1
|
Chauhan P, Wadhwa K, Singh G, Gupta S, Iqbal D, Abomughaid MM, Almutary AG, Mishra PC, Nelson VK, Jha NK. Exploring complexities of Alzheimer's disease: New insights into molecular and cellular mechanisms of neurodegeneration and targeted therapeutic interventions. Ageing Res Rev 2024:102548. [PMID: 39419399 DOI: 10.1016/j.arr.2024.102548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
Alzheimer's disease (AD), the common form of dementia globally, is a complex condition including neurodegeneration; shares incompletely known pathogenesis. Signal transduction and biological activities, including cell metabolism, growth, and death are regulated by different signaling pathways including AKT/MAPK, Wnt, Leptin, mTOR, ubiquitin, Sirt1, and insulin. Absolute evidence linking specific molecular pathways with the genesis and/or progression of AD is still lacking. Changes in gut microbiota and blood-brain barrier also cause amyloid β aggregation in AD. The current review reports significant characteristics of various signaling pathways, their relationship with each other, and how they interact in disease genesis and/or progression. Nevertheless, due to the enormous complexity of the brain and numerous chemical linkages between these pathways, the use of signaling pathways as possible targets for drug development against AD is minimal. Currently, there is no permanent cure for AD, and there is no way to stop brain cell loss. This review also aimed to draw attention to the role of a novel group of signaling pathways, which can be collectively dubbed "anti-AD pathways", in multi-target therapy for AD, where cellular metabolic functions are severely impaired. Thus, different hypotheses have been formulated and elaborated to explain the genesis of AD, which can be further explored for drug development too.
Collapse
Affiliation(s)
- Payal Chauhan
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Karan Wadhwa
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India.
| | - Saurabh Gupta
- Deparment of Biotechnology, GLA University, Mathura, India
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, 61922, Saudi Arabia
| | - Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi P.O. Box 59911, United Arab Emirates
| | - Prabhu Chandra Mishra
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Vinod Kumar Nelson
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India
| | - Niraj Kumar Jha
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Centre of Research Impact and Outreach, Chitkara University Institute of Engineering and Technology, Chitkara University, Punjab, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India.
| |
Collapse
|
2
|
Moawad MHED, Serag I, Alkhawaldeh IM, Abbas A, Sharaf A, Alsalah S, Sadeq MA, Shalaby MMM, Hefnawy MT, Abouzid M, Meshref M. Exploring the Mechanisms and Therapeutic Approaches of Mitochondrial Dysfunction in Alzheimer's Disease: An Educational Literature Review. Mol Neurobiol 2024:10.1007/s12035-024-04468-y. [PMID: 39254911 DOI: 10.1007/s12035-024-04468-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024]
Abstract
Alzheimer's disease (AD) presents a significant challenge to global health. It is characterized by progressive cognitive deterioration and increased rates of morbidity and mortality among older adults. Among the various pathophysiologies of AD, mitochondrial dysfunction, encompassing conditions such as increased reactive oxygen production, dysregulated calcium homeostasis, and impaired mitochondrial dynamics, plays a pivotal role. This review comprehensively investigates the mechanisms of mitochondrial dysfunction in AD, focusing on aspects such as glucose metabolism impairment, mitochondrial bioenergetics, calcium signaling, protein tau and amyloid-beta-associated synapse dysfunction, mitophagy, aging, inflammation, mitochondrial DNA, mitochondria-localized microRNAs, genetics, hormones, and the electron transport chain and Krebs cycle. While lecanemab is the only FDA-approved medication to treat AD, we explore various therapeutic modalities for mitigating mitochondrial dysfunction in AD, including antioxidant drugs, antidiabetic agents, acetylcholinesterase inhibitors (FDA-approved to manage symptoms), nutritional supplements, natural products, phenylpropanoids, vaccines, exercise, and other potential treatments.
Collapse
Affiliation(s)
- Mostafa Hossam El Din Moawad
- Faculty of Pharmacy, Clinical Department, Alexandria Main University Hospital, Alexandria, Egypt
- Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Ibrahim Serag
- Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | | | - Abdallah Abbas
- Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| | - Abdulrahman Sharaf
- Department of Clinical Pharmacy, Salmaniya Medical Complex, Government Hospital, Manama, Bahrain
| | - Sumaya Alsalah
- Ministry of Health, Primary Care, Governmental Health Centers, Manama, Bahrain
| | | | | | | | - Mohamed Abouzid
- Department of Physical Pharmacy and Pharmacokinetics, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3 St., 60-806, Poznan, Poland.
- Doctoral School, Poznan University of Medical Sciences, 60-812, Poznan, Poland.
| | - Mostafa Meshref
- Department of Neurology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
3
|
Targa Dias Anastacio H, Matosin N, Ooi L. Familial Alzheimer's Disease Neurons Bearing Mutations in PSEN1 Display Increased Calcium Responses to AMPA as an Early Calcium Dysregulation Phenotype. Life (Basel) 2024; 14:625. [PMID: 38792645 PMCID: PMC11123496 DOI: 10.3390/life14050625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/18/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Familial Alzheimer's disease (FAD) can be caused by mutations in PSEN1 that encode presenilin-1, a component of the gamma-secretase complex that cleaves amyloid precursor protein. Alterations in calcium (Ca2+) homeostasis and glutamate signaling are implicated in the pathogenesis of FAD; however, it has been difficult to assess in humans whether or not these phenotypes are the result of amyloid or tau pathology. This study aimed to assess the early calcium and glutamate phenotypes of FAD by measuring the Ca2+ response of induced pluripotent stem cell (iPSC)-derived neurons bearing PSEN1 mutations to glutamate and the ionotropic glutamate receptor agonists NMDA, AMPA, and kainate compared to isogenic control and healthy lines. The data show that in early neurons, even in the absence of amyloid and tau phenotypes, FAD neurons exhibit increased Ca2+ responses to glutamate and AMPA, but not NMDA or kainate. Together, this suggests that PSEN1 mutations alter Ca2+ and glutamate signaling as an early phenotype of FAD.
Collapse
Affiliation(s)
- Helena Targa Dias Anastacio
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia;
| | - Natalie Matosin
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia;
| | - Lezanne Ooi
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia;
| |
Collapse
|
4
|
Xicota L, Cosentino S, Vardarajan B, Mayeux R, Perls TT, Andersen SL, Zmuda JM, Thyagarajan B, Yashin A, Wojczynski MK, Krinsky‐McHale S, Handen BL, Christian BT, Head E, Mapstone ME, Schupf N, Lee JH, Barral S. Whole genome-wide sequence analysis of long-lived families (Long-Life Family Study) identifies MTUS2 gene associated with late-onset Alzheimer's disease. Alzheimers Dement 2024; 20:2670-2679. [PMID: 38380866 PMCID: PMC11032545 DOI: 10.1002/alz.13718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/17/2023] [Accepted: 01/04/2024] [Indexed: 02/22/2024]
Abstract
INTRODUCTION Late-onset Alzheimer's disease (LOAD) has a strong genetic component. Participants in Long-Life Family Study (LLFS) exhibit delayed onset of dementia, offering a unique opportunity to investigate LOAD genetics. METHODS We conducted a whole genome sequence analysis of 3475 LLFS members. Genetic associations were examined in six independent studies (N = 14,260) with a wide range of LOAD risk. Association analysis in a sub-sample of the LLFS cohort (N = 1739) evaluated the association of LOAD variants with beta amyloid (Aβ) levels. RESULTS We identified several single nucleotide polymorphisms (SNPs) in tight linkage disequilibrium within the MTUS2 gene associated with LOAD (rs73154407, p = 7.6 × 10-9). Association of MTUS2 variants with LOAD was observed in the five independent studies and was significantly stronger within high levels of Aβ42/40 ratio compared to lower amyloid. DISCUSSION MTUS2 encodes a microtubule associated protein implicated in the development and function of the nervous system, making it a plausible candidate to investigate LOAD biology. HIGHLIGHTS Long-Life Family Study (LLFS) families may harbor late onset Alzheimer's dementia (LOAD) variants. LLFS whole genome sequence analysis identified MTUS2 gene variants associated with LOAD. The observed LLFS variants generalized to cohorts with wide range of LOAD risk. The association of MTUS2 with LOAD was stronger within high levels of beta amyloid. Our results provide evidence for MTUS2 gene as a novel LOAD candidate locus.
Collapse
Affiliation(s)
- Laura Xicota
- Department of NeurologyColumbia University Irving Medical CenterNew York CityNew YorkUSA
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical CenterNew York CityNew YorkUSA
| | - Stephanie Cosentino
- Department of NeurologyColumbia University Irving Medical CenterNew York CityNew YorkUSA
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical CenterNew York CityNew YorkUSA
| | - Badri Vardarajan
- Department of NeurologyColumbia University Irving Medical CenterNew York CityNew YorkUSA
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical CenterNew York CityNew YorkUSA
- Gertrude H. Sergievsky CenterColumbia University Irving Medical CenterNew York CityNew YorkUSA
| | - Richard Mayeux
- Department of NeurologyColumbia University Irving Medical CenterNew York CityNew YorkUSA
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical CenterNew York CityNew YorkUSA
- Gertrude H. Sergievsky CenterColumbia University Irving Medical CenterNew York CityNew YorkUSA
| | - Thomas T. Perls
- Section of GeriatricsDepartment of MedicineBoston University School of MedicineBostonMassachusettsUSA
| | - Stacy L. Andersen
- Section of GeriatricsDepartment of MedicineBoston University School of MedicineBostonMassachusettsUSA
| | - Joseph M. Zmuda
- Department of EpidemiologyGraduate School of Public Health, University of PittsburghPittsburghPennsylvaniaUSA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and PathologyUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Anatoli Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke UniversityDurhamNorth CarolinaUSA
| | - Mary K. Wojczynski
- Division of Statistical GenomicsDepartment of GeneticsWashington University School of MedicineSt. LouisMissouriUSA
| | - Sharon Krinsky‐McHale
- Gertrude H. Sergievsky CenterColumbia University Irving Medical CenterNew York CityNew YorkUSA
- Department of PsychologyNew York Institute for Basic Research in Developmental DisabilitiesStaten IslandNew YorkUSA
| | - Benjamin L. Handen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Bradley T. Christian
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin‐Madison School of Medicine and Public HealthMadisonWisconsinUSA
- Department of Medical PhysicsUniversity of Wisconsin‐Madison School of Medicine, and Public HealthMadisonWisconsinUSA
| | - Elizabeth Head
- Department of Pathology and Laboratory MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Mark E. Mapstone
- Department of NeurologyInstitute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| | - Nicole Schupf
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical CenterNew York CityNew YorkUSA
| | - Joseph H. Lee
- Department of NeurologyColumbia University Irving Medical CenterNew York CityNew YorkUSA
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical CenterNew York CityNew YorkUSA
- Gertrude H. Sergievsky CenterColumbia University Irving Medical CenterNew York CityNew YorkUSA
| | - Sandra Barral
- Department of NeurologyColumbia University Irving Medical CenterNew York CityNew YorkUSA
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical CenterNew York CityNew YorkUSA
- Gertrude H. Sergievsky CenterColumbia University Irving Medical CenterNew York CityNew YorkUSA
| | | |
Collapse
|
5
|
Hewawasam C, Wickramasinghe A, Caldera MC, Dassanayake TL. Subclinical memory impairment in unaffected siblings of patients with dementia. Clin Neuropsychol 2023; 37:1669-1685. [PMID: 36866972 DOI: 10.1080/13854046.2023.2182832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 02/15/2023] [Indexed: 03/04/2023]
Abstract
Objective: Family history of dementia is a known risk factor for dementia. The cognitive performance of unaffected siblings of dementia patients has been poorly studied. We aimed to determine whether clinically unaffected siblings of dementia patients have significant cognitive impairment compared to individuals who do not have first-degree relatives with dementia. Methods: We compared the cognitive performance of 67 patients with dementia (24 males; mean age 69.5), 90 healthy siblings of those patients (34 males; mean age 61.56) and 92 healthy adults (35 males; mean age 60.96) who have no first-degree relatives with dementia. We assessed learning and memory (Rey Auditory Verbal Learning Test (RAVLT)), short-term/working memory (Digit Span) executive functions (Stroop Test) and general intelligence (Raven Progressive Matrices). Test scores were compared among three groups, with regression-based adjustments for age, sex, and education. Results: As expected, the patients with dementia were impaired in all cognitive domains. In the Sibling Group, RAVLT total learning was significantly lower compared to controls (B = -3.192, p = .005). In a subgroup analysis, compared to controls, RAVLT delayed recall was poorer in the siblings of patients with early-onset (<65 years) dementia. No significant differences were observed in other cognitive domains. Conclusion: Clinically unaffected siblings of dementia patients seem to have a selective subclinical impairment in memory encoding. This impairment seems to be more prominent in siblings of patients with early-onset dementia who also have deficits in delayed recall. Future studies are needed to determine if the observed cognitive impairment deteriorates to dementia.
