1
|
Labonte J, Sugarman MA, Pettway E, Zetterberg H, Blennow K, Ashton NJ, Karikari TK, Aparicio HJ, Frank B, Tripodis Y, Martin B, Palmisano JN, Steinberg EG, Simkin I, Farrer LA, Jun GR, Turk KW, Budson AE, O'Connor MK, Au R, Goldstein LE, Stern RA, Stein TD, McKee AC, Qiu WQ, Mez J, Banks SJ, Alosco ML. Sex differences on tau, astrocytic, and neurodegenerative plasma biomarkers. J Alzheimers Dis 2025:13872877251329468. [PMID: 40151917 DOI: 10.1177/13872877251329468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
BackgroundSex differences have consistently been identified on autopsy, neuroimaging, and cerebrospinal fluid outcomes related to Alzheimer's disease (AD), but the exact mechanisms for these associations are unclear. Blood-based biomarkers are practical alternatives for the investigation of mechanisms of AD, in addition to accurate disease detection and monitoring.ObjectiveThe objective of this study was to examine sex differences across a panel of blood-based plasma biomarkers in participants with and without cognitive impairment due to AD.MethodsPlasma samples were collected from 567 participants from across the AD diagnostic continuum (i.e., normal cognition (NC), mild cognitive impairment (MCI), and dementia) and analyzed for glial fibrillary acidic protein (GFAP), neurofilament light (NfL), phosphorylated tau at threonine 181 (p-tau181), and total tau (t-tau). Baseline and longitudinal analyses evaluated for any significant associations between sex and AD-related plasma biomarkers.ResultsFemales were found to have higher plasma GFAP compared to males at baseline regardless of cognitive diagnosis. Among those with AD dementia, females were also found to have higher NfL levels compared to males. Longitudinal analyses found that higher plasma NfL at baseline was associated with an increased risk of worsening AD dementia status only in females. No significant findings were observed for p-tau181 or t-tau.ConclusionsThis study found significant sex differences in plasma biomarkers of GFAP and NfL. Further research is needed to better understand the underlying mechanisms mediating these differences.
Collapse
Affiliation(s)
- Jacob Labonte
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Michael A Sugarman
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Medical University of South Carolina, Charleston, SC, USA
| | - Erika Pettway
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, UCL Institute of Neurology, University College London, London, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Division of Life Sciences and Medicine, and Department of Neurology, Neurodegenerative Disorder Research Center, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Thomas K Karikari
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hugo J Aparicio
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Brandon Frank
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- US Department of Veteran Affairs, VA Boston Healthcare System, Jamaica Plain, MA, USA
| | - Yorghos Tripodis
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Brett Martin
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
| | - Joseph N Palmisano
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
| | - Eric G Steinberg
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Irene Simkin
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Lindsay A Farrer
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Gyungah R Jun
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Katherine W Turk
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- US Department of Veteran Affairs, VA Boston Healthcare System, Jamaica Plain, MA, USA
| | - Andrew E Budson
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- US Department of Veteran Affairs, VA Boston Healthcare System, Jamaica Plain, MA, USA
| | - Maureen K O'Connor
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neuropsychology, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, USA
| | - Rhoda Au
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Lee E Goldstein
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biomedical, Electrical, and Computer Engineering, Boston University College of Engineering, Boston, MA, USA
| | - Robert A Stern
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurosurgery, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Thor D Stein
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- US Department of Veteran Affairs, VA Boston Healthcare System, Jamaica Plain, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- US Department of Veteran Affairs, VA Bedford Healthcare System, Bedford, MA, USA
| | - Ann C McKee
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- US Department of Veteran Affairs, VA Boston Healthcare System, Jamaica Plain, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- US Department of Veteran Affairs, VA Bedford Healthcare System, Bedford, MA, USA
| | - Wei Qiao Qiu
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jesse Mez
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sarah J Banks
- Departments of Neuroscience and Psychiatry, University of California, San Diego, CA, USA
| | - Michael L Alosco
- Boston University Alzheimer's Disease Research Center and BU CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
2
|
Bangs MC, Gadhavi J, Carter EK, Ping L, Duong DM, Dammer EB, Wu F, Shantaraman A, Fox EJ, Johnson EC, Lah JJ, Levey AI, Seyfried NT. Proteomic Subtyping of Alzheimer's Disease CSF links Blood-Brain Barrier Dysfunction to Reduced levels of Tau and Synaptic Biomarkers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643332. [PMID: 40161719 PMCID: PMC11952530 DOI: 10.1101/2025.03.14.643332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD) is characterized by significant clinical and molecular heterogeneity, influenced by genetic and demographic factors. Using an unbiased, network-driven approach, we analyzed the cerebrospinal fluid (CSF) proteome from 431 individuals (483 samples), including 111 African American participants, to identify core protein modules associated with AD, race, sex, and age. Our analysis revealed ten co-expression modules linked to distinct biological pathways and cell types, many of which correlated with established AD biomarkers such as β-amyloid, tau, and phosphorylated tau. To further resolve disease heterogeneity, we applied a proteomic subtyping approach, identifying six distinct CSF subtypes spanning the clinical and pathological spectrum. These subtypes were validated across independent cohorts, with many aligning with previously defined AD subtypes, including those linked to neuronal hyperplasticity, immune activation, and blood-brain barrier (BBB) integrity. Notably, the BBB subtype, enriched with African Americans and men, was characterized by low CSF tau, high CSF/serum albumin ratios, and reduced synaptic protein levels. This subtype also exhibited increased levels of proteolytic enzymes, including thrombin and matrix metalloproteases, that cleave tau. Plasma dilution into the neuronal hyperplastic AD subtype CSF led to reduced tau and synaptic protein module levels, indicating that plasma protease activity contributes to tau and synaptic protein depletion independent of underlying brain pathology. These findings highlight the impact of BBB integrity on CSF tau levels, particularly in men and African Americans, and underscore the need for diversity-informed AD biomarker strategies to improve diagnostics and therapeutic targeting across populations.
Collapse
Affiliation(s)
- Madison C. Bangs
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Joshna Gadhavi
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - E. Kathleen Carter
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lingyan Ping
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Duc M. Duong
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Eric B. Dammer
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Fang Wu
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Anantharaman Shantaraman
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Edward J. Fox
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Erik C.B. Johnson
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA 30322, USA
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - James J. Lah
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA 30322, USA
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Allan I. Levey
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA 30322, USA
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nicholas T. Seyfried
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA 30322, USA
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
3
|
Neal SJ, Chitta S, Magden ER, Simmons JH. Characterizing plasma and cerebrospinal fluid biomarkers relevant to neurodegeneration in captive olive baboons (Papio anubis). PLoS One 2025; 20:e0318173. [PMID: 39946349 PMCID: PMC11825030 DOI: 10.1371/journal.pone.0318173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/12/2025] [Indexed: 02/16/2025] Open
Abstract
Alzheimer's disease and related dementias (ADRD) present a significant global disease burden that is only expected to grow in the future. As such, there is a need to develop and investigate biomarkers that identify individuals at risk of developing ADRD with the goal of providing early interventions and treatments. Non-human primate (NHP) models of neurodegeneration present opportunities to examine such biomarkers in a preclinical model with the ability to control several confounding factors present in research with humans. Baboons naturally develop several ADRD-related neuropathologies that humans also exhibit, including age-related tau and amyloid deposition. However, to our knowledge, there are no data characterizing fluid biomarkers relevant to neurodegeneration or ADRD in baboons. We collected plasma (N = 139) and cerebrospinal fluid (CSF, N = 44) from captive baboons ranging in age from 3-19 years old. We characterized biomarkers as a function of age, sex, and rearing status in baboons using a bead-based bioplex human assay (Thermo Fisher Scientific's Neuroscience 18-Plex Human ProcartaPlex™ Panel). Fluid biomarkers were more detectable in CSF compared to plasma. Additionally, while sex and rearing did not significantly predict biomarkers in baboons, age significantly predicted levels of eight of the 12 biomarkers detected in the assay. Linear regressions showed that CSF levels of total tau, pTau181, NGF-beta, GFAP, NF-H, and S100B were higher in older baboons, as were plasma levels of NGF-beta. Lastly, older baboons showed a higher incidence of co-occurrence of multiple biomarkers as measured in CSF, but not in plasma. These data show that baboons exhibit age-dependent changes in biomarkers used in humans for clinical screening, diagnosis, and prognosis of ADRD, thereby further demonstrating the value of baboons as a model of aging and, possibly, ADRD.
Collapse
Affiliation(s)
- Sarah J. Neal
- Department of Comparative Medicine, Michale E. Keeling Center for Comparative Medicine and Research, The University of Texas MD Anderson Cancer Center, Bastrop, Texas, United States of America
| | - Sriram Chitta
- Department of Comparative Medicine, Michale E. Keeling Center for Comparative Medicine and Research, The University of Texas MD Anderson Cancer Center, Bastrop, Texas, United States of America
| | - Elizabeth R. Magden
- Department of Comparative Medicine, Michale E. Keeling Center for Comparative Medicine and Research, The University of Texas MD Anderson Cancer Center, Bastrop, Texas, United States of America
| | - Joe H. Simmons
- Department of Comparative Medicine, Michale E. Keeling Center for Comparative Medicine and Research, The University of Texas MD Anderson Cancer Center, Bastrop, Texas, United States of America
| |
Collapse
|
4
|
Choi YJ, Haddadnezhad M, Baek SJ, Lee CN, Park S, Sim SJ. Plasmonic Nanogap-Enhanced Tunable Three-Dimensional Nanoframes in Application to Clinical Diagnosis of Alzheimer's Disease. ACS Sens 2024; 9:5587-5595. [PMID: 39356173 DOI: 10.1021/acssensors.4c02037] [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] [Indexed: 10/03/2024]
Abstract
Advancements in nanotechnology led to significant improvements in synthesizing plasmon-enhanced nanoarchitectures for biosensor applications, and high-yield productivity at low cost is vital to step further into medical commerce. Metal nanoframes via wet chemistry are gaining attention for their homogeneous structure and outstanding catalytic and optical properties. However, nanoframe morphology should be considered delicately when brought to biosensors to utilize its superior characteristics thoroughly, and the need to prove its clinical applicability still remains. Herein, we controlled the frameworks of double-walled nanoframes (DWFs) precisely via wet chemistry to construct a homogeneous plasmon-enhanced nanotransducer for localized surface plasmon resonance biosensors. By tuning the physical properties considering the finite-difference time-domain simulation results, biomolecular interactions were feasible in the electromagnetic field-enhanced nanospace. As a result, DWF10 exhibited a 10-fold lower detection limit of 2.21 fM compared to DWF14 for tau detection. Further application into blood-based clinical and Alzheimer's disease (AD) diagnostics, notable improvement in classifying mild cognitive impairment patients against healthy controls and AD patients, was demonstrated along with impressive AUC values. Thus, in response to diverse detection methods, optimizing nanoframe dimensions such as nanogap and frame thickness to maximize sensor performance is critical to realize future POCT diagnosis.
