1
|
Ankeny SE, Bacci JR, Decourt B, Sabbagh MN, Mielke MM. Navigating the Landscape of Plasma Biomarkers in Alzheimer's Disease: Focus on Past, Present, and Future Clinical Applications. Neurol Ther 2024; 13:1541-1557. [PMID: 39244522 PMCID: PMC11541985 DOI: 10.1007/s40120-024-00658-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 08/20/2024] [Indexed: 09/09/2024] Open
Abstract
As the prevalence of Alzheimer's disease (AD) and its impact on healthcare systems increase, developing tools for accurate diagnosis and monitoring of disease progression is a priority. Recent technological advancements have allowed for the development of blood-based biomarkers (BBMs) to aid in the diagnosis of AD, but many questions remain regarding the clinical implementation of these BBMs. This review outlines the historical timeline of AD BBM development. It highlights key breakthroughs that have transformed the perspective of AD BBMs from theoretically ideal but unattainable markers, to clinically valid and reliable BBMs with potential for implementation in healthcare settings. Technological advancements like single-molecule detection and mass spectrometry methods have significantly improved assay sensitivity and accuracy. High-throughput, fully automated platforms have potential for clinical use. Despite these advancements, however, significant work is needed before AD BBMs can be implemented in widespread clinical practice. Cutpoints must be established, the influence of chronic conditions and medications on BBM levels must be better understood, and guidelines must be created for healthcare providers related to interpreting and communicating information obtained from AD BBMs. Additionally, the development of BBMs for synaptic dysfunction, inflammation, and cerebrovascular disease may provide better precision medicine approaches to treating AD and related dementia. Future research and collaboration between scientists and physicians are essential to addressing these challenges and further advancing AD BBMs, with the goal of integration in clinical practice.
Collapse
Affiliation(s)
- Sarrah E Ankeny
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Julia R Bacci
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Boris Decourt
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Marwan N Sabbagh
- Alzheimer's and Memory Disorders Division, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Michelle M Mielke
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
2
|
Hsu YP, Hsu CW, Chen LF, Liu YK. Methodological flaws in"diagnostic accuracy of blood biomarkers for Alzheimer's disease and amnestic mild cognitive impairment: A meta-analysis". Ageing Res Rev 2023; 88:101938. [PMID: 37088230 DOI: 10.1016/j.arr.2023.101938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 02/19/2023] [Accepted: 04/19/2023] [Indexed: 04/25/2023]
Abstract
We read with interest the review by Chen et al. They intended to examine the diagnostic accuracy of blood-based biomarkers for detecting Alzheimer's disease and amnestic mild cognitive impairment. We believe that there were substantial methodological flaws in their meta-analysis. These methodological flaws included no comprehensive literature search details, neglect of the negative result research, no prespecified cut-off values, erroneous data input in their meta-analysis, and the issue of prevalence determined by the included studies. These factors potentially contributed to overestimation of the discriminative accuracy of blood-based biomarkers. Subsequently, the conclusion that blood-based biomarkers are effective tools for detecting Alzheimer's disease is debatable without correction of these methodological flaws and providing robust and trustworthy estimates.
Collapse
Affiliation(s)
- Yuan-Pin Hsu
- Department of Emergency, School of Medicine, College of Medicine, Taipei Medical University, Taiwan; Emergency Department, Department of Emergency and Critical Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan
| | - Chin-Wang Hsu
- Department of Emergency, School of Medicine, College of Medicine, Taipei Medical University, Taiwan; Emergency Department, Department of Emergency and Critical Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan
| | - Liang-Fu Chen
- Department of Emergency, School of Medicine, College of Medicine, Taipei Medical University, Taiwan; Emergency Department, Department of Emergency and Critical Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan
| | - Ying-Kuo Liu
- Department of Emergency, School of Medicine, College of Medicine, Taipei Medical University, Taiwan; Emergency Department, Department of Emergency and Critical Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan.
| |
Collapse
|
3
|
Mei Y, Li Y, Cheng Y, Gao L. The effect of gastric bypass surgery on cognitive function of Alzheimer's disease and the role of GLP1-SGLT1 pathway. Exp Neurol 2023; 363:114377. [PMID: 36893833 DOI: 10.1016/j.expneurol.2023.114377] [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: 12/13/2022] [Revised: 02/20/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023]
Abstract
OBJECTIVE Gastric bypass surgery has been shown to improve metabolic profiles via GLP1, which may also have cognitive benefits for Alzheimer's disease (AD) patients. However, the exact mechanism requires further investigation. METHODS Roux-en-Y gastric bypass or sham surgery was performed on APP/PS1/Tau triple transgenic mice (an AD mice model) or wild type C57BL/6 mice. Morris Water Maze (MWM) test was used to evaluate the cognitive function of mice and animal tissue samples were obtained for measurements two months after the surgery. Additionally, STC-1 intestine cells were treated with siTAS1R2 and siSGLT1, and HT22 nerve cells were treated with Aβ, siGLP1R, GLP1 and siSGLT1 in vitro to explore the role of GLP1-SGLT1 related signaling pathway in cognitive function. RESULTS The MWM test showed that bypass surgery significantly improved cognitive function in AD mice as measured by navigation and spatial probe tests. Moreover, bypass surgery reversed neurodegeneration, down-regulated hyperphosphorylation of Tau protein and Aβ deposition, improved glucose metabolism, and up-regulated the expression of GLP1, SGLT1, and TAS1R2/3 in the hippocampus. Furthermore, GLP1R silencing down-regulated SGLT1 expression, whereas SGLT1 silencing increased Tau protein deposition and exacerbated dysregulated of glucose metabolism in HT22 cells. However, RYGB did not alter the level of GLP1 secretion in the brainstem (where central GLP1 is mainly produced). Additionally, GLP1 expression was upregulated by RYGB via TAS1R2/3-SGLT1 activation sequentially in the small intestine. CONCLUSION RYGB surgery could improve cognition function in AD mice through facilitating glucose metabolism and reducing Tau phosphorylation and Aβ deposition in the hippocampus, mediated by peripheral serum GLP1 activation of SGLT1 in the brain. Furthermore, RYGB increased GLP1 expression through sequential activation of TAS1R2/TAS1R3 and SGLT1 in the small intestine.
Collapse
Affiliation(s)
- Yingna Mei
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, China
| | - Yubing Li
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, China
| | - Yanxiang Cheng
- Department of Obstetrics & Gynecology, Renmin Hospital of Wuhan University, China.
| | - Ling Gao
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, China.
| |
Collapse
|
4
|
Brand AL, Lawler PE, Bollinger JG, Li Y, Schindler SE, Li M, Lopez S, Ovod V, Nakamura A, Shaw LM, Zetterberg H, Hansson O, Bateman RJ. The performance of plasma amyloid beta measurements in identifying amyloid plaques in Alzheimer's disease: a literature review. Alzheimers Res Ther 2022; 14:195. [PMID: 36575454 PMCID: PMC9793600 DOI: 10.1186/s13195-022-01117-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/06/2022] [Indexed: 12/28/2022]
Abstract
The extracellular buildup of amyloid beta (Aβ) plaques in the brain is a hallmark of Alzheimer's disease (AD). Detection of Aβ pathology is essential for AD diagnosis and for identifying and recruiting research participants for clinical trials evaluating disease-modifying therapies. Currently, AD diagnoses are usually made by clinical assessments, although detection of AD pathology with positron emission tomography (PET) scans or cerebrospinal fluid (CSF) analysis can be used by specialty clinics. These measures of Aβ aggregation, e.g. plaques, protofibrils, and oligomers, are medically invasive and often only available at specialized medical centers or not covered by medical insurance, and PET scans are costly. Therefore, a major goal in recent years has been to identify blood-based biomarkers that can accurately detect AD pathology with cost-effective, minimally invasive procedures.To assess the performance of plasma Aβ assays in predicting amyloid burden in the central nervous system (CNS), this review compares twenty-one different manuscripts that used measurements of 42 and 40 amino acid-long Aβ (Aβ42 and Aβ40) in plasma to predict CNS amyloid status. Methodologies that quantitate Aβ42 and 40 peptides in blood via immunoassay or immunoprecipitation-mass spectrometry (IP-MS) were considered, and their ability to distinguish participants with amyloidosis compared to amyloid PET and CSF Aβ measures as reference standards was evaluated. Recent studies indicate that some IP-MS assays perform well in accurately and precisely measuring Aβ and detecting brain amyloid aggregates.
Collapse
Affiliation(s)
- Abby L Brand
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Paige E Lawler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - James G Bollinger
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Melody Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Samir Lopez
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Vitaliy Ovod
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Akinori Nakamura
- Department of Biomarker Research, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Cognition and Behavior Science, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
- Memory Clinic, Skåne University Hospital, Lund, Sweden
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA.
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
5
|
Uchida K. Waste Clearance in the Brain and Neuroinflammation: A Novel Perspective on Biomarker and Drug Target Discovery in Alzheimer's Disease. Cells 2022; 11:cells11050919. [PMID: 35269541 PMCID: PMC8909773 DOI: 10.3390/cells11050919] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/26/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is a multifactorial disease with a heterogeneous etiology. The pathology of Alzheimer’s disease is characterized by amyloid-beta and hyperphosphorylated tau, which are necessary for disease progression. Many clinical trials on disease-modifying drugs for AD have failed to indicate their clinical benefits. Recent advances in fundamental research have indicated that neuroinflammation plays an important pathological role in AD. Damage- and pathogen-associated molecular patterns in the brain induce neuroinflammation and inflammasome activation, causing caspase-1-dependent glial and neuronal cell death. These waste products in the brain are eliminated by the glymphatic system via perivascular spaces, the blood-brain barrier, and the blood–cerebrospinal fluid barrier. Age-related vascular dysfunction is associated with an impairment of clearance and barrier functions, leading to neuroinflammation. The proteins involved in waste clearance in the brain and peripheral circulation may be potential biomarkers and drug targets in the early stages of cognitive impairment. This short review focuses on waste clearance dysfunction in AD pathobiology and discusses the improvement of waste clearance as an early intervention in prodromal AD and preclinical stages of dementia.
Collapse
Affiliation(s)
- Kazuhiko Uchida
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba 305-8575, Ibaraki, Japan; ; Tel.: +81-29-853-3210; Fax: +81-50-3730-7456
- Institute for Biomedical Research, MCBI, 4-9-29 Matsushiro, Tsukuba 305-0035, Ibaraki, Japan
| |
Collapse
|
6
|
Fan Q, Gao Y, Mazur F, Chandrawati R. Nanoparticle-based colorimetric sensors to detect neurodegenerative disease biomarkers. Biomater Sci 2021; 9:6983-7007. [PMID: 34528639 DOI: 10.1039/d1bm01226f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neurodegenerative disorders (NDDs) are progressive, incurable health conditions that primarily affect brain cells, and result in loss of brain mass and impaired function. Current sensing technologies for NDD detection are limited by high cost, long sample preparation, and/or require skilled personnel. To overcome these limitations, optical sensors, specifically colorimetric sensors, have garnered increasing attention towards the development of a cost-effective, simple, and rapid alternative approach. In this review, we evaluate colorimetric sensing strategies of NDD biomarkers (e.g. proteins, neurotransmitters, bio-thiols, and sulfide), address the limitations and challenges of optical sensor technologies, and provide our outlook on the future of this field.
Collapse
Affiliation(s)
- Qingqing Fan
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia.
| | - Yuan Gao
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia.
| | - Federico Mazur
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia.
| | - Rona Chandrawati
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia.
| |
Collapse
|
7
|
Koychev I, Jansen K, Dette A, Shi L, Holling H. Blood-Based ATN Biomarkers of Alzheimer's Disease: A Meta-Analysis. J Alzheimers Dis 2021; 79:177-195. [PMID: 33252080 DOI: 10.3233/jad-200900] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The Amyloid Tau Neurodegeneration (ATN) framework was proposed to define the biological state underpinning Alzheimer's disease (AD). Blood-based biomarkers offer a scalable alternative to the costly and invasive currently available biomarkers. OBJECTIVE In this meta-analysis we sought to assess the diagnostic performance of plasma amyloid (Aβ40, Aβ42, Aβ42/40 ratio), tangle (p-tau181), and neurodegeneration (total tau [t-tau], neurofilament light [NfL]) biomarkers. METHODS Electronic databases were screened for studies reporting biomarker concentrations for AD and control cohorts. Biomarker performance was examined by random-effect meta-analyses based on the ratio between biomarker concentrations in patients and controls. RESULTS 83 studies published between 1996 and 2020 were included in the analyses. Aβ42/40 ratio as well as Aβ42 discriminated AD patients from controls when using novel platforms such as immunomagnetic reduction (IMR). We found significant differences in ptau-181 concentration for studies based on single molecule array (Simoa), but not for studies based on IMR or ELISA. T-tau was significantly different between AD patients and control in IMR and Simoa but not in ELISA-based studies. In contrast, NfL differentiated between groups across platforms. Exosome studies showed strong separation between patients and controls for Aβ42, t-tau, and p-tau181. CONCLUSION Currently available assays for sampling plasma ATN biomarkers appear to differentiate between AD patients and controls. Novel assay methodologies have given the field a significant boost for testing these biomarkers, such as IMR for Aβ, Simoa for p-tau181. Enriching samples through extracellular vesicles shows promise but requires further validation.