Collapse
Affiliation(s)
- Chandana Hewawasam
- Department of Physiology, Faculty of Medicine and Allied Sciences, Rajarata University of Sri Lanka, Saliyapura, Sri Lanka
| | - Anuprabha Wickramasinghe
- Department of Psychiatry, Faculty of Medicine and Allied Sciences, Rajarata University of Sri Lanka, Saliyapura, Sri Lanka
- Department of Psychiatry, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Manjula C Caldera
- Neurology Unit, Teaching Hospital Anuradhapura, Anuradhapura, Sri Lanka
| | - Tharaka L Dassanayake
- Department of Physiology, Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
- School of Psychological Sciences, The University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
6
|
Leisher S, Bohorquez A, Gay M, Garcia V, Jones R, Baldaranov D, Rafii MS. Amyloid-Lowering Monoclonal Antibodies for the Treatment of Early Alzheimer's Disease. CNS Drugs 2023; 37:671-677. [PMID: 37470978 PMCID: PMC10439019 DOI: 10.1007/s40263-023-01021-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 07/21/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia worldwide. Numerous biomarker studies have clearly demonstrated that AD has a long asymptomatic phase, with the development of pathology occurring at least 2 decades prior to the development of any symptoms. These pathological changes include a stepwise development of amyloid-β (Aβ) plaques, followed by tau neurofibrillary tangles and subsequently extensive neurodegeneration in the brain. In this review, we discuss the first class of drugs intended to be disease modifying to be approved by the US Food and Drug Administration (FDA) for AD-anti-Aβ monoclonal antibodies-and the scientific rationale with which they were developed.
Collapse
Affiliation(s)
- Solana Leisher
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of University of Southern California, San Diego, CA, 92130, USA
| | - Adriana Bohorquez
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of University of Southern California, San Diego, CA, 92130, USA
| | - Marcus Gay
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of University of Southern California, San Diego, CA, 92130, USA
| | - Victoria Garcia
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of University of Southern California, San Diego, CA, 92130, USA
| | - Renarda Jones
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of University of Southern California, San Diego, CA, 92130, USA
| | - Dobri Baldaranov
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of University of Southern California, San Diego, CA, 92130, USA
| | - Michael S Rafii
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of University of Southern California, San Diego, CA, 92130, USA.
| |
Collapse
|
7
|
Sundarrajan S, Venkatesan A, Kumar S U, Gopikrishnan M, Tayubi IA, Aditya M, Siddaiah GB, George Priya Doss C, Zayed H. Exome sequence analysis of rare frequency variants in Late-Onset Alzheimer Disease. Metab Brain Dis 2023; 38:2025-2036. [PMID: 37162726 PMCID: PMC10348954 DOI: 10.1007/s11011-023-01221-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/19/2023] [Indexed: 05/11/2023]
Abstract
Alzheimer disease (AD) is a leading cause of dementia in elderly patients who continue to live between 3 and 11 years of diagnosis. A steep rise in AD incidents is observed in the elderly population in East-Asian countries. The disease progresses through several changes, including memory loss, behavioural issues, and cognitive impairment. The etiology of AD is hard to determine because of its complex nature. The whole exome sequences of late-onset AD (LOAD) patients of Korean origin are investigated to identify rare genetic variants that may influence the complex disorder. Computational annotation was performed to assess the function of candidate variants in LOAD. The in silico pathogenicity prediction tools such as SIFT, Polyphen-2, Mutation Taster, CADD, LRT, PROVEAN, DANN, VEST3, fathmm-MKL, GERP + + , SiPhy, phastCons, and phyloP identified around 17 genes harbouring deleterious variants. The variants in the ALDH3A2 and RAD54B genes were pathogenic, while in 15 other genes were predicted to be variants of unknown significance. These variants can be potential risk candidates contributing to AD. In silico computational techniques such as molecular docking, molecular dynamic simulation and steered molecular dynamics were carried out to understand the structural insights of RAD54B with ATP. The simulation of mutant (T459N) RAD54B with ATP revealed reduced binding strength of ATP at its binding site. In addition, lower binding free energy was observed when compared to the wild-type RAD54B. Our study shows that the identified uncommon variants are linked to AD and could be probable predisposing genetic factors of LOAD.
Collapse
Affiliation(s)
| | - Arthi Venkatesan
- BIOVIA Specialist, VIAS 3D, MG Road, Bengaluru, 560001, Karnataka, India
| | - Udhaya Kumar S
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Mohanraj Gopikrishnan
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Iftikhar Aslam Tayubi
- Department of Computer Science, Faculty of Computing and Information Technology, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - M Aditya
- Department of Biotechnology, Siddaganga Institute of Technology, Tumkur, Karnataka, 572103, India
| | | | - C George Priya Doss
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| | - Hatem Zayed
- Department of Biomedical Sciences College of Health Sciences, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
8
|
Targa Dias Anastacio H, Matosin N, Ooi L. Neuronal hyperexcitability in Alzheimer's disease: what are the drivers behind this aberrant phenotype? Transl Psychiatry 2022; 12:257. [PMID: 35732622 PMCID: PMC9217953 DOI: 10.1038/s41398-022-02024-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/01/2022] [Accepted: 06/08/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder leading to loss of cognitive abilities and ultimately, death. With no cure available, limited treatments mostly focus on symptom management. Identifying early changes in the disease course may provide new therapeutic targets to halt or reverse disease progression. Clinical studies have shown that cortical and hippocampal hyperactivity are a feature shared by patients in the early stages of disease, progressing to hypoactivity during later stages of neurodegeneration. The exact mechanisms causing neuronal excitability changes are not fully characterized; however, animal and cell models have provided insights into some of the factors involved in this phenotype. In this review, we summarize the evidence for neuronal excitability changes over the course of AD onset and progression and the molecular mechanisms underpinning these differences. Specifically, we discuss contributors to aberrant neuronal excitability, including abnormal levels of intracellular Ca2+ and glutamate, pathological amyloid β (Aβ) and tau, genetic risk factors, including APOE, and impaired inhibitory interneuron and glial function. In light of recent research indicating hyperexcitability could be a predictive marker of cognitive dysfunction, we further argue that the hyperexcitability phenotype could be leveraged to improve the diagnosis and treatment of AD, and present potential targets for future AD treatment development.
Collapse
Affiliation(s)
- Helena Targa Dias Anastacio
- grid.510958.0Illawarra Health and Medical Research Institute, Wollongong, NSW 2522 Australia ,grid.1007.60000 0004 0486 528XMolecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522 Australia
| | - Natalie Matosin
- grid.510958.0Illawarra Health and Medical Research Institute, Wollongong, NSW 2522 Australia ,grid.1007.60000 0004 0486 528XMolecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522 Australia
| | - Lezanne Ooi
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia. .,Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
9
|
de Godoy LL, Alves CAPF, Saavedra JSM, Studart-Neto A, Nitrini R, da Costa Leite C, Bisdas S. Understanding brain resilience in superagers: a systematic review. Neuroradiology 2020; 63:663-683. [PMID: 32995945 DOI: 10.1007/s00234-020-02562-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/16/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE Superagers are older adults presenting excellent memory performance that may reflect resilience to the conventional pathways of aging. Our contribution aims to shape the evidence body of the known distinctive biomarkers of superagers and their connections with the Brain and Cognitive Reserve and Brain Maintenance concepts. METHODS We performed a systematic literature search in PubMed and ScienceDirect with no limit on publication date for studies that evaluated potential biomarkers in superagers classified by validated neuropsychological tests. Methodological quality was assessed using the QUADAS-2 tool. RESULTS Twenty-one studies were included, the majority in neuroimaging, followed by histological, genetic, cognition, and a single one on blood plasma analysis. Superagers exhibited specific regions of cortical preservation, rather than global cortical maintenance, standing out the anterior cingulate and hippocampus regions. Both superagers and controls showed similar levels of amyloid deposition. Moreover, the functional oscillation patterns in superagers resembled those described in young adults. Most of the quality assessment for the included studies showed medium risks of bias. CONCLUSION This systematic review supports selective cortical preservation in superagers, comprehending regions of the default mode, and salience networks, overlapped by stronger functional connectivity. In this context, the anterior cingulate cortex is highlighted as an imaging and histologic signature of these subjects. Besides, the biomarkers included pointed out that the Brain and Cognitive Reserve and Brain Maintenance concepts are independent and complementary in the superagers' setting.
Collapse
Affiliation(s)
- Laiz Laura de Godoy
- The National Hospital of Neurology and Neurosurgery, University College London, London, UK. .,Department of Radiology and Oncology, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, Brazil.
| | | | | | - Adalberto Studart-Neto
- Department of Radiology and Oncology, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, Brazil
| | - Ricardo Nitrini
- Department of Radiology and Oncology, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, Brazil
| | - Claudia da Costa Leite
- Department of Radiology and Oncology, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, Brazil
| | - Sotirios Bisdas
- The National Hospital of Neurology and Neurosurgery, University College London, London, UK
| |
Collapse
|
10
|
Li Y, Li D, Zhao P, Nandakumar K, Wang L, Song Y. Microfluidics-Based Systems in Diagnosis of Alzheimer's Disease and Biomimetic Modeling. MICROMACHINES 2020; 11:mi11090787. [PMID: 32825153 PMCID: PMC7569794 DOI: 10.3390/mi11090787] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/13/2020] [Accepted: 08/13/2020] [Indexed: 12/21/2022]
Abstract
Early detection and accurate diagnosis of Alzheimer’s disease (AD) is essential for patient care and disease treatment. Microfluidic technology is emerging as an economical and versatile platform in disease detection and diagnosis. It can be conveniently integrated with nanotechnology and/or biological models for biomedical functional and pre-clinical treatment study. These strengths make it advantageous in disease biomarker detection and functional analysis against a wide range of biological backgrounds. This review highlights the recent developments and trends of microfluidic applications in AD research. The first part looks at the principles and methods for AD diagnostic biomarker detection and profiling. The second part discusses how microfluidic chips, especially organ-on-a-chip platforms, could be used as an independent approach and/or integrated with other technologies in AD biomimetic functional analysis.
