tommy Andersson
- Department of Clinical Sciences, Karolinska Institutet at Danderyd University Hospital, Storskogsvägen 5, Stockholm, Sweden
| | - Mårten Rosenqvist
- Department of Clinical Sciences, Karolinska Institutet at Danderyd University Hospital, Storskogsvägen 5, Stockholm, Sweden
| |
Collapse
|
135
|
Krell-Roesch J, Syrjanen JA, Rakusa M, Vemuri P, Machulda MM, Kremers WK, Mielke MM, Lowe VJ, Jack CR, Knopman DS, Stokin GB, Petersen RC, Vassilaki M, Geda YE. Association of Cortical and Subcortical β-Amyloid With Standardized Measures of Depressive and Anxiety Symptoms in Adults Without Dementia. J Neuropsychiatry Clin Neurosci 2020; 33:64-71. [PMID: 33086924 PMCID: PMC7856245 DOI: 10.1176/appi.neuropsych.20050103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The purpose of this study was to test the hypothesis that subcortical β-amyloid (Aβ) deposition was associated with elevated scores on standardized measures of depressive and anxiety symptoms when compared with cortical (Aβ) deposition in persons without dementia. METHODS The authors performed a cross-sectional study, derived from the population-based Mayo Clinic Study of Aging, comprising participants aged ≥70 years (N=1,022; 55% males; 28% apolipoprotein E [APOE] ε4 carriers; without cognitive impairment, N=842; mild cognitive impairment; N=180). To assess Aβ deposition in cortical and subcortical (the amygdala, striatum, and thalamus) regions, participants underwent Pittsburgh Compound B positron emission tomography (PiB-PET) and completed the Beck Depression Inventory-II (BDI-II) and the Beck Anxiety Inventory (BAI). The investigators ran linear regression models to examine the association between PiB-PET standardized uptake value ratios (SUVRs) in the neocortex and subcortical regions and depressive and anxiety symptoms (BDI-II and BAI total scores). Models were adjusted for age, sex, education level, and APOE ε4 carrier status and stratified by cognitive status (without cognitive impairment, mild cognitive impairment). RESULTS Cortical PiB-PET SUVRs were associated with depressive symptoms (β=0.57 [SE=0.13], p<0.001) and anxiety symptoms (β=0.34 [SE=0.13], p=0.011). PiB-PET SUVRs in the amygdala were associated only with depressive symptoms (β=0.80 [SE=0.26], p=0.002). PiB-PET SUVRs in the striatum and thalamus were associated with depressive symptoms (striatum: β=0.69 [SE=0.18], p<0.001; thalamus: β=0.61 [SE=0.24], p=0.011) and anxiety symptoms (striatum: β=0.56 [SE=0.18], p=0.002; thalamus: β=0.65 [SE=0.24], p=0.008). In the mild cognitive impairment subsample, Aβ deposition, regardless of neuroanatomic location, was associated with depressive symptoms but not anxiety symptoms. CONCLUSIONS Elevated amyloid deposition in cortical and subcortical brain regions was associated with higher depressive and anxiety symptoms, although these findings did not significantly differ by cortical versus subcortical Aβ deposition. This cross-sectional observation needs to be confirmed by a longitudinal study.
Collapse
Affiliation(s)
- Janina Krell-Roesch
- Departments of Health Sciences Research (Krell-Roesch, Syrjanen, Rakusa, Kremers, Mielke, Vassilaki), Radiology (Vemuri, Lowe, Jack), Psychiatry and Psychology (Machulda), and Neurology (Mielke, Knopman, Petersen), Mayo Clinic, Rochester, Minn.; Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany (Krell-Roesch); Department of Neurology, University Medical Center, Maribor, Slovenia (Rakusa); International Clinical Research Center, St. Anne Hospital, Brno, Czech Republic (Stokin); and Department of Neurology, Barrow Neurological Institute, Phoenix (Geda)
| | - Jeremy A Syrjanen
- Departments of Health Sciences Research (Krell-Roesch, Syrjanen, Rakusa, Kremers, Mielke, Vassilaki), Radiology (Vemuri, Lowe, Jack), Psychiatry and Psychology (Machulda), and Neurology (Mielke, Knopman, Petersen), Mayo Clinic, Rochester, Minn.; Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany (Krell-Roesch); Department of Neurology, University Medical Center, Maribor, Slovenia (Rakusa); International Clinical Research Center, St. Anne Hospital, Brno, Czech Republic (Stokin); and Department of Neurology, Barrow Neurological Institute, Phoenix (Geda)
| | - Martin Rakusa
- Departments of Health Sciences Research (Krell-Roesch, Syrjanen, Rakusa, Kremers, Mielke, Vassilaki), Radiology (Vemuri, Lowe, Jack), Psychiatry and Psychology (Machulda), and Neurology (Mielke, Knopman, Petersen), Mayo Clinic, Rochester, Minn.; Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany (Krell-Roesch); Department of Neurology, University Medical Center, Maribor, Slovenia (Rakusa); International Clinical Research Center, St. Anne Hospital, Brno, Czech Republic (Stokin); and Department of Neurology, Barrow Neurological Institute, Phoenix (Geda)
| | - Prashanthi Vemuri
- Departments of Health Sciences Research (Krell-Roesch, Syrjanen, Rakusa, Kremers, Mielke, Vassilaki), Radiology (Vemuri, Lowe, Jack), Psychiatry and Psychology (Machulda), and Neurology (Mielke, Knopman, Petersen), Mayo Clinic, Rochester, Minn.; Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany (Krell-Roesch); Department of Neurology, University Medical Center, Maribor, Slovenia (Rakusa); International Clinical Research Center, St. Anne Hospital, Brno, Czech Republic (Stokin); and Department of Neurology, Barrow Neurological Institute, Phoenix (Geda)
| | - Mary M Machulda
- Departments of Health Sciences Research (Krell-Roesch, Syrjanen, Rakusa, Kremers, Mielke, Vassilaki), Radiology (Vemuri, Lowe, Jack), Psychiatry and Psychology (Machulda), and Neurology (Mielke, Knopman, Petersen), Mayo Clinic, Rochester, Minn.; Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany (Krell-Roesch); Department of Neurology, University Medical Center, Maribor, Slovenia (Rakusa); International Clinical Research Center, St. Anne Hospital, Brno, Czech Republic (Stokin); and Department of Neurology, Barrow Neurological Institute, Phoenix (Geda)
| | - Walter K Kremers
- Departments of Health Sciences Research (Krell-Roesch, Syrjanen, Rakusa, Kremers, Mielke, Vassilaki), Radiology (Vemuri, Lowe, Jack), Psychiatry and Psychology (Machulda), and Neurology (Mielke, Knopman, Petersen), Mayo Clinic, Rochester, Minn.; Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany (Krell-Roesch); Department of Neurology, University Medical Center, Maribor, Slovenia (Rakusa); International Clinical Research Center, St. Anne Hospital, Brno, Czech Republic (Stokin); and Department of Neurology, Barrow Neurological Institute, Phoenix (Geda)
| | - Michelle M Mielke