Collapse
Affiliation(s)
- Ivan Koychev
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Katrin Jansen
- Department of Psychology, University of Münster, Münster, Germany
| | - Alina Dette
- Department of Psychology, University of Münster, Münster, Germany
| | - Liu Shi
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Heinz Holling
- Department of Psychology, University of Münster, Münster, Germany
| |
Collapse
|
8
|
Alawode DOT, Heslegrave AJ, Ashton NJ, Karikari TK, Simrén J, Montoliu‐Gaya L, Pannee J, O´Connor A, Weston PSJ, Lantero‐Rodriguez J, Keshavan A, Snellman A, Gobom J, Paterson RW, Schott JM, Blennow K, Fox NC, Zetterberg H. Transitioning from cerebrospinal fluid to blood tests to facilitate diagnosis and disease monitoring in Alzheimer's disease. J Intern Med 2021; 290:583-601. [PMID: 34021943 PMCID: PMC8416781 DOI: 10.1111/joim.13332] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/18/2021] [Accepted: 03/30/2021] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is increasingly prevalent worldwide, and disease-modifying treatments may soon be at hand; hence, now, more than ever, there is a need to develop techniques that allow earlier and more secure diagnosis. Current biomarker-based guidelines for AD diagnosis, which have replaced the historical symptom-based guidelines, rely heavily on neuroimaging and cerebrospinal fluid (CSF) sampling. While these have greatly improved the diagnostic accuracy of AD pathophysiology, they are less practical for application in primary care, population-based and epidemiological settings, or where resources are limited. In contrast, blood is a more accessible and cost-effective source of biomarkers in AD. In this review paper, using the recently proposed amyloid, tau and neurodegeneration [AT(N)] criteria as a framework towards a biological definition of AD, we discuss recent advances in biofluid-based biomarkers, with a particular emphasis on those with potential to be translated into blood-based biomarkers. We provide an overview of the research conducted both in CSF and in blood to draw conclusions on biomarkers that show promise. Given the evidence collated in this review, plasma neurofilament light chain (N) and phosphorylated tau (p-tau; T) show particular potential for translation into clinical practice. However, p-tau requires more comparisons to be conducted between its various epitopes before conclusions can be made as to which one most robustly differentiates AD from non-AD dementias. Plasma amyloid beta (A) would prove invaluable as an early screening modality, but it requires very precise tests and robust pre-analytical protocols.
Collapse
Affiliation(s)
- D. O. T. Alawode
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - A. J. Heslegrave
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - N. J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational MedicineDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Department of Old Age PsychiatryInstitute of Psychiatry, Psychology & NeuroscienceKing’s College LondonLondonUK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS FoundationLondonUK
| | - T. K. Karikari
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - J. Simrén
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - L. Montoliu‐Gaya
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - J. Pannee
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - A. O´Connor
- UK Dementia Research Institute at UCLLondonUK
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - P. S. J. Weston
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - J. Lantero‐Rodriguez
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - A. Keshavan
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - A. Snellman
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Turku PET CentreUniversity of TurkuTurkuFinland
| | - J. Gobom
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - R. W. Paterson
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - J. M. Schott
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - K. Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - N. C. Fox
- UK Dementia Research Institute at UCLLondonUK
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - H. Zetterberg
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| |
Collapse
|
9
|
Castillo-Mendieta T, Arana-Lechuga Y, Campos-Peña V, Sosa AL, Orozco-Suarez S, Pinto-Almazán R, Segura-Uribe J, Javier Rodríguez-Sánchez de Tagle A, Ruiz-Sánchez E, Guerra-Araiza C. Plasma Levels of Amyloid-β Peptides and Tau Protein in Mexican Patients with Alzheimer's Disease. J Alzheimers Dis 2021; 82:S271-S281. [PMID: 34151786 DOI: 10.3233/jad-200912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) causes memory deficit and alterations in other cognitive functions, mainly in adults over 60 years of age. As the diagnosis confirmation is performed by a postmortem neuropathological examination of the brain, this disease can be confused with other types of dementia at early stages. About 860,000 Mexicans are affected by dementia, most of them with insufficient access to adequate comprehensive health care services. Plasma biomarkers could be a rapid option for early diagnosis of the disease. OBJECTIVE This study aimed to analyze some plasma biomarkers (amyloid-β, tau, and lipids) in Mexican AD patients and control subjects with no associated neurodegenerative diseases. METHODS Plasma amyloid-β peptides (Aβ40 and Aβ42), total and phosphorylated tau protein (T-tau and P-tau), and cholesterol and triglyceride levels were quantified by enzyme-linked immunosorbent assay in AD patients and control subjects. RESULTS In Mexican AD patients, we found significantly lower levels of Aβ42 (p < 0.05) compared to the control group. In contrast, significantly higher levels of P-tau (p < 0.05) and triglycerides (p < 0.05) were observed in AD patients compared to controls. Furthermore, a significant correlation was found between the severity of dementia and plasma P-tau levels, Aβ42/Aβ40 and P-tau/T-tau ratios, and triglycerides concentrations. This correlation increased gradually with cognitive decline. CONCLUSION The detection of these plasma biomarkers is an initial step in searching for a timely, less invasive, and cost-efficient diagnosis in Mexicans.
Collapse
Affiliation(s)
- Tzayaka Castillo-Mendieta
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico.,Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Yoaly Arana-Lechuga
- Sleep Disorders Clinic, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Victoria Campos-Peña
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
| | - Ana Luisa Sosa
- Clínica de Demencia, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
| | - Sandra Orozco-Suarez
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Rodolfo Pinto-Almazán
- Laboratorio de Biología Molecular en Enfermedades Metabólicas y Neurodegenerativas, Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, Ixtapaluca, State of Mexico, Mexico
| | - Julia Segura-Uribe
- Subdirección de Gestión de la Investigación, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Mexico City, Mexico
| | - Aldo Javier Rodríguez-Sánchez de Tagle
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico.,Coordinación de QFBT, Universidad del Valle de México-Chapultepec, México City, México
| | - Elizabeth Ruiz-Sánchez
- Laboratorio de Neurotoxicología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
| | - Christian Guerra-Araiza
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| |
Collapse
|
10
|
Amponsah AE, Guo R, Kong D, Feng B, He J, Zhang W, Liu X, Du X, Ma Z, Liu B, Ma J, Cui H. Patient-derived iPSCs, a reliable in vitro model for the investigation of Alzheimer's disease. Rev Neurosci 2021; 32:379-402. [PMID: 33550785 DOI: 10.1515/revneuro-2020-0065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 11/07/2020] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease and a common cause of dementia among elderly individuals. The disease is characterized by progressive cognitive decline, accumulation of senile amyloid plaques and neurofibrillary tangles, oxidative stress, and inflammation. Human-derived cell models of AD are scarce, and over the years, non-human-derived models have been developed to recapitulate clinical AD, investigate the disease's pathogenesis and develop therapies for the disease. Several pharmacological compounds have been developed for AD based on findings from non-human-derived cell models; however, these pharmacological compounds have failed at different phases of clinical trials. This necessitates the application of human-derived cell models, such as induced pluripotent stem cells (iPSCs) in their optimized form in AD mechanistic studies and preclinical drug testing. This review provides an overview of AD and iPSCs. The AD-relevant phenotypes of iPSC-derived AD brain cells and the usefulness of iPSCs in AD are highlighted. Finally, the various recommendations that have been made to enhance iPSC/AD modelling are discussed.
Collapse
Affiliation(s)
- Asiamah Ernest Amponsah
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Ruiyun Guo
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Desheng Kong
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Baofeng Feng
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Jingjing He
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Wei Zhang
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Xin Liu
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Xiaofeng Du
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Zhenhuan Ma
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Boxin Liu
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Jun Ma
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China.,Human Anatomy Department, Hebei Medical University, Shijiazhuang, Hebei Province050017, China
| | - Huixian Cui
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China.,Human Anatomy Department, Hebei Medical University, Shijiazhuang, Hebei Province050017, China
| |
Collapse
|
11
|
Rehiman SH, Lim SM, Neoh CF, Majeed ABA, Chin AV, Tan MP, Kamaruzzaman SB, Ramasamy K. Proteomics as a reliable approach for discovery of blood-based Alzheimer's disease biomarkers: A systematic review and meta-analysis. Ageing Res Rev 2020; 60:101066. [PMID: 32294542 DOI: 10.1016/j.arr.2020.101066] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/03/2020] [Accepted: 04/03/2020] [Indexed: 02/08/2023]
Abstract
In order to gauge the impact of proteomics in discovery of Alzheimer's disease (AD) blood-based biomarkers, this study had systematically reviewed articles published between 1984-2019. Articles that fulfilled the inclusion criteria were assessed for risk of bias. A meta-analysis was performed for replicable candidate biomarkers (CB). Of the 1651 articles that were identified, 17 case-control and two cohort studies, as well as three combined case-control and longitudinal designs were shortlisted. A total of 207 AD and mild cognitive impairment (MCI) CB were discovered, with 48 reported in >2 studies. This review highlights six CB, namely alpha-2-macroglobulin (α2M)ps, pancreatic polypeptide (PP)ps, apolipoprotein A-1 (ApoA-1)ps, afaminp, insulin growth factor binding protein-2 (IGFBP-2)ps and fibrinogen-γ-chainp, all of which exhibited consistent pattern of regulation in >three independent cohorts. They are involved in AD pathogenesis via amyloid-beta (Aβ), neurofibrillary tangles, diabetes and cardiovascular diseases (CVD). Meta-analysis indicated that ApoA-1ps was significantly downregulated in AD (SMD = -1.52, 95% CI: -1.89, -1.16, p < 0.00001), with low inter-study heterogeneity (I2 = 0%, p = 0.59). α2Mps was significantly upregulated in AD (SMD = 0.83, 95% CI: 0.05, 1.62, p = 0.04), with moderate inter-study heterogeneity (I2 = 41%, p = 0.19). Both CB are involved in Aβ formation. These findings provide important insights into blood-based AD biomarkers discovery via proteomics.
Collapse
|
12
|
Wongchitrat P, Pakpian N, Kitidee K, Phopin K, Dharmasaroja PA, Govitrapong P. Alterations in the Expression of Amyloid Precursor Protein Cleaving Enzymes mRNA in Alzheimer Peripheral Blood. Curr Alzheimer Res 2020; 16:29-38. [PMID: 30411686 DOI: 10.2174/1567205015666181109103742] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/19/2018] [Accepted: 11/01/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common cause of dementia in elderly populations. Changes in the expression of the Amyloid Precursor Protein (APP)-cleaving enzymes directly affect the formation of Amyloid Beta (Aβ) plaques, a neuropathological hallmark of AD. OBJECTIVE We used peripheral blood from AD patients to investigate the expression of genes related to APP-processing [(β-site APP-cleaving enzyme 1 (BACE1), presenilin1 (PSEN1), and a disintegrin and metalloproteinase family 10 (ADAM10) and 17 (ADAM17)] and the epigenetic genes sirtuin (SIRT)1-3, which regulate Aβ production. METHOD Real-time polymerase chain reactions were performed to determine the specific mRNA levels in plasma. The mRNA levels in AD patients were compared to those in healthy persons and assessed in relation to the subjects' cognitive performance. RESULTS BACE1 mRNA level in AD subjects was significantly higher than those of healthy controls, whereas ADAM10 level was significantly lower in the AD subjects. The SIRT1 level was significantly decreased, while that of SIRT2 was increased in AD subjects and elderly controls compared to levels in healthy young control. In addition, correlations were found between the expression levels of BACE1, ADAM10 and SIRT1 and cognitive performance scores. Total Aβ (Aβ40+Aβ42) levels and the Aβ40/Aβ42 ratio were significantly increased in the AD subjects, whereas decrease in plasma Aβ42 was found in AD subjects. There was a negative correlation between Aβ40 or total Aβ and Thai Mental State Examination (TMSE) while there was no correlation between Aβ40/Aβ42 ratio or Aβ42 and TMSE. CONCLUSION The present findings provide evidence and support for the potential roles of these enzymes that drive Aβ synthesis and for epigenetic regulation in AD progression and development, which can possibly be considered peripheral markers of AD.