Collapse
Affiliation(s)
- Young Jae Choi
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| | - MohammadNavid Haddadnezhad
- Department of Chemistry, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003-9303, United States
| | - Seung Jong Baek
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Chan-Nyoung Lee
- Korea University Anam Hospital, Seoul 02841, Republic of Korea
| | - Sungho Park
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Sang Jun Sim
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
5
|
Sharma M, Pal P, Gupta SK. Advances in Alzheimer's disease: A multifaceted review of potential therapies and diagnostic techniques for early detection. Neurochem Int 2024; 177:105761. [PMID: 38723902 DOI: 10.1016/j.neuint.2024.105761] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 04/20/2024] [Accepted: 05/06/2024] [Indexed: 06/04/2024]
Abstract
Alzheimer's disease (AD) remains one of the most formidable neurological disorders, affecting millions globally. This review provides a holistic overview of the therapeutic strategies, both conventional and novel, aimed at mitigating the impact of AD. Initially, we delve into the conventional approach, emphasizing the role of Acetylcholinesterase (AChE) inhibition, which has been a cornerstone in AD management. As our understanding of AD evolves, several novel potential approaches emerge. We discuss the promising roles of Butyrylcholinesterase (BChE) inhibition, Tau Protein inhibitors, COX-2 inhibition, PPAR-γ agonism, and FAHH inhibition, among others. The potential of the endocannabinoids (eCB) system, cholesterol-lowering drugs, metal chelators, and MMPs inhibitors are also explored, culminating in the exploration of the pivotal role of microRNA in AD progression. Parallel to these therapeutic insights, we shed light on the novel tools and methodologies revolutionizing AD research. From the quantitative analysis of gene expression by qRTPCR to the evaluation of mitochondrial function using induced pluripotent stem cells (iPSCs), the advances in diagnostic and research tools offer renewed hope. Moreover, we explore the current landscape of clinical trials, highlighting the leading drug interventions and their respective stages of development. This comprehensive review concludes with a look into the future perspectives, capturing the potential breakthroughs and innovations on the horizon. Through a synthesis of current knowledge and emerging research, this article aims to provide a consolidated resource for clinicians, researchers, and academicians in the realm of Alzheimer's disease.
Collapse
Affiliation(s)
- Monika Sharma
- Faculty of Pharmacy, Department of Pharmacology, Swami Vivekanand Subharti University, Meerut, Uttar Pradesh, India
| | - Pankaj Pal
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, India.
| | - Sukesh Kumar Gupta
- KIET School of Pharmacy, KIET Group of Institutions, Ghaziabad, Uttar Pradesh, India; Department of Ophthalmology, Visual and Anatomical Sciences (OVAS), School of Medicine, Wayne State University, USA.
| |
Collapse
|
6
|
Yamada K, Iwatsubo T. Involvement of the glymphatic/meningeal lymphatic system in Alzheimer's disease: insights into proteostasis and future directions. Cell Mol Life Sci 2024; 81:192. [PMID: 38652179 PMCID: PMC11039514 DOI: 10.1007/s00018-024-05225-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 01/29/2024] [Accepted: 04/01/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is pathologically characterized by the abnormal accumulation of Aβ and tau proteins. There has long been a keen interest among researchers in understanding how Aβ and tau are ultimately cleared in the brain. The discovery of this glymphatic system introduced a novel perspective on protein clearance and it gained recognition as one of the major brain clearance pathways for clearing these pathogenic proteins in AD. This finding has sparked interest in exploring the potential contribution of the glymphatic/meningeal lymphatic system in AD. Furthermore, there is a growing emphasis and discussion regarding the possibility that activating the glymphatic/meningeal lymphatic system could serve as a novel therapeutic strategy against AD. OBJECTIVES Given this current research trend, the primary focus of this comprehensive review is to highlight the role of the glymphatic/meningeal lymphatic system in the pathogenesis of AD. The discussion will encompass future research directions and prospects for treatment in relation to the glymphatic/meningeal lymphatic system.
Collapse
Affiliation(s)
- Kaoru Yamada
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| |
Collapse
|
7
|
Gao L, Wang D, Ren J, Tan X, Chen J, Kong Z, Nie Y, Yan M. Acteoside ameliorates learning and memory impairment in APP/PS1 transgenic mice by increasing Aβ degradation and inhibiting tau hyperphosphorylation. Phytother Res 2024; 38:1735-1744. [PMID: 37661763 DOI: 10.1002/ptr.8006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 06/27/2023] [Accepted: 08/20/2023] [Indexed: 09/05/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease. Senile plaques and intracellular neurofibrillary tangles are pathological hallmarks of AD. Recent studies have described the improved cognitive and neuroprotective functions of acteoside (AS). This study aimed to investigate whether the improved cognition of AS was mediated by Aβ degradation and tau phosphorylation in APP/PS1 mice. The open field, Y maze, and novel object recognition tests were used to assess cognitive behavioral changes. We evaluated the levels of Aβ40 and Aβ42 in serum, cortex, and hippocampus, and Aβ-related scavenging enzymes, phosphorylated GSK3β and hyperphosphorylated tau in the cortex and hippocampus of APP/PS1 mice by western blotting. Our results revealed that AS treatment ameliorated anxious behaviors, spatial learning, and memory impairment in APP/PS1 mice and significantly reduced Aβ deposition in their serum, cortex, and hippocampus. AS significantly increased Aβ degradation, inhibited the hyperphosphorylation of tau, and significantly decreased the activity of GSK3β, which is involved in tau phosphorylation. Altogether, these findings indicated that the beneficial effects of AS on AD-associated anxious behaviors and cognitive impairments could be attributed to promoting Aβ degradation and inhibiting tau hyperphosphorylation, which might be partly mediated by GSK3β.
Collapse
Affiliation(s)
- Li Gao
- Prescription Laboratory of Xinjiang Traditional Uyghur Medicine, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, China
| | - Dongqing Wang
- Prescription Laboratory of Xinjiang Traditional Uyghur Medicine, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, China
| | - Jia Ren
- Prescription Laboratory of Xinjiang Traditional Uyghur Medicine, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, China
| | - Xue Tan
- Prescription Laboratory of Xinjiang Traditional Uyghur Medicine, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, China
| | - Jiayuan Chen
- Prescription Laboratory of Xinjiang Traditional Uyghur Medicine, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, China
| | - Zheng Kong
- Prescription Laboratory of Xinjiang Traditional Uyghur Medicine, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, China
| | - Yunan Nie
- Prescription Laboratory of Xinjiang Traditional Uyghur Medicine, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, China
| | - Ming Yan
- Prescription Laboratory of Xinjiang Traditional Uyghur Medicine, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, China
| |
Collapse
|
8
|
Ramos-Cejudo J, Scott MR, Tanner JA, Pase MP, McGrath ER, Ghosh S, Osorio RS, Thibault E, El Fakhri G, Johnson KA, Beiser A, Seshadri S. Associations of Plasma Tau with Amyloid and Tau PET: Results from the Community-Based Framingham Heart Study. J Alzheimers Dis 2024; 100:487-494. [PMID: 38875034 DOI: 10.3233/jad-231320] [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] [Indexed: 06/16/2024]
Abstract
Background Associations of plasma total tau levels with future risk of AD have been described. Objective To examine the extent to which plasma tau reflects underlying AD brain pathology in cognitively healthy individuals. Methods We examined cross-sectional associations of plasma total tau with 11C-Pittsburgh Compound-B (PiB)-PET and 18F-Flortaucipir (FTP)-PET in middle-aged participants at the community-based Framingham Heart Study. Results Our final sample included 425 participants (mean age 57.6± 9.9, 50% F). Plasma total tau levels were positively associated with amyloid-β deposition in the precuneus region (β±SE, 0.11±0.05; p = 0.025). A positive association between plasma total tau and tau PET in the rhinal cortex was suggested in participants with higher amyloid-PET burden and in APOEɛ4 carriers. Conclusions Our study highlights that plasma total tau is a marker of amyloid deposition as early as in middle-age.