- Departments of Health Sciences Research (Krell-Roesch, Syrjanen, Rakusa, Kremers, Mielke, Vassilaki), Radiology (Vemuri, Lowe, Jack), Psychiatry and Psychology (Machulda), and Neurology (Mielke, Knopman, Petersen), Mayo Clinic, Rochester, Minn.; Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany (Krell-Roesch); Department of Neurology, University Medical Center, Maribor, Slovenia (Rakusa); International Clinical Research Center, St. Anne Hospital, Brno, Czech Republic (Stokin); and Department of Neurology, Barrow Neurological Institute, Phoenix (Geda)
| | - Val J Lowe
- Departments of Health Sciences Research (Krell-Roesch, Syrjanen, Rakusa, Kremers, Mielke, Vassilaki), Radiology (Vemuri, Lowe, Jack), Psychiatry and Psychology (Machulda), and Neurology (Mielke, Knopman, Petersen), Mayo Clinic, Rochester, Minn.; Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany (Krell-Roesch); Department of Neurology, University Medical Center, Maribor, Slovenia (Rakusa); International Clinical Research Center, St. Anne Hospital, Brno, Czech Republic (Stokin); and Department of Neurology, Barrow Neurological Institute, Phoenix (Geda)
| | - Clifford R Jack
- Departments of Health Sciences Research (Krell-Roesch, Syrjanen, Rakusa, Kremers, Mielke, Vassilaki), Radiology (Vemuri, Lowe, Jack), Psychiatry and Psychology (Machulda), and Neurology (Mielke, Knopman, Petersen), Mayo Clinic, Rochester, Minn.; Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany (Krell-Roesch); Department of Neurology, University Medical Center, Maribor, Slovenia (Rakusa); International Clinical Research Center, St. Anne Hospital, Brno, Czech Republic (Stokin); and Department of Neurology, Barrow Neurological Institute, Phoenix (Geda)
| | - David S Knopman
- Departments of Health Sciences Research (Krell-Roesch, Syrjanen, Rakusa, Kremers, Mielke, Vassilaki), Radiology (Vemuri, Lowe, Jack), Psychiatry and Psychology (Machulda), and Neurology (Mielke, Knopman, Petersen), Mayo Clinic, Rochester, Minn.; Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany (Krell-Roesch); Department of Neurology, University Medical Center, Maribor, Slovenia (Rakusa); International Clinical Research Center, St. Anne Hospital, Brno, Czech Republic (Stokin); and Department of Neurology, Barrow Neurological Institute, Phoenix (Geda)
| | - Gorazd B Stokin
- Departments of Health Sciences Research (Krell-Roesch, Syrjanen, Rakusa, Kremers, Mielke, Vassilaki), Radiology (Vemuri, Lowe, Jack), Psychiatry and Psychology (Machulda), and Neurology (Mielke, Knopman, Petersen), Mayo Clinic, Rochester, Minn.; Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany (Krell-Roesch); Department of Neurology, University Medical Center, Maribor, Slovenia (Rakusa); International Clinical Research Center, St. Anne Hospital, Brno, Czech Republic (Stokin); and Department of Neurology, Barrow Neurological Institute, Phoenix (Geda)
| | - Ronald C Petersen
- Departments of Health Sciences Research (Krell-Roesch, Syrjanen, Rakusa, Kremers, Mielke, Vassilaki), Radiology (Vemuri, Lowe, Jack), Psychiatry and Psychology (Machulda), and Neurology (Mielke, Knopman, Petersen), Mayo Clinic, Rochester, Minn.; Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany (Krell-Roesch); Department of Neurology, University Medical Center, Maribor, Slovenia (Rakusa); International Clinical Research Center, St. Anne Hospital, Brno, Czech Republic (Stokin); and Department of Neurology, Barrow Neurological Institute, Phoenix (Geda)
| | - Maria Vassilaki
- Departments of Health Sciences Research (Krell-Roesch, Syrjanen, Rakusa, Kremers, Mielke, Vassilaki), Radiology (Vemuri, Lowe, Jack), Psychiatry and Psychology (Machulda), and Neurology (Mielke, Knopman, Petersen), Mayo Clinic, Rochester, Minn.; Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany (Krell-Roesch); Department of Neurology, University Medical Center, Maribor, Slovenia (Rakusa); International Clinical Research Center, St. Anne Hospital, Brno, Czech Republic (Stokin); and Department of Neurology, Barrow Neurological Institute, Phoenix (Geda)
| | - Yonas E Geda
- Departments of Health Sciences Research (Krell-Roesch, Syrjanen, Rakusa, Kremers, Mielke, Vassilaki), Radiology (Vemuri, Lowe, Jack), Psychiatry and Psychology (Machulda), and Neurology (Mielke, Knopman, Petersen), Mayo Clinic, Rochester, Minn.; Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany (Krell-Roesch); Department of Neurology, University Medical Center, Maribor, Slovenia (Rakusa); International Clinical Research Center, St. Anne Hospital, Brno, Czech Republic (Stokin); and Department of Neurology, Barrow Neurological Institute, Phoenix (Geda)
| |
Collapse
|
136
|
Ejaz HW, Wang W, Lang M. Copper Toxicity Links to Pathogenesis of Alzheimer's Disease and Therapeutics Approaches. Int J Mol Sci 2020; 21:E7660. [PMID: 33081348 PMCID: PMC7589751 DOI: 10.3390/ijms21207660] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/24/2020] [Accepted: 09/29/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is an irreversible, age-related progressive neurological disorder, and the most common type of dementia in aged people. Neuropathological lesions of AD are neurofibrillary tangles (NFTs), and senile plaques comprise the accumulated amyloid-beta (Aβ), loaded with metal ions including Cu, Fe, or Zn. Some reports have identified metal dyshomeostasis as a neurotoxic factor of AD, among which Cu ions seem to be a central cationic metal in the formation of plaque and soluble oligomers, and have an essential role in the AD pathology. Cu-Aβ complex catalyzes the generation of reactive oxygen species (ROS) and results in oxidative damage. Several studies have indicated that oxidative stress plays a crucial role in the pathogenesis of AD. The connection of copper levels in AD is still ambiguous, as some researches indicate a Cu deficiency, while others show its higher content in AD, and therefore there is a need to increase and decrease its levels in animal models, respectively, to study which one is the cause. For more than twenty years, many in vitro studies have been devoted to identifying metals' roles in Aβ accumulation, oxidative damage, and neurotoxicity. Towards the end, a short review of the modern therapeutic approach in chelation therapy, with the main focus on Cu ions, is discussed. Despite the lack of strong proofs of clinical advantage so far, the conjecture that using a therapeutic metal chelator is an effective strategy for AD remains popular. However, some recent reports of genetic-regulating copper transporters in AD models have shed light on treating this refractory disease. This review aims to succinctly present a better understanding of Cu ions' current status in several AD features, and some conflicting reports are present herein.