Collapse
Affiliation(s)
- Prapimpun Wongchitrat
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakon Pathom, Thailand
| | - Nattaporn Pakpian
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakon Pathom, Thailand
| | - Kuntida Kitidee
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakon Pathom, Thailand
| | - Kamonrat Phopin
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakon Pathom, Thailand
| | - Pornpatr A Dharmasaroja
- Stroke and Neurodegenerative Diseases Research Unit, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Piyarat Govitrapong
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakon Pathom, Thailand.,Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, Thailand
| |
Collapse
|
13
|
Medical Histories of Control Subjects Influence the Biomarker Potential of Plasma Aβ in Alzheimer's Disease: a Meta-analysis. J Mol Neurosci 2020; 70:861-870. [PMID: 32125624 DOI: 10.1007/s12031-020-01510-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/19/2020] [Indexed: 12/19/2022]
Abstract
Whether blood amyloid-β (Aβ) could be a peripheral biomarker of Alzheimer's disease (AD) remains in dispute. In the present study, we conducted a meta-analysis with 19 citations searched from Embase, PubMed, and the Cochrane Library database. Weighted mean difference (WMD) with 95% confidence intervals (CIs) was used to estimate the effect size. We firstly analyzed the plasma Aβ40, Aβ42, and Aβ42/Aβ40 ratio in AD and control group subjects. However, only a lower level of plasma Aβ42 was figured out in AD group subjects with weak statistical significance (WMD 1.82; 95% CI 0.59, 3.06; P = 0.004; I2 = 84%). We considered that the medical histories of control subjects could influence the biomarker ability of plasma Aβ. Therefore, subgroup analyses were then carried out based on a new recruiting criterion for control subjects, defining as no afflictions of any Aβ-related diseases. Surprisingly, AD group subjects showed a significant decrease in plasma Aβ42/Aβ40 ratio with low heterogeneity among studies (WMD 0.02; 95% CI 0.02, 0.02; P < 0.00001; I2 = 0%). Moreover, not only the Aβ42/Aβ40 ratio but also Aβ42 and Aβ40 were indifferent between AD and pseudo-control subjects which might be afflicted with Aβ-related diseases. This meta-analysis demonstrated that medical histories of control subjects were interference factors impeding plasma Aβ to be a biomarker of AD.
Collapse
|
14
|
Chatterjee P, Elmi M, Goozee K, Shah T, Sohrabi HR, Dias CB, Pedrini S, Shen K, Asih PR, Dave P, Taddei K, Vanderstichele H, Zetterberg H, Blennow K, Martins RN. Ultrasensitive Detection of Plasma Amyloid-β as a Biomarker for Cognitively Normal Elderly Individuals at Risk of Alzheimer’s Disease. J Alzheimers Dis 2019; 71:775-783. [DOI: 10.3233/jad-190533] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Pratishtha Chatterjee
- Department of Biomedical Sciences, Macquarie University, North Ryde, NSW, Australia
- School of Medical Health and Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Mitra Elmi
- Department of Biomedical Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Kathryn Goozee
- Department of Biomedical Sciences, Macquarie University, North Ryde, NSW, Australia
- School of Medical Health and Sciences, Edith Cowan University, Joondalup, WA, Australia
- KaRa Institute of Neurological Disease, Sydney, Macquarie Park, Australia
- Anglicare, Sydney, Castle Hill, NSW, Australia
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, WA, Australia
| | - Tejal Shah
- Department of Biomedical Sciences, Macquarie University, North Ryde, NSW, Australia
- School of Medical Health and Sciences, Edith Cowan University, Joondalup, WA, Australia
- Australian Alzheimer’s Research Foundation, Nedlands, WA, Australia
| | - Hamid R. Sohrabi
- Department of Biomedical Sciences, Macquarie University, North Ryde, NSW, Australia
- School of Medical Health and Sciences, Edith Cowan University, Joondalup, WA, Australia
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, WA, Australia
- Australian Alzheimer’s Research Foundation, Nedlands, WA, Australia
| | - Cintia B. Dias
- Department of Biomedical Sciences, Macquarie University, North Ryde, NSW, Australia
- School of Biomedical Sciences & Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Steve Pedrini
- School of Medical Health and Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Kaikai Shen
- Australian eHealth Research Centre, CSIRO, Floreat, Australia
| | - Prita R. Asih
- Department of Biomedical Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Preeti Dave
- Department of Biomedical Sciences, Macquarie University, North Ryde, NSW, Australia
- Anglicare, Sydney, Castle Hill, NSW, Australia
- John Curtin School of Medical research, Canberra, Australia
| | - Kevin Taddei
- School of Medical Health and Sciences, Edith Cowan University, Joondalup, WA, Australia
- Australian Alzheimer’s Research Foundation, Nedlands, WA, Australia
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ralph N. Martins
- Department of Biomedical Sciences, Macquarie University, North Ryde, NSW, Australia
- School of Medical Health and Sciences, Edith Cowan University, Joondalup, WA, Australia
- KaRa Institute of Neurological Disease, Sydney, Macquarie Park, Australia
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, WA, Australia
- Australian Alzheimer’s Research Foundation, Nedlands, WA, Australia
| |
Collapse
|
15
|
Kim Y, Yoo YK, Kim HY, Roh JH, Kim J, Baek S, Lee JC, Kim HJ, Chae MS, Jeong D, Park D, Lee S, Jang H, Kim K, Lee JH, Byun BH, Park SY, Ha JH, Lee KC, Cho WW, Kim JS, Koh JY, Lim SM, Hwang KS. Comparative analyses of plasma amyloid-β levels in heterogeneous and monomerized states by interdigitated microelectrode sensor system. SCIENCE ADVANCES 2019; 5:eaav1388. [PMID: 31001580 PMCID: PMC6469948 DOI: 10.1126/sciadv.aav1388] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 02/25/2019] [Indexed: 05/31/2023]
Abstract
Detection of amyloid-β (Aβ) aggregates contributes to the diagnosis of Alzheimer disease (AD). Plasma Aβ is deemed a less invasive and more accessible hallmark of AD, as Aβ can penetrate blood-brain barriers. However, correlations between biofluidic Aβ concentrations and AD progression has been tenuous. Here, we introduce a diagnostic technique that compares the heterogeneous and the monomerized states of Aβ in plasma. We used a small molecule, EPPS [4-(2-hydroxyethyl)-1-piperazinepropanesulfonic acid], to dissociate aggregated Aβ into monomers to enhance quantification accuracy. Subsequently, Aβ levels of EPPS-treated plasma were compared to those of untreated samples to minimize inter- and intraindividual variations. The interdigitated microelectrode sensor system was used to measure plasma Aβ levels on a scale of 0.1 pg/ml. The implementation of this self-standard blood test resulted in substantial distinctions between patients with AD and individuals with normal cognition (NC), with selectivity and sensitivity over 90%.
Collapse
Affiliation(s)
- YoungSoo Kim
- Integrated Science and Engineering Division, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Department of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Yong Kyoung Yoo
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Electrical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| | - Hye Yun Kim
- Department of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Jee Hoon Roh
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Jinsik Kim
- Department of Medical Biotechnology, Dongguk University, 30 Pildong-ro 1-gil, Jung-gu, Seoul 04620, Republic of Korea
| | - Seungyeop Baek
- Department of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro Seodaemun-gu, Seoul 03722, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Jinny Claire Lee
- Integrated Science and Engineering Division, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Department of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Hye Jin Kim
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Myung-Sic Chae
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Dahye Jeong
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Dongsung Park
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Sejin Lee
- Department of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - HoChung Jang
- Department of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Kyeonghwan Kim
- Department of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Jeong Hoon Lee
- Department of Electrical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| | - Byung Hyun Byun
- Department of Nuclear Medicine, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Su Yeon Park
- Department of Neurology of Korea Cancer Center Hospital, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Jeong Ho Ha
- Department of Neurology of Korea Cancer Center Hospital, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Kyo Chul Lee
- Division of RI-Convergence Research, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Won Woo Cho
- Cantis, Sangnok-gu, Ansan-si, Gyeonggi-do 15588, Republic of Korea
| | - Jae-Seung Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Jae-Young Koh
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Sang Moo Lim
- Department of Neurology of Korea Cancer Center Hospital, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Kyo Seon Hwang
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| |
Collapse
|
16
|
Next-generation biomarker discovery in Alzheimer's disease using metabolomics - from animal to human studies. Bioanalysis 2018; 10:1525-1546. [PMID: 30198770 DOI: 10.4155/bio-2018-0135] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is a complex disease driven mainly by neuronal loss due to accumulation of intracellular neurofibrillary tangles and amyloid β aggregates in the brain. The diagnosis of AD currently relies on clinical symptoms while the disease can only be confirmed at autopsy. The few available biomarkers allowing for diagnosis are typically detected many years after the onset of the disease. New diagnostic approaches, particularly in easily-accessible biofluids, are essential. By providing an exhaustive information of the phenotype, metabolomics is an ideal approach for identification of new biomarkers. This review investigates the current position of metabolomics in the field of AD research, focusing on animal and human studies, and discusses the improvements carried out over the past decade.
Collapse
|
17
|
Yao F, Hong X, Li S, Zhang Y, Zhao Q, Du W, Wang Y, Ni J. Urine-Based Biomarkers for Alzheimer’s Disease Identified Through Coupling Computational and Experimental Methods. J Alzheimers Dis 2018; 65:421-431. [DOI: 10.3233/jad-180261] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Fang Yao
- College of Life Science and Oceanography, Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Shenzhen University, Shenzhen, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China
| | - Xiaoyu Hong
- College of Life Science and Oceanography, Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Shenzhen University, Shenzhen, China
| | - Shuiming Li
- College of Life Science and Oceanography, Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Shenzhen University, Shenzhen, China
| | - Yan Zhang
- College of Life Science and Oceanography, Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Shenzhen University, Shenzhen, China
| | - Qing Zhao
- Department of Neurology, China-Japan Union Hospital, Changchun, China
| | - Wei Du
- College of Computer Science and Technology, Jilin University, Changchun, China
| | - Yong Wang
- College of Life Science and Oceanography, Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Shenzhen University, Shenzhen, China
| | - Jiazuan Ni
- College of Life Science and Oceanography, Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Shenzhen University, Shenzhen, China
| |
Collapse
|
18
|
Misfolded Protein Linked Strategies Toward Biomarker Development for Neurodegenerative Diseases. Mol Neurobiol 2018; 56:2559-2578. [DOI: 10.1007/s12035-018-1232-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/10/2018] [Indexed: 12/14/2022]
|
19
|
Peripheral Biomarkers for Early Detection of Alzheimer's and Parkinson's Diseases. Mol Neurobiol 2018; 56:2256-2277. [PMID: 30008073 DOI: 10.1007/s12035-018-1151-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 05/24/2018] [Indexed: 01/18/2023]
Abstract
Neurological disorders are found to be influencing the peripheral tissues outside CNS. Recent developments in biomarkers for CNS have emerged with various diagnostic and therapeutic shortcomings. The role of central biomarkers including CSF-based and molecular imaging-based probes are still unclear for early diagnosis of major neurological diseases. Current trends show that early detection of neurodegenerative diseases with non-invasive methods is a major focus of researchers, and the development of biomarkers aiming peripheral tissues is in demand. Alzheimer's and Parkinson's diseases are known for the progressive loss in neural structures or functions, including the neural death. Various dysfunctions of metabolic and biochemical pathways are associated with early occurrence of neuro-disorders in peripheral tissues including skin, blood cells, and eyes. This article reviews the peripheral biomarkers explored for early detection of Alzheimer's and Parkinson's diseases including blood cells, skin fibroblast, proteomics, saliva, olfactory, stomach and colon, heart and peripheral nervous system, and others. Graphical Abstract.