Collapse
Affiliation(s)
- Yan Li
- Energy Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; (Y.L.); (P.Z.); (K.N.)
- School of Energy and Power Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Danni Li
- Department of Neurology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250013, China;
| | - Pei Zhao
- Energy Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; (Y.L.); (P.Z.); (K.N.)
- School of Energy and Power Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Krishnaswamy Nandakumar
- Energy Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; (Y.L.); (P.Z.); (K.N.)
- Cain Department of Chemical Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Liqiu Wang
- Department of Mechanical Engineering, Faculty of Engineering, The University of Hong Kong, Hong Kong, China
- Correspondence: (L.W.); (Y.S.)
| | - Youqiang Song
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
- State Key Laboratory for Cognitive and Brain Sciences, The University of Hong Kong, Hong Kong, China
- Correspondence: (L.W.); (Y.S.)
| |
Collapse
|
11
|
Swerdlow RH. The mitochondrial hypothesis: Dysfunction, bioenergetic defects, and the metabolic link to Alzheimer's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:207-233. [PMID: 32739005 PMCID: PMC8493961 DOI: 10.1016/bs.irn.2020.01.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) features mitochondrial dysfunction and altered metabolism. Other pathologies could drive these changes, or alternatively these changes could drive other pathologies. In considering this question, it is worth noting that perturbed AD patient mitochondrial and metabolism dysfunction extend beyond the brain and to some extent define a systemic phenotype. It is difficult to attribute this systemic phenotype to brain beta-amyloid or tau proteins. Conversely, mitochondria increasingly appear to play a critical role in cell proteostasis, which suggests that mitochondrial dysfunction may promote protein aggregation. Mitochondrial and metabolism-related characteristics also define AD endophenotypes in cognitively normal middle-aged individuals, which suggests that mitochondrial and metabolism-related AD characteristics precede clinical decline. Genetic analyses increasingly implicate mitochondria and metabolism-relevant genes in AD risk. Collectively these factors suggest that mitochondria are more relevant to the causes of AD than its consequences, and support the view that a mitochondrial cascade features prominently in AD. This chapter reviews the case for mitochondrial and metabolism dysfunction in AD and the challenges of proving that a primary mitochondrial cascade is pertinent to the disease.
Collapse
Affiliation(s)
- Russell H Swerdlow
- University of Kansas Alzheimer's Disease Center, University of Kansas Medical Center, Kansas City, KS, United States.
| |
Collapse
|
12
|
Weigand AJ, Bangen KJ, Thomas KR, Delano-Wood L, Gilbert PE, Brickman AM, Bondi MW. Is tau in the absence of amyloid on the Alzheimer's continuum?: A study of discordant PET positivity. Brain Commun 2019; 2:fcz046. [PMID: 32051933 PMCID: PMC7001143 DOI: 10.1093/braincomms/fcz046] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/15/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
The amyloid cascade model of Alzheimer’s disease posits the primacy of amyloid beta deposition preceding tau-mediated neurofibrillary tangle formation. The amyloid-tau-neurodegeneration biomarker-only diagnostic framework similarly requires the presence of amyloid beta for a diagnosis on the Alzheimer’s continuum. However, medial temporal lobe tau pathology in the absence of amyloid beta is frequently observed at autopsy in cognitively normal individuals, a phenomenon that may reflect a consequence of aging and has been labelled ‘primary age-related tauopathy’. Alternatively, others argue that this tauopathy reflects an early stage of the developmental continuum leading to Alzheimer’s disease. We used positron emission tomography imaging to investigate amyloid beta and tau positivity and associations with cognition to better inform the conceptualization of biomarker changes in Alzheimer’s pathogenesis. Five hundred twenty-three individuals from the Alzheimer’s Disease Neuroimaging Initiative who had undergone flortaucipir positron emission tomography imaging were selected to derive positron emission tomography positivity thresholds using conditional inference decision tree regression. A subsample of 301 individuals without dementia (i.e. those with normal cognition or mild cognitive impairment) had also undergone florbetapir positron emission tomography imaging within 12 months and were categorized into one of the four groups based on cortical amyloid and Braak stage I/II tau positivity: A−/T−, A+/T−, A−/T+, or A+/T+. Tau positivity in the absence of amyloid beta positivity (i.e. A−/T+) comprised the largest group, representing 45% of the sample. In contrast, only 6% of the sample was identified as A+/T−, and the remainder of the sample fell into A−/T− (22%) or A+/T+ (27%) categories. A−/T− and A+/T− groups had the best cognitive performances across memory, language and executive function; the A−/T+ group showed small-to-moderate relative decreases in cognition; and the A+/T+ group had the worst cognitive performances. Furthermore, there were negative associations between Braak stage I/II tau values and all cognitive domains only in the A−/T+ and A+/T+ groups, with strongest associations for the A+/T+ group. Among our sample of older adults across the Alzheimer’s pathological spectrum, 7-fold fewer individuals have positron emission tomography evidence of amyloid beta pathology in the absence of tau pathology than the converse, challenging prevailing models of amyloid beta’s primacy in Alzheimer’s pathogenesis. Given that cognitive performance in the A−/T+ group was poorer than in individuals without either pathology, our results suggest that medial temporal lobe tau without cortical amyloid beta may reflect an early stage on the Alzheimer’s pathological continuum.
Collapse
Affiliation(s)
- Alexandra J Weigand
- San Diego State University/University of California San Diego Joint Doctoral Program, San Diego, CA 92182, USA
| | - Katherine J Bangen
- VA San Diego Healthcare System, San Diego, CA 92161, USA.,Department of Psychiatry, University of California, San Diego, CA 92161, USA
| | - Kelsey R Thomas
- VA San Diego Healthcare System, San Diego, CA 92161, USA.,Department of Psychiatry, University of California, San Diego, CA 92161, USA
| | - Lisa Delano-Wood
- VA San Diego Healthcare System, San Diego, CA 92161, USA.,Department of Psychiatry, University of California, San Diego, CA 92161, USA
| | - Paul E Gilbert
- Department of Psychology, San Diego State University, San Diego, CA 92182, USA
| | - Adam M Brickman
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Mark W Bondi
- VA San Diego Healthcare System, San Diego, CA 92161, USA.,Department of Psychiatry, University of California, San Diego, CA 92161, USA
| | | |
Collapse
|
13
|
Parnell N, Rye K, Greenberg N. Health and well-being management in the military: a systematic review of genetic studies. J ROY ARMY MED CORPS 2017; 164:302-308. [PMID: 28939699 DOI: 10.1136/jramc-2017-000765] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 07/04/2017] [Accepted: 07/04/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Genetic research may have therapeutic value for mental and physical disorders and could have an indicative or preventative capacity. Little is known about the extent, form and utility of military-specific genetic research. METHOD A systematic review was conducted to evaluate existing genetic well-being studies of service personnel. The review specifically aimed to ascertain the current state of knowledge and feasibility of using genetics to aid recruitment and health management within military populations. Databases searched included MEDLINE, Embase, PsycINFO and Web of Science for relevant studies. Papers were rated using a genetics-specific quality assessment framework. RESULTS Ten papers were included within the final review, with seven mental-health-focused and three physical-health-focused genetic studies found within military populations. Eight papers considered candidate genes, one gene expression and one study was an outline of a future study of significant interest. Genetic commonalties were derived to yield shared physiological pathways. The 10 reviewed papers revealed moderate quality based on quality assessment. CONCLUSIONS Current genetic research within military populations is limited. Further studies on genetics, cost effectiveness, ethics and continual monitoring need to be explored before considering any movement toward clinical translation.
Collapse
Affiliation(s)
- Nathan Parnell
- King's Centre for Military Health Research, King's College London, London, UK
| | - K Rye
- King's Centre for Military Health Research, King's College London, London, UK
| | - N Greenberg
- Academic Department for Military Mental Health, King's College London, London, UK
| |
Collapse
|
14
|
Dekhtyar M, Papp KV, Buckley R, Jacobs HIL, Schultz AP, Johnson KA, Sperling RA, Rentz DM. Neuroimaging markers associated with maintenance of optimal memory performance in late-life. Neuropsychologia 2017; 100:164-170. [PMID: 28472627 PMCID: PMC5522601 DOI: 10.1016/j.neuropsychologia.2017.04.037] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 04/27/2017] [Accepted: 04/29/2017] [Indexed: 11/21/2022]
Abstract
BACKGROUND Age-related memory decline has been well-documented; however, some individuals reach their 8th-10th decade while maintaining strong memory performance. OBJECTIVE To determine which demographic and biomarker factors differentiated top memory performers (aged 75+, top 20% for memory) from their peers and whether top memory performance was maintained over 3 years. METHODS Clinically normal adults (n=125, CDR=0; age: 79.5±3.57 years) from the Harvard Aging Brain Study underwent cognitive testing and neuroimaging (amyloid PET, MRI) at baseline and 3-year follow-up. Participants were grouped into Optimal (n=25) vs. Typical (n=100) performers using performance on 3 challenging memory measures. Non-parametric tests were used to compare groups. RESULTS There were no differences in age, sex, or education between Optimal vs. Typical performers. The Optimal group performed better in Processing Speed (p=0.016) and Executive Functioning (p<0.001). Optimal performers had larger hippocampal volumes at baseline compared with Typical Performers (p=0.027) but no differences in amyloid burden (p=0.442). Twenty-three of the 25 Optimal performers had longitudinal data and16 maintained top memory performance while 7 declined. Non-Maintainers additionally declined in Executive Functioning but not Processing Speed. Longitudinally, there were no hippocampal volume differences between Maintainers and Non-Maintainers, however Non-Maintainers exhibited higher amyloid burden at baseline in contrast with Maintainers (p=0.008). CONCLUSIONS Excellent memory performance in late life does not guarantee protection against cognitive decline. Those who maintain an optimal memory into the 8th and 9th decades may have lower levels of AD pathology.
Collapse
Affiliation(s)
- Maria Dekhtyar
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Kathryn V Papp
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States.
| | - Rachel Buckley
- Department of Neurology, Massachusetts General Hospital, Boston, MA, United States; Florey Institutes of Neuroscience and Mental Health, Melbourne, Australia; Melbourne School of Psychological Science, University of Melbourne, Australia
| | - Heidi I L Jacobs
- Department of Neurology, Massachusetts General Hospital, Boston, MA, United States; Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Boston, MA, United States; Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Boston, MA, United States; Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States
| | - Reisa A Sperling
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States; Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States
| | - Dorene M Rentz
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
15
|
Quaid KA, Murrell JR, Hake AM, Farlow MR, Ghetti B. Presymptomatic Genetic Testing with an APP Mutation in Early-Onset Alzheimer Disease: A Descriptive Study of Sibship Dynamics. J Genet Couns 2015; 9:327-41. [PMID: 26141474 DOI: 10.1023/a:1009406229745] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Early-onset Alzheimer disease (AD) accounts for only 5% of all cases of Alzheimer disease. To date, mutations in three different genes, the Amyloid precursor protein (APP), Presenilin 1 (PS1), and Presenilin 2 (PS2), have been identified as causative in early-onset AD, making predictive testing possible. Predictive testing for early-onset Alzheimer disease is a relatively new phenomenon. This paper describes the process of identifying a new mutation in the APP gene associated with early-onset AD, notifying family members, and offering participation in research as well as predictive testing. The goal is to share the complexities of predictive testing in a sibship newly identified as being at risk for an adult-onset, incurable neurodegenerative disease.