Collapse
Affiliation(s)
- Hafza Wajeeha Ejaz
- CAS Center for Excellence in Biotic Interactions, College of Life Science, University of Chinese Academy of Sciences, Yuquan Road 19, Beijing 100049, China;
| | - Wei Wang
- School of Medical and Health Sciences, Edith Cowan University, Perth WA6027, Australia;
| | - Minglin Lang
- CAS Center for Excellence in Biotic Interactions, College of Life Science, University of Chinese Academy of Sciences, Yuquan Road 19, Beijing 100049, China;
- College of Life Science, Agricultural University of Hebei, Baoding 071000, China
| |
Collapse
|
137
|
Springer A, Monsch AU, Dutilh G, Coslovsky M, Kievit RA, Bonati LH, Conen D, Aeschbacher S, Beer JH, Schwenkglenks M, Fischer U, Meyer-Zuern CS, Conte G, Moutzouri E, Moschovitis G, Kühne M, Osswald S. A factor score reflecting cognitive functioning in patients from the Swiss Atrial Fibrillation Cohort Study (Swiss-AF). PLoS One 2020; 15:e0240167. [PMID: 33035257 PMCID: PMC7546506 DOI: 10.1371/journal.pone.0240167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 09/21/2020] [Indexed: 11/18/2022] Open
Abstract
Background Atrial fibrillation (AF), the most common sustained cardiac arrhythmia, is considered as risk factor for the development of mild cognitive impairment (MCI) and dementia. However, dynamics of cognitive functions are subtle, and neurocognitive assessments largely differ in detecting these changes. We aimed to develop and evaluate a score which represents the common aspects of the cognitive functions measured by validated tests (i.e., “general cognitive construct”), while reducing overlap between tests and be more sensitive to identify changes in overall cognitive functioning. Methods We developed the CoCo (cognitive construct) score to reflect the cognitive performance obtained by all items of four neurocognitive assessments (Montreal Cognitive Assessment (MoCA); Trail Making Test; Semantic Fluency, animals; Digital Symbol Substitution Test). The sample comprised 2,415 AF patients from the Swiss Atrial Fibrillation Cohort Study (Swiss-AF), 87% aged at least 65 years. Psychometric statistics were calculated for two cognitive measures based on (i) the full set of items from the neurocognitive test battery administered in the Swiss-AF study (i.e., CoCo item set) and (ii) the items from the widely used MoCA test. For the CoCo item set, a factor score was derived based on a principal component analysis, and its measurement properties were analyzed. Results Both the MoCA item set and the full neurocognitive test battery revealed good psychometric properties, especially the full battery. A one-factor model with good model fit and performance across time and groups was identified and used to generate the CoCo score, reflecting for each patient the common cognitive skill performance measured across the full neurocognitive test battery. The CoCo score showed larger effect sizes compared to the MoCA score in relation to relevant clinical variables. Conclusion The derived factor score allows summarizing AF patients’ cognitive performance as a single score. Using this score in the Swiss-AF project increases measurement sensitivity and decreases the number of statistical tests needed, which will be helpful in future studies addressing how AF affects the risk of developing cognitive impairment.
Collapse
Affiliation(s)
- Anne Springer
- Cardiology Division, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel (CRIB), Basel, Switzerland
- * E-mail:
| | - Andreas U. Monsch
- Memory Clinic, University Department of Geriatric Medicine FELIX PLATTER, Basel, Switzerland
- Faculty of Psychology, University of Basel, Basel, Switzerland
| | - Gilles Dutilh
- Department of Clinical Research, Clinical Trial Unit, University of Basel Hospital, Basel, Switzerland
| | - Michael Coslovsky
- Cardiology Division, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel (CRIB), Basel, Switzerland
- Department of Clinical Research, Clinical Trial Unit, University of Basel Hospital, Basel, Switzerland
| | - Rogier A. Kievit
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| | - Leo H. Bonati
- Department of Neurology and Stroke Center, University Hospital Basel, University of Basel, Basel, Switzerland
| | - David Conen
- Cardiovascular Research Institute Basel (CRIB), Basel, Switzerland
- Population Health Research Institute, McMaster University, Hamilton, Canada
| | - Stefanie Aeschbacher
- Cardiology Division, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel (CRIB), Basel, Switzerland
| | - Juerg H. Beer
- Department of Medicine, Cantonal Hospital of Baden and Molecular Cardiology, University Hospital of Zurich, Zurich, Switzerland
| | - Matthias Schwenkglenks
- Epidemiology, Biostatistics, and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Urs Fischer
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Christine S. Meyer-Zuern
- Cardiology Division, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel (CRIB), Basel, Switzerland
| | | | - Elisavet Moutzouri
- Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
- Department of General Internal Medicine, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
| | - Giorgio Moschovitis
- Population Health Research Institute, McMaster University, Hamilton, Canada
- Division of Cardiology, Ospedale Regionale di Lugano EOC, Lugano, Switzerland
| | - Michael Kühne
- Cardiology Division, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel (CRIB), Basel, Switzerland
| | - Stefan Osswald
- Cardiology Division, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel (CRIB), Basel, Switzerland
| | | |
Collapse
|
138
|
Vancampfort D, Solmi M, Firth J, Vandenbulcke M, Stubbs B. The Impact of Pharmacologic and Nonpharmacologic Interventions to Improve Physical Health Outcomes in People With Dementia: A Meta-Review of Meta-Analyses of Randomized Controlled Trials. J Am Med Dir Assoc 2020; 21:1410-1414.e2. [DOI: 10.1016/j.jamda.2020.01.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/03/2020] [Accepted: 01/08/2020] [Indexed: 12/18/2022]
|
139
|
Bhatt DL, Hull MA, Song M, Van Hulle C, Carlsson C, Chapman MJ, Toth PP. Beyond cardiovascular medicine: potential future uses of icosapent ethyl. Eur Heart J Suppl 2020; 22:J54-J64. [PMID: 33061868 PMCID: PMC7537800 DOI: 10.1093/eurheartj/suaa119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The REDUCE-IT trial demonstrated that icosapent ethyl, an ethyl ester of eicosapentaenoic acid (EPA), reduced cardiovascular events in an at-risk population by a substantial degree. While the cardiovascular protective properties of this compound are now proven, several other potential uses are being actively explored in clinical studies. These areas of investigation include cancer, inflammatory bowel disease, infections, Alzheimer's disease, dementia, and depression. The next decade promises to deepen our understanding of the beneficial effects that EPA may offer beyond cardiovascular risk reduction.