Collapse
Affiliation(s)
- Jaime Ramos-Cejudo
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Matthew R Scott
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Jeremy A Tanner
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Matthew P Pase
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Emer R McGrath
- HRB Clinical Research Facility, University of Galway, Galway, Ireland
- The Framingham Study, Boston, MA, USA
- School of Medicine, University of Galway, Galway, Ireland
| | | | - Ricardo S Osorio
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Emma Thibault
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | | | - Keith A Johnson
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Alexa Beiser
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- The Framingham Study, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
- The Framingham Study, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
9
|
Li Z, Fan Z, Zhang Q. The Associations of Phosphorylated Tau 181 and Tau 231 Levels in Plasma and Cerebrospinal Fluid with Cognitive Function in Alzheimer's Disease: A Systematic Review and Meta-Analysis. J Alzheimers Dis 2024; 98:13-32. [PMID: 38339929 DOI: 10.3233/jad-230799] [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] [Indexed: 02/12/2024]
Abstract
Background Cerebrospinal fluid (CSF) or blood biomarkers like phosphorylated tau proteins (p-tau) are used to detect Alzheimer's disease (AD) early. Increasing studies on cognitive function and blood or CSF p-tau levels are controversial. Objective Our study examined the potential of p-tau as a biomarker of cognitive status in normal control (NC), mild cognitive impairment (MCI), and AD patients. Methods We searched PubMed, Cochrane, Embase, and Web of Science for relevant material through 12 January 2023. 5,017 participants from 20 studies-1,033 AD, 2,077 MCI, and 1,907 NC-were evaluated. Quantitative analysis provided continuous outcomes as SMDs with 95% CIs. Begg tested publication bias. Results MCI patients had lower CSF p-tau181 levels than AD patients (SMD =-0.60, 95% CI (-0.85, -0.36)) but higher than healthy controls (SMD = 0.67). AD/MCI patients had greater plasma p-tau181 levels than healthy people (SMD =-0.73, 95% CI (-1.04, -0.43)). MCI patients had significantly lower p-tau231 levels than AD patients in plasma and CSF (SMD =-0.90, 95% CI (-0.82, -0.45)). MCI patients showed greater CSF and plasma p-tau231 than healthy controls (SMD = 1.34, 95% CI (0.89, 1.79) and 0.43, (0.23, 0.64)). Plasma p-tau181/231 levels also distinguished the three categories. MCI patients had higher levels than healthy people, while AD patients had higher levels than MCI patients. Conclusions CSF p-tau181 and p-tau231 biomarkers distinguished AD, MCI, and healthy populations. Plasma-based p-tau181 and p-tau231 biomarkers for AD and MCI need further study.
Collapse
Affiliation(s)
- Zhirui Li
- Department of Disease Control and Prevention, Sichuan Provincial Center for Disease Control and Prevention, Sichuan Chengdu, China
| | - Zixuan Fan
- School of Health Policy and Management, Peking Union Medical College, Beijing, China
| | - Qian Zhang
- Department of Oncology, Xiamen Fifth Hospital, Fujian Xiamen, China
| |
Collapse
|
10
|
Wojdała AL, Bellomo G, Gaetani L, Toja A, Chipi E, Shan D, Chiasserini D, Parnetti L. Trajectories of CSF and plasma biomarkers across Alzheimer's disease continuum: disease staging by NF-L, p-tau181, and GFAP. Neurobiol Dis 2023; 189:106356. [PMID: 37977432 DOI: 10.1016/j.nbd.2023.106356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/02/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023] Open
Abstract
CSF-to-plasma transition will open new avenues for molecular phenotyping of Alzheimer's disease (AD). Here we evaluated a panel of AD biomarkers in matched CSF and plasma samples across the AD continuum, from preclinical AD to dementia. The aims were to: 1) compare diagnostic performance of the two biofluids, 2) evaluate trajectories of the biomarkers along AD progression. We analyzed CSF and plasma Aβ42/40, p-tau181, p-tau231, t-tau, NF-L, GFAP, UCHL-1 and CSF SNAP-25 in a cohort (n = 173) of preclinical AD, MCI-AD, AD dementia, frontotemporal dementia patients, and controls. We found a significant correlation between CSF and plasma levels of Aβ42/40, p-tau181, p-tau231, NF-L, and GFAP, while no CSF-plasma correlation was observed for t-tau and UCHL-1. Next to the core CSF biomarkers (Aβ42/40, p-tau181, t-tau), those providing the best discrimination between controls and preclinical AD were CSF p-tau231 and SNAP-25 and plasma Aβ42/40, p-tau231, and GFAP. Among plasma biomarkers, we found Aβ42/Aβ40, GFAP, and p-tau231 to show the largest rate of change at the CSF biomarker-defined cut-offs for amyloidosis and tauopathy. Finally, we identified GFAP, NF-L, and p-tau181 as the biomarkers most significantly associated with disease progression in both CSF and plasma. We suggest that a well-standardized and validated panel of selected plasma markers can facilitate early AD diagnosis, even at the asymptomatic disease stage. We propose that both CSF and plasma measurement of NF-L, p-tau181, and GFAP may play a significant role in disease staging and monitoring.
Collapse
Affiliation(s)
- Anna Lidia Wojdała
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy
| | - Giovanni Bellomo
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy
| | - Andrea Toja
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy
| | - Elena Chipi
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy
| | - Dandan Shan
- Quanterix Corporation, Billerica, MA 01821, United States of America
| | - Davide Chiasserini
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy.
| | - Lucilla Parnetti
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy; Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy.
| |
Collapse
|
11
|
Gokce M, Velioglu HA, Bektay MY, Guler EM. Evaluating the Clinical Significance of Diazepam Binding Inhibitor in Alzheimer's Disease: A Comparison with Inflammatory, Oxidative, and Neurodegenerative Biomarkers. Gerontology 2023; 69:1104-1112. [PMID: 37607528 DOI: 10.1159/000531849] [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: 09/27/2022] [Accepted: 06/26/2023] [Indexed: 08/24/2023] Open
Abstract
INTRODUCTION Alzheimer's disease (AD) is one of the pathologies that the scientific world is still desperate for. The aim of this study was the investigation of diazepam binding inhibitor (DBI) as a prognostic factor for AD prognosis. METHODS A total of 120 participants were divided into 3 groups. Forty new diagnosed Alzheimer patients (NDG) who have been diagnosed but have not started AD treatment, 40 patients who diagnosed 5 years ago (D5YG), and 40 healthy control groups (CG) were included in the study. Levels of DBI, oxidative stress, inflammatory, and neurodegenerative biomarkers were compared between 3 groups. RESULTS Plasma levels of DBI, oligomeric Aβ, total tau, glial fibrillary acidic protein, α-synuclein, interleukin (IL) 1β, IL6, tumor necrosis factor α, oxidative stress index, high-sensitive C-reactive protein, and DNA damage were found higher in D5YG and NDG as compared to CG (p < 0.001). On the contrary, plasma levels of total thiol, native thiol, vitamin D and vitamin B12 were lower in D5YG and NDG as compared to CG (p < 0.001). DISCUSSION DBI may be a potential plasma biomarker and promising drug target for AD. It could help physicians make a comprehensive evaluation with cognitive and neurodegenerative tests.
Collapse
Affiliation(s)
- Mustafa Gokce
- Department of Pharmacology, Bezmialem Vakif University School of Pharmacy, Istanbul, Turkey
- Department of Pharmacology, Institute of Health Sciences, Istanbul University, Istanbul, Turkey
| | - Halil Aziz Velioglu
- Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, New York, USA
- Functional Imaging and Cognitive-Affective Neuroscience Lab (fINCAN), Health Sciences and Technology Research Institute (SABITA), Regenerative and Restorative Medicine Research Center (REMER), Istanbul Medipol University, Istanbul, Turkey
| | - Muhammed Yunus Bektay
- Department of Clinical Pharmacy, Bezmialem Vakif University School of Pharmacy, Istanbul, Turkey
| | - Eray Metin Guler
- Department of Medical Biochemistry, Faculty of Hamidiye Medicine, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
12
|
Villalobos D, Reese M, Wright MC, Wong M, Syed A, Park J, Hall A, Browndyke JN, Martucci KT, Devinney MJ, Acker L, Moretti EW, Talbot L, Colin B, Ohlendorf B, Waligorska T, Shaw LM, Whitson HE, Cohen HJ, Mathew JP, Berger M. Perioperative changes in neurocognitive and Alzheimer's disease-related cerebrospinal fluid biomarkers in older patients randomised to isoflurane or propofol for anaesthetic maintenance. Br J Anaesth 2023; 131:328-337. [PMID: 37271721 PMCID: PMC10375507 DOI: 10.1016/j.bja.2023.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 06/06/2023] Open
Abstract
BACKGROUND Animal studies have shown that isoflurane and propofol have differential effects on Alzheimer's disease (AD) pathology and memory, although it is unclear whether this occurs in humans. METHODS This was a nested randomised controlled trial within a prospective cohort study; patients age ≥60 yr undergoing noncardiac/non-neurological surgery were randomised to isoflurane or propofol for anaesthetic maintenance. Cerebrospinal fluid (CSF) was collected via lumbar puncture before, 24 h, and 6 weeks after surgery. Cognitive testing was performed before and 6 weeks after surgery. Nonparametric methods and linear regression were used to evaluate CSF biomarkers and cognitive function, respectively. RESULTS There were 107 subjects (54 randomised to isoflurane and 53 to propofol) who completed the 6-week follow-up and were included in the analysis. There was no significant effect of anaesthetic treatment group, time, or group-by-time interaction for CSF amyloid-beta (Aβ), tau, or phospho-tau181p levels, or on the tau/Aβ or p-tau181p/Aβ ratios (all P>0.05 after Bonferroni correction). In multivariable-adjusted intention-to-treat analyses, there were no significant differences between the isoflurane and propofol groups in 6-week postoperative change in overall cognition (mean difference [95% confidence interval]: 0.01 [-0.12 to 0.13]; P=0.89) or individual cognitive domains (P>0.05 for each). Results remained consistent across as-treated and per-protocol analyses. CONCLUSIONS Intraoperative anaesthetic maintenance with isoflurane vs propofol had no significant effect on postoperative cognition or CSF Alzheimer's disease-related biomarkers within 6 weeks after noncardiac, non-neurological surgery in older adults. CLINICAL TRIAL REGISTRATION NCT01993836.