Collapse
Affiliation(s)
- K A Quaid
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, 975 West Walnut Street, Indianapolis, IN, 46202-5251
| | | | | | | | | |
Collapse
|
16
|
Neuropathologically mixed Alzheimer's and Lewy body disease: burden of pathological protein aggregates differs between clinical phenotypes. Acta Neuropathol 2015; 129:729-48. [PMID: 25758940 DOI: 10.1007/s00401-015-1406-3] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 03/03/2015] [Accepted: 03/03/2015] [Indexed: 12/31/2022]
Abstract
Multiple different pathological protein aggregates are frequently seen in human postmortem brains and hence mixed pathology is common. Mixed dementia on the other hand is less frequent and neuropathologically should only be diagnosed if criteria for more than one full blown disease are met. We quantitatively measured the amount of hyperphosphorylated microtubule associated tau (HP-τ), amyloid-β protein (Aβ) and α-synuclein (α-syn) in cases that were neuropathologically diagnosed as mixed Alzheimer's disease (AD) and neocortical Lewy body disease (LBD) but clinically presented either as dementia due to AD or LBD, the latter including dementia with Lewy bodies (DLB) and Parkinson's disease dementia (PDD). Our study group consisted of 28 cases (mean age, 76.11 SE: ±1.29 years; m:f, 17:11) of which 19 were neuropathologically diagnosed as mixed AD/DLB. Clinically, 8 mixed AD/DLB cases were diagnosed as AD (cAD), 8 as DLB (cDLB) and 3 as PDD (cPDD). In addition, we investigated cases that were both clinically and neuropathologically diagnosed as either AD (pure AD; n = 5) or DLB/neocortical LBD (pure DLB; n = 4). Sections from neocortical, limbic and subcortical areas were stained with antibodies against HP-τ, Aβ and α-syn. The area covered by immunopositivity was measured using image analysis. cAD cases had higher HP-τ loads than both cDLB and cPDD and the distribution of HP-τ in cAD was similar to the one observed in pure AD whilst cDLB showed comparatively less hippocampal HP-τ load. cPDD cases showed lower HP-τ and Aβ loads and higher α-syn loads. Here, we show that in neuropathologically mixed AD/DLB cases both the amount and the topographical distribution of pathological protein aggregates differed between distinct clinical phenotypes. Large-scale clinicopathological correlative studies using a quantitative methodology are warranted to further elucidate the neuropathological correlate of clinical symptoms in cases with mixed pathology.
Collapse
|
17
|
Cavedo E, Pievani M, Boccardi M, Galluzzi S, Bocchetta M, Bonetti M, Thompson PM, Frisoni GB. Medial temporal atrophy in early and late-onset Alzheimer's disease. Neurobiol Aging 2014; 35:2004-12. [PMID: 24721821 PMCID: PMC4053814 DOI: 10.1016/j.neurobiolaging.2014.03.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 02/14/2014] [Accepted: 03/11/2014] [Indexed: 01/01/2023]
Abstract
Late-onset and early-onset Alzheimer's disease (LOAD, EOAD) affect different neural systems and may be separate nosographic entities. The most striking differences are in the medial temporal lobe, severely affected in LOAD and relatively spared in EOAD. We assessed amygdalar morphology and volume in 18 LOAD and 18 EOAD patients and 36 aged-matched controls and explored their relationship with the hippocampal volume. Three-dimensional amygdalar shape was reconstructed with the radial atrophy mapping technique, hippocampal volume was measured using a manual method. Atrophy was greater in LOAD than EOAD: 25% versus 17% in the amygdala and 20% versus 13% in the hippocampus. In the amygdala, LOAD showed significantly greater tissue loss than EOAD in the right dorsal central, lateral, and basolateral nuclei (20%-30% loss, p < 0.03), all known to be connected to limbic regions. In LOAD but not EOAD, greater hippocampal atrophy was associated with amygdalar atrophy in the left dorsal central and medial nuclei (r = 0.6, p < 0.05) also part of the limbic system. These findings support the notion that limbic involvement is a prominent feature of LOAD but not EOAD.
Collapse
Affiliation(s)
- Enrica Cavedo
- LENITEM Laboratory Q1 of Epidemiology, Neuroimaging, and Telemedicine IRCCS Istituto Centro San Giovanni di Dio-FBF, Brescia, Italy; Cognition, Neuroimaging and Brain Diseases Laboratory, Centre de Recherche de l'Institut du Cerveau et de la Moelle Épiniére (CRICM-UMRS 975), Université Pierre et Marie Curie-Paris 6, France
| | - Michela Pievani
- LENITEM Laboratory Q1 of Epidemiology, Neuroimaging, and Telemedicine IRCCS Istituto Centro San Giovanni di Dio-FBF, Brescia, Italy
| | - Marina Boccardi
- LENITEM Laboratory Q1 of Epidemiology, Neuroimaging, and Telemedicine IRCCS Istituto Centro San Giovanni di Dio-FBF, Brescia, Italy
| | - Samantha Galluzzi
- LENITEM Laboratory Q1 of Epidemiology, Neuroimaging, and Telemedicine IRCCS Istituto Centro San Giovanni di Dio-FBF, Brescia, Italy
| | - Martina Bocchetta
- LENITEM Laboratory Q1 of Epidemiology, Neuroimaging, and Telemedicine IRCCS Istituto Centro San Giovanni di Dio-FBF, Brescia, Italy; Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Matteo Bonetti
- Service of Neuroradiology, Istituto Clinico Citta' di Brescia, Brescia, Italy
| | - Paul M Thompson
- Laboratory of NeuroImaging (LoNI), University of Southern California, Los Angeles, CA, USA; Department of Psychiatry & Biobehavioral Sciences, Semel Institute, UCLA School of Medicine, Los Angeles, CA, USA
| | - Giovanni B Frisoni
- LENITEM Laboratory Q1 of Epidemiology, Neuroimaging, and Telemedicine IRCCS Istituto Centro San Giovanni di Dio-FBF, Brescia, Italy; Memory Clinic and LANVIE - Laboratory of Neuroimaging of Aging, University Hospitals and University of Geneva, Geneva, Switzerland.
| |
Collapse
|
18
|
Binnewijzend MAA, Adriaanse SM, Van der Flier WM, Teunissen CE, de Munck JC, Stam CJ, Scheltens P, van Berckel BNM, Barkhof F, Wink AM. Brain network alterations in Alzheimer's disease measured by eigenvector centrality in fMRI are related to cognition and CSF biomarkers. Hum Brain Mapp 2013; 35:2383-93. [PMID: 24039033 DOI: 10.1002/hbm.22335] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 04/18/2013] [Accepted: 05/09/2013] [Indexed: 01/23/2023] Open
Abstract
Recent imaging studies have demonstrated functional brain network changes in patients with Alzheimer's disease (AD). Eigenvector centrality (EC) is a graph analytical measure that identifies prominent regions in the brain network hierarchy and detects localized differences between patient populations. This study used voxel-wise EC mapping (ECM) to analyze individual whole-brain resting-state functional magnetic resonance imaging (MRI) scans in 39 AD patients (age 67 ± 8) and 43 healthy controls (age 69 ± 7). Between-group differences were assessed by a permutation-based method. Associations of EC with biomarkers for AD pathology in cerebrospinal fluid (CSF) and Mini Mental State Examination (MMSE) scores were assessed using Spearman correlation analysis. Decreased EC was found bilaterally in the occipital cortex in AD patients compared to controls. Regions of increased EC were identified in the anterior cingulate and paracingulate gyrus. Across groups, frontal and occipital EC changes were associated with pathological concentrations of CSF biomarkers and with cognition. In controls, decreased EC values in the occipital regions were related to lower MMSE scores. Our main finding is that ECM, a hypothesis-free and computationally efficient analysis method of functional MRI (fMRI) data, identifies changes in brain network organization in AD patients that are related to cognition and underlying AD pathology. The relation between AD-like EC changes and cognitive performance suggests that resting-state fMRI measured EC is a potential marker of disease severity for AD.
Collapse
Affiliation(s)
- Maja A A Binnewijzend
- Department of Radiology and Nuclear Medicine, VU University Medical Center and Neuroscience Campus Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Frontotemporal dementia, manifested as schizophrenia, with decreased heterochromatin on chromosome 1. Case Rep Psychiatry 2012; 2012:937518. [PMID: 23082270 PMCID: PMC3467798 DOI: 10.1155/2012/937518] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 09/04/2012] [Indexed: 01/03/2023] Open
Abstract
Introduction. Frontotemporal dementia is a disorder of complex etiology, with genetic components contributing to the disease. The aim of this report is to describe a young patient suffering from frontotemporal dementia, misdiagnosed as schizophrenia, related to a genetic defect on chromosome 1. Case Presentation. A 29-year-old female patient, previously diagnosed as having schizophrenia, was hospitalized with severe behavioural disturbances. She demonstrated severe sexual disinhibition, hyperphagia, lack of motivation, apathy, psychotic symptoms, suicidal thoughts, and cognitive deterioration. Focal atrophy of frontal and anterior temporal structures bilaterally was found on brain MRI, as well as bifrontal hypo perfusion of the brain on SPECT scan. The diagnosis of frontotemporal dementia was made clinically, according to Lund and Manchester groups and Neary diagnostic criteria. Chromosomal analysis was conducted and revealed decrease in length of heterochromatin on the long arm of chromosome 1 (46, XX, 1qh-). Parental karyotypes were normal. Discussion. Frontotemporal dementia, and particularly early-onset cases, can be often misdiagnosed as schizophrenia, with negative impact on case management. Genetic testing could be an aid to the correct diagnosis, which is crucial for optimal patient care.
Collapse
|
20
|
Mosconi L, de Leon M, Murray J, E L, Lu J, Javier E, McHugh P, Swerdlow RH. Reduced mitochondria cytochrome oxidase activity in adult children of mothers with Alzheimer's disease. J Alzheimers Dis 2012; 27:483-90. [PMID: 21841246 DOI: 10.3233/jad-2011-110866] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Biomarker studies demonstrate inheritance of glucose hypometabolism and increased amyloid-β deposition in adult offspring of mothers, but not fathers, affected by late-onset Alzheimer's disease (LOAD). The underlying genetic mechanisms are unknown. We investigated whether cognitively normal (NL) individuals with a maternal history of LOAD (MH) have reduced platelet mitochondrial cytochrome oxidase activity (COX, electron transport chain complex IV) compared to those with paternal (PH) or negative family history (NH). Thirty-six consecutive NL individuals (age 55 ± 15 y, range 27-71 y, 56% female, CDR = 0, MMSE ≥28, 28% APOE-4 carriers), including 12 NH, 12 PH, and 12 MH, received a blood draw to measure platelet mitochondrial COX activity. Citrate synthase activity (CS) was measured as a reference. Groups were comparable for clinical and neuropsychological measures. We found that after correcting for CS, COX activity was reduced by 29% in MH compared to NH, and by 30% in MH compared to PH (p ≤ 0.006). Results remained significant controlling for age, gender, education, and APOE. No differences were found between PH and NH. COX measures discriminated MH from the other groups with accuracy ≥75%, and relative risk ≥3 (p ≤ 0.005). Among NL with LOAD-parents, only those with MH showed reduced COX activity in platelet mitochondria compared to PH and NH. The association between maternal history of LOAD and systemic COX reductions suggests transmission via mitochondrial DNA, which is exclusively maternally inherited in humans.