Collapse
Affiliation(s)
- Deepak L Bhatt
- Brigham and Women’s Hospital, Heart & Vascular Center and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Mark A Hull
- Division of Gastrointestinal and Surgical Sciences, Leeds Institute of Medical Research, St James’s University Hospital, University of Leeds, Leeds, LS9 7TF, UK
| | - Mingyang Song
- Departments of Epidemiology and Nutrition, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA 02115, USA
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital and Harvard Medical School, 100 Cambridge Street, Boston, MA 02114, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, 50 Fruit Street, Boston, MA 02114, USA
| | - Carol Van Hulle
- University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Cindy Carlsson
- William S. Middleton Memorial Veterans Hospital, Madison VA Geriatric Research, Education and Clinical Center (GRECC), 2500 Overlook Terrace, Madison, WI 53705, USA
- Division of Geriatrics and Gerontology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center (ADRC), 600 Highland Ave, J5/1 Mezzanine, Madison, WI 53792, USA
- Wisconsin Alzheimer’s Institute (WAI), 610 Walnut St Suite 957, Madison, WI 53726, USA
| | - M John Chapman
- Sorbonne University, 21, Rue de l'Ecole de Medicine, 75006 Paris, France
- Endocrinology-Metabolism Division, Pitie-Salpetriere University Hospital, 47-83, Boulevard de lopital, 75651 Paris Cedex, France
| | - Peter P Toth
- CGH Medical Center, 101 East Miller Road, Sterling, IL 61081, USA
- Cicarrone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
140
|
Gliozzi M, Musolino V, Bosco F, Scicchitano M, Scarano F, Nucera S, Zito MC, Ruga S, Carresi C, Macrì R, Guarnieri L, Maiuolo J, Tavernese A, Coppoletta AR, Nicita C, Mollace R, Palma E, Muscoli C, Belzung C, Mollace V. Cholesterol homeostasis: Researching a dialogue between the brain and peripheral tissues. Pharmacol Res 2020; 163:105215. [PMID: 33007421 DOI: 10.1016/j.phrs.2020.105215] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023]
Abstract
Cholesterol homeostasis is a highly regulated process in human body because of its several functions underlying the biology of cell membranes, the synthesis of all steroid hormones and bile acids and the need of trafficking lipids destined to cell metabolism. In particular, it has been recognized that peripheral and central nervous system cholesterol metabolism are separated by the blood brain barrier and are regulated independently; indeed, peripherally, it depends on the balance between dietary intake and hepatic synthesis on one hand and its degradation on the other, whereas in central nervous system it is synthetized de novo to ensure brain physiology. In view of this complex metabolism and its relevant functions in mammalian, impaired levels of cholesterol can induce severe cellular dysfunction leading to metabolic, cardiovascular and neurodegenerative diseases. The aim of this review is to clarify the role of cholesterol homeostasis in health and disease highlighting new intriguing aspects of the cross talk between its central and peripheral metabolism.
Collapse
Affiliation(s)
- Micaela Gliozzi
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Vincenzo Musolino
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Francesca Bosco
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Miriam Scicchitano
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Federica Scarano
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Saverio Nucera
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Maria Caterina Zito
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Stefano Ruga
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Cristina Carresi
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Roberta Macrì
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Lorenza Guarnieri
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Jessica Maiuolo
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Annamaria Tavernese
- Division of Cardiology, University Hospital Policlinico Tor Vergata, Rome, Italy.
| | - Anna Rita Coppoletta
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Caterina Nicita
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Rocco Mollace
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Ernesto Palma
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Carolina Muscoli
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy; IRCCS San Raffaele Pisana, Via di Valcannuta, Rome, Italy.
| | | | - Vincenzo Mollace
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy; IRCCS San Raffaele Pisana, Via di Valcannuta, Rome, Italy.
| |
Collapse
|
141
|
Anstey KJ, Cherbuin N, Kim S, McMaster M, D'Este C, Lautenschlager N, Rebok G, McRae I, Torres SJ, Cox KL, Pond CD. An Internet-Based Intervention Augmented With a Diet and Physical Activity Consultation to Decrease the Risk of Dementia in At-Risk Adults in a Primary Care Setting: Pragmatic Randomized Controlled Trial. J Med Internet Res 2020; 22:e19431. [PMID: 32969833 PMCID: PMC7545332 DOI: 10.2196/19431] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/28/2020] [Accepted: 08/03/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND There is a need to develop interventions to reduce the risk of dementia in the community by addressing lifestyle factors and chronic diseases over the adult life course. OBJECTIVE This study aims to evaluate a multidomain dementia risk reduction intervention, Body Brain Life in General Practice (BBL-GP), targeting at-risk adults in primary care. METHODS A pragmatic, parallel, three-arm randomized trial involving 125 adults aged 18 years or older (86/125, 68.8% female) with a BMI of ≥25 kg/m2 or a chronic health condition recruited from general practices was conducted. The arms included (1) BBL-GP, a web-based intervention augmented with an in-person diet and physical activity consultation; (2) a single clinician-led group, Lifestyle Modification Program (LMP); and (3) a web-based control. The primary outcome was the Australian National University Alzheimer Disease Risk Index Short Form (ANU-ADRI-SF). RESULTS Baseline assessments were conducted on 128 participants. A total of 125 participants were randomized to 3 groups (BBL-GP=42, LMP=41, and control=42). At immediate, week 18, week 36, and week 62 follow-ups, the completion rates were 43% (18/42), 57% (24/42), 48% (20/42), and 48% (20/42), respectively, for the BBL-GP group; 71% (29/41), 68% (28/41), 68% (28/41), and 51% (21/41), respectively, for the LMP group; and 62% (26/42), 69% (29/42), 60% (25/42), and 60% (25/42), respectively, for the control group. The primary outcome of the ANU-ADRI-SF score was lower for the BBL-GP group than the control group at all follow-ups. These comparisons were all significant at the 5% level for estimates adjusted for baseline differences (immediate: difference in means -3.86, 95% CI -6.81 to -0.90, P=.01; week 18: difference in means -4.05, 95% CI -6.81 to -1.28, P<.001; week 36: difference in means -4.99, 95% CI -8.04 to -1.94, P<.001; and week 62: difference in means -4.62, 95% CI -7.62 to -1.62, P<.001). CONCLUSIONS A web-based multidomain dementia risk reduction program augmented with allied health consultations administered within the general practice context can reduce dementia risk exposure for at least 15 months. This study was limited by a small sample size, and replication on a larger sample with longer follow-up will strengthen the results. TRIAL REGISTRATION Australian clinical trials registration number (ACTRN): 12616000868482; https://anzctr.org.au/ACTRN12616000868482.aspx.