Collapse
|
20
|
Crane A, Brubaker WD, Johansson JU, Trigunaite A, Ceballos J, Bradt B, Glavis-Bloom C, Wallace TL, Tenner AJ, Rogers J. Peripheral complement interactions with amyloid β peptide in Alzheimer's disease: 2. Relationship to amyloid β immunotherapy. Alzheimers Dement 2018; 14:243-252. [PMID: 28755839 PMCID: PMC5881571 DOI: 10.1016/j.jalz.2017.04.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/27/2017] [Accepted: 04/30/2017] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Our previous studies have shown that amyloid β peptide (Aβ) is subject to complement-mediated clearance from the peripheral circulation, and that this mechanism is deficient in Alzheimer's disease. The mechanism should be enhanced by Aβ antibodies that form immune complexes (ICs) with Aβ, and therefore may be relevant to current Aβ immunotherapy approaches. METHODS Multidisciplinary methods were employed to demonstrate enhanced complement-mediated capture of Aβ antibody immune complexes compared with Aβ alone in both erythrocytes and THP1-derived macrophages. RESULTS Aβ antibodies dramatically increased complement activation and opsonization of Aβ, followed by commensurately enhanced Aβ capture by human erythrocytes and macrophages. These in vitro findings were consistent with enhanced peripheral clearance of intravenously administered Aβ antibody immune complexes in nonhuman primates. DISCUSSION Together with our previous results, showing significant Alzheimer's disease deficits in peripheral Aβ clearance, the present findings strongly suggest that peripheral mechanisms should not be ignored as contributors to the effects of Aβ immunotherapy.
Collapse
Affiliation(s)
- Andrés Crane
- Biosciences Division, SRI International, Menlo Park, CA, USA
| | | | | | | | | | - Bonnie Bradt
- Biosciences Division, SRI International, Menlo Park, CA, USA
| | | | - Tanya L Wallace
- Biosciences Division, SRI International, Menlo Park, CA, USA
| | - Andrea J Tenner
- Departments of Molecular Biology and Biochemistry and Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
| | - Joseph Rogers
- Biosciences Division, SRI International, Menlo Park, CA, USA.
| |
Collapse
|
21
|
Peripheral complement interactions with amyloid β peptide: Erythrocyte clearance mechanisms. Alzheimers Dement 2017; 13:1397-1409. [PMID: 28475854 DOI: 10.1016/j.jalz.2017.03.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 02/28/2017] [Accepted: 03/27/2017] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Although amyloid β peptide (Aβ) is cleared from the brain to cerebrospinal fluid and the peripheral circulation, mechanisms for its removal from blood remain unresolved. Primates have uniquely evolved a highly effective peripheral clearance mechanism for pathogens, immune adherence, in which erythrocyte complement receptor 1 (CR1) plays a major role. METHODS Multidisciplinary methods were used to demonstrate immune adherence capture of Aβ by erythrocytes and its deficiency in Alzheimer's disease (AD). RESULTS Aβ was shown to be subject to immune adherence at every step in the pathway. Aβ dose-dependently activated serum complement. Complement-opsonized Aβ was captured by erythrocytes via CR1. Erythrocytes, Aβ, and hepatic Kupffer cells were colocalized in the human liver. Significant deficits in erythrocyte Aβ levels were found in AD and mild cognitive impairment patients. DISCUSSION CR1 polymorphisms elevate AD risk, and >80% of human CR1 is vested in erythrocytes to subserve immune adherence. The present results suggest that this pathway is pathophysiologically relevant in AD.
Collapse
|
22
|
Lerche S, Brockmann K, Pilotto A, Wurster I, Sünkel U, Hobert MA, von Thaler AK, Schulte C, Stoops E, Vanderstichele H, Herbst V, Brix B, Eschweiler GW, Metzger FG, Maetzler W, Berg D. Prospective longitudinal course of cognition in older subjects with mild parkinsonian signs. ALZHEIMERS RESEARCH & THERAPY 2016; 8:42. [PMID: 27724983 PMCID: PMC5057460 DOI: 10.1186/s13195-016-0209-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/07/2016] [Indexed: 11/10/2022]
Abstract
Background Mild parkinsonian signs (MPS) are common in older people and are associated with an increased risk of different neurodegenerative diseases. This study prospectively evaluates the longitudinal course of cognitive performance in older individuals with MPS. Methods From the TREND study, 480 individuals neurologically healthy at baseline, aged between 50 and 80 years, with complete follow-up data for three assessments within a mean of 43.8 months, were included in this analysis. Participants underwent a detailed cognitive test battery, evaluation of prodromal markers for neurodegenerative diseases and history of vascular diseases at each study visit. In addition, plasma levels of amyloid-beta (Aβ)1–40 and Aβ1–42 were evaluated longitudinally. Results In 52 (11 %) of the 480 participants, MPS could be detected at baseline. These individuals had cognitive deficits significantly more often compared with controls at each time point and their cognitive performance showed a steeper decline during follow-up. In addition, their levels of plasma Aβ1–42 were significantly lower than those of controls, and declined more rapidly over time. Conclusions This longitudinal study shows that MPS are associated with cognitive decline and decrease in plasma Aβ1–42, possibly indicating an ongoing neurodegenerative process. Electronic supplementary material The online version of this article (doi:10.1186/s13195-016-0209-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stefanie Lerche
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Hoppe Seyler-Strasse 3, Tuebingen, 72076, Germany. .,German Center for Neurodegenerative Diseases, University of Tuebingen, Otfried-Müller-Straße 27, Tuebingen, 72076, Germany.
| | - Kathrin Brockmann
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Hoppe Seyler-Strasse 3, Tuebingen, 72076, Germany.,German Center for Neurodegenerative Diseases, University of Tuebingen, Otfried-Müller-Straße 27, Tuebingen, 72076, Germany
| | - Andrea Pilotto
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Hoppe Seyler-Strasse 3, Tuebingen, 72076, Germany.,Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Isabel Wurster
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Hoppe Seyler-Strasse 3, Tuebingen, 72076, Germany.,German Center for Neurodegenerative Diseases, University of Tuebingen, Otfried-Müller-Straße 27, Tuebingen, 72076, Germany
| | - Ulrike Sünkel
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Hoppe Seyler-Strasse 3, Tuebingen, 72076, Germany.,German Center for Neurodegenerative Diseases, University of Tuebingen, Otfried-Müller-Straße 27, Tuebingen, 72076, Germany
| | - Markus A Hobert
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Hoppe Seyler-Strasse 3, Tuebingen, 72076, Germany.,German Center for Neurodegenerative Diseases, University of Tuebingen, Otfried-Müller-Straße 27, Tuebingen, 72076, Germany
| | - Anna-Katharina von Thaler
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Hoppe Seyler-Strasse 3, Tuebingen, 72076, Germany.,German Center for Neurodegenerative Diseases, University of Tuebingen, Otfried-Müller-Straße 27, Tuebingen, 72076, Germany
| | - Claudia Schulte
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Hoppe Seyler-Strasse 3, Tuebingen, 72076, Germany.,German Center for Neurodegenerative Diseases, University of Tuebingen, Otfried-Müller-Straße 27, Tuebingen, 72076, Germany
| | | | | | - Victor Herbst
- EUROIMMUN Medizinische Labordiagnostika AG, Lübeck, Germany
| | - Britta Brix
- EUROIMMUN Medizinische Labordiagnostika AG, Lübeck, Germany
| | - Gerhard W Eschweiler
- Geriatric Center at the University Hospital of Tübingen, Tübingen, Germany.,Department of Psychiatry and Psychotherapy, University Hospital of Tübingen, Tübingen, Germany
| | - Florian G Metzger
- Geriatric Center at the University Hospital of Tübingen, Tübingen, Germany.,Department of Psychiatry and Psychotherapy, University Hospital of Tübingen, Tübingen, Germany
| | - Walter Maetzler
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Hoppe Seyler-Strasse 3, Tuebingen, 72076, Germany.,German Center for Neurodegenerative Diseases, University of Tuebingen, Otfried-Müller-Straße 27, Tuebingen, 72076, Germany
| | - Daniela Berg
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Hoppe Seyler-Strasse 3, Tuebingen, 72076, Germany.,German Center for Neurodegenerative Diseases, University of Tuebingen, Otfried-Müller-Straße 27, Tuebingen, 72076, Germany
| |
Collapse
|
23
|
Kleinschmidt M, Schoenfeld R, Göttlich C, Bittner D, Metzner JE, Leplow B, Demuth HU. Characterizing Aging, Mild Cognitive Impairment, and Dementia with Blood-Based Biomarkers and Neuropsychology. J Alzheimers Dis 2016; 50:111-26. [PMID: 26639953 DOI: 10.3233/jad-143189] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Current treatment in Alzheimer's disease (AD) is initiated at a stage where the brain already has irreversible structural deteriorations. Therefore, the concept of treatment prior to obvious cognitive deficits has become widely accepted, and simple biochemical tests to discriminate normal aging from prodromal or demented stages are now common practice. OBJECTIVE The objective of the study was the differentiation of controls, mild cognitive impairment (MCI) and AD patients by novel blood-based assays in combination with neuropsychological tests. METHODS In a cross-sectional study, 143 subjects aged 18 to 85 years were recruited. All participants were classified by a comprehensive neuropsychological assessment. Blood samples were analyzed for several amyloid-β (Aβ) species, pro-inflammatory markers, anti-Aβ autoantibodies, and ApoE allele status, respectively. RESULTS Plasma Aβ1-42 was significantly decreased in MCI and AD compared to age-matched controls, whereas Aβ1-40 did not differ, but increases with age in healthy controls. The Aβ1-42 to Aβ1-40 ratio was stepwise decreased from age-matched controls via MCI to AD, and shows a clear correlation with memory scores. Reduced Aβ1-42 and Aβ1-42 to Aβ1-40 ratio have strongly correlated with carrying ApoE ɛ4 allele. Autoantibodies against pyroglutamate-modified Aβ, but only a certain subclass, were significantly decreased in AD compared to MCI and age-matched controls, whereas autoantibodies against the unmodified N-terminus of Aβ did not differ. CONCLUSION Comprehensive sample preparation and assay standardization enable reliable usage of plasma Aβ for diagnosis of MCI and AD. Anti-pGlu-Aβ autoantibodies correlate with cognition, but not with ApoE, supporting the associated plasma Aβ analysis with additional and independent information.
Collapse
Affiliation(s)
- Martin Kleinschmidt
- Probiodrug AG, Halle (Saale), Germany.,Current address: Fraunhofer Institute of Cell Therapy and Immunology, Department of Drug Design and Target Validation, Halle (Saale), Germany
| | - Robby Schoenfeld
- Martin-Luther-University Halle-Wittenberg, Department of Psychology, Halle (Saale), Germany
| | - Claudia Göttlich
- Probiodrug AG, Halle (Saale), Germany.,Department Tissue Engineering & Regenerative Medicine (TERM), University Hospital Wuerzburg, Germany
| | - Daniel Bittner
- Clinic for Neurology, Otto-von-Guericke University Magdeburg, Germany
| | | | - Bernd Leplow
- Martin-Luther-University Halle-Wittenberg, Department of Psychology, Halle (Saale), Germany
| | - Hans-Ulrich Demuth
- Probiodrug AG, Halle (Saale), Germany.,Current address: Fraunhofer Institute of Cell Therapy and Immunology, Department of Drug Design and Target Validation, Halle (Saale), Germany
| |
Collapse
|
24
|
Martins IJ. The Role of Clinical Proteomics, Lipidomics, and Genomics in the Diagnosis of Alzheimer's Disease. Proteomes 2016; 4:proteomes4020014. [PMID: 28248224 PMCID: PMC5217345 DOI: 10.3390/proteomes4020014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/24/2016] [Accepted: 03/28/2016] [Indexed: 02/07/2023] Open
Abstract
The early diagnosis of Alzheimer’s disease (AD) has become important to the reversal and treatment of neurodegeneration, which may be relevant to premature brain aging that is associated with chronic disease progression. Clinical proteomics allows the detection of various proteins in fluids such as the urine, plasma, and cerebrospinal fluid for the diagnosis of AD. Interest in lipidomics has accelerated with plasma testing for various lipid biomarkers that may with clinical proteomics provide a more reproducible diagnosis for early brain aging that is connected to other chronic diseases. The combination of proteomics with lipidomics may decrease the biological variability between studies and provide reproducible results that detect a community’s susceptibility to AD. The diagnosis of chronic disease associated with AD that now involves genomics may provide increased sensitivity to avoid inadvertent errors related to plasma versus cerebrospinal fluid testing by proteomics and lipidomics that identify new disease biomarkers in body fluids, cells, and tissues. The diagnosis of AD by various plasma biomarkers with clinical proteomics may now require the involvement of lipidomics and genomics to provide interpretation of proteomic results from various laboratories around the world.