Collapse
Affiliation(s)
| | - Melody Reese
- Department of Anaesthesiology, Duke University Medical Centre, Durham, NC, USA; Center for the Study of Aging and Human Development, Duke University Medical Centre, Durham, NC, USA
| | - Mary Cooter Wright
- Department of Anaesthesiology, Duke University Medical Centre, Durham, NC, USA
| | - Megan Wong
- Duke University School of Medicine, Durham, NC, USA
| | - Ayesha Syed
- Department of Anaesthesiology, Duke University Medical Centre, Durham, NC, USA; Trinity College, Duke University, Durham, NC, USA
| | - John Park
- Duke University School of Medicine, Durham, NC, USA; Department of Anaesthesiology, Duke University Medical Centre, Durham, NC, USA
| | - Ashley Hall
- Department of Anaesthesiology, Duke University Medical Centre, Durham, NC, USA
| | - Jeffrey N Browndyke
- Department of Psychiatry and Behavioural Medicine, Division of Behavioral Medicine & Neurosciences, Duke University Medical Center, Durham, NC, USA; Center for Cognitive Neuroscience, Duke University, Durham, NC, USA; Duke Brain Imaging and Analysis Center, Durham, NC, USA; Duke Institute for Brain Sciences, Durham, NC, USA
| | - Katherine T Martucci
- Department of Anaesthesiology, Duke University Medical Centre, Durham, NC, USA; Center for Cognitive Neuroscience, Duke University, Durham, NC, USA; Duke Brain Imaging and Analysis Center, Durham, NC, USA; Duke Institute for Brain Sciences, Durham, NC, USA
| | - Michael J Devinney
- Department of Anaesthesiology, Duke University Medical Centre, Durham, NC, USA
| | - Leah Acker
- Department of Anaesthesiology, Duke University Medical Centre, Durham, NC, USA
| | - Eugene W Moretti
- Department of Anaesthesiology, Duke University Medical Centre, Durham, NC, USA
| | - Leonard Talbot
- Department of Anaesthesiology, Duke University Medical Centre, Durham, NC, USA
| | - Brian Colin
- Department of Anaesthesiology, Duke University Medical Centre, Durham, NC, USA
| | - Brian Ohlendorf
- Department of Anaesthesiology, Duke University Medical Centre, Durham, NC, USA
| | - Teresa Waligorska
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Heather E Whitson
- Center for the Study of Aging and Human Development, Duke University Medical Centre, Durham, NC, USA; Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Harvey J Cohen
- Center for the Study of Aging and Human Development, Duke University Medical Centre, Durham, NC, USA; Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Joseph P Mathew
- Department of Anaesthesiology, Duke University Medical Centre, Durham, NC, USA
| | - Miles Berger
- Duke University School of Medicine, Durham, NC, USA; Department of Anaesthesiology, Duke University Medical Centre, Durham, NC, USA; Center for the Study of Aging and Human Development, Duke University Medical Centre, Durham, NC, USA; Center for Cognitive Neuroscience, Duke University, Durham, NC, USA; Duke Institute for Brain Sciences, Durham, NC, USA.
| |
Collapse
|
13
|
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
|
14
|
Pais MV, Forlenza OV, Diniz BS. Plasma Biomarkers of Alzheimer's Disease: A Review of Available Assays, Recent Developments, and Implications for Clinical Practice. J Alzheimers Dis Rep 2023; 7:355-380. [PMID: 37220625 PMCID: PMC10200198 DOI: 10.3233/adr-230029] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 04/03/2023] [Indexed: 05/25/2023] Open
Abstract
Recently, low-sensitive plasma assays have been replaced by new ultra-sensitive assays such as single molecule enzyme-linked immunosorbent assay (Simoa), the Mesoscale Discovery (MSD) platform, and immunoprecipitation-mass spectrometry (IP-MS) with higher accuracy in the determination of plasma biomarkers of Alzheimer's disease (AD). Despite the significant variability, many studies have established in-house cut-off values for the most promising available biomarkers. We first reviewed the most used laboratory methods and assays to measure plasma AD biomarkers. Next, we review studies focused on the diagnostic performance of these biomarkers to identify AD cases, predict cognitive decline in pre-clinical AD cases, and differentiate AD cases from other dementia. We summarized data from studies published until January 2023. A combination of plasma Aβ42/40 ratio, age, and APOE status showed the best accuracy in diagnosing brain amyloidosis with a liquid chromatography-mass spectrometry (LC-MS) assay. Plasma p-tau217 has shown the best accuracy in distinguishing Aβ-PET+ from Aβ-PET-even in cognitively unimpaired individuals. We also summarized the different cut-off values for each biomarker when available. Recently developed assays for plasma biomarkers have undeniable importance in AD research, with improved analytical and diagnostic performance. Some biomarkers have been extensively used in clinical trials and are now clinically available. Nonetheless, several challenges remain to their widespread use in clinical practice.
Collapse
Affiliation(s)
- Marcos V. Pais
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Faculdade de Medicina, Universidade de Sao Paulo (FMUSP), Sao Paulo, SP, Brazil
| | - Orestes V. Forlenza
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Faculdade de Medicina, Universidade de Sao Paulo (FMUSP), Sao Paulo, SP, Brazil
| | - Breno S. Diniz
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
15
|
Weigand AJ, Ortiz G, Walker KS, Galasko DR, Bondi MW, Thomas KR. APOE differentially moderates cerebrospinal fluid and plasma phosphorylated tau181 associations with multi-domain cognition. Neurobiol Aging 2023; 125:1-8. [PMID: 36780762 DOI: 10.1016/j.neurobiolaging.2022.10.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 01/19/2023]
Abstract
Biofluid markers of phosphorylated tau181 (p-tau181) are increasingly popular for the detection of early Alzheimer's pathologic changes. However, the differential dynamics of cerebrospinal fluid (CSF) and plasma p-tau181 remain under investigation. We studied 727 participants from the Alzheimer's Disease Neuroimaging Initiative with plasma and CSF p-tau181 data, apolipoprotein (APOE) ε4 carrier status, amyloid positron emission tomography (PET) imaging, and neuropsychological data. Higher levels of plasma and CSF p-tau181 were observed among APOE ε4 carriers. CSF and plasma p-tau181 were significantly associated with memory, and this effect was greater in APOE ε4 carriers. However, whereas CSF p-tau181 was not significantly associated with language or attention/executive function among ε4 carriers or non-carriers, APOE ε4 status moderated the association of plasma p-tau181 with both language and attention/executive function. These findings lend support to the notion that p-tau181 biofluid markers are useful in measuring AD pathologic changes but also suggest that CSF and plasma p-tau181 have unique properties and dynamics that should be considered when using these markers in research and clinical practice.
Collapse
Affiliation(s)
- Alexandra J Weigand
- San Diego State University/University of California San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
| | - Gema Ortiz
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Kayla S Walker
- San Diego State University, Department of Psychology, San Diego, CA, USA
| | - Douglas R Galasko
- VA San Diego Healthcare System, San Diego, CA, USA; University of California San Diego, Department of Neurosciences, La Jolla, CA, USA
| | - Mark W Bondi
- VA San Diego Healthcare System, San Diego, CA, USA; University of California San Diego, Department of Psychiatry, La Jolla, CA, USA
| | - Kelsey R Thomas
- VA San Diego Healthcare System, San Diego, CA, USA; University of California San Diego, Department of Psychiatry, La Jolla, CA, USA.
| | | |
Collapse
|
16
|
Huang X, Li Y, Fowler C, Doecke JD, Lim YY, Drysdale C, Zhang V, Park K, Trounson B, Pertile K, Rumble R, Pickering JW, Rissman RA, Sarsoza F, Abdel‐Latif S, Lin Y, Doré V, Villemagne V, Rowe CC, Fripp J, Martins R, Wiley JS, Maruff P, Mintzer JE, Masters CL, Gu BJ. Leukocyte surface biomarkers implicate deficits of innate immunity in sporadic Alzheimer's disease. Alzheimers Dement 2023; 19:2084-2094. [PMID: 36349985 PMCID: PMC10166765 DOI: 10.1002/alz.12813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 08/24/2022] [Accepted: 09/02/2022] [Indexed: 11/10/2022]
Abstract
INTRODUCTION Blood-based diagnostics and prognostics in sporadic Alzheimer's disease (AD) are important for identifying at-risk individuals for therapeutic interventions. METHODS In three stages, a total of 34 leukocyte antigens were examined by flow cytometry immunophenotyping. Data were analyzed by logistic regression and receiver operating characteristic (ROC) analyses. RESULTS We identified leukocyte markers differentially expressed in the patients with AD. Pathway analysis revealed a complex network involving upregulation of complement inhibition and downregulation of cargo receptor activity and Aβ clearance. A proposed panel including four leukocyte markers - CD11c, CD59, CD91, and CD163 - predicts patients' PET Aβ status with an area under the curve (AUC) of 0.93 (0.88 to 0.97). CD163 was the top performer in preclinical models. These findings have been validated in two independent cohorts. CONCLUSION Our finding of changes on peripheral leukocyte surface antigens in AD implicates the deficit in innate immunity. Leukocyte-based biomarkers prove to be both sensitive and practical for AD screening and diagnosis.