Collapse
Affiliation(s)
- Kaarin J Anstey
- School of Psychology, University of New South Wales, Sydney, Australia.,Neuroscience Research Australia, Sydney, Australia.,Centre for Research on Ageing, Health and Wellbeing, Australian National University, Canberra, Australia
| | - Nicolas Cherbuin
- Centre for Research on Ageing, Health and Wellbeing, Australian National University, Canberra, Australia
| | - Sarang Kim
- Wicking Dementia Resaerch & Education Centre, University of Tasmania, Hobart, Australia
| | - Mitchell McMaster
- Centre for Research on Ageing, Health and Wellbeing, Australian National University, Canberra, Australia
| | - Catherine D'Este
- National Centre for Epidemiology and Public Health, Australian National University, Canberra, Australia.,School of Medicine and Public Health, University of Newcastle, Newcastle, Australia
| | - Nicola Lautenschlager
- Academic Unit for Psychiatry of Old Age, University of Melbourne, Melbourne, Australia
| | - George Rebok
- Johns Hopkins Centre on Aging and Health, Johns Hopkins University, Baltimore, MD, United States
| | - Ian McRae
- Centre for Research on Ageing, Health and Wellbeing, Australian National University, Canberra, Australia
| | - Susan J Torres
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Melbourne, Australia
| | - Kay L Cox
- Medical School, University of Western Australia, Perth, Australia
| | | |
Collapse
|
142
|
Endothelium-Independent Vasodilatory Effect of Sailuotong (SLT) on Rat Isolated Tail Artery. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:8125805. [PMID: 33029174 PMCID: PMC7527950 DOI: 10.1155/2020/8125805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/21/2020] [Accepted: 09/07/2020] [Indexed: 01/24/2023]
Abstract
Background Sailuotong (SLT) is a standardized three-herb formulation consisting of extracts of Panax ginseng, Ginkgo biloba, and Crocus sativus for the treatment of vascular dementia (VaD). Although SLT has been shown to increase cerebral blood flow, the direct effects of SLT on vascular reactivity have not been explored. This study aims to examine the vasodilatory effects of SLT and the underlying mechanisms in rat isolated tail artery. Methods Male (250-300 g) Wistar Kyoto (WKY) rat tail artery was isolated for isometric tension measurement. The effects of SLT on the influx of calcium through the cell membrane calcium channels were determined in Ca2+-free solution experiments. Results SLT (0.1-5,000 μg/ml) caused a concentration-dependent relaxation in rat isolated tail artery precontracted by phenylephrine. In the contraction experiments, SLT (500, 1,000, and 5,000 μg/mL) significantly inhibited phenylephrine (0.001 to 10 μM)- and KCl (10-80 mM)-induced contraction, in a concentration-dependent manner. In Ca2+-free solution, SLT (500, 1,000, and 5,000 μg/mL) markedly suppressed Ca2+-induced (0.001-3 mM) vasoconstriction in a concentration-dependent manner in both phenylephrine (10 μM) or KCl (80 mM) stimulated tail arteries. L-type calcium channel blocker nifedipine (10 μM) inhibited PE-induced contraction. Furthermore, SLT significantly reduced phenylephrine-induced transient vasoconstriction in the rat isolated tail artery. Conclusion SLT induces relaxation of rat isolated tail artery through endothelium-independent mechanisms. The SLT-induced vasodilatation appeared to be jointly meditated by blockages of extracellular Ca2+ influx via receptor-gated and voltage-gated Ca2+ channels and inhibition of the release of Ca2+ from the sarcoplasmic reticulum.
Collapse
|
143
|
Ikram M, Park TJ, Ali T, Kim MO. Antioxidant and Neuroprotective Effects of Caffeine against Alzheimer's and Parkinson's Disease: Insight into the Role of Nrf-2 and A2AR Signaling. Antioxidants (Basel) 2020; 9:antiox9090902. [PMID: 32971922 PMCID: PMC7554764 DOI: 10.3390/antiox9090902] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 12/15/2022] Open
Abstract
This paper reviews the results of studies conducted on the role of caffeine in the management of different neurological disorders, such as Parkinson's disease (PD) and Alzheimer's disease (AD). To highlight the potential role of caffeine in managing different neurodegenerative diseases, we identified studies by searching PubMed, Web of Science, and Google Scholar by scrutinizing the lists of pertinent publications. According to the collected overall findings, caffeine may reduce the elevated oxidative stress; inhibit the activation of adenosine A2A, thereby regulating the accumulation of Aβ; reduce the hyperphosphorylation of tau; and reduce the accumulation of misfolded proteins, such as α-synuclein, in Alzheimer's and Parkinson's diseases. The studies have suggested that caffeine has promising protective effects against different neurodegenerative diseases and that these effects may be used to tackle the neurological diseases and/or their consequences. Here, we review the ongoing research on the role of caffeine in the management of different neurodegenerative disorders, focusing on AD and PD. The current findings suggest that caffeine produces potent antioxidant, inflammatory, and anti-apoptotic effects against different models of neurodegenerative disease, including AD, PD, and other neurodegenerative disorders. Caffeine has shown strong antagonistic effects against the adenosine A2A receptor, which is a microglial receptor, and strong agonistic effects against nuclear-related factor-2 (Nrf-2), thereby regulating the cellular homeostasis at the brain by reducing oxidative stress, neuroinflammation, regulating the accumulation of α-synuclein in PD and tau hyperphosphorylation, amyloidogenesis, and synaptic deficits in AD, which are the cardinal features of these neurodegenerative diseases.
Collapse
Affiliation(s)
- Muhammad Ikram
- Division of Life Science and Applied Life Science (BK21 plus), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (M.I.); (T.A.)
| | - Tae Ju Park
- Paul O’Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow 0747 657 5394, UK;
| | - Tahir Ali
- Division of Life Science and Applied Life Science (BK21 plus), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (M.I.); (T.A.)
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 plus), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (M.I.); (T.A.)