Collapse
Affiliation(s)
- Lisa Mosconi
- Center for Brain Health, Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Alzheimer's disease and vascular deficiency: lessons from imaging studies and down syndrome. Curr Gerontol Geriatr Res 2012; 2012:929734. [PMID: 22400025 PMCID: PMC3286884 DOI: 10.1155/2012/929734] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 11/30/2011] [Indexed: 11/30/2022] Open
Abstract
Down syndrome (DS) individuals are at high risk for developing Alzheimer's disease (AD) and consequently provide a unique opportunity to examine the factors leading to the onset of AD. This paper focuses on the neglected vascular parallels between AD and DS that can readily be examined in DS. Several recent AD studies provide evidence that internal jugular vein (IJV) reflux may result in white matter lesions and a 30% decrease in cerebrospinal fluid (CSF) clearance of amyloid-β. At the same time, studies analyzing the synthesis of amyloid-β in DS showed greater than expected amounts of Aβ than would be predicted by the increase in gene dosage, perhaps due to slower clearance. These studies are discussed along with the possibility that the venous and CSF dysfunction found in AD patients may be present early in life in persons with DS, leaving them particularly vulnerable to early onset AD. Studying IJV function in DS provides an opportunity to understand the role of vascular function in the initiation of AD.
Collapse
|
22
|
Corbo CP, Alonso ADC. Therapeutic targets in Alzheimer's disease and related tauopathies. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 98:47-83. [PMID: 21199770 DOI: 10.1016/b978-0-12-385506-0.00002-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease is a progressive neurodegenerative disease that is characterized histopathologically by the presence of plaques, mainly composed of Abeta amyloid and the tangles, mainly composed of hyperphosphorylated tau. To date, there is no treatment that can reverse the disease, and all the current therapeutics is directed to cope with the symptoms of the disease. Here we describe the efforts dedicated to attack the plaques and, in more detail, the process of neurofibrillary degeneration, linked to the presence of the hyperphosphorylated microtubule associated protein tau. We have identified the different putative targets for therapeutics and the current knowledge on them.
Collapse
Affiliation(s)
- Christopher P Corbo
- College of Staten Island, Program in evelopmental Neuroscience, The Graduate Center, City University of New York (CUNY), Staten Island, New York, USA
| | | |
Collapse
|
23
|
Devadhasan JP, Kim S, An J. Fish-on-a-chip: a sensitive detection microfluidic system for Alzheimer's disease. J Biomed Sci 2011; 18:33. [PMID: 21619660 PMCID: PMC3125339 DOI: 10.1186/1423-0127-18-33] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 05/28/2011] [Indexed: 01/09/2023] Open
Abstract
Microfluidics has become an important tool in diagnosing many diseases, including neurological and genetic disorders. Alzheimer's disease (AD) is a neurodegenerative disease that irreversibly and progressively destroys memory, language ability, and thinking skills. Commonly, detection of AD is expensive and complex. Fluorescence in situ hybridization (FISH)-based microfluidic chip platform is capable of diagnosing AD at an early stage and they are effective tools for the diagnosis with low cost, high speed, and high sensitivity. In this review, we tried to provide basic information on the diagnosis of AD via FISH-based microfluidics. Different sample preparations using a microfluidic chip for diagnosis of AD are highlighted. Moreover, rapid innovations in nanotechnology for diagnosis are explained. This review will provide information on dynamic quantification methods for the diagnosis and treatment of AD. The knowledge provided in this review will help develop new integration diagnostic techniques based on FISH and microfluidics.
Collapse
Affiliation(s)
- Jasmine P Devadhasan
- College of Bionanotechnology, Kyungwon University, San 65, Bokjeong-Dong, Sujeong-Gu, Seongnam-Si, Gyeonggi-Do 461-701, Republic of Korea
| | | | | |
Collapse
|
24
|
BACE1 gene promoter single-nucleotide polymorphisms in Alzheimer's disease. J Mol Neurosci 2010; 42:127-33. [PMID: 20455082 DOI: 10.1007/s12031-010-9381-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Accepted: 04/20/2010] [Indexed: 10/19/2022]
Abstract
Alzheimer's disease (AD) is the most neurodegenerative disorder leading to dementia. Neuritic plaque formation in brains is a hallmark of AD pathogenesis. Amyloid beta protein (Abeta) is the central component of neuritic plaques. Processing beta-amyloid precursor protein (APP) at the beta-secretase site by the beta-site APP cleaving enzyme 1 (BACE1) is essential for generation of Abeta. Elevation of BACE1 activity and expression has been reported in AD brains. However, no mutation in the BACE1 coding sequence has been identified in AD cases. Human BACE1 expression is tightly regulated at the transcription and translation level. To determine whether there is any single-nucleotide polymorphisms in the BACE1 gene promoter region affecting BACE1 expression in AD pathogenesis, in this study, we screened 2.6 kb of the human BACE1 gene promoter region from late-onset AD patients and found that there was no significant association between single-nucleotide polymorphisms and AD cases.
Collapse
|
25
|
Liu GT, Volpe NJ, Galetta SL. Disorders of higher cortical visual function. Neuroophthalmology 2010. [DOI: 10.1016/b978-1-4160-2311-1.00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
26
|
Deb S, McHugh R. Dementia among Persons with Down Syndrome. ACTA ACUST UNITED AC 2010. [DOI: 10.1016/s0074-7750(10)39008-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
|
27
|
Xue S, Jia L, Jia J. Association between somatostatin gene polymorphisms and sporadic Alzheimer's disease in Chinese population. Neurosci Lett 2009; 465:181-3. [PMID: 19733630 DOI: 10.1016/j.neulet.2009.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 08/29/2009] [Accepted: 09/01/2009] [Indexed: 10/20/2022]
Abstract
Recent evidence has suggested that down-regulation of somatostatin (SST) expression in the human brain during early stages of aging leads to an elevation in the steady-state levels of Abeta and therefore may be involved in Alzheimer's disease (AD) progression. We hypothesized that alterations in the SST gene might alter its expression or function and also play a role in the pathogenesis of sporadic AD (SAD). First, we sequenced the entire SST gene in 25 randomly selected controls and 25 SAD patients and then screened for C/T polymorphisms (rs4988514) in the 3' un-translated region. We genotyped rs4988514 polymorphisms as well as apolipoprotein epsilon4 (APOE epsilon4) status in 309 SAD patients and 276 normal controls with restriction fragment length polymorphism (RFLP) analysis. Results showed that the C allele of the rs4988514 polymorphism had an increased incidence in the SAD group compared to the control group (p=0.042). In subjects with the APOE epsilon4 allele, the presence of both the CC genotype and the C allele of this polymorphism were elevated in the SAD group compared to the control group (genotype p=0.027, allele p=0.011). In the whole study group, the age, sex, and APOE epsilon4 adjusted OR for the risk of AD in C allele carriers was 1.313 (95%CI=1.068-2.234, p=0.027) whereas within only APOE epsilon4 allele carriers, the adjusted OR increased to 2.734 (95%CI=1.236-5.862, p=0.012). Our results supported the notion that the C allele of the rs4988514 polymorphism may increase the risk for AD in the Chinese population and possibly have additive effect with the APOE epsilon4 allele.
Collapse
Affiliation(s)
- Sufang Xue
- Department of Neurology, Xuan Wu Hospital of the Capital Medical University, Key Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, 45 Changchun Street, Beijing 100053, PR China
| | | | | |
Collapse
|
28
|
Spremo-Potparević B, Zivković L, Djelić N, Plećas-Solarović B, Smith MA, Bajić V. Premature centromere division of the X chromosome in neurons in Alzheimer's disease. J Neurochem 2008; 106:2218-23. [PMID: 18624923 DOI: 10.1111/j.1471-4159.2008.05555.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Premature centromere division (PCD) represents a loss of control over the sequential separation and segregation of chromosome centromeres. Although first described in aging women, PCD on the X chromosome (PCD,X) is markedly elevated in peripheral blood lymphocytes of individuals suffering from Alzheimer disease (AD). The present study evaluated PCD,X, using a fluorescent in situ hybridization method, in interphase nuclei of frontal cerebral cortex neurons from sporadic AD patients and age-matched controls. The average frequency of PCD,X in AD patients (8.60 +/- 1.20%) was almost three times higher (p < 0.01) than in the control group (2.96 +/- 1.20). However, consistent with previous studies, no mitotic cells were found in neurons in either AD or control brain, suggesting an intrinsic inability of post-mitotic neurons to divide. In view of the fact that it has been well-documented that neurons in AD can re-enter into the cell division cycle, the findings presented here of increased PCD advance the hypothesis that deregulation of the cell cycle may contribute to neuronal degeneration and subsequent cognitive deficits in AD.
Collapse
|
29
|
|
30
|
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease. To rationally develop novel therapeutic and/or preventative agents for AD, an understanding of the etiology and pathogenesis of this complex disease is necessary. This article examines the evidence for the amyloid hypothesis of AD pathogenesis and discusses how it relates to the neurological and neuropathological features of AD, the known genetic risk factors and causative mutations, and the heightened risk associated with advanced age.
Collapse
Affiliation(s)
- Christopher B Eckman
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA.
| | | |
Collapse
|
31
|
Matsui T, Ingelsson M, Fukumoto H, Ramasamy K, Kowa H, Frosch MP, Irizarry MC, Hyman BT. Expression of APP pathway mRNAs and proteins in Alzheimer's disease. Brain Res 2007; 1161:116-23. [PMID: 17586478 DOI: 10.1016/j.brainres.2007.05.050] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Revised: 05/29/2007] [Accepted: 05/31/2007] [Indexed: 12/17/2022]
Abstract
In both trisomy 21 and rare cases of triplication of amyloid precursor protein (APP) Alzheimer's disease (AD) pathological changes are believed to be secondary to increased expression of APP. We hypothesized that sporadic AD may also be associated with changes in transcription of APP or its metabolic partners. To address this issue, temporal neocortex of 27 AD and 21 non-demented control brains was examined to assess mRNA levels of APP isoforms (total APP, APP containing the Kunitz protease inhibitor domain [APP-KPI] and APP770) and APP metabolic enzymatic partners (the APP cleaving enzymes beta-secretase [BACE] and presenilin-1 [PS-1], and putative clearance molecules, low-density lipoprotein receptor protein [LRP] and apolipoprotein E [apoE]). Furthermore, we evaluated how changes in APP at the mRNA level affect the amount of Tris buffer extractable APP protein and Abeta40 and 42 peptides in AD and control brains. As assessed by quantitative PCR, APP-KPI (p=0.007), APP770 (p=0.004), PS-1 (p=0.004), LRP (p=0.003), apoE (p=0.0002) and GFAP (p<0.0001) mRNA levels all increased in AD, and there was a shift from APP695 (a neuronal isoform) towards KPI containing isoforms that are present in glia as well. APP-KPI mRNA levels correlated with soluble APPalpha-KPI protein (sAPPalpha-KPI) levels measured by ELISA (tau=0.33, p=0.015 by Kendall's rank correlation); in turn, soluble APPalpha-KPI protein levels positively correlated with Tris-extractable, soluble Abeta40 (p=0.046) and 42 levels (p=0.007). The ratio of soluble APPalpha-KPI protein levels to total APP protein increased in AD, and also correlated with GFAP protein levels in AD. These results suggest that altered transcription of APP in AD is proportionately associated with Abeta peptide, may occur in the context of gliosis, and may contribute to Abeta deposition in sporadic AD.