Collapse
Affiliation(s)
- Xin Huang
- The Florey Institute of Neurosciencethe University of MelbourneParkvilleVictoriaAustralia
| | - Yihan Li
- The Florey Institute of Neurosciencethe University of MelbourneParkvilleVictoriaAustralia
| | - Christopher Fowler
- The Florey Institute of Neurosciencethe University of MelbourneParkvilleVictoriaAustralia
| | - James D. Doecke
- The Australian e‐Health Research CentreCSIROBrisbaneQueenslandAustralia
| | - Yen Ying Lim
- Turner Institute for Brain and Mental HealthSchool of Psychological SciencesMonash UniversityClaytonVictoriaAustralia
| | - Candace Drysdale
- The Florey Institute of Neurosciencethe University of MelbourneParkvilleVictoriaAustralia
| | - Vicky Zhang
- The Florey Institute of Neurosciencethe University of MelbourneParkvilleVictoriaAustralia
| | - Keunha Park
- The Florey Institute of Neurosciencethe University of MelbourneParkvilleVictoriaAustralia
| | - Brett Trounson
- The Florey Institute of Neurosciencethe University of MelbourneParkvilleVictoriaAustralia
| | - Kelly Pertile
- The Florey Institute of Neurosciencethe University of MelbourneParkvilleVictoriaAustralia
| | - Rebecca Rumble
- The Florey Institute of Neurosciencethe University of MelbourneParkvilleVictoriaAustralia
| | - John W. Pickering
- Department of MedicineUniversity of OtagoNew Zealand and Department of Emergency MedicineChristchurch HospitalChristchurchNew Zealand
| | - Robert A. Rissman
- Department of NeurosciencesUniversity of CaliforniaSan DiegoCaliforniaUSA
| | - Floyd Sarsoza
- Department of NeurosciencesUniversity of CaliforniaSan DiegoCaliforniaUSA
| | - Sara Abdel‐Latif
- Alzheimer's Therapeutic Research InstituteUniversity of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Yong Lin
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
| | - Vincent Doré
- The Australian e‐Health Research CentreCSIROBrisbaneQueenslandAustralia
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Australia, and Department of Medicinethe University of MelbourneMelbourneAustralia
| | - Victor Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Australia, and Department of Medicinethe University of MelbourneMelbourneAustralia
| | - Christopher C. Rowe
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Australia, and Department of Medicinethe University of MelbourneMelbourneAustralia
| | - Jurgen Fripp
- The Australian e‐Health Research CentreCSIROBrisbaneQueenslandAustralia
| | - Ralph Martins
- Centre of Excellence for Alzheimer's Disease Research and CareSchool of Medical and Health SciencesEdith Cowan UniversityJoondalupWestern AustraliaAustralia
| | - James S. Wiley
- The Florey Institute of Neurosciencethe University of MelbourneParkvilleVictoriaAustralia
| | - Paul Maruff
- The Florey Institute of Neurosciencethe University of MelbourneParkvilleVictoriaAustralia
- CogState Ltd.MelbourneVictoriaAustralia
| | | | - Colin L. Masters
- The Florey Institute of Neurosciencethe University of MelbourneParkvilleVictoriaAustralia
| | - Ben J. Gu
- The Florey Institute of Neurosciencethe University of MelbourneParkvilleVictoriaAustralia
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
17
|
Min J, Rouanet J, Martini AC, Nashiro K, Yoo HJ, Porat S, Cho C, Wan J, Cole SW, Head E, Nation DA, Thayer JF, Mather M. Modulating heart rate oscillation affects plasma amyloid beta and tau levels in younger and older adults. Sci Rep 2023; 13:3967. [PMID: 36894565 PMCID: PMC9998394 DOI: 10.1038/s41598-023-30167-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 02/16/2023] [Indexed: 03/11/2023] Open
Abstract
Slow paced breathing via heart rate variability (HRV) biofeedback stimulates vagus-nerve pathways that counter noradrenergic stress and arousal pathways that can influence production and clearance of Alzheimer's disease (AD)-related proteins. Thus, we examined whether HRV biofeedback intervention affects plasma Αβ40, Αβ42, total tau (tTau), and phosphorylated tau-181 (pTau-181) levels. We randomized healthy adults (N = 108) to use slow-paced breathing with HRV biofeedback to increase heart rate oscillations (Osc+) or to use personalized strategies with HRV biofeedback to decrease heart rate oscillations (Osc-). They practiced 20-40 min daily. Four weeks of practicing the Osc+ and Osc- conditions produced large effect size differences in change in plasma Aβ40 and Aβ42 levels. The Osc+ condition decreased plasma Αβ while the Osc- condition increased Αβ. Decreases in Αβ were associated with decreases in gene transcription indicators of β-adrenergic signaling, linking effects to the noradrenergic system. There were also opposing effects of the Osc+ and Osc- interventions on tTau for younger adults and pTau-181 for older adults. These results provide novel data supporting a causal role of autonomic activity in modulating plasma AD-related biomarkers.Trial registration: NCT03458910 (ClinicalTrials.gov); first posted on 03/08/2018.
Collapse
Affiliation(s)
- Jungwon Min
- University of Southern California, Los Angeles, CA, USA
| | | | | | - Kaoru Nashiro
- University of Southern California, Los Angeles, CA, USA
| | - Hyun Joo Yoo
- University of Southern California, Los Angeles, CA, USA
| | - Shai Porat
- University of Southern California, Los Angeles, CA, USA
| | - Christine Cho
- University of Southern California, Los Angeles, CA, USA
| | - Junxiang Wan
- University of Southern California, Los Angeles, CA, USA
| | - Steve W Cole
- University of California, Los Angeles, Los Angeles, CA, USA
| | | | | | | | - Mara Mather
- University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
18
|
Kováč A, Majerová P, Nytka M, Cechová MZ, Bednář P, Hájek R, Cooper-Shepherd DA, Muck A, Lemr K. Separation of Isomeric Tau Phosphopeptides from Alzheimer's Disease Brain by Cyclic Ion Mobility Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:394-400. [PMID: 36706338 PMCID: PMC10017020 DOI: 10.1021/jasms.2c00289] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 06/18/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder of increasing concern. It belongs to diseases termed tauopathies which are characterized by inclusions of abnormally hyperphosphorylated and truncated forms of the protein tau. Studies of tauopathies often focus on detection and characterization of these aberrant tau proteoforms, in particular the phosphorylation sites, which represent a significant analytical challenge for example when several phosphosites can be present on the same peptide. Such isomers can even be difficult to fully separate chromatographically. Since recently introduced cyclic ion mobility-mass spectrometry can offer different selectivity, we have investigated the closely positioned phosphorylation sites S214, T212, and T217 of a tryptic peptide from proline rich region of tau-TPSLPTPPTREPK. The conformational heterogeneity of the isomeric peptides in the gas phase hindered their separation due to their overlapping arrival time distributions. Increasing the resolution of the analysis alone is insufficient to distinguish the peptides in a mixture typical of patient samples. We therefore developed a method based on a combination of collision-induced dissociation, isomeric product ions (m/z 677) mobility separation and post-mobility dissociation to aid in analyzing the isomeric phosphopeptides of tau in diseased brain extract. For all three isomers (T212, S214, and T217), the ion mobility signal of the ion at m/z 677 was still observable at the concentration of 0.1 nmol/L. This work not only offers insights into the phosphorylation of tau protein in AD but also provides an analytical workflow for the characterization of challenging pathological protein modifications in neurodegenerative diseases.
Collapse
Affiliation(s)
- Andrej Kováč
- Institute
of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 10 Bratislava, Slovak Republic
| | - Petra Majerová
- Axon
Neuroscience R&D Services SE, Dvořákovo nábrežie 10, 811 02 Bratislava, Slovak Republic
| | - Marianna Nytka
- Department
of Analytical Chemistry, Faculty of Science, Palacky University, 17 listopadu 12, 771 46 Olomouc, Czech Republic
| | - Monika Zajacová Cechová
- Department
of Analytical Chemistry, Faculty of Science, Palacky University, 17 listopadu 12, 771 46 Olomouc, Czech Republic
| | - Petr Bednář
- Department
of Analytical Chemistry, Faculty of Science, Palacky University, 17 listopadu 12, 771 46 Olomouc, Czech Republic
| | - Roman Hájek
- Waters
Corporation, Stamford Avenue, Altrincham Road, Wilmslow SK9 4AX, United Kingdom
| | | | - Alexander Muck
- Waters
Corporation, Stamford Avenue, Altrincham Road, Wilmslow SK9 4AX, United Kingdom
| | - Karel Lemr
- Department
of Analytical Chemistry, Faculty of Science, Palacky University, 17 listopadu 12, 771 46 Olomouc, Czech Republic
- Institute
of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| |
Collapse
|
19
|
Role of Tau in Various Tauopathies, Treatment Approaches, and Emerging Role of Nanotechnology in Neurodegenerative Disorders. Mol Neurobiol 2023; 60:1690-1720. [PMID: 36562884 DOI: 10.1007/s12035-022-03164-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
A few protein kinases and phosphatases regulate tau protein phosphorylation and an imbalance in their enzyme activity results in tau hyper-phosphorylation. Aberrant tau phosphorylation causes tau to dissociate from the microtubules and clump together in the cytosol to form neurofibrillary tangles (NFTs), which lead to the progression of neurodegenerative disorders including Alzheimer's disease (AD) and other tauopathies. Hence, targeting hyperphosphorylated tau protein is a restorative approach for treating neurodegenerative tauopathies. The cyclin-dependent kinase (Cdk5) and the glycogen synthase kinase (GSK3β) have both been implicated in aberrant tau hyperphosphorylation. The limited transport of drugs through the blood-brain barrier (BBB) for reaching the central nervous system (CNS) thus represents a significant problem in the development of drugs. Drug delivery systems based on nanocarriers help solve this problem. In this review, we discuss the tau protein, regulation of tau phosphorylation and abnormal hyperphosphorylation, drugs in use or under clinical trials, and treatment strategies for tauopathies based on the critical role of tau hyperphosphorylation in the pathogenesis of the disease. Pathology of neurodegenerative disease due to hyperphosphorylation and various therapeutic approaches including nanotechnology for its treatment.
Collapse
|
20
|
Abed SS, Hamdan FB, Hussein MM, Al-Mayah QS. Plasma tau and neurofilament light chain as biomarkers of Alzheimer's disease and their relation to cognitive functions. J Med Life 2023; 16:284-289. [PMID: 36937471 PMCID: PMC10015560 DOI: 10.25122/jml-2022-0251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/03/2023] [Indexed: 03/21/2023] Open
Abstract
Alzheimer's disease (AD) dementia is the most frequent cause of neurodegenerative dementia. The cognitive and behavioral symptoms associated with this disorder often have overlapping characteristics, potentially resulting in delayed diagnosis or misdiagnosis. This study aimed to assess the level of peripheral blood neurofilament light chain (NfL) and total tau (t-tau) protein in AD patients and investigate their relationship with cognitive impairment. The study included 80 participants of both sexes between the ages of 60 to 85 years. The participants were divided into two groups, consisting of 40 individuals in the control group (mean age 75±6.6 years) who had no cognitive or functional impairments and 40 AD patients (mean age 74.98±5.03 years). This study utilized the DSM-5 diagnostic criteria for major or mild neurocognitive disorder attributed to Alzheimer's disease (AD). The clinical and biochemical features of all participants were documented, and the Alzheimer's disease Assessment Scale cognitive subscale (ADAS-cog) scores were evaluated. Sandwich ELISA was employed to determine serum NfL and t-tau protein levels. The median serum NfL and t-tau protein levels in AD patients were significantly higher than those of the controls (47.84 pg/ml versus 17.66 pg/ml and 12.05 pg/ml versus 11.13 pg/ml, respectively). Age was positively correlated with NfL, t-tau levels, and ADAS-cog. Although elevated NfL and t-tau protein levels may play a role in disease progression, their diagnostic value for AD was limited.