- Correspondence: ; Tel.: +82-55-772-1345; Fax: +82-55-772-2656
| |
Collapse
|
144
|
Sharma VK, Singh TG. Insulin resistance and bioenergetic manifestations: Targets and approaches in Alzheimer's disease. Life Sci 2020; 262:118401. [PMID: 32926928 DOI: 10.1016/j.lfs.2020.118401] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/04/2020] [Accepted: 09/05/2020] [Indexed: 12/15/2022]
Abstract
AIM Insulin has a well-established role in cognition, neuronal detoxification and synaptic plasticity. Insulin transduction affect neurotransmitter functions, influence bioenergetics and regulate neuronal survival through regulating glucose energy metabolism and downward pathways. METHODS A systematic literature review of PubMed, Medline, Bentham, Scopus and EMBASE (Elsevier) databases was carried out with the help of the keywords like "Alzheimer's disease; Hypometabolism; Oxidative stress; energy failure in AD, Insulin; Insulin resistance; Bioenergetics" till June 2020. The review was conducted using the above keywords to collect the latest articles and to understand the nature of the extensive work carried out on insulin resistance and bioenergetic manifestations in Alzheimer's disease. KEY FINDINGS The article sheds light on insulin resistance mediated hypometabolic state on pathological progression of AD. The disrupted insulin signaling has pathological outcome in form of disturbed glucose homeostasis, altered bioenergetic state which increases build-up of senile plaques (Aβ), neurofibrillary tangles (τ), decline in transportation of glucose and activation of inflammatory pathways. The mechanistic link of insulin resistant state with therapeutically explorable potential transduction pathways is the focus of the reviewed work. SIGNIFICANCE The present work opines that the mechanism by which the insulin resistance mediates dysregulation of bioenergetics and progresses to neurodegenerative state holds the tangible potential to succeed in the development of novel dementia therapies. Further, hypometabolic complications and altered insulin signaling may be explored as a mechanistic relation between bioenergetic deficits and AD.
Collapse
Affiliation(s)
- Vivek Kumar Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India; Govt. College of Pharmacy, Rohru, District Shimla, Himachal Pradesh 171207, India
| | | |
Collapse
|
145
|
McGurran H, Glenn JM, Madero EN, Bott NT. Prevention and Treatment of Alzheimer's Disease: Biological Mechanisms of Exercise. J Alzheimers Dis 2020; 69:311-338. [PMID: 31104021 DOI: 10.3233/jad-180958] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. With an aging population and no disease modifying treatments available, AD is quickly becoming a global pandemic. A substantial body of research indicates that lifestyle behaviors contribute to the development of AD, and that it may be worthwhile to approach AD like other chronic diseases such as cardiovascular disease, in which prevention is paramount. Exercise is an important lifestyle behavior that may influence the course and pathology of AD, but the biological mechanisms underpinning these effects remain unclear. This review focuses on how exercise can modify four possible mechanisms which are involved with the pathology of AD: oxidative stress, inflammation, peripheral organ and metabolic health, and direct interaction with AD pathology. Exercise is just one of many lifestyle behaviors that may assist in preventing AD, but understanding the systemic and neurobiological mechanisms by which exercise affects AD could help guide the development of novel pharmaceutical agents and non-pharmacological personalized lifestyle interventions for at-risk populations.
Collapse
Affiliation(s)
- Hugo McGurran
- Research Master's Programme Brain and Cognitive Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | - Nicholas T Bott
- Neurotrack Technologies Inc., Redwood City, CA, USA.,Clinical Excellence Research Center, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Department of Psychology, PGSP-Stanford Consortium, Palo Alto University, Palo Alto, CA, USA
| |
Collapse
|
146
|
Lee H, Kim E. Repositioning medication for cardiovascular and cerebrovascular disease to delay the onset and prevent progression of Alzheimer's disease. Arch Pharm Res 2020; 43:932-960. [PMID: 32909178 DOI: 10.1007/s12272-020-01268-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 08/31/2020] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is a complex, progressive, neurodegenerative disorder. As with other common chronic diseases, multiple risk factors contribute to the onset and progression of AD. Many researchers have evaluated the epidemiologic and pathophysiological association between AD, cardiovascular diseases (CVDs), and cerebrovascular diseases (CBVDs), including commonly reported risk factors such as diabetes, hypertension, and dyslipidemia. Relevant therapies of CVDs/CBVDs for the attenuation of AD have also been empirically investigated. Considering the challenges of new drug development, in terms of cost and time, multifactorial approaches such as therapeutic repositioning of CVD/CBVD medication should be explored to delay the onset and progression of AD. Thus, in this review, we discuss our current understanding of the association between cardiovascular risk factors and AD, as revealed by clinical and non-clinical studies, as well as the therapeutic implications of CVD/CBVD medication that may attenuate AD. Furthermore, we discuss future directions by evaluating ongoing trials in the field.
Collapse
Affiliation(s)
- Heeyoung Lee
- Department of Clinical Medicinal Sciences, Konyang University, 121 Daehakro, Nonsan, 32992, Republic of Korea
| | - EunYoung Kim
- Evidence-Based Research Laboratory, Division of Clinical Pharmacotherapy, College of Pharmacy, Chung-Ang University, Seoul, 156-756, Republic of Korea.
| |
Collapse
|
147
|
Cerebral Small Vessel Disease Influences Hippocampal Subfield Atrophy in Mild Cognitive Impairment. Transl Stroke Res 2020; 12:284-292. [PMID: 32894401 DOI: 10.1007/s12975-020-00847-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/15/2020] [Accepted: 08/31/2020] [Indexed: 01/04/2023]
Abstract
To investigate patterns of hippocampal subfield atrophy among patients with amnestic mild cognitive impairment, stratified by severity of small vessel disease (SVD) and corresponding associations with cognitive domains. One hundred seventy-six MCI subjects (mean age = 65.56 years, SD = 8.77) underwent neuropsychological assessments and magnetic resonance imaging. SVD was rated 0 (no SVD), 1 (mild SVD) and 2 (moderate to severe SVD) based on load of white matter hyperintensities (WMH) and lacunes. Demographics, cerebrovascular risk factors, grey and white matter volumes and hippocampal subfield atrophies were compared across SVD severity through ANCOVA analyses. Subjects were categorized into positive or negative SVD-hippocampal subfield atrophy (HSA) and influence of positive SVD-HSA on episodic memory and frontal executive function was evaluated through ANCOVA analyses. All analyses corrected for covariates and bias-corrected bootstrap estimation with 1000 resamples was applied with Bonferroni correction. Hippocampal subfield atrophy worsened with increasing SVD severity. Positive SVD-HSA was characterised by significant atrophy in the subiculum, CA1, CA4, molecular layer and dentate gyrus. Greater atrophy was seen with moderate to severe SVD compared to mild SVD in these subfields. Atrophy in the five subfields of SVD-HSA was significantly associated with poor episodic memory and frontal executive function. Presence and burden of SVD influences the pattern and severity of hippocampal subfield atrophy. SVD-related hippocampal subfield atrophy is associated with poorer episodic memory and frontal executive function in mild cognitive impairment.