Collapse
Affiliation(s)
- Toshifumi Matsui
- Alzheimer Disease Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Perry RT, Wiener H, Harrell LE, Blacker D, Tanzi RE, Bertram L, Bassett SS, Go RCP. Follow-up mapping supports the evidence for linkage in the candidate region at 9q22 in the NIMH Alzheimer's disease Genetics Initiative cohort. Am J Med Genet B Neuropsychiatr Genet 2007; 144B:220-7. [PMID: 17034007 DOI: 10.1002/ajmg.b.30433] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Other than the APOE peak at 19q13, the 9q22 region was identified in our original genomic scan as the candidate region with the highest multipoint lod score (MLS) in the subset of late onset Alzheimer's Disease (AD) families (MLS = 2.9 at 101 cM) from the NIMH Genetics Initiative sample. We have now genotyped an additional 12 short tandem repeats (STR) in this region. Multipoint analysis shows the region remains significant with an increase in the peak MLS from 2.9 to 3.8 at 95 cM near marker D9S1815, and the 1 LOD interval narrows from 21.5 to 11 cM. HLOD scores also provide evidence for significant linkage (4.5 with an alpha = 31%) with a further narrowing of the region to 6.6 cM (92.2-98.8 cM). Single nucleotide polymorphisms (SNPs) in the Ubiquilin1 gene (UBQLN1), located at 83.3 cM, have been reported to be significantly associated to AD, accounting for a substantial portion of the original linkage signal [Bertram et al., 2005]. Our analyses of the higher resolution genotype data generated here provide further support for the existence of a least one additional locus on chromosome 9q22. In an effort to pinpoint this putative AD susceptibility gene, we have begun to analyze SNPs in other candidate genes in and around this narrowed region to test for additional associations to AD.
Collapse
Affiliation(s)
- Rodney T Perry
- Department of Epidemiology and International Health, University of Alabama at Birmingham, Birmingham, Alabama 35294-0022, USA.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Zivković L, Spremo-Potparević B, Djelić N, Bajić V. Analysis of premature centromere division (PCD) of the chromosome 18 in peripheral blood lymphocytes in Alzheimer disease patients. Mech Ageing Dev 2006; 127:892-6. [PMID: 17069875 DOI: 10.1016/j.mad.2006.09.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2005] [Revised: 02/28/2006] [Accepted: 09/20/2006] [Indexed: 11/16/2022]
Abstract
Premature centromere division (PCD) of the chromosome 18 was analyzed by using fluorescent in situ hybridization (FISH) on interphase peripheral blood lymphocytes isolated from six sporadic Alzheimer disease (AD) patients and six healthy elderly controls. Results of FISH analysis revealed that chromosome 18 expressed PCD in 5.18% interphase nuclei of AD patients, and in 2.59% interphase nuclei of age-matched controls (p<0.05). Our study also showed that hypoploidy and hyperploidy frequency for chromosome 18 exhibited a statistically significant increase in the AD group compared to the control one. The increase in spontaneous aneuploidy of chromosome 18 in AD patients which is correlated with PCD shows that deregulation of the time of centromere separation can be considered as a manifestation of chromosome instability leading to aneuploidy.
Collapse
Affiliation(s)
- Lada Zivković
- Department of Biology and Human Genetics, Institute of Physiology, Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia and Montenegro.
| | | | | | | |
Collapse
|
34
|
Jia L, Zhou C, Lv H, Wang W, Ye J, Zhang X, Zhou W, Xu J, Wang L, Jia J. Association between presenilin 1 intronic polymorphism and late onset Alzheimer's disease in the North Chinese population. Brain Res 2006; 1116:201-5. [PMID: 16938285 DOI: 10.1016/j.brainres.2006.07.096] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2006] [Revised: 07/26/2006] [Accepted: 07/27/2006] [Indexed: 11/17/2022]
Abstract
The association between presenilin 1 intronic polymorphism (rs165932) and late onset Alzheimer's disease (LOAD) has been a matter of controversy. Within China, varied results have been reported. Therefore, we collected a large sample from the North Chinese population to test the association of the PS1 polymorphism with LOAD. AD patients (467 total, mean age=75.3+/-7.3, age at onset=70.2+/-5.1) and age-matched normal elderly controls (480 total) were recruited. Genotypes of PS1 and apolipoprotein E (APOE) were determined by PCR and RFLP. The results showed that there were significant differences in the distributions of both alleles (chi(2)=45.305, P<10(-5)) and genotypes (chi(2)=53.055, P<10(-5)) of PS1 gene between the AD and control groups. The APOE epsilon4 allele was more prevalent in patients than in controls (chi(2)=46.389, P<10(-5)). It was significantly different when PS1 alleles and genotypes were compared between AD and controls with APOE epsilon4 negative. However, no significance was found when PS1 alleles or genotypes were compared between AD and controls with APOE epsilon4 positive. Furthermore, with PS1 2/2 genotype as a reference, the odds ratios (ORs) of LOAD with PS1 1/2, 1/1+1/2 and 1/1 genotypes gradually increased allele 1 copy number, suggesting that allele 1 is a crucial risk for LOAD. In summary, we found an association between presenilin 1 intronic polymorphism and LOAD, but no influence of APOE epsilon4 on the distribution of the PS1 intronic polymorphism. In addition, the larger sample size raises the possibility that ethnic and regional differences in China may explain the differences in reported results.
Collapse
Affiliation(s)
- Longfei Jia
- Department of Neurology, Xuanwu Hospital, Capital University of Medical Sciences, Beijing, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Swerdlow RH. Is aging part of Alzheimer's disease, or is Alzheimer's disease part of aging? Neurobiol Aging 2006; 28:1465-80. [PMID: 16876913 DOI: 10.1016/j.neurobiolaging.2006.06.021] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2006] [Revised: 06/05/2006] [Accepted: 06/22/2006] [Indexed: 01/11/2023]
Abstract
For 70 years after Alois Alzheimer described a disorder of tangle-and-plaque dementia, Alzheimer's disease was a condition of the relatively young. Definitions of Alzheimer's disease (AD) have, however, changed over the past 30 years and under the revised view AD has truly become an age-related disease. Most now diagnosed with AD are elderly and would not have been diagnosed with AD as originally conceived. Accordingly, younger patients that qualify for a diagnosis of AD under both original and current Alzheimer's disease constructs now represent an exceptionally small percentage of the diagnosed population. The question of whether pathogenesis of the "early" and "late" onset cases is similar enough to qualify as a single disease was previously raised although not conclusively settled. Interestingly, debate on this issue has not kept pace with advancing knowledge about the molecular, biochemical and clinical underpinnings of tangle-and-plaque dementias. Since the question of whether both forms of AD share a common pathogenesis could profoundly impact diagnostic and treatment development efforts, it seems worthwhile to revisit this debate.
Collapse
Affiliation(s)
- Russell H Swerdlow
- Department of Neurology, University of Virginia Health System, McKim Hall, 1 Hospital Drive, P.O. Box 800394, Charlottesville, VA 22908, United States.
| |
Collapse
|
36
|
Giedraitis V, Hedlund M, Skoglund L, Blom E, Ingvast S, Brundin R, Lannfelt L, Glaser A. New Alzheimer's disease locus on chromosome 8. J Med Genet 2006; 43:931-5. [PMID: 16825432 PMCID: PMC2563209 DOI: 10.1136/jmg.2006.043000] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Family history is one of the most consistent risk factors for dementia. Therefore, analysis of families with a distinct inheritance pattern of disease can be a powerful approach for the identification of previously unknown disease genes. OBJECTIVE To map susceptibility regions for Alzheimer's disease. METHODS A complete genome scan with 369 microsatellite markers was carried out in 12 extended families collected in Sweden. Age at disease onset ranged from 53 to 78 years, but in 10 of the families there was at least one member with age at onset of < or =65 years. Mutations in known early-onset Alzheimer's disease susceptibility genes have been excluded. All people were genotyped for APOE, but no clear linkage with the epsilon4 allele was observed. RESULTS Although no common disease locus could be found in all families, in two families an extended haplotype was identified on chromosome 8q shared by all affected members. In one of the families, a non-parametric multimarker logarithm of the odds (LOD) score of 4.2 (p = 0.004) was obtained and analysis based on a dominant model showed a parametric LOD score of 2.4 for this region. All six affected members of this family shared a haplotype of 10 markers spanning about 40 cM. Three affected members in another family also shared a haplotype in the same region. CONCLUSION On the basis of our data, we propose the existence of a dominantly acting Alzheimer's disease susceptibility locus on chromosome 8.
Collapse
Affiliation(s)
- V Giedraitis
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden .
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Fernández Del Pozo V, Alvarez Alvarez M, Fernández Martínez M, Galdós Alcelay L, Gómez Busto F, Peña JA, Alfonso-Sánchez MA, Zarranz Imirizaldu JJ, de Pancorbo MM. Polymorphism in the cholesterol 24S-hydroxylase gene (CYP46A1) associated with the APOEpsilon3 allele increases the risk of Alzheimer's disease and of mild cognitive impairment progressing to Alzheimer's disease. Dement Geriatr Cogn Disord 2006; 21:81-7. [PMID: 16340204 DOI: 10.1159/000090215] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/03/2005] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Late-onset Alzheimer's disease (LOAD) is associated with changes in certain proteins, such as ApoE and Cyp46A1, of the elimination route for cerebral cholesterol. The main lipoprotein involved in its transport is ApoE whose Epsilon4 allele is the least efficient. However, the presence or absence of this allele does not determine the development of LOAD, which implies the existence of other susceptibility factors associated with the disease, such as the CYP46A1 gene that encodes the enzyme cholesterol 24S-hydroxylase. OBJECTIVE To find new data to contribute to the evaluation of whether the presence of the T allele in the polymorphic site rs754203 of the CYP46A1 gene leads to a greater risk of developing mild cognitive impairment (MCI) and LOAD. Furthermore, given the link between APOE and CYP46A1, we proceeded to relate both genotypes in each of the patient groups studied. METHODS We studied MCI and LOAD patients and also carried out an analysis of those MCI patients who progressed from a mild cognitive deterioration to a clinically evident Alzheimer's disease during the study. RESULTS The frequency of the CYP46A1-T allele in the LOAD patients with APOEpsilon3 alleles is significantly higher with respect to the control group; the same occurs in the group made up of LOAD patients together with the MCI patients who progressed to LOAD. The risk of developing LOAD when this allelic combination exists is 2.262 times higher (95% CI 1.337-4.202). However, having the CYP46A1-T allele does not increase the risk of suffering from LOAD in carriers of the APOEpsilon4 allele, probably because the transport of cholesterol is already affected in such patients and possibly masks the effect of the CYP46A1-T allele. CONCLUSIONS The CYP46A1-T allele increases the risk of suffering from LOAD in persons carrying the APOEpsilon3 allele.