Collapse
Affiliation(s)
- Ian James Martins
- School of Medical Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup 6027, Australia.
| |
Collapse
|
25
|
Shanthi KB, Krishnan S, Rani P. A systematic review and meta-analysis of plasma amyloid 1-42 and tau as biomarkers for Alzheimer's disease. SAGE Open Med 2015; 3:2050312115598250. [PMID: 26770797 PMCID: PMC4679337 DOI: 10.1177/2050312115598250] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 06/27/2015] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Amyloid 1-42 (Aβ42) and tau in cerebrospinal fluid are currently used as markers for diagnosis of Alzheimer's disease. Conflicting reports exist regarding their plasma levels in Alzheimer's disease patients. A meta-analysis was performed to statistically validate the use of plasma Aβ42 and tau as biomarkers for Alzheimer's disease. METHODS Different databases were searched using the search key: (amyloid OR amyloid1-42 OR Aβ42) AND (tau OR total tau) AND plasma AND (alzheimer's OR alzheimer's disease), and for databases not accepting boolean search, records were retrieved using the search key: plasma + amyloid + tau + alzheimer's. A total of 1880 articles for Aβ42 and 1508 articles for tau were shortlisted. The abstracts were screened, and 69 articles reporting plasma Aβ42 levels and 6 articles reporting plasma tau were identified. After exclusion, 25 studies reporting plasma Aβ42 and 6 studies reporting total tau were analysed in Review Manager version 5.2 using weighted mean difference method, and the bias between studies was assessed using the funnel plot. RESULTS Plasma Aβ42 and tau did not vary significantly between Alzheimer's disease patients and controls. The funnel plot showed that there was no bias between studies for Aβ42, while possible bias existed for tau due to availability of limited studies. CONCLUSION This analysis pinpoints that plasma Aβ42 and tau could not serve as reliable markers independently for diagnosis of Alzheimer's disease and a cohort study with age, sex and apolipoprotein E correction is warranted for their possible use as Alzheimer's disease markers.
Collapse
Affiliation(s)
| | - Sreeram Krishnan
- Department of Biotechnology, PSG College of Technology, Coimbatore, India
| | - P Rani
- Department of Biotechnology, PSG College of Technology, Coimbatore, India
| |
Collapse
|
26
|
Galozzi S, Marcus K, Barkovits K. Amyloid-β as a biomarker for Alzheimer’s disease: quantification methods in body fluids. Expert Rev Proteomics 2015; 12:343-54. [DOI: 10.1586/14789450.2015.1065183] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
27
|
Hallé M, Tribout-Jover P, Lanteigne AM, Boulais J, St-Jean JR, Jodoin R, Girouard MP, Constantin F, Migneault A, Renaud F, Didierlaurent AM, Mallett CP, Burkhart D, Pilorget A, Palmantier R, Larocque D. Methods to monitor monocytes-mediated amyloid-beta uptake and phagocytosis in the context of adjuvanted immunotherapies. J Immunol Methods 2015; 424:64-79. [PMID: 26002154 DOI: 10.1016/j.jim.2015.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 04/11/2015] [Accepted: 05/06/2015] [Indexed: 12/14/2022]
Abstract
Antibody-mediated capture of amyloid-beta (Aβ) in peripheral blood was identified as an attractive strategy to eliminate cerebral toxic amyloid in Alzheimer's disease (AD) patients and murine models. Alternatively, defective capacity of peripheral monocytes to engulf Aβ was reported in individuals with AD. In this report, we developed different approaches to investigate cellular uptake and phagocytosis of Aβ, and to examine how two immunological devices--an immunostimulatory Adjuvant System and different amyloid specific antibodies--may affect these biological events. Between one and thirteen months of age, APPswe X PS1.M146V (TASTPM) AD model mice had decreasing concentrations of Aβ in their plasma. In contrast, the proportion of blood monocytes containing Aβ tended to increase with age. Importantly, the TLR-agonist containing Adjuvant System AS01B primed monocytes to promote de novo Aβ uptake capacity, particularly in the presence of anti-Aβ antibodies. Biochemical experiments demonstrated that cells achieved Aβ uptake and internalization followed by Aβ degradation via mechanisms that required effective actin polymerization and proteolytic enzymes such as insulin-degrading enzyme. We further demonstrated that both Aβ-specific monoclonal antibodies and plasma from Aβ-immunized mice enhanced the phagocytosis of 1 μm Aβ-coated particles. Together, our data highlight a new biomarker testing to follow amyloid clearance within the blood and a mechanism of Aβ uptake by peripheral monocytes in the context of active or passive immunization, and emphasize on novel approaches to investigate this phenomenon.
Collapse
Affiliation(s)
- Maxime Hallé
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8; Neuroscience Laboratory, 2705, Boulevard Laurier, T-2-50, Department of Molecular Medicine, Centre Hospitalier Universitaire de Québec Research Center, Université Laval, Quebec City, Quebec, Canada, G1V 4G2
| | | | | | - Jonathan Boulais
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - Julien R St-Jean
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - Rachel Jodoin
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | | | - Florin Constantin
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - Annik Migneault
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - Frédéric Renaud
- GSK Vaccines, Rue de l'Institut 89, B-1330 Rixensart, Belgium
| | | | - Corey P Mallett
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - David Burkhart
- GSK Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA
| | - Anthony Pilorget
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - Rémi Palmantier
- GSK Vaccines, Rue de l'Institut 89, B-1330 Rixensart, Belgium
| | - Daniel Larocque
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8.
| |
Collapse
|
28
|
Ishii R, Tokuda T, Tatebe H, Ohmichi T, Kasai T, Nakagawa M, Mizuno T, El-Agnaf OMA. Decrease in plasma levels of α-synuclein is evident in patients with Parkinson's disease after elimination of heterophilic antibody interference. PLoS One 2015; 10:e0123162. [PMID: 25849645 PMCID: PMC4388641 DOI: 10.1371/journal.pone.0123162] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 02/18/2015] [Indexed: 12/15/2022] Open
Abstract
There is substantial biochemical, pathological, and genetic evidence that α-synuclein (A-syn) is a principal molecule in the pathogenesis of Parkinson disease (PD). We previously reported that total A-syn levels in cerebrospinal fluid (CSF), measured with the specific enzyme-linked immunosorbent assay (ELISA) developed by ourselves, were decreased in patients with PD, and suggested the usefulness of A-syn in CSF and plasma as a biomarker for the diagnosis of PD. After our report, a considerable number of studies have investigated the levels A-syn in CSF and in blood, but have reported inconclusive results. Such discrepancies have often been attributed not only to the use of different antibodies in the ELISAs but also to interference from hemolysis. In this study we measured the levels of A-syn in CSF and plasma by using our own sandwich ELISA with or without heterophilic antibody (HA) inhibitor in 30 patients with PD and 58 age-matched controls. We thereby revealed that HA interfered with ELISA measurements of A-syn and are accordingly considered to be an important confounder in A-syn ELISAs. HA produced falsely exaggerated signals in A-syn ELISAs more prominently in plasma samples than in CSF samples. After elimination of HA interference, it was found that hemolysis did not have a significant effect on the signals obtained using our A-syn ELISA. Furthermore, plasma levels of A-syn were significantly lower in the PD group compared with the control group following elimination of HA interference with an HA inhibitor. Our results demonstrate that HA was a major confounder that should be controlled in A-syn ELISAs, and that plasma A-syn could be a useful biomarker for the diagnosis of PD if adequately quantified following elimination of HA interference.
Collapse
Affiliation(s)
- Ryotaro Ishii
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takahiko Tokuda
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Molecular Pathobiology of Brain Diseases, Kyoto Prefectural University of Medicine, Kyoto, Japan
- * E-mail:
| | - Harutsugu Tatebe
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Zaitaku (Homecare), Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Medical Education and Primary Care, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takuma Ohmichi
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takashi Kasai
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masanori Nakagawa
- North Medical Center, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshiki Mizuno
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Omar M. A. El-Agnaf
- College of Science, Engineering and Technology, HBKU, Education City, Qatar Foundation, Doha, Qatar
| |
Collapse
|
29
|
Zhao Z, Zhu L, Bu X, Ma H, Yang S, Yang Y, Hu Z. Label-free detection of Alzheimer’s disease through the ADP3 peptoid recognizing the serum amyloid-beta42 peptide. Chem Commun (Camb) 2015; 51:718-21. [DOI: 10.1039/c4cc07037b] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Surface plasmon resonance imaging in combination with the ADP3 peptoid was used to identify Alzheimer’s disease through detecting amyloid-beta42 in the serum.
Collapse
Affiliation(s)
- Zijian Zhao
- National Center for Nanoscience and Technology
- Beijing
- P.R. China
| | - Ling Zhu
- National Center for Nanoscience and Technology
- Beijing
- P.R. China
| | - Xiangli Bu
- National Center for Nanoscience and Technology
- Beijing
- P.R. China
| | - Huailei Ma
- National Center for Nanoscience and Technology
- Beijing
- P.R. China
| | - Shu Yang
- National Center for Nanoscience and Technology
- Beijing
- P.R. China
| | - Yanlian Yang
- National Center for Nanoscience and Technology
- Beijing
- P.R. China
| | - Zhiyuan Hu
- National Center for Nanoscience and Technology
- Beijing
- P.R. China
| |
Collapse
|
30
|
Correlations of amyloid-β concentrations between CSF and plasma in acute Alzheimer mouse model. Sci Rep 2014; 4:6777. [PMID: 25345439 PMCID: PMC4209448 DOI: 10.1038/srep06777] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 10/07/2014] [Indexed: 12/27/2022] Open
Abstract
Amyloid-β (Aβ) is one of the few neuropathological biomarkers associated with transporters of the blood-brain barrier (BBB). Despite the well-characterized clinical indication of decreasing Aβ levels in the cerebrospinal fluid (CSF) during the development of Alzheimer's disease (AD), the link between the alternation of Aβ level in the blood and the progress of the disorder is still controversial. Here, we report a direct correlation of Aβ(1-42) levels between CSF and plasma in AD mouse model. We injected monomeric Aβ(1-42) directly into the intracerebroventricular (ICV) region of normal adult mouse brains to induce AD-like phenotypes. Using sandwich enzyme-linked immunosorbent assays, we observed proportional elevation of Aβ(1-42) levels in both CSF and plasma in a dose-dependent manner. Our findings that plasma Aβ(1-42) reflects the condition of CSF Aβ(1-42) warrant further investigation as a biomarker for the blood diagnosis of AD.
Collapse
|
31
|
Nicholson AM, Finch NA, Thomas CS, Wojtas A, Rutherford NJ, Mielke MM, Roberts RO, Boeve BF, Knopman DS, Petersen RC, Rademakers R. Progranulin protein levels are differently regulated in plasma and CSF. Neurology 2014; 82:1871-8. [PMID: 24771538 DOI: 10.1212/wnl.0000000000000445] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE We aimed to investigate the relationship between plasma and CSF progranulin (PGRN) levels. METHODS Plasma and CSF PGRN were measured in a cohort of 345 subjects from the Mayo Clinic Study of Aging by ELISA. Single nucleotide polymorphism genotyping was performed using TaqMan assays. Associations between PGRN and sex, age at sample collection, diagnosis, single nucleotide polymorphism genotypes (GRN, SORT1, and APOE), and Pittsburgh compound B score were explored separately in CSF and plasma using single variable linear regression models. Pearson partial correlation coefficient was used to estimate the correlation of PGRN in CSF and plasma. RESULTS Plasma (p = 0.0031) and CSF (p = 0.0044) PGRN significantly increased with age, whereas plasma PGRN levels were 7% lower (p = 0.0025) and CSF PGRN levels 5% higher (p = 0.0024) in male compared with female participants. Correcting for age and sex, higher plasma PGRN was associated with higher CSF PGRN (partial r = 0.17, p = 0.004). In plasma, both rs5848 (GRN; p = 0.002) and rs646776 (SORT1; p = 3.56E-7) were associated with PGRN, while only rs5848 showed highly significant association in CSF (p = 5.59E-14). Age, sex, rs5848 genotype, and plasma PGRN together accounted for only 18% of the variability observed in CSF PGRN. CONCLUSIONS While some correlation exists between plasma and CSF PGRN, age, sex, and genetic factors differently affect PGRN levels. Therefore, caution should be taken when using plasma PGRN to predict PGRN changes in the brain. These findings further highlight that plasma PGRN levels may not accurately predict clinical features or response to future frontotemporal lobar degeneration therapies.