Collapse
|
148
|
Hay M, Barnes C, Huentelman M, Brinton R, Ryan L. Hypertension and Age-Related Cognitive Impairment: Common Risk Factors and a Role for Precision Aging. Curr Hypertens Rep 2020; 22:80. [PMID: 32880739 PMCID: PMC7467861 DOI: 10.1007/s11906-020-01090-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Purpose of Review Precision Aging® is a novel concept that we have recently employed to describe how the model of precision medicine can be used to understand and define the multivariate risks that drive age-related cognitive impairment (ARCI). Hypertension and cardiovascular disease are key risk factors for both brain function and cognitive aging. In this review, we will discuss the common mechanisms underlying the risk factors for both hypertension and ARCI and how the convergence of these mechanisms may be amplified in an individual to drive changes in brain health and accelerate cognitive decline. Recent Findings Currently, our cognitive health span does not match our life span. Age-related cognitive impairment and preventing and treating ARCI will require an in-depth understanding of the interrelated risk factors, including individual genetic profiles, that affect brain health and brain aging. Hypertension and cardiovascular disease are important risk factors for ARCI. And, many of the risk factors for developing hypertension, such as diabetes, smoking, stress, viral infection, and age, are shared with the development of ARCI. We must first understand the mechanisms common to the converging risk factors in hypertension and ARCI and then design person-specific therapies to optimize individual brain health. Summary The understanding of the convergence of shared risk factors between hypertension and ARCI is required to develop individualized interventions to optimize brain health across the life span. We will conclude with a discussion of possible steps that may be taken to decrease ARCI and optimize an individual’s cognitive life span.
Collapse
Affiliation(s)
- Meredith Hay
- Department of Physiology, University of Arizona, 1501 N Campbell Rd, Room 4103, Tucson, AZ, 85724, USA.
- Psychology Department, University of Arizona, Tucson, AZ, USA.
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA.
| | - Carol Barnes
- Psychology Department, University of Arizona, Tucson, AZ, USA
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
| | - Matt Huentelman
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
- Neurogenomics Division, TGen, Phoenix, AZ, USA
| | - Roberta Brinton
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
- Center for Innovative Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Lee Ryan
- Psychology Department, University of Arizona, Tucson, AZ, USA
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
149
|
Falkenhain K, Ruiz-Uribe NE, Haft-Javaherian M, Ali M, Michelucci PE, Schaffer CB, Bracko O. A pilot study investigating the effects of voluntary exercise on capillary stalling and cerebral blood flow in the APP/PS1 mouse model of Alzheimer's disease. PLoS One 2020; 15:e0235691. [PMID: 32857763 PMCID: PMC7455035 DOI: 10.1371/journal.pone.0235691] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/19/2020] [Indexed: 11/18/2022] Open
Abstract
Exercise exerts a beneficial effect on the major pathological and clinical symptoms associated with Alzheimer’s disease in humans and mouse models of the disease. While numerous mechanisms for such benefits from exercise have been proposed, a clear understanding of the causal links remains elusive. Recent studies also suggest that cerebral blood flow in the brain of both Alzheimer’s patients and mouse models of the disease is decreased and that the cognitive symptoms can be improved when blood flow is restored. We therefore hypothesized that the mitigating effect of exercise on the development and progression of Alzheimer’s disease may be mediated through an increase in the otherwise reduced brain blood flow. To test this idea, we performed a pilot study to examine the impact of three months of voluntary wheel running in a small cohort of ~1-year-old APP/PS1 mice on short-term memory function, brain inflammation, amyloid deposition, and baseline cerebral blood flow. Our findings that exercise led to a trend toward improved spatial short-term memory, reduced brain inflammation, markedly increased neurogenesis in the dentate gyrus, and a reduction in hippocampal amyloid-beta deposits are consistent with other reports on the impact of exercise on the progression of Alzheimer’s related symptoms in mouse models. Notably, we did not observe any impact of wheel running on overall baseline blood flow nor on the incidence of non-flowing capillaries, a mechanism we recently identified as one contributing factor to cerebral blood flow deficits in mouse models of Alzheimer’s disease. Overall, our findings add to the emerging picture of differential effects of exercise on cognition and blood flow in Alzheimer’s disease pathology by showing that capillary stalling is not decreased following exercise.
Collapse
Affiliation(s)
- Kaja Falkenhain
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
- Institute of Cognitive Science, Osnabrück University, Osnabrück, Germany
| | - Nancy E. Ruiz-Uribe
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Mohammad Haft-Javaherian
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Muhammad Ali
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | | | | | - Chris B. Schaffer
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Oliver Bracko
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
- * E-mail:
| |
Collapse
|
150
|
Snitz BE, Chang Y, Tudorascu DL, Lopez OL, Lopresti BJ, DeKosky ST, Carlson MC, Cohen AD, Kamboh MI, Aizenstein HJ, Klunk WE, Kuller LH. Predicting resistance to amyloid-beta deposition and cognitive resilience in the oldest-old. Neurology 2020; 95:e984-e994. [PMID: 32699143 PMCID: PMC7668550 DOI: 10.1212/wnl.0000000000010239] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 02/20/2020] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE To explore long-term predictors of avoiding β-amyloid (Aβ) deposition and maintaining unimpaired cognition as outcomes in the oldest old. METHODS In a longitudinal observational cohort study, 100 former participants of the Ginkgo Evaluation of Memory Study (GEMS; 2000-2008) completed biannual Pittsburgh compound B-PET imaging and annual clinical-cognitive evaluations beginning in 2010. Most recent Aβ status and cognitive status were selected for each participant. Longitudinal outcomes included change in serial Aβ and cognitive tests. Baseline predictors from GEMS included neuropsychological tests, daily functioning, APOE genotype, lifestyle variables, occupational measures, health history, sleep, subjective memory, physical and cognitive activities, depressive symptoms, and physical performance and health indices, among others. RESULTS Mean age at the last cognitive evaluation was 92.0 (range 86-100) years. Mean follow-up time from baseline to last measured Aβ status was 12.3 (SD 1.9) years and to last cognitive evaluation was 14.1 (SD 1.9) years. The APOE*2 allele predicted last Aβ status (n = 34 Aβ negative vs n = 66 Aβ positive). Baseline cognition predicted cognitive status (n = 30 unimpaired vs n = 70 impaired). Predictors of cognitive status among Aβ-positive participants only (n = 14 normal cognition vs n = 52 impaired) were baseline cognitive test scores and smoking history. Baseline pulse pressure predicted longitudinal Aβ increase; paid work engagement and life satisfaction predicted less cognitive decline. CONCLUSIONS The APOE*2 allele and lower pulse pressure predict resistance to Aβ deposition in advanced aging. Cognitive test scores 14 years prior, likely reflecting premorbid abilities, predict cognitive status and maintenance of unimpaired cognition in the presence of Aβ. Several lifestyle factors appear protective.