Collapse
Affiliation(s)
- Sadiruldeen Sami Abed
- Department of Pharmacy, Osol Aldeen University College, Baghdad, Iraq
- Corresponding Author: Sadiruldeen Sami Abed, Department of Pharmacy, Osol Aldeen University College, Baghdad, Iraq. E-mail:
| | - Farqad Bader Hamdan
- Department of Physiology, College of Medicine, Al-Nahrain University, Baghdad, Iraq
| | | | | |
Collapse
|
21
|
Hayes JP, Pierce ME, Brown E, Salat D, Logue MW, Constantinescu J, Valerio K, Miller MW, Sherva R, Huber BR, Milberg W, McGlinchey R. Genetic Risk for Alzheimer Disease and Plasma Tau Are Associated With Accelerated Parietal Cortex Thickness Change in Middle-Aged Adults. Neurol Genet 2023; 9:e200053. [PMID: 36742995 PMCID: PMC9893442 DOI: 10.1212/nxg.0000000000200053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/21/2022] [Indexed: 02/04/2023]
Abstract
Background and Objectives Neuroimaging and biomarker studies in Alzheimer disease (AD) have shown well-characterized patterns of cortical thinning and altered biomarker concentrations of tau and β-amyloid (Aβ). However, earlier identification of AD has great potential to advance clinical care and determine candidates for drug trials. The extent to which AD risk markers relate to cortical thinning patterns in midlife is unknown. The first objective of this study was to examine cortical thickness change associated with genetic risk for AD among middle-aged military veterans. The second objective was to determine the relationship between plasma tau and Aβ and change in brain cortical thickness among veterans stratified by genetic risk for AD. Methods Participants consisted of post-9/11 veterans (N = 155) who were consecutively enrolled in the Translational Research Center for TBI and Stress Disorders prospective longitudinal cohort and were assessed for mild traumatic brain injury (TBI) and posttraumatic disorder (PTSD). Genome-wide polygenic risk scores (PRSs) for AD were calculated using summary results from the International Genomics of Alzheimer's Disease Project. T-tau and Aβ40 and Aβ42 plasma assays were run using Simoa technology. Whole-brain MRI cortical thickness change estimates were obtained using the longitudinal stream of FreeSurfer. Follow-up moderation analyses examined the AD PRS × plasma interaction on change in cortical thickness in AD-vulnerable regions. Results Higher AD PRS, signifying greater genetic risk for AD, was associated with accelerated cortical thickness change in a right hemisphere inferior parietal cortex cluster that included the supramarginal gyrus, angular gyrus, and intraparietal sulcus. Higher tau, but not Aβ42/40 ratio, was associated with greater cortical thickness change among those with higher AD PRS. Mild TBI and PTSD were not associated with cortical thickness change. Discussion Plasma tau, particularly when combined with genetic stratification for AD risk, can be a useful indicator of brain change in midlife. Accelerated inferior parietal cortex changes in midlife may be an important factor to consider as a marker of AD-related brain alterations.
Collapse
Affiliation(s)
- Jasmeet Pannu Hayes
- Department of Psychology (J.P.H., K.V.), The Ohio State University, & Chronic Brain Injury Program, The Ohio State University, Columbus; Translational Research Center for TBI and Stress Disorders (TRACTS) (M.E.P., E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Department of Psychiatry (M.E.P., M.W.L., M.W.M., B.R.H.), Boston University School of Medicine, MA; Neuroimaging Research for Veterans (NeRVe) Center (E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Brain Aging and Dementia (BAnD) Laboratory (D.S.), A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown; National Center for PTSD (M.W.L., M.W.M., B.R.H.), Behavioral Sciences Division, VA Boston Healthcare System, MA; Boston University School of Medicine (M.W.L., R.S.), Biomedical Genetics, MA; Boston University School of Public Health (M.W.L.), Department of Biostatistics, MA; Department of Neurology (B.R.H.), Boston University School of Medicine, MA; Geriatric Research (W.M., R.M.), Education, and Clinical Center (GRECC), VA Boston Healthcare System, MA; and Department of Psychiatry (W.M., R.M.), Harvard Medical School, Boston, MA
| | - Meghan E Pierce
- Department of Psychology (J.P.H., K.V.), The Ohio State University, & Chronic Brain Injury Program, The Ohio State University, Columbus; Translational Research Center for TBI and Stress Disorders (TRACTS) (M.E.P., E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Department of Psychiatry (M.E.P., M.W.L., M.W.M., B.R.H.), Boston University School of Medicine, MA; Neuroimaging Research for Veterans (NeRVe) Center (E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Brain Aging and Dementia (BAnD) Laboratory (D.S.), A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown; National Center for PTSD (M.W.L., M.W.M., B.R.H.), Behavioral Sciences Division, VA Boston Healthcare System, MA; Boston University School of Medicine (M.W.L., R.S.), Biomedical Genetics, MA; Boston University School of Public Health (M.W.L.), Department of Biostatistics, MA; Department of Neurology (B.R.H.), Boston University School of Medicine, MA; Geriatric Research (W.M., R.M.), Education, and Clinical Center (GRECC), VA Boston Healthcare System, MA; and Department of Psychiatry (W.M., R.M.), Harvard Medical School, Boston, MA
| | - Emma Brown
- Department of Psychology (J.P.H., K.V.), The Ohio State University, & Chronic Brain Injury Program, The Ohio State University, Columbus; Translational Research Center for TBI and Stress Disorders (TRACTS) (M.E.P., E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Department of Psychiatry (M.E.P., M.W.L., M.W.M., B.R.H.), Boston University School of Medicine, MA; Neuroimaging Research for Veterans (NeRVe) Center (E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Brain Aging and Dementia (BAnD) Laboratory (D.S.), A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown; National Center for PTSD (M.W.L., M.W.M., B.R.H.), Behavioral Sciences Division, VA Boston Healthcare System, MA; Boston University School of Medicine (M.W.L., R.S.), Biomedical Genetics, MA; Boston University School of Public Health (M.W.L.), Department of Biostatistics, MA; Department of Neurology (B.R.H.), Boston University School of Medicine, MA; Geriatric Research (W.M., R.M.), Education, and Clinical Center (GRECC), VA Boston Healthcare System, MA; and Department of Psychiatry (W.M., R.M.), Harvard Medical School, Boston, MA
| | - David Salat
- Department of Psychology (J.P.H., K.V.), The Ohio State University, & Chronic Brain Injury Program, The Ohio State University, Columbus; Translational Research Center for TBI and Stress Disorders (TRACTS) (M.E.P., E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Department of Psychiatry (M.E.P., M.W.L., M.W.M., B.R.H.), Boston University School of Medicine, MA; Neuroimaging Research for Veterans (NeRVe) Center (E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Brain Aging and Dementia (BAnD) Laboratory (D.S.), A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown; National Center for PTSD (M.W.L., M.W.M., B.R.H.), Behavioral Sciences Division, VA Boston Healthcare System, MA; Boston University School of Medicine (M.W.L., R.S.), Biomedical Genetics, MA; Boston University School of Public Health (M.W.L.), Department of Biostatistics, MA; Department of Neurology (B.R.H.), Boston University School of Medicine, MA; Geriatric Research (W.M., R.M.), Education, and Clinical Center (GRECC), VA Boston Healthcare System, MA; and Department of Psychiatry (W.M., R.M.), Harvard Medical School, Boston, MA
| | - Mark W Logue
- Department of Psychology (J.P.H., K.V.), The Ohio State University, & Chronic Brain Injury Program, The Ohio State University, Columbus; Translational Research Center for TBI and Stress Disorders (TRACTS) (M.E.P., E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Department of Psychiatry (M.E.P., M.W.L., M.W.M., B.R.H.), Boston University School of Medicine, MA; Neuroimaging Research for Veterans (NeRVe) Center (E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Brain Aging and Dementia (BAnD) Laboratory (D.S.), A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown; National Center for PTSD (M.W.L., M.W.M., B.R.H.), Behavioral Sciences Division, VA Boston Healthcare System, MA; Boston University School of Medicine (M.W.L., R.S.), Biomedical Genetics, MA; Boston University School of Public Health (M.W.L.), Department of Biostatistics, MA; Department of Neurology (B.R.H.), Boston University School of Medicine, MA; Geriatric Research (W.M., R.M.), Education, and Clinical Center (GRECC), VA Boston Healthcare System, MA; and Department of Psychiatry (W.M., R.M.), Harvard Medical School, Boston, MA
| | - Julie Constantinescu
- Department of Psychology (J.P.H., K.V.), The Ohio State University, & Chronic Brain Injury Program, The Ohio State University, Columbus; Translational Research Center for TBI and Stress Disorders (TRACTS) (M.E.P., E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Department of Psychiatry (M.E.P., M.W.L., M.W.M., B.R.H.), Boston University School of Medicine, MA; Neuroimaging Research for Veterans (NeRVe) Center (E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Brain Aging and Dementia (BAnD) Laboratory (D.S.), A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown; National Center for PTSD (M.W.L., M.W.M., B.R.H.), Behavioral Sciences Division, VA Boston Healthcare System, MA; Boston University School of Medicine (M.W.L., R.S.), Biomedical Genetics, MA; Boston University School of Public Health (M.W.L.), Department of Biostatistics, MA; Department of Neurology (B.R.H.), Boston University School of Medicine, MA; Geriatric Research (W.M., R.M.), Education, and Clinical Center (GRECC), VA Boston Healthcare System, MA; and Department of Psychiatry (W.M., R.M.), Harvard Medical School, Boston, MA
| | - Kate Valerio