Collapse
Affiliation(s)
- Alexandra M Nicholson
- From the Department of Neuroscience (A.M.N., N.A.F., A.W., N.J.R., R.R.) and Division of Biomedical Statistics and Informatics (C.S.T.), Mayo Clinic, Jacksonville, FL; and Division of Epidemiology, Department of Health Sciences Research (M.M.M., R.O.R.) and Department of Neurology (R.O.R., B.F.B., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - NiCole A Finch
- From the Department of Neuroscience (A.M.N., N.A.F., A.W., N.J.R., R.R.) and Division of Biomedical Statistics and Informatics (C.S.T.), Mayo Clinic, Jacksonville, FL; and Division of Epidemiology, Department of Health Sciences Research (M.M.M., R.O.R.) and Department of Neurology (R.O.R., B.F.B., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Colleen S Thomas
- From the Department of Neuroscience (A.M.N., N.A.F., A.W., N.J.R., R.R.) and Division of Biomedical Statistics and Informatics (C.S.T.), Mayo Clinic, Jacksonville, FL; and Division of Epidemiology, Department of Health Sciences Research (M.M.M., R.O.R.) and Department of Neurology (R.O.R., B.F.B., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Aleksandra Wojtas
- From the Department of Neuroscience (A.M.N., N.A.F., A.W., N.J.R., R.R.) and Division of Biomedical Statistics and Informatics (C.S.T.), Mayo Clinic, Jacksonville, FL; and Division of Epidemiology, Department of Health Sciences Research (M.M.M., R.O.R.) and Department of Neurology (R.O.R., B.F.B., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Nicola J Rutherford
- From the Department of Neuroscience (A.M.N., N.A.F., A.W., N.J.R., R.R.) and Division of Biomedical Statistics and Informatics (C.S.T.), Mayo Clinic, Jacksonville, FL; and Division of Epidemiology, Department of Health Sciences Research (M.M.M., R.O.R.) and Department of Neurology (R.O.R., B.F.B., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Michelle M Mielke
- From the Department of Neuroscience (A.M.N., N.A.F., A.W., N.J.R., R.R.) and Division of Biomedical Statistics and Informatics (C.S.T.), Mayo Clinic, Jacksonville, FL; and Division of Epidemiology, Department of Health Sciences Research (M.M.M., R.O.R.) and Department of Neurology (R.O.R., B.F.B., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Rosebud O Roberts
- From the Department of Neuroscience (A.M.N., N.A.F., A.W., N.J.R., R.R.) and Division of Biomedical Statistics and Informatics (C.S.T.), Mayo Clinic, Jacksonville, FL; and Division of Epidemiology, Department of Health Sciences Research (M.M.M., R.O.R.) and Department of Neurology (R.O.R., B.F.B., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Bradley F Boeve
- From the Department of Neuroscience (A.M.N., N.A.F., A.W., N.J.R., R.R.) and Division of Biomedical Statistics and Informatics (C.S.T.), Mayo Clinic, Jacksonville, FL; and Division of Epidemiology, Department of Health Sciences Research (M.M.M., R.O.R.) and Department of Neurology (R.O.R., B.F.B., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - David S Knopman
- From the Department of Neuroscience (A.M.N., N.A.F., A.W., N.J.R., R.R.) and Division of Biomedical Statistics and Informatics (C.S.T.), Mayo Clinic, Jacksonville, FL; and Division of Epidemiology, Department of Health Sciences Research (M.M.M., R.O.R.) and Department of Neurology (R.O.R., B.F.B., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Ronald C Petersen
- From the Department of Neuroscience (A.M.N., N.A.F., A.W., N.J.R., R.R.) and Division of Biomedical Statistics and Informatics (C.S.T.), Mayo Clinic, Jacksonville, FL; and Division of Epidemiology, Department of Health Sciences Research (M.M.M., R.O.R.) and Department of Neurology (R.O.R., B.F.B., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Rosa Rademakers
- From the Department of Neuroscience (A.M.N., N.A.F., A.W., N.J.R., R.R.) and Division of Biomedical Statistics and Informatics (C.S.T.), Mayo Clinic, Jacksonville, FL; and Division of Epidemiology, Department of Health Sciences Research (M.M.M., R.O.R.) and Department of Neurology (R.O.R., B.F.B., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN.
| |
Collapse
|
32
|
Role of copper and cholesterol association in the neurodegenerative process. Int J Alzheimers Dis 2013; 2013:414817. [PMID: 24288650 PMCID: PMC3830777 DOI: 10.1155/2013/414817] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Revised: 09/05/2013] [Accepted: 09/05/2013] [Indexed: 01/22/2023] Open
Abstract
Age is one of the main factors involved in the development of neurological illnesses, in particular, Alzheimer, and it is widely held that the rapid aging of the world population is accompanied by a rise in the prevalence and incidence of Alzheimer disease. However, evidence from recent decades indicates that Cu and Cho overload are emerging causative factors in neurodegeneration, a hypothesis that has been partially investigated in experimental models. The link between these two variables and the onset of Alzheimer disease has opened up interesting new possibilities requiring more in-depth analysis. The aim of the present study was therefore to investigate the effect of the association of Cu + Cho (CuCho) as a possible synergistic factor in the development of an Alzheimer-like pathology in Wistar rats. We measured total- and nonceruloplasmin-bound Cu and Cho (free and sterified) contents in plasma and brain zones (cortex and hippocampus), markers of oxidative stress damage, inflammation, and programmed cell death (caspase-3 and calpain isoforms). The ratio beta-amyloid (1-42)/(1-40) was determined in plasma and brain as neurodegenerative biomarker. An evaluation of visuospatial memory (Barnes maze test) was also performed. The results demonstrate the establishment of a prooxidative and proinflammatory environment after CuCho treatment, hallmarked by increased TBARS, protein carbonyls, and nitrite plus nitrate levels in plasma and brain zones (cortex and hippocampus) with a consequent increase in the activity of calpains and no significant changes in caspase-3. A simultaneous increase in the plasma Aβ1-42/Aβ1-40 ratio was found. Furthermore, a slight but noticeable change in visuospatial memory was observed in rats treated with CuCho. We conclude that our model could reflect an initial stage of neurodegeneration in which Cu and Cho interact with one another to exacerbate neurological damage.
Collapse
|
33
|
Erickson MA, Banks WA. Blood-brain barrier dysfunction as a cause and consequence of Alzheimer's disease. J Cereb Blood Flow Metab 2013; 33:1500-13. [PMID: 23921899 PMCID: PMC3790938 DOI: 10.1038/jcbfm.2013.135] [Citation(s) in RCA: 399] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 07/05/2013] [Accepted: 07/09/2013] [Indexed: 12/14/2022]
Abstract
The blood-brain barrier (BBB) plays critical roles in the maintenance of central nervous system (CNS) homeostasis. Dysfunction of the BBB occurs in a number of CNS diseases, including Alzheimer's disease (AD). A prevailing hypothesis in the AD field is the amyloid cascade hypothesis that states that amyloid-β (Aβ) deposition in the CNS initiates a cascade of molecular events that cause neurodegeneration, leading to AD onset and progression. In this review, the participation of the BBB in the amyloid cascade and in other mechanisms of AD neurodegeneration will be discussed. We will specifically focus on three aspects of BBB dysfunction: disruption, perturbation of transporters, and secretion of neurotoxic substances by the BBB. We will also discuss the interaction of the BBB with components of the neurovascular unit in relation to AD and the potential contribution of AD risk factors to aspects of BBB dysfunction. From the results discussed herein, we conclude that BBB dysfunction contributes to AD through a number of mechanisms that could be initiated in the presence or absence of Aβ pathology.
Collapse
Affiliation(s)
- Michelle A Erickson
- 1] GRECC, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA [2] Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington School of Medicine, Seattle, Washington, USA [3] Department of Pathology, School of Dental Medicine, University of Pennsylvania, Seattle, Washington, USA
| | | |
Collapse
|
34
|
Yamamoto K, Shimada H, Koh H, Ataka S, Miki T. Serum levels of albumin-amyloid beta complexes are decreased in Alzheimer's disease. Geriatr Gerontol Int 2013; 14:716-23. [DOI: 10.1111/ggi.12147] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2013] [Indexed: 11/29/2022]
Affiliation(s)
- Keiichi Yamamoto
- Department of Geriatric Medicine and Neurology; Osaka City University Graduate School of Medicine; Osaka Japan
| | - Hiroyuki Shimada
- Department of Geriatric Medicine and Neurology; Osaka City University Graduate School of Medicine; Osaka Japan
| | - Hideo Koh
- Department of Hematology; Osaka City University Graduate School of Medicine; Osaka Japan
| | - Suzuka Ataka
- Department of Geriatric Medicine and Neurology; Osaka City University Graduate School of Medicine; Osaka Japan
| | - Takami Miki
- Department of Geriatric Medicine and Neurology; Osaka City University Graduate School of Medicine; Osaka Japan
| |
Collapse
|
35
|
Rembach A, Ryan TM, Roberts BR, Doecke JD, Wilson WJ, Watt AD, Barnham KJ, Masters CL. Progress towards a consensus on biomarkers for Alzheimer’s disease: a review of peripheral analytes. Biomark Med 2013; 7:641-62. [DOI: 10.2217/bmm.13.59] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia in the elderly population and attempts to develop therapies have been unsuccessful because there is no means to target an effective therapeutic window. CNS biomarkers are insightful but impractical for high-throughput population-based screening. Therefore, a peripheral, blood-based biomarker for AD would significantly improve early diagnosis, potentially enable presymptomatic detection and facilitate effective targeting of disease-modifying treatments. The various constituents of blood, including plasma, platelets and cellular fractions, are now being systematically explored as a pool of putative peripheral biomarkers for AD. In this review we cover some less known peripheral biomarkers and highlight the latest developments for their clinical application.
Collapse
Affiliation(s)
- Alan Rembach
- The Mental Health Research Institute, The University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria, 3010, Australia.
| | - Tim M Ryan
- The Mental Health Research Institute, The University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria, 3010, Australia
| | - Blaine R Roberts
- The Mental Health Research Institute, The University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria, 3010, Australia
| | - James D Doecke
- The Australian e-Health Research Centre, Herston, Queensland, 4029, Australia
- CSIRO Preventative Health National Research Flagship, North Ryde, New South Wales, 2113, Australia
| | - William J Wilson
- CSIRO Preventative Health National Research Flagship, North Ryde, New South Wales, 2113, Australia
| | - Andrew D Watt
- The Mental Health Research Institute, The University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria, 3010, Australia
| | - Kevin J Barnham
- The Mental Health Research Institute, The University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria, 3010, Australia
| | - Colin L Masters
- The Mental Health Research Institute, The University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria, 3010, Australia
| |
Collapse
|
36
|
Maftei M, Thurm F, Schnack C, Tumani H, Otto M, Elbert T, Kolassa IT, Przybylski M, Manea M, von Arnim CAF. Increased levels of antigen-bound β-amyloid autoantibodies in serum and cerebrospinal fluid of Alzheimer's disease patients. PLoS One 2013; 8:e68996. [PMID: 23874844 PMCID: PMC3715516 DOI: 10.1371/journal.pone.0068996] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Accepted: 06/04/2013] [Indexed: 12/11/2022] Open
Abstract
Recent studies have suggested a protective role of physiological β-amyloid autoantibodies (Aβ-autoantibodies) in Alzheimer’s disease (AD). However, the determination of both free and dissociated Aβ-autoantibodies in serum hitherto has yielded inconsistent results regarding their function and possible biomarker value. Here we report the application of a new sandwich enzyme-linked immunosorbent assay (ELISA) for the determination of antigen-bound Aβ-autoantibodies (intact Aβ-IgG immune complexes) in serum and cerebrospinal fluid (CSF) of a total number of 112 AD patients and age- and gender-matched control subjects. Both serum and CSF levels of Aβ-IgG immune complexes were found to be significantly higher in AD patients compared to control subjects. Moreover, the levels of Aβ-IgG complexes were negatively correlated with the cognitive status across the groups, increasing with declining cognitive test performance of the subjects. Our results suggest a contribution of IgG-type autoantibodies to Aβ clearance in vivo and an increased immune response in AD, which may be associated with deficient Aβ-IgG removal. These findings may contribute to elucidating the role of Aβ-autoantibodies in AD pathophysiology and their potential application in AD diagnosis.