Collapse
|
38
|
Yescas P, Huertas-Vazquez A, Villarreal-Molina MT, Rasmussen A, Tusié-Luna MT, López M, Canizales-Quinteros S, Alonso ME. Founder effect for the Ala431Glu mutation of the presenilin 1 gene causing early-onset Alzheimer's disease in Mexican families. Neurogenetics 2006; 7:195-200. [PMID: 16628450 DOI: 10.1007/s10048-006-0043-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2005] [Accepted: 03/23/2006] [Indexed: 10/24/2022]
Abstract
The etiology of Alzheimer's disease (AD) is complex. To date, molecular genetic studies in several families affected with AD have identified three genes associated with highly penetrant early-onset AD: Presenilin 1 (PSEN1), Presenilin 2 (PSEN2) and beta-amyloid precursor protein (APP); and one gene (apolipoprotein E) associated with late-onset AD. Molecular analysis of the PSEN1 gene was performed by direct sequencing of genomic DNA. The possible founder effect was investigated analyzing two highly polymorphic microsatellite markers flanking the PSEN1 gene. Twelve unrelated Mexican families with early-onset AD were analyzed. The Ala431Glu mutation in exon 12 of PSEN1 was found in nine (75%) of these families, which segregated showing autosomal dominant inheritance. Because all families bearing the mutation are from the State of Jalisco (located in Western Mexico), a founder effect was hypothesized. Microsatellite haplotype analysis suggested a common ancestor in these nine kindreds. In conclusion, the Ala431Glu mutation is a prevalent cause of early-onset familial Alzheimer's disease in families from the State of Jalisco, Mexico. Genetic evidence supports that it is a founder mutation descending from a single common ancestor. These findings have important implications for prompt diagnosis and genetic counseling for Mexican patients with familial AD from Jalisco.
Collapse
Affiliation(s)
- Petra Yescas
- Departamento de Neurogenética y Biología Molecular, lnstituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, México D.F., México
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Xue S, Jia J. Genetic association between Ubiquitin Carboxy-terminal Hydrolase-L1 gene S18Y polymorphism and sporadic Alzheimer's disease in a Chinese Han population. Brain Res 2006; 1087:28-32. [PMID: 16626667 DOI: 10.1016/j.brainres.2006.02.121] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2005] [Revised: 11/17/2005] [Accepted: 02/26/2006] [Indexed: 11/30/2022]
Abstract
Increasing evidence indicates that the dysfunction of ubiquitin-proteasome system (UPS) is associated with Alzheimer's disease (AD). In the ubiquitin-proteasome pathway, Ubiquitin Carboxy-terminal Hydrolase-L1 (UCH-L1) plays an important role for the cellular clearance of abnormal proteins. Since a substitution of serine by tyrosine at codon 18, exon 3 (S18Y polymorphism) of the UCH-L1 gene exhibits a protective effect against the development of degenerative disease such as sporadic Parkinson's disease (PD) in several different ethnic groups, we hypothesized that UCH-L1 gene S18Y polymorphism may have that same effect on the pathologic process of AD. We examined UCH-L1 S18Y polymorphism genotypes of 116 sporadic AD patients and 123 healthy subjects in Chinese Han population using PCR-restriction fragment length polymorphism (RFLP) analysis. The allele and genotype data as well as data after stratification by age of onset failed to demonstrate any association between AD and S18Y polymorphism. However, after stratification by gender, female AD patients showed significantly less frequencies of Y allele and YY genotype in S18Y polymorphism than female controls (P = 0.003 and P = 0.015 respectively). We conclude that Y allele and YY genotype of S18Y in the UCH-L1 gene may have a protective effect against sporadic AD in female subjects, probably due to altering the function of UCH-L1 and the interactions among different risk factors.
Collapse
Affiliation(s)
- Sufang Xue
- Department of Neurology, Xuanwu Hospital, Capital University of Medical Sciences, 45 Changchun Street, 100053 Beijing, China
| | | |
Collapse
|
40
|
Haroun N, Dunn L, Haroun A, Cadenhead KS. Risk and protection in prodromal schizophrenia: ethical implications for clinical practice and future research. Schizophr Bull 2006; 32:166-78. [PMID: 16207892 PMCID: PMC2632176 DOI: 10.1093/schbul/sbj007] [Citation(s) in RCA: 173] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Over the last decade schizophrenia researchers have turned their attention to earlier identification in the prodromal period of illness. A greater understanding of both risk and protective factors can lead to improved prevention and treatment strategies in this vulnerable population. This research, however, has far-reaching ethical implications. One year follow-up data from 50 individuals who met established criteria for a prodromal state is used to illustrate ethical issues that directly affect clinicians and future research strategies. At 1-year follow-up, the psychotic transition rate was 13%, but it increased in subsequent years with smaller sample sizes. One-half developed an affective psychosis. The converted sample was older (p > 0.05) than the nonconverted sample and more likely to have a premorbid history of substance abuse, as well as higher clinical ratings on "subsyndromal" psychotic items (delusional thinking, suspiciousness, and thought disorder). Despite a lack of conversion, the nonconverted sample remained symptomatic and had a high rate of affective and anxiety disorders with evidence of functional disability. This conversion rate is relatively low compared to similar studies at 1 year. Specific risk factors were identified, but these findings need to be replicated in a larger cohort. By examining the rate of conversion and nonconversion in this sample as an example, we hope to contribute to the discussion of implications for clinical practice and the direction of future research in the schizophrenia prodrome. Finally, our data strengthen the evidence base available to inform the discussion of ethical issues relevant to this important research area.
Collapse
Affiliation(s)
| | | | | | - Kristin S Cadenhead
- To whom correspondence should be addressed; Department of Psychiatry, 0810, University of California, San Diego, 9500 Gilman Drive, La Jolla CA 92093-0810; tel: 619-725-3537, fax: 619-260-8437, e-mail:
| |
Collapse
|
41
|
Bauduceau B, Bourdel-Marchasson I, Brocker P, Taillia H. The brain of the elderly diabetic patient. DIABETES & METABOLISM 2005; 31 Spec No 2:5S92-5S97. [PMID: 16415771 DOI: 10.1016/s1262-3636(05)73657-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
All available estimations agree that the French population is aging and that the proportion of diabetics in the elderly population is increasing. The prevalence of diabetes could be about 10% in the over 65 y population. The fact that diabetes has an effect on brain function is widely accepted, but there are very few studies providing pertinent details. Diabetes is known to affect brain function, potential consequences including cognitive decline, dementia, depression, and stroke. These complications are frequently associated, leading to poor quality-of-life with considerable social and economic impact. While the results of different studies can be contradictory, there is an overall trend towards the conclusion that diabetes, often associated with high blood pressure, contributes to cognitive decline in elderly diabetics as well as to an increased frequency and severity of cerebral vascular events. These considerations point out the importance of proper management of diabetes in the elderly population and the need for cooperative studies to determine the role of diabetes and different cardiovascular risk factors in the development of dementia, stroke, and depressive syndromes whose consequences are probably underestimated.
Collapse
Affiliation(s)
- B Bauduceau
- Service d'Endocrinologie, Hôpital d'Instruction des Armées Bégin, Saint-Mandé, France.
| | | | | | | |
Collapse
|
42
|
Roberts JS, Cupples LA, Relkin NR, Whitehouse PJ, Green RC. Genetic risk assessment for adult children of people with Alzheimer's disease: the Risk Evaluation and Education for Alzheimer's Disease (REVEAL) study. J Geriatr Psychiatry Neurol 2005; 18:250-5. [PMID: 16306249 DOI: 10.1177/0891988705281883] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
As genetic risk factors continue to be identified for common, complex adult-onset diseases, it will become increasingly important to understand if, how, and when to translate these discoveries into clinical practice. This article provides an overview of and results to date from the REVEAL study, a multisite randomized clinical trial (n = 162) examining the impact of a genetic risk assessment program, including apolipoprotein E genotype disclosure, for adult children of people with Alzheimer's disease. The study's rationale and procedures are described, including the generation of numerical lifetime risk curves for use in the education and counseling protocol. Findings are summarized across numerous study questions, including (1) who seeks genetic risk assessment and why, (2) how apolipoprotein E results affect risk perceptions, (3) the psychological impact of genetic risk assessment, and (4) how risk information affects participants' subsequent health and insurance behaviors.
Collapse
Affiliation(s)
- J Scott Roberts
- Department of Neurology, Boston University School of Medicine, MA 02118, USA.
| | | | | | | | | |
Collapse
|
43
|
El Mouedden M, Haseldonckx M, Mackie C, Meert T, Mercken M. Method for the determination of the levels of β-amyloid peptide in the CSF sampled from freely moving rats. J Pharmacol Toxicol Methods 2005; 52:229-33. [PMID: 16125620 DOI: 10.1016/j.vascn.2005.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2004] [Accepted: 02/11/2005] [Indexed: 11/19/2022]
Abstract
INTRODUCTION In the present study, a model was developed to determine the effect of secretase inhibition on beta-amyloid peptide (Abeta) levels in the cerebrospinal fluid (CSF) of freely moving adult rats. METHODS Rats were chronically implanted with a cannula into the cisterna magna and CSF samples were collected at different time points from the same animal without anaesthesia. The levels of CSF Abeta were measured by a sandwich ELISA assay. RESULTS Administration of DAPT, a functional gamma-secretase inhibitor, resulted in a substantial reduction of Abeta40 and Abeta42, confirming the in vivo functionality of the CSF as a biomarker source for endogenous APP processing modulation by secretase inhibitors. DISCUSSION Thus, the present work provides clear evidence for the usefulness of CSF sampling from the freely moving rat model for testing the effectiveness of small molecule inhibitors of Abeta production.
Collapse
Affiliation(s)
- Mohammed El Mouedden
- Johnson & Johnson Pharmaceutical Research & Development, a Division of Janssen Pharmaceutica, Turnhoutseweg 30, B-2340 Beerse, Belgium.
| | | | | | | | | |
Collapse
|
44
|
Pietrzik C, Behl C. Concepts for the treatment of Alzheimer's disease: molecular mechanisms and clinical application. Int J Exp Pathol 2005; 86:173-85. [PMID: 15910551 PMCID: PMC2517414 DOI: 10.1111/j.0959-9673.2005.00435.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
To date, various strategies have been developed in order to prevent or to slow down the progression of Alzheimer's disease (AD). Despite the medical need for an effective therapeutic treatment of AD, progress towards this goal is painstakingly slow. Although AD is the most common neurodegenerative disorder and a large amount of primary basic and clinical research has been performed already, it appears very difficult to identify appropriate targets, which would promise fast, effective and safe strategies to combat the disease onset and progression. In this review, we present some of clinically applied treatment options, which may improve AD symptoms for a short period but so far lack the ability to prevent or halt this devastating disease. Additionally, we summarize some of the experimental approaches in AD therapy, which might lead to the development of more promising drugs in the future.