- Department of Psychology (J.P.H., K.V.), The Ohio State University, & Chronic Brain Injury Program, The Ohio State University, Columbus; Translational Research Center for TBI and Stress Disorders (TRACTS) (M.E.P., E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Department of Psychiatry (M.E.P., M.W.L., M.W.M., B.R.H.), Boston University School of Medicine, MA; Neuroimaging Research for Veterans (NeRVe) Center (E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Brain Aging and Dementia (BAnD) Laboratory (D.S.), A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown; National Center for PTSD (M.W.L., M.W.M., B.R.H.), Behavioral Sciences Division, VA Boston Healthcare System, MA; Boston University School of Medicine (M.W.L., R.S.), Biomedical Genetics, MA; Boston University School of Public Health (M.W.L.), Department of Biostatistics, MA; Department of Neurology (B.R.H.), Boston University School of Medicine, MA; Geriatric Research (W.M., R.M.), Education, and Clinical Center (GRECC), VA Boston Healthcare System, MA; and Department of Psychiatry (W.M., R.M.), Harvard Medical School, Boston, MA
| | - Mark W Miller
- Department of Psychology (J.P.H., K.V.), The Ohio State University, & Chronic Brain Injury Program, The Ohio State University, Columbus; Translational Research Center for TBI and Stress Disorders (TRACTS) (M.E.P., E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Department of Psychiatry (M.E.P., M.W.L., M.W.M., B.R.H.), Boston University School of Medicine, MA; Neuroimaging Research for Veterans (NeRVe) Center (E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Brain Aging and Dementia (BAnD) Laboratory (D.S.), A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown; National Center for PTSD (M.W.L., M.W.M., B.R.H.), Behavioral Sciences Division, VA Boston Healthcare System, MA; Boston University School of Medicine (M.W.L., R.S.), Biomedical Genetics, MA; Boston University School of Public Health (M.W.L.), Department of Biostatistics, MA; Department of Neurology (B.R.H.), Boston University School of Medicine, MA; Geriatric Research (W.M., R.M.), Education, and Clinical Center (GRECC), VA Boston Healthcare System, MA; and Department of Psychiatry (W.M., R.M.), Harvard Medical School, Boston, MA
| | - Richard Sherva
- Department of Psychology (J.P.H., K.V.), The Ohio State University, & Chronic Brain Injury Program, The Ohio State University, Columbus; Translational Research Center for TBI and Stress Disorders (TRACTS) (M.E.P., E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Department of Psychiatry (M.E.P., M.W.L., M.W.M., B.R.H.), Boston University School of Medicine, MA; Neuroimaging Research for Veterans (NeRVe) Center (E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Brain Aging and Dementia (BAnD) Laboratory (D.S.), A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown; National Center for PTSD (M.W.L., M.W.M., B.R.H.), Behavioral Sciences Division, VA Boston Healthcare System, MA; Boston University School of Medicine (M.W.L., R.S.), Biomedical Genetics, MA; Boston University School of Public Health (M.W.L.), Department of Biostatistics, MA; Department of Neurology (B.R.H.), Boston University School of Medicine, MA; Geriatric Research (W.M., R.M.), Education, and Clinical Center (GRECC), VA Boston Healthcare System, MA; and Department of Psychiatry (W.M., R.M.), Harvard Medical School, Boston, MA
| | - Bertrand Russell Huber
- Department of Psychology (J.P.H., K.V.), The Ohio State University, & Chronic Brain Injury Program, The Ohio State University, Columbus; Translational Research Center for TBI and Stress Disorders (TRACTS) (M.E.P., E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Department of Psychiatry (M.E.P., M.W.L., M.W.M., B.R.H.), Boston University School of Medicine, MA; Neuroimaging Research for Veterans (NeRVe) Center (E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Brain Aging and Dementia (BAnD) Laboratory (D.S.), A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown; National Center for PTSD (M.W.L., M.W.M., B.R.H.), Behavioral Sciences Division, VA Boston Healthcare System, MA; Boston University School of Medicine (M.W.L., R.S.), Biomedical Genetics, MA; Boston University School of Public Health (M.W.L.), Department of Biostatistics, MA; Department of Neurology (B.R.H.), Boston University School of Medicine, MA; Geriatric Research (W.M., R.M.), Education, and Clinical Center (GRECC), VA Boston Healthcare System, MA; and Department of Psychiatry (W.M., R.M.), Harvard Medical School, Boston, MA
| | - William Milberg
- Department of Psychology (J.P.H., K.V.), The Ohio State University, & Chronic Brain Injury Program, The Ohio State University, Columbus; Translational Research Center for TBI and Stress Disorders (TRACTS) (M.E.P., E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Department of Psychiatry (M.E.P., M.W.L., M.W.M., B.R.H.), Boston University School of Medicine, MA; Neuroimaging Research for Veterans (NeRVe) Center (E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Brain Aging and Dementia (BAnD) Laboratory (D.S.), A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown; National Center for PTSD (M.W.L., M.W.M., B.R.H.), Behavioral Sciences Division, VA Boston Healthcare System, MA; Boston University School of Medicine (M.W.L., R.S.), Biomedical Genetics, MA; Boston University School of Public Health (M.W.L.), Department of Biostatistics, MA; Department of Neurology (B.R.H.), Boston University School of Medicine, MA; Geriatric Research (W.M., R.M.), Education, and Clinical Center (GRECC), VA Boston Healthcare System, MA; and Department of Psychiatry (W.M., R.M.), Harvard Medical School, Boston, MA
| | - Regina McGlinchey
- Department of Psychology (J.P.H., K.V.), The Ohio State University, & Chronic Brain Injury Program, The Ohio State University, Columbus; Translational Research Center for TBI and Stress Disorders (TRACTS) (M.E.P., E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Department of Psychiatry (M.E.P., M.W.L., M.W.M., B.R.H.), Boston University School of Medicine, MA; Neuroimaging Research for Veterans (NeRVe) Center (E.B., D.S., J.C., W.M., R.M.), VA Boston Healthcare System, MA; Brain Aging and Dementia (BAnD) Laboratory (D.S.), A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown; National Center for PTSD (M.W.L., M.W.M., B.R.H.), Behavioral Sciences Division, VA Boston Healthcare System, MA; Boston University School of Medicine (M.W.L., R.S.), Biomedical Genetics, MA; Boston University School of Public Health (M.W.L.), Department of Biostatistics, MA; Department of Neurology (B.R.H.), Boston University School of Medicine, MA; Geriatric Research (W.M., R.M.), Education, and Clinical Center (GRECC), VA Boston Healthcare System, MA; and Department of Psychiatry (W.M., R.M.), Harvard Medical School, Boston, MA
| |
Collapse
|
22
|
Hsiao WWW, Angela S, Le TN, Ku CC, Hu PS, Chiang WH. Evolution of Detecting Early Onset of Alzheimer's Disease: From Neuroimaging to Optical Immunoassays. J Alzheimers Dis 2023; 93:821-845. [PMID: 37125550 DOI: 10.3233/jad-221202] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Alzheimer's disease (AD) is a pathological disorder defined by the symptoms of memory loss and deterioration of cognitive abilities over time. Although the etiology is complex, it is mainly associated with the accumulation of toxic amyloid-β peptide (Aβ) aggregates and tau protein-induced neurofibrillary tangles (NFTs). Even now, creating non-invasive, sensitive, specific, and cost-effective diagnostic methods for AD remains challenging. Over the past few decades, polymers, and nanomaterials (e.g., nanodiamonds, nanogold, quantum dots) have become attractive and practical tools in nanomedicine for diagnosis and treatment. This review focuses on current developments in sensing methods such as enzyme-linked immunosorbent assay (ELISA) and surface-enhanced Raman scattering (SERS) to boost the sensitivity in detecting related biomarkers for AD. In addition, optical analysis platforms such as ELISA and SERS have found increasing popularity among researchers due to their excellent sensitivity and specificity, which may go as low as the femtomolar range. While ELISA offers easy technological usage and high throughput, SERS has the advantages of improved mobility, simple electrical equipment integration, and lower cost. Both portable optical sensing techniques are highly superior in terms of sensitivity, specificity, human application, and practicality, enabling the early identification of AD biomarkers.
Collapse
Affiliation(s)
- Wesley Wei-Wen Hsiao
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan, R.O.C
| | - Stefanny Angela
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan, R.O.C
| | - Trong-Nghia Le
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, Taiwan, R.O.C
| | - Chia-Chi Ku
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan, R.O.C
| | - Po-Sheng Hu
- College of Photonics, National Yang Ming Chiao Tung University, Tainan City, Taiwan, R.O.C
| | - Wei-Hung Chiang
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan, R.O.C
| |
Collapse
|
23
|
Rodrigues Martins D, Vandermeeren M, Van Kolen K, Brepoels E, Borgers M, Wintmolders C, Delay C, Bottelbergs A, Mercken M, Theunis C. Development and Characterization of Mouse-Specific Anti-Tau Monoclonal Antibodies: Relevance for Analysis of Murine Tau in Cerebrospinal Fluid. J Alzheimers Dis 2023; 93:151-167. [PMID: 36970909 DOI: 10.3233/jad-221266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
BACKGROUND Clearance of tau seeds by immunization with tau antibodies is currently evaluated as therapeutic strategy to block the spreading of tau pathology in Alzheimer's disease and other tauopathies. Preclinical evaluation of passive immunotherapy is performed in different cellular culture systems and in wild-type and human tau transgenic mouse models. Depending on the preclinical model used, tau seeds or induced aggregates can either be of mouse, human or mixed origin. OBJECTIVE We aimed to develop human and mouse tau-specific antibodies to discriminate between the endogenous tau and the introduced form in preclinical models. METHODS Using hybridoma technology, we developed human and mouse tau-specific antibodies that were then used to develop several assays to specifically detect mouse tau. RESULTS Four antibodies, mTau3, mTau5, mTau8, and mTau9, with a high degree of specificity for mouse tau were identified. Additionally, their potential application in highly sensitive immunoassays to measure tau in mouse brain homogenate and cerebrospinal fluid is illustrated, as well as their application for specific endogenous mouse tau aggregation detection. CONCLUSION The antibodies reported here can be very important tools to better interpret the results obtained from different model systems as well as to study the role of endogenous tau in tau aggregation and pathology observed in the diverse mouse models available.