Collapse
Affiliation(s)
- Madalina Maftei
- Laboratory of Analytical Chemistry and Biopolymer Structure Analysis, Department of Chemistry, University of Konstanz, Konstanz, Germany
- Steinbeis Research Center for Biopolymer Analysis, University of Konstanz, Konstanz, Germany
| | - Franka Thurm
- Department of Psychology, University of Konstanz, Konstanz, Germany
- Clinical and Biological Psychology, Institute of Psychology and Education, University of Ulm, Ulm, Germany
- Department of Psychology, TU Dresden, Dresden, Germany
| | | | | | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Thomas Elbert
- Department of Psychology, University of Konstanz, Konstanz, Germany
| | - Iris-Tatjana Kolassa
- Clinical and Biological Psychology, Institute of Psychology and Education, University of Ulm, Ulm, Germany
- Zukunftskolleg, University of Konstanz, Konstanz, Germany
- * E-mail: (MM); (I-TK); (CAFVA); (MP)
| | - Michael Przybylski
- Laboratory of Analytical Chemistry and Biopolymer Structure Analysis, Department of Chemistry, University of Konstanz, Konstanz, Germany
- * E-mail: (MM); (I-TK); (CAFVA); (MP)
| | - Marilena Manea
- Laboratory of Analytical Chemistry and Biopolymer Structure Analysis, Department of Chemistry, University of Konstanz, Konstanz, Germany
- Zukunftskolleg, University of Konstanz, Konstanz, Germany
- * E-mail: (MM); (I-TK); (CAFVA); (MP)
| | | |
Collapse
|
37
|
Gupta VB, Sundaram R, Martins RN. Multiplex biomarkers in blood. ALZHEIMERS RESEARCH & THERAPY 2013; 5:31. [PMID: 23795953 PMCID: PMC3707019 DOI: 10.1186/alzrt185] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Advances in the field of blood biomarker discovery will help in identifying Alzheimer's disease in its preclinical stage, allowing treatment to be initiated before irreversible damage occurs. This review discusses some recent past and current approaches being taken by researchers in the field. Individual blood biomarkers have been unsuccessful in defining the disease pathology, progression and thus diagnosis. This directs to the need for discovering a multiplex panel of blood biomarkers as a promising approach with high sensitivity and specificity for early diagnosis. However, it is a great challenge to standardize a worldwide blood biomarker panel due to the innate differences in the population tested, nature of the samples and methods utilised in different studies across the globe. We highlight several issues that result in the lack of reproducibility in this field of research currently faced by researchers. Several important measures are summarized towards the end of the review that can be taken to minimize the variability among various centres.
Collapse
Affiliation(s)
- Veer Bala Gupta
- Centre of Excellence for Alzheimer's disease Research & Care, School of Medical Sciences, Edith Cowan University, Joondalup, WA, 6027 Australia
| | - Ramani Sundaram
- Centre for Ageing and Alzheimer's, Nightingales, Bangalore, 560043 India
| | - Ralph N Martins
- Centre of Excellence for Alzheimer's disease Research & Care, School of Medical Sciences, Edith Cowan University, Joondalup, WA, 6027 Australia ; Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), Perth, WA, 6009 Australia ; Suite 22, Hollywood Medical Centre, 85 Monash Ave, Nedlands, WA, 6009 Australia
| |
Collapse
|
38
|
Alzheimer's disease biomarkers: correspondence between human studies and animal models. Neurobiol Dis 2013; 56:116-30. [PMID: 23631871 DOI: 10.1016/j.nbd.2013.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 04/11/2013] [Accepted: 04/18/2013] [Indexed: 01/05/2023] Open
Abstract
Alzheimer's disease (AD) represents an escalating global threat as life expectancy and disease prevalence continue to increase. There is a considerable need for earlier diagnoses to improve clinical outcomes. Fluid biomarkers measured from cerebrospinal fluid (CSF) and blood, or imaging biomarkers have considerable potential to assist in the diagnosis and management of AD. An additional important utility of biomarkers is in novel therapeutic development and clinical trials to assess efficacy and side effects of therapeutic interventions. Because many biomarkers are initially examined in animal models, the extent to which markers translate from animals to humans is an important issue. The current review highlights many existing and pipeline biomarker approaches, focusing on the degree of correspondence between AD patients and animal models. The review also highlights the need for greater translational correspondence between human and animal biomarkers.
Collapse
|
39
|
Takeda S, Sato N, Uchio-Yamada K, Yu H, Moriguchi A, Rakugi H, Morishita R. Oral glucose loading modulates plasma β-amyloid level in alzheimer's disease patients: potential diagnostic method for Alzheimer's disease. Dement Geriatr Cogn Disord 2013; 34:25-30. [PMID: 22889768 DOI: 10.1159/000338704] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/04/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Although plasma β-amyloid (Aβ) has been suggested to be a noninvasive diagnostic biomarker for Alzheimer's disease (AD), its significance and validity have been inconclusive. Thus, it is quite important to establish a novel diagnostic method related to plasma Aβ. METHODS As our previous animal studies demonstrated a relation of glucose with plasma Aβ, we examined the effect of glucose loading on plasma Aβ levels in AD patients. After fasting, an oral glucose load was administered to AD patients and non-AD dementia patients, and subsequently, blood glucose, plasma insulin, and plasma Aβ levels were measured. RESULTS The plasma levels of baseline blood glucose, plasma insulin, and plasma Aβ were not different between the two groups. However, immediately after glucose loading, a significant increase in plasma Aβ40 and Aβ42 levels was observed in AD patients, whereas a mild decrease in plasma Aβ40 and Aβ42 levels was detected in non-AD dementia patients. CONCLUSION The present study clearly demonstrated a different response in plasma Aβ40 and Aβ42 levels after glucose loading between AD and non-AD dementia patients, which is consistent with our previous animal studies. These findings suggest a novel diagnostic tool for AD using the elevation of plasma Aβ level after glucose loading, although further studies are necessary.
Collapse
Affiliation(s)
- Shuko Takeda
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Suita, Japan
| | | | | | | | | | | | | |
Collapse
|
40
|
Zhou L, Chan KH, Chu LW, Kwan JSC, Song YQ, Chen LH, Ho PWL, Cheng OY, Ho JWM, Lam KSL. Plasma amyloid-β oligomers level is a biomarker for Alzheimer's disease diagnosis. Biochem Biophys Res Commun 2012; 423:697-702. [PMID: 22704931 DOI: 10.1016/j.bbrc.2012.06.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 06/05/2012] [Indexed: 10/28/2022]
Abstract
Amyloid beta (Aβ), especially Aβ oligomers, is important in Alzheimer's disease (AD) pathogenesis. We studied plasma Aβ(40), Aβ(42), and Aβ oligomers levels in 44 AD patients and 22 non-demented controls. Cognitive functions were assessed by Chinese version of mini-mental state examination (MMSE), Abbreviated Metal Test (AMT), Alzheimer's Disease Assessment Scale Cognitive Subscale (ADAS-cog). Plasma Aβ monomers and oligomers levels were measured by ELISA. We found that the median plasma Aβ(40) and Aβ(42) levels were similar between AD and controls, and without significant correlation with cognition. Plasma Aβ oligomers level was higher in AD than controls (642.54 ng/ml [range 103.33-2676.93] versus 444.18 ng/ml [range 150.19-1311.18], p=0.047), and negatively correlated with cognition. In multivariate logistic regression analysis, the highest tertile of Aβ oligomers levels showed an increased risk of AD than the combined group of middle and lowest tertiles (OR=8.85, p=0.013), after adjustment of gender, age and APOE4 genotype. Increased plasma Aβ oligomers level was associated with decreased MMSE and AMT scores (p=0.037, p=0.043, respectively) and increased ADAS-cog score (p=0.036), suggesting negative correlation with cognitive function. We concluded that plasma Aβ oligomers level is an useful biomarker for AD diagnosis.
Collapse
Affiliation(s)
- L Zhou
- University Department of Medicine, The University of Hong Kong, Hong Kong
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Reale M, Kamal MA, Velluto L, Gambi D, Di Nicola M, Greig NH. Relationship between inflammatory mediators, Aβ levels and ApoE genotype in Alzheimer disease. Curr Alzheimer Res 2012; 9:447-57. [PMID: 22272623 PMCID: PMC5215089 DOI: 10.2174/156720512800492549] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 11/05/2011] [Accepted: 11/09/2011] [Indexed: 11/22/2022]
Abstract
Activation of inflammatory processes is observed within the brain as well as periphery of subjects with Alzheimer's disease (AD). Whether or not inflammation represents a possible cause of AD or occurs as a consequence of the disease process, or, alternatively, whether the inflammatory response might be beneficial to slow the disease progression remains to be elucidated. The cytokine IL-18 shares with IL-1 the same pro-inflammatory features. Consequent to these similarities, IL-18 and its endogenous inhibitor, IL-18BP, were investigated in the plasma of AD patients versus healthy controls (HC). An imbalance of IL-18 and IL-18BP was observed in AD, with an elevated IL-18/IL-18BP ratio that might be involved in disease pathogenesis. As part of the inflammatory response, altered levels of RANTES, MCP-1 and ICAM- 1, molecules involved in cell recruitment to inflammatory sites, were observed in AD. Hence, correlations between IL-18 and other inflammatory plasma markers were analyzed. A negative correlation was observed between IL-18 and IL-18BP in both AD and HC groups. A positive correlation was observed between IL-18 and ICAM-1 in AD patients, whereas a negative correlation was evident in the HC group. IL-18 positively correlated with Aβ in both groups, and no significant correlations were observed between IL-18, RANTES and MCP-1. An important piece of evidence supporting a pathophysiologic role for inflammation in AD is the number of inflammatory mediators that have been found to be differentially regulated in AD patients, and specific ones may provide utility as part of a biomarker panel to not only aid early AD diagnosis, but follow its progression.
Collapse
Affiliation(s)
- M Reale
- Dept. of Oncology and Experimental Medicine, Unit of Immunodiagnostic, University "G. D'Annunzio", Chieti, Italy.
| | | | | | | | | | | |
Collapse
|
42
|
Mazumdar M, Xia W, Hofmann O, Gregas M, Ho Sui S, Hide W, Yang T, Needleman HL, Bellinger DC. Prenatal lead levels, plasma amyloid β levels, and gene expression in young adulthood. ENVIRONMENTAL HEALTH PERSPECTIVES 2012; 120:702-7. [PMID: 22313790 PMCID: PMC3346789 DOI: 10.1289/ehp.1104474] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Accepted: 02/07/2012] [Indexed: 05/06/2023]
Abstract
BACKGROUND Animal studies suggest that early-life lead exposure influences gene expression and production of proteins associated with Alzheimer's disease (AD). OBJECTIVES We attempted to assess the relationship between early-life lead exposure and potential biomarkers for AD among young men and women. We also attempted to assess whether early-life lead exposure was associated with changes in expression of AD-related genes. METHODS We used sandwich enzyme-linked immunosorbent assays (ELISA) to measure plasma concentrations of amyloid β proteins Aβ40 and Aβ42 among 55 adults who had participated as newborns and young children in a prospective cohort study of the effects of lead exposure on development. We used RNA microarray techniques to analyze gene expression. RESULTS Mean plasma Aβ42 concentrations were lower among 13 participants with high umbilical cord blood lead concentrations (≥ 10 μg/dL) than in 42 participants with lower cord blood lead concentrations (p = 0.08). Among 10 participants with high prenatal lead exposure, we found evidence of an inverse relationship between umbilical cord lead concentration and expression of ADAM metallopeptidase domain 9 (ADAM9), reticulon 4 (RTN4), and low-density lipoprotein receptor-related protein associated protein 1 (LRPAP1) genes, whose products are believed to affect Aβ production and deposition. Gene network analysis suggested enrichment in gene sets involved in nerve growth and general cell development. CONCLUSIONS Data from our exploratory study suggest that prenatal lead exposure may influence Aβ-related biological pathways that have been implicated in AD onset. Gene network analysis identified further candidates to study the mechanisms of developmental lead neurotoxicity.