Collapse
Affiliation(s)
- Claus Pietrzik
- Department of Molecular Neurodegeneration, Johannes Gutenberg-University Mainz, Medical SchoolMainz, Germany
| | - Christian Behl
- Department of Molecular Neuroprotection and Aging Research, Institute for Physiological Chemistry and Pathobiochemistry, Johannes Gutenberg-University Mainz, Medical SchoolMainz, Germany
| |
Collapse
|
45
|
Du AT, Schuff N, Chao LL, Kornak J, Jagust WJ, Kramer JH, Reed BR, Miller BL, Norman D, Chui HC, Weiner MW. Age effects on atrophy rates of entorhinal cortex and hippocampus. Neurobiol Aging 2005; 27:733-40. [PMID: 15961190 PMCID: PMC1779763 DOI: 10.1016/j.neurobiolaging.2005.03.021] [Citation(s) in RCA: 157] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2004] [Revised: 03/16/2005] [Accepted: 03/30/2005] [Indexed: 01/16/2023]
Abstract
The effects of age, subcortical vascular disease, apolipoprotein E (APOE) epsilon4 allele and hypertension on entorhinal cortex (ERC) and hippocampal atrophy rates were explored in a longitudinal MRI study with 42 cognitively normal (CN) elderly subjects from 58 to 87 years old. The volumes of the ERC, hippocampus, and white matter hyperintensities (WMH) and the presence of lacunes were assessed on MR images. Age was significantly associated with increased atrophy rates of 0.04+/-0.02% per year for ERC and 0.05+/-0.02% per year for hippocampus. Atrophy rates of hippocampus, but not that of ERC increased with presence of lacunes, in addition to age. WMH, APOE epsilon4 and hypertension had no significant effect on atrophy rates. In conclusion, age and presence of lacunes should be taken into consideration in imaging studies of CN subjects and AD patients to predict AD progression and assess the response to treatment trials.
Collapse
Affiliation(s)
- An-Tao Du
- Magnetic Resonance Unit (114M), Department of Veterans Affairs
Medical Center, 4150, Clement Street, San Francisco, CA 94121, USA
| | - Norbert Schuff
- Magnetic Resonance Unit (114M), Department of Veterans Affairs
Medical Center, 4150, Clement Street, San Francisco, CA 94121, USA
- Department of Radiology, University of California, San
Francisco, CA, USA
| | - Linda L. Chao
- Magnetic Resonance Unit (114M), Department of Veterans Affairs
Medical Center, 4150, Clement Street, San Francisco, CA 94121, USA
- Department of Radiology, University of California, San
Francisco, CA, USA
- Department of Psychiatry, University of California, San
Francisco, CA, USA
| | - John Kornak
- Magnetic Resonance Unit (114M), Department of Veterans Affairs
Medical Center, 4150, Clement Street, San Francisco, CA 94121, USA
- Department of Radiology, University of California, San
Francisco, CA, USA
| | | | - Joel H. Kramer
- Department of Psychiatry, University of California, San
Francisco, CA, USA
| | - Bruce R. Reed
- Department of Neurology, University of California, Davis, CA,
USA
| | - Bruce L. Miller
- Department of Neurology, University of California, San
Francisco, CA, USA
| | - David Norman
- Department of Radiology, University of California, San
Francisco, CA, USA
| | - Helena C. Chui
- Department of Neurology, University of Southern California, Los
Angeles, CA, USA
| | - Michael W. Weiner
- Magnetic Resonance Unit (114M), Department of Veterans Affairs
Medical Center, 4150, Clement Street, San Francisco, CA 94121, USA
- Department of Radiology, University of California, San
Francisco, CA, USA
- Department of Psychiatry, University of California, San
Francisco, CA, USA
- Department of Neurology, University of California, San
Francisco, CA, USA
- Department of Medicine, University of California, San
Francisco, CA, USA
- * Corresponding author. Tel.: +1 415 221
4810x3642; fax: +1 415 668 2864. E-mail address: (M.W. Weiner)
| |
Collapse
|
46
|
Benaki D, Zikos C, Evangelou A, Livaniou E, Vlassi M, Mikros E, Pelecanou M. Solution structure of humanin, a peptide against Alzheimer's disease-related neurotoxicity. Biochem Biophys Res Commun 2005; 329:152-60. [PMID: 15721287 DOI: 10.1016/j.bbrc.2005.01.100] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2005] [Indexed: 11/18/2022]
Abstract
Humanin is a newly identified 24-residue peptide that suppresses neuronal cell death caused by a wide spectrum of familial Alzheimer's disease genes and the beta-amyloid peptide. In this study, NMR and circular dichroism studies of synthetic humanin in aqueous and 30% 2,2,2-trifluoroethanol (TFE) solutions are reported. In aqueous solution, humanin exists predominantly in an unstructured conformation in equilibrium with turn-like structures involving residues Gly5 to Leu10 and Glu15 to Leu18, providing indication of nascent helix. In the less polar environment of 30% TFE, humanin readily adopts helical structure with long-range order spanning residues Gly5 to Leu18. Comparative 3D modeling studies and topology predictions are in qualitative agreement with the experimental findings in both environments. Our studies reveal a flexible peptide in aqueous environment, which is free to interact with possible receptors that mediate its action, but may also acquire a helical conformation necessary for specific interactions and/or passage through membranes.
Collapse
Affiliation(s)
- Dimitra Benaki
- Institute of Biology, NCSR Demokritos, 153 10 Athens, Greece
| | | | | | | | | | | | | |
Collapse
|
47
|
Fernandez LL, Scheibe RM. Is MTHFR polymorphism a risk factor for Alzheimer's disease like APOE? ARQUIVOS DE NEURO-PSIQUIATRIA 2005; 63:1-6. [PMID: 15830056 DOI: 10.1590/s0004-282x2005000100001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND The role of methylenetetrahydrofolate reductase (MTHFR) gene polymorphisms as risk factors for the occurrence of Alzheimer's disease (AD) is still controversial. OBJECTIVE To verify the association between MTHFR and apolipoprotein E (APOE) polymorphisms and Alzheimer's disease. METHOD This work was conducted as a case-control study. Cases included thirty patients with probable AD. Controls were constituted by 29 individuals without dementia according to neuropsychological tests paired to age, sex, race and educational level. DNA was isolated from peripheral leukocytes of anticoagulated venous blood. Genotyping of APOE and MTHFR were performed by DNA amplification and digestion. The frequences of APOE and MTHFR genotypes were submitted by chi-square test corrected by Fisher test; the APOE genotypes, to chi-square linear tendency test and the frequences of MTHFR mutant and AD, by stratificated analysis adjust by Mantel-Haenszel method. RESULTS There was significant difference about APOE4 and APOE2 in the groups. (p=0.002) The odds ratio increased exponentially with the increased number of E4 allele (chi2 linear tendency test). No significant difference was detected on MTHFR genotypes in both case and control groups. CONCLUSION The APOE4 is a risk factor and demonstrated a dose-dependent effect while APOE2 allele conferred a protection to AD. The MTHFR mutation had no correlation with AD.
Collapse
|
48
|
Christoforidis M, Schober R, Krohn K. Genetic-morphologic association study: association between the low density lipoprotein-receptor related protein (LRP) and cerebral amyloid angiopathy. Neuropathol Appl Neurobiol 2005; 31:11-9. [PMID: 15634227 DOI: 10.1111/j.1365-2990.2004.00614.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Accumulating evidence suggests that genetic factors such as apolipoprotein E (APOE), can act in different ways in the pathogenesis of cerebral amyloid angiopathy (CAA) and Alzheimer's disease (AD). The role of the low-density lipoprotein-receptor related protein (LRP), the major cerebral APOE receptor, in AD has been discussed controversially depending on data from different populations and methodological approaches. We examined the influence of LRP polymorphisms on CAA in 125 post-mortem cases genotyped for APOE and classified according to the neurofibrillary Braak and Braak staging of AD (indicating neurodegeneration grade). CAA was assessed separately for leptomeningeal (CAAlep.), noncapillary cortical (CAAcort.) and capillary cortical (CAAcap.) vessels in beta-amyloid stained sections. Our results suggest: (i) the 87 bp allele of LRP5' polymorphism (LRP5') is an independent predictive factor for CAAcort. and CAAlep.; (ii) the C/C genotype (C allele) of the LRP exon 3 polymorphism is positively associated with the severity of CAAlep. and CAAcort., implicating a younger age of CAA onset and/or faster CAA progression; (iii) as CAAcort. and CAAlep. showed different genetic associations in contrast to CAAcap., we can underscore the hypothesis that different molecular mechanisms are involved in CAA pathogenesis of noncapillary and capillary cerebral vessels. Our results lead us to postulate that the LRP5'87 bp and the LRP exon 3 C alleles of the LRP gene (or another locus that might be in linkage disequilibrium with these LRP polymorphic sites) could modify cerebrovascular LRP function or expression in noncapillary cerebral vessels, leading to an increased cerebrovascular amyloid deposition.
Collapse
|
49
|
Sun Y, Shi J, Zhang S, Tang M, Han H, Guo Y, Ma C, Liu X, Li T. The APOC3 SstI polymorphism is weakly associated with sporadic Alzheimer's disease in a Chinese population. Neurosci Lett 2005; 380:219-22. [PMID: 15862889 DOI: 10.1016/j.neulet.2005.01.038] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2004] [Revised: 12/26/2004] [Accepted: 01/13/2005] [Indexed: 02/05/2023]
Abstract
In order to clarify the relationship of apolipoprotein CIII (APOC3) polymorphism and sporadic Alzheimer's disease (AD) in Chinese, 165 sporadic AD patients and 174 age-matched elderly individuals were genotyped for the APOC3 SstI and apolipoprotein E (APOE) HhaI polymorphisms. As the result, the APOC3 3017G allele was found to be associated with AD in APOE epsilon4 allele noncarriers (chi2=4.433, P=0.035), and the risk estimate of allele C versus G resulted in an OR of 1.56 (95% CI: 1.03-2.37), although in total no significant differences of allelic or genotypic frequencies between patients and controls were found. Assessment of interaction between APOE epsilon4 and APOC3 3017G status presented an adjusted odds ratio of 0.62 (95% CI: 0.37-1.03) with a borderline significant P-value (P=0.066). Therefore, we conclude that the rare APOC3 G allele may offer some protection against the development of sporadic AD in APOE epsilon4 noncarriers in Chinese.
Collapse
Affiliation(s)
- Yan Sun
- Department of Medical Genetics, West China Hospital, Sichuan University and Division of Human Morbid Genomics, State Key Laboratory of Biotherapy of Human Diseases, Chengdu 610041, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Lung FW, Yen YC, Chou LJ, Hong CJ, Wu CK. The allele interaction between apolipoprotein epsilon2 and epsilon4 in Taiwanese Alzheimer's disease patients. Acta Psychiatr Scand 2005; 111:38-43. [PMID: 15636592 DOI: 10.1111/j.1600-0447.2004.00394.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE This study aimed to determine the impact of the present of apolipoprotein epsilon (Apoepsilon) 2 on the relationship between Apoepsilon4 and Alzheimer's disease (AD). METHOD We examined ApoE genotypes in 428 Taiwanese patients with AD and 807 controls; all participants were older than 65 years. RESULTS The allele frequency of Apoepsilon4 was greater in AD patients than controls, but significantly lower than in Caucasians. The presence of an epsilon2 allele alone was not associated with lower risk for AD, but the presence of an epsilon2 allele was associated with an epsilon4 allele frequency similar to that of controls. CONCLUSION The low allele frequency of epsilon4 in persons with an epsilon2 allele suggests that this may be part of the protective effect of epsilon2 against AD.
Collapse
Affiliation(s)
- F-W Lung
- Department of Psychiatry, Military Kaohsiung General Hospital, Chung Cheng, Kaohsiung, Taiwan.
| | | | | | | | | |
Collapse
|