Collapse
Affiliation(s)
- Dina Rodrigues Martins
- Neuroscience Department, Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Marc Vandermeeren
- Neuroscience Department, Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Kristof Van Kolen
- Neuroscience Department, Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Eddy Brepoels
- Biologics Research, Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Marianne Borgers
- Neuroscience Department, Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Cindy Wintmolders
- Neuroscience Department, Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Charlotte Delay
- Neuroscience Department, Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Astrid Bottelbergs
- Neuroscience Department, Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Marc Mercken
- Neuroscience Department, Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Clara Theunis
- Neuroscience Department, Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| |
Collapse
|
24
|
Extra-Virgin Olive Oil Enhances the Blood-Brain Barrier Function in Mild Cognitive Impairment: A Randomized Controlled Trial. Nutrients 2022; 14:nu14235102. [PMID: 36501136 PMCID: PMC9736478 DOI: 10.3390/nu14235102] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Mild cognitive impairment (MCI) and early Alzheimer's disease (AD) are characterized by blood-brain barrier (BBB) breakdown leading to abnormal BBB permeability ahead of brain atrophy or dementia. Previous findings in AD mouse models have reported the beneficial effect of extra-virgin olive oil (EVOO) against AD, which improved BBB and memory functions and reduced brain amyloid-β (Aβ) and related pathology. This work aimed to translate these preclinical findings to humans in individuals with MCI. We examined the effect of daily consumption of refined olive oil (ROO) and EVOO for 6 months in MCI subjects on BBB permeability (assessed by contrast-enhanced MRI), and brain function (assessed using functional-MRI) as the primary outcomes. Cognitive function and AD blood biomarkers were also assessed as the secondary outcomes. Twenty-six participants with MCI were randomized with 25 participants completed the study. EVOO significantly improved clinical dementia rating (CDR) and behavioral scores. EVOO also reduced BBB permeability and enhanced functional connectivity. While ROO consumption did not alter BBB permeability or brain connectivity, it improved CDR scores and increased functional brain activation to a memory task in cortical regions involved in perception and cognition. Moreover, EVOO and ROO significantly reduced blood Aβ42/Aβ40 and p-tau/t-tau ratios, suggesting that both altered the processing and clearance of Aβ. In conclusion, EVOO and ROO improved CDR and behavioral scores; only EVOO enhanced brain connectivity and reduced BBB permeability, suggesting EVOO biophenols contributed to such an effect. This proof-of-concept study justifies further clinical trials to assess olive oil's protective effects against AD and its potential role in preventing MCI conversion to AD and related dementias.
Collapse
|
25
|
Ramesh M, Govindaraju T. Multipronged diagnostic and therapeutic strategies for Alzheimer's disease. Chem Sci 2022; 13:13657-13689. [PMID: 36544728 PMCID: PMC9710308 DOI: 10.1039/d2sc03932j] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/13/2022] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and a major contributor to dementia cases worldwide. AD is clinically characterized by learning, memory, and cognitive deficits. The accumulation of extracellular amyloid β (Aβ) plaques and neurofibrillary tangles (NFTs) of tau are the pathological hallmarks of AD and are explored as targets for clinical diagnosis and therapy. AD pathology is poorly understood and there are no fully approved diagnosis and treatments. Notwithstanding the gap, decades of research in understanding disease mechanisms have revealed the multifactorial nature of AD. As a result, multipronged and holistic approaches are pertinent to targeting multiple biomarkers and targets for developing effective diagnosis and therapeutics. In this perspective, recent developments in Aβ and tau targeted diagnostic and therapeutic tools are discussed. Novel indirect, combination, and circulating biomarkers as potential diagnostic targets are highlighted. We underline the importance of multiplexing and multimodal detection of multiple biomarkers to generate biomarker fingerprints as a reliable diagnostic strategy. The classical therapeutics targeting Aβ and tau aggregation pathways are described with bottlenecks in the strategy. Drug discovery efforts targeting multifaceted toxicity involving protein aggregation, metal toxicity, oxidative stress, mitochondrial damage, and neuroinflammation are highlighted. Recent efforts focused on multipronged strategies to rationally design multifunctional modulators targeting multiple pathological factors are presented as future drug development strategies to discover potential therapeutics for AD.
Collapse
Affiliation(s)
- Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru Karnataka 560064 India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru Karnataka 560064 India
| |
Collapse
|
26
|
Hier DB, Azizi S, Thimgan MS, Wunsch DC. Tau kinetics in Alzheimer's disease. Front Aging Neurosci 2022; 14:1055170. [PMID: 36437992 PMCID: PMC9682289 DOI: 10.3389/fnagi.2022.1055170] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/27/2022] [Indexed: 07/20/2023] Open
Abstract
The cytoskeletal protein tau is implicated in the pathogenesis of Alzheimer's disease which is characterized by intra-neuronal neurofibrillary tangles containing abnormally phosphorylated insoluble tau. Levels of soluble tau are elevated in the brain, the CSF, and the plasma of patients with Alzheimer's disease. To better understand the causes of these elevated levels of tau, we propose a three-compartment kinetic model (brain, CSF, and plasma). The model assumes that the synthesis of tau follows zero-order kinetics (uncorrelated with compartmental tau levels) and that the release, absorption, and clearance of tau is governed by first-order kinetics (linearly related to compartmental tau levels). Tau that is synthesized in the brain compartment can be released into the interstitial fluid, catabolized, or retained in neurofibrillary tangles. Tau released into the interstitial fluid can mix with the CSF and eventually drain to the plasma compartment. However, losses of tau in the drainage pathways may be significant. The kinetic model estimates half-life of tau in each compartment (552 h in the brain, 9.9 h in the CSF, and 10 h in the plasma). The kinetic model predicts that an increase in the neuronal tau synthesis rate or a decrease in tau catabolism rate best accounts for observed increases in tau levels in the brain, CSF, and plasma found in Alzheimer's disease. Furthermore, the model predicts that increases in brain half-life of tau in Alzheimer's disease should be attributed to decreased tau catabolism and not to increased tau synthesis. Most clearance of tau in the neuron occurs through catabolism rather than release to the CSF compartment. Additional experimental data would make ascertainment of the model parameters more precise.
Collapse
Affiliation(s)
- Daniel B. Hier
- Applied Computational Intelligence Laboratory, Department of Electrical & Computer Engineering, Missouri University of Science & Technology, Rolla, MO, United States
- Department of Neurology and Rehabilitation, University of Illinois at Chicago, Chicago, IL, United States
| | - Sima Azizi
- Applied Computational Intelligence Laboratory, Department of Electrical & Computer Engineering, Missouri University of Science & Technology, Rolla, MO, United States
| | - Matthew S. Thimgan
- Department of Biological Sciences, Missouri University of Science & Technology, Rolla, MO, United States
| | - Donald C. Wunsch
- Applied Computational Intelligence Laboratory, Department of Electrical & Computer Engineering, Missouri University of Science & Technology, Rolla, MO, United States
- ECCS Division, National Science Foundation, Alexandria, VA, United States
| |
Collapse
|
27
|
Sapkota S, Erickson K, Harvey D, Tomaszewski‐Farias SE, Olichney JM, Johnson DK, Dugger BN, Mungas DM, Fletcher E, Maillard P, Seshadri S, Satizabal CL, Kautz T, Parent D, Tracy RP, Maezawa I, Jin L, DeCarli C. Plasma biomarkers predict cognitive trajectories in an ethnoracially and clinically diverse cohort: Mediation with hippocampal volume. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12349. [PMID: 36092690 PMCID: PMC9434579 DOI: 10.1002/dad2.12349] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/25/2022] [Accepted: 06/26/2022] [Indexed: 11/11/2022]
Abstract
Introduction We examine whether the association between key plasma biomarkers (amyloid β [aβ] 42/40, total tau (t-tau), neurofilament light [NfL]) and cognitive trajectories (executive function [EF] and episodic memory [EM]) is mediated through neurodegeneration. Methods All participants were recruited from the University of California, Davis-Alzheimer's Disease Research Center (n = 473; baseline age range = 49-95 years, 60% women). We applied an accelerated longitudinal design to test latent growth models for EF and EM, and path and mediation analyses. Age was centered at 75 years, and all models were adjusted for sex, education, and ethnicity. Results HV differentially mediated the association aβ 42/40 and NfL on EF and EM level and change. Hippocampal volume (HV) did not mediate the association between t-tau and cognitive performance. Discussion Neurodegeneration as represented with HV selectively mediates the association between key non-invasive plasma biomarkers and cognitive trajectories in an ethnoracially and clinically diverse community-based sample.
Collapse
Affiliation(s)
- Shraddha Sapkota
- Department of NeurologyUniversity of CaliforniaDavisCaliforniaUSA
| | - Kelsey Erickson
- Department of Pathology and Laboratory MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Danielle Harvey
- Department of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | | | - John M. Olichney
- Department of NeurologyUniversity of CaliforniaDavisCaliforniaUSA
| | - David K. Johnson
- Department of NeurologyUniversity of CaliforniaDavisCaliforniaUSA
| | - Brittany N. Dugger
- Department of Pathology and Laboratory MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Dan M. Mungas
- Department of NeurologyUniversity of CaliforniaDavisCaliforniaUSA
| | - Evan Fletcher
- Department of NeurologyUniversity of CaliforniaDavisCaliforniaUSA
| | - Pauline Maillard
- Department of NeurologyUniversity of CaliforniaDavisCaliforniaUSA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases and Department of Population Health SciencesUT Health San AntonioSan AntonioTexasUSA
| | - Claudia L. Satizabal
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases and Department of Population Health SciencesUT Health San AntonioSan AntonioTexasUSA
- Department of NeurologyBoston University School of MedicineBostonMassachusettsUSA
- The Framingham Heart StudyFraminghamMassachusettsUSA
| | - Tiffany Kautz
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases and Department of Population Health SciencesUT Health San AntonioSan AntonioTexasUSA
| | - Danielle