Collapse
Affiliation(s)
- Maitreyi Mazumdar
- Department of Neurology, Children's Hospital Boston, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Song F, Poljak A, Valenzuela M, Mayeux R, Smythe GA, Sachdev PS. Meta-analysis of plasma amyloid-β levels in Alzheimer's disease. J Alzheimers Dis 2012; 26:365-75. [PMID: 21709378 DOI: 10.3233/jad-2011-101977] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Plasma amyloid-β (Aβ) levels have been proposed as biomarkers of Alzheimer's disease (AD), but studies have produced inconsistent results. We present a meta-analytic review of cross-sectional studies that examined plasma Aβ levels in AD and cognitively normal subjects, and longitudinal studies that used baseline plasma Aβ levels to predict conversion from normal cognition to AD. Medline and EMBASE databases were searched to generate an initial list of relevant studies, and selected authors approached for additional data. Twelve cross- sectional studies (n = 1483) and seven longitudinal (n = 3920) met the inclusion criteria for meta-analysis. Random effects model was used to calculate the weighted mean difference (WMD) by Review Manager Version 4.2. In longitudinal studies, cognitively normal individuals who converted to AD had higher baseline Aβ1-40 and Aβ1-42 levels (WMD: 10.29, z = 3.80, p = 0.0001 and WMD: 8.01, z = 2.76, p = 0.006, respectively), and non-significantly increased Aβ1-42/Aβ1-40 ratio (WMD: 0.03, z = 1.65, p = 0.10). In cross sectional studies, compared to cognitively normal individuals, AD patients had marginally but non-significantly lower Aβ1-42 levels (WMD:-2.84, z = 1.73, p = 0.08), but Aβ1-40 levels were not significantly different (WMD: 3.43, z = 0.40, p = 0.69). Our systematic review suggests a model of differential longitudinal changes in plasma Aβ levels in cognitively stable individuals versus those who go on to develop AD dementia. Baseline Aβ1-40 and Aβ1-42 levels in cognitively normal elderly individuals might be predictors of higher rates of progression to AD, and should be further explored as potential biomarkers.
Collapse
Affiliation(s)
- Fei Song
- Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, Australia
| | | | | | | | | | | |
Collapse
|
44
|
Humpel C, Hochstrasser T. Cerebrospinal fluid and blood biomarkers in Alzheimer’s disease. World J Psychiatry 2011; 1:8-18. [PMID: 24175162 PMCID: PMC3782169 DOI: 10.5498/wjp.v1.i1.8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 10/14/2011] [Accepted: 12/26/2011] [Indexed: 02/05/2023] Open
Abstract
Due to an ever aging society and growing prevalence of Alzheimer’s disease (AD), the challenge to meet social and health care system needs will become increasingly difficult. Unfortunately, a definite ante mortem diagnosis is not possible. Thus, an early diagnosis and identification of AD patients is critical for promising, early pharmacological interventions as well as addressing health care needs. The most advanced and most reliable markers are β-amyloid, total tau and phosphorylated tau in cerebrospinal fluid (CSF). In blood, no single biomarker has been identified despite an intense search over the last decade. The most promising approaches consist of a combination of several blood-based markers increasing the reliability, sensitivity and specificity of the AD diagnosis. However, contradictory data make standardized testing methods in longitudinal and multi-center studies extremely difficult. In this review, we summarize a range of the most promising CSF and blood biomarkers for diagnosing AD.
Collapse
Affiliation(s)
- Christian Humpel
- Christian Humpel, Tanja Hochstrasser, Laboratory for Psychiatry and Experimental Alzheimer's Research, Department of Psychiatry and Psychotherapy, Innsbruck Medical University, 6020 Innsbruck, Austria
| | | |
Collapse
|
45
|
Blood-based protein biomarkers for diagnosis and classification of neurodegenerative diseases: current progress and clinical potential. Mol Diagn Ther 2011; 15:83-102. [PMID: 21623645 DOI: 10.1007/bf03256398] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Biomarker research is a rapidly advancing field in medicine. Recent advances in genomic, genetic, epigenetic, neuroscientific, proteomic, and metabolomic knowledge and technologies have opened the way to thriving research. In the most general sense, a biomarker refers to any useful characteristic that can be measured and used as an indicator of a normal biologic process, a pathogenic process, or a pharmacologic response to a therapeutic agent. Despite the extensive resources concentrated on this area, there are very few biomarkers currently available that qualify and are satisfactorily validated for mental disorders, and there is still a major lack of biomarkers for typifying neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease. This article provides an overview of this field of research and focuses on recent advances in biomarker research in Alzheimer's disease and Parkinson's disease.
Collapse
|
46
|
Zhang Y, Lee DHS. Sink Hypothesis and Therapeutic Strategies for Attenuating Aβ Levels. Neuroscientist 2011; 17:163-173. [DOI: 10.1177/1073858410381532] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Amyloid β (Aβ) plaque, comprised mainly by Aβ peptides, is an important pathology of Alzheimer’s brains. Major efforts have been devoted to targeting this neurotoxic Aβ peptide for discovering disease-modifying treatments for Alzheimer’s disease. Inasmuch as Aβ is found in both the brain and the periphery, it is hypothesized that there is some form of equilibrium for the Aβ in the brain and the periphery such that Aβ can be transported across the blood-brain barrier. By modulating the periphery Aβ levels, it is predicted that the brain Aβ levels will undergo concomitant changes, forming the basis of the “sink hypothesis” for Aβ lowering strategies. In this review, the significance and implication of this sink hypothesis as well as how the sink hypothesis may contribute to the recent Aβ-based drug discovery in AD are discussed. Ultimately, the validity of the sink hypothesis will be resolved when the appropriate Aβ agents are being tested in the clinic.
Collapse
Affiliation(s)
- Yan Zhang
- Laboratory of Neurobiology and State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, 100871, China,
| | - Daniel H. S. Lee
- Translational Medicine, GlaxoSmithKline R&D, Shanghai, 20001, China
| |
Collapse
|
47
|
Seppälä TT, Herukka SK, Hänninen T, Tervo S, Hallikainen M, Soininen H, Pirttilä T. Plasma Abeta42 and Abeta40 as markers of cognitive change in follow-up: a prospective, longitudinal, population-based cohort study. J Neurol Neurosurg Psychiatry 2010; 81:1123-7. [PMID: 20478847 PMCID: PMC2976614 DOI: 10.1136/jnnp.2010.205757] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Single measurements of plasma Aβ are not useful in the diagnostics of Alzheimer's disease (AD). However, changes in plasma Aβ levels during repeated testing may be helpful in the prediction and evaluation of progression of the incipient AD or mild cognitive impairment. OBJECTIVE To examine the relation of baseline and serial plasma Aβ levels to cognitive change in follow-up. METHODS 269 subjects (52 cognitively impaired and 217 controls) from a population-based cohort were clinically followed up from 3 to 6 years. Serial plasma samples were available from 70 subjects who were followed up for 3 years and 43 subjects followed for 6 years. The plasma Aβ levels were measured using ELISA. RESULTS Subjects who declined cognitively during the follow-up had lower levels of plasma Aβ42 at the baseline. Plasma Aβ42 and the Aβ42/Aβ40 ratio decreased (-2.4 pg/ml for Aβ42 in 6 years) in those who declined in follow-up, whereas Aβ42 and the Aβ42/Aβ40 ratio increased in the subjects who remained cognitively stable or improved in follow-up. Subjects using acetylsalicylic acid, dipyridamole, antidiabetic or anticoagulant drugs as well as subjects with coronary heart disease had higher levels of Aβ40. CONCLUSIONS Low or decreasing plasma Aβ42 during the follow-up is associated with cognitive decline. Serial measurement of plasma Aβ42 may be useful in the detection of the subjects who are at risk for cognitive decline.
Collapse
Affiliation(s)
- T T Seppälä
- Department of Neurology, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | | | | | | | | | | | | |
Collapse
|
48
|
Schipper HM. Biological markers and Alzheimer disease: a canadian perspective. Int J Alzheimers Dis 2010; 2010. [PMID: 20811568 PMCID: PMC2929634 DOI: 10.4061/2010/978182] [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: 02/16/2010] [Accepted: 07/11/2010] [Indexed: 01/14/2023] Open
Abstract
Decreased β-amyloid1-42 and increased phospho-tau protein levels in the cerebrospinal fluid (CSF) are currently the most accurate chemical neurodiagnostics of sporadic Alzheimer disease (AD). A report (2007) of the Third Canadian Consensus Conference on the Diagnosis and Treatment of Dementia (2006) recommended that biological markers should not be currently requisitioned by primary care physicians in the routine investigation of subjects with memory complaints. Consideration for such testing should prompt patient referral to a specialist engaged in dementia evaluations or a Memory Clinic. The specialist should consider having CSF biomarkers (β-amyloid1-42 and phospho-tau) measured at a reputable facility in restricted cases presenting with atypical features and diagnostic confusion, but not as a routine procedure in all individuals with typical sporadic AD phenotypes. We submit that developments in the field of AD biomarker discovery since publication of the 3rd CCCDTD consensus data do not warrant revision of the 2007 recommendations.
Collapse
Affiliation(s)
- Hyman M Schipper
- Department of Neurology and Neurosurgery, Centre for Neurotranslational Research, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, 3755 Cote St. Catherine Rd. Montreal, QC, Canada H3T 1E2
| |
Collapse
|
49
|
Takeda S, Sato N, Rakugi H, Morishita R. Plasma beta-amyloid as potential biomarker of Alzheimer disease: possibility of diagnostic tool for Alzheimer disease. MOLECULAR BIOSYSTEMS 2010; 6:1760-6. [PMID: 20567751 DOI: 10.1039/c003148h] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alzheimer disease (AD), which is characterized by progressive cognitive and behavioral deficit, is the most common form of dementia. The incidence of AD is increasing at an alarming rate, and has become a major public health concern in many countries. It is well known that the onset of AD is preceded by a long preclinical period. It is thus critical to establish diagnostic biomarkers that can predict the risk of developing AD prior to clinical manifestation of dementia, for effective prevention and early intervention. With the emergence of potential promising approaches to treat AD targeting the beta-amyloid (Abeta) pathway, such as gamma-secretase inhibitors and vaccine therapy, there is an urgent need for such diagnostic markers. Although cerebrospinal fluid (CSF) Abeta and tau protein levels are candidate biomarkers for AD, the invasive sampling procedure with associated complications limits their use in routine clinical practice. Plasma Abeta has been suggested as an inexpensive and non-invasive biomarker for AD. Although most previous cross-sectional studies on plasma Abeta level in humans failed to show a significant difference between individuals with AD compared to healthy older adults, many strategies are under investigation to improve the diagnostic potential of plasma Abeta. One promising approach is to modify the plasma Abeta level using some potential modulators. It is possible that a difference in plasma Abeta level might be unmasked by evaluating the response to stimulation by a modulator. Anti-Abeta antibody and Abeta binding proteins have been reported to be such modulators of plasma Abeta. In addition, the glucometabolic or hormonal status appears to modulate the plasma Abeta level. Our recent study has shown the possibility that glucose loading could be a novel simple strategy to modulate the plasma Abeta level, making it better suited for early diagnosis. This review summarizes the utility and limitations of current biomarkers of AD and discusses future strategies to improve the diagnostic potential of plasma Abeta.
Collapse
Affiliation(s)
- Shuko Takeda
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | | | |
Collapse
|
50
|
Zheng Y, He J, Hong T. Biomarkers of Alzheimer's disease in body fluids. SCIENCE CHINA-LIFE SCIENCES 2010; 53:490-6. [PMID: 20596916 DOI: 10.1007/s11427-010-0081-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Accepted: 07/23/2009] [Indexed: 10/19/2022]
Abstract
Various innovative diagnostic methods for Alzheimer's disease (AD) have been developed in view of the increasing prevalence and consequences of later-life dementia. Biomarkers in cerebrospinal fluid (CSF) and blood for AD are primarily based on the detection of components derived from amyloid plaques and neurofibrillary tangles (NFTs). Published reports on CSF and blood biomarkers in AD indicate that although biomarkers in body fluids may be utilized in the clinical diagnosis of AD, there are no specific markers that permit accurate and reliable diagnosis of early-stage AD or the monitoring of disease progression.
Collapse
Affiliation(s)
- YanPeng Zheng
- College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| | | | | |
Collapse
|