Collapse
Affiliation(s)
- Beth E Snitz
- From the Departments of Neurology (B.E.S., O.L.L., W.E.K.), Neurological Surgery (Y.C.), Medicine (D.L.T.), Radiology (B.J.L.), Psychiatry (A.D.C., H.J.A., W.E.K.), Human Genetics (M.I.K.), and Epidemiology (L.H.K.), University of Pittsburgh, PA; Department of Neurology (S.T.D.), University of Florida, Gainesville; and Department of Mental Health (M.C.C.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD.
| | - Yuefang Chang
- From the Departments of Neurology (B.E.S., O.L.L., W.E.K.), Neurological Surgery (Y.C.), Medicine (D.L.T.), Radiology (B.J.L.), Psychiatry (A.D.C., H.J.A., W.E.K.), Human Genetics (M.I.K.), and Epidemiology (L.H.K.), University of Pittsburgh, PA; Department of Neurology (S.T.D.), University of Florida, Gainesville; and Department of Mental Health (M.C.C.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Dana L Tudorascu
- From the Departments of Neurology (B.E.S., O.L.L., W.E.K.), Neurological Surgery (Y.C.), Medicine (D.L.T.), Radiology (B.J.L.), Psychiatry (A.D.C., H.J.A., W.E.K.), Human Genetics (M.I.K.), and Epidemiology (L.H.K.), University of Pittsburgh, PA; Department of Neurology (S.T.D.), University of Florida, Gainesville; and Department of Mental Health (M.C.C.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Oscar L Lopez
- From the Departments of Neurology (B.E.S., O.L.L., W.E.K.), Neurological Surgery (Y.C.), Medicine (D.L.T.), Radiology (B.J.L.), Psychiatry (A.D.C., H.J.A., W.E.K.), Human Genetics (M.I.K.), and Epidemiology (L.H.K.), University of Pittsburgh, PA; Department of Neurology (S.T.D.), University of Florida, Gainesville; and Department of Mental Health (M.C.C.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Brian J Lopresti
- From the Departments of Neurology (B.E.S., O.L.L., W.E.K.), Neurological Surgery (Y.C.), Medicine (D.L.T.), Radiology (B.J.L.), Psychiatry (A.D.C., H.J.A., W.E.K.), Human Genetics (M.I.K.), and Epidemiology (L.H.K.), University of Pittsburgh, PA; Department of Neurology (S.T.D.), University of Florida, Gainesville; and Department of Mental Health (M.C.C.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Steven T DeKosky
- From the Departments of Neurology (B.E.S., O.L.L., W.E.K.), Neurological Surgery (Y.C.), Medicine (D.L.T.), Radiology (B.J.L.), Psychiatry (A.D.C., H.J.A., W.E.K.), Human Genetics (M.I.K.), and Epidemiology (L.H.K.), University of Pittsburgh, PA; Department of Neurology (S.T.D.), University of Florida, Gainesville; and Department of Mental Health (M.C.C.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Michelle C Carlson
- From the Departments of Neurology (B.E.S., O.L.L., W.E.K.), Neurological Surgery (Y.C.), Medicine (D.L.T.), Radiology (B.J.L.), Psychiatry (A.D.C., H.J.A., W.E.K.), Human Genetics (M.I.K.), and Epidemiology (L.H.K.), University of Pittsburgh, PA; Department of Neurology (S.T.D.), University of Florida, Gainesville; and Department of Mental Health (M.C.C.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Ann D Cohen
- From the Departments of Neurology (B.E.S., O.L.L., W.E.K.), Neurological Surgery (Y.C.), Medicine (D.L.T.), Radiology (B.J.L.), Psychiatry (A.D.C., H.J.A., W.E.K.), Human Genetics (M.I.K.), and Epidemiology (L.H.K.), University of Pittsburgh, PA; Department of Neurology (S.T.D.), University of Florida, Gainesville; and Department of Mental Health (M.C.C.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - M Ilyas Kamboh
- From the Departments of Neurology (B.E.S., O.L.L., W.E.K.), Neurological Surgery (Y.C.), Medicine (D.L.T.), Radiology (B.J.L.), Psychiatry (A.D.C., H.J.A., W.E.K.), Human Genetics (M.I.K.), and Epidemiology (L.H.K.), University of Pittsburgh, PA; Department of Neurology (S.T.D.), University of Florida, Gainesville; and Department of Mental Health (M.C.C.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Howard J Aizenstein
- From the Departments of Neurology (B.E.S., O.L.L., W.E.K.), Neurological Surgery (Y.C.), Medicine (D.L.T.), Radiology (B.J.L.), Psychiatry (A.D.C., H.J.A., W.E.K.), Human Genetics (M.I.K.), and Epidemiology (L.H.K.), University of Pittsburgh, PA; Department of Neurology (S.T.D.), University of Florida, Gainesville; and Department of Mental Health (M.C.C.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - William E Klunk
- From the Departments of Neurology (B.E.S., O.L.L., W.E.K.), Neurological Surgery (Y.C.), Medicine (D.L.T.), Radiology (B.J.L.), Psychiatry (A.D.C., H.J.A., W.E.K.), Human Genetics (M.I.K.), and Epidemiology (L.H.K.), University of Pittsburgh, PA; Department of Neurology (S.T.D.), University of Florida, Gainesville; and Department of Mental Health (M.C.C.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Lewis H Kuller
- From the Departments of Neurology (B.E.S., O.L.L., W.E.K.), Neurological Surgery (Y.C.), Medicine (D.L.T.), Radiology (B.J.L.), Psychiatry (A.D.C., H.J.A., W.E.K.), Human Genetics (M.I.K.), and Epidemiology (L.H.K.), University of Pittsburgh, PA; Department of Neurology (S.T.D.), University of Florida, Gainesville; and Department of Mental Health (M.C.C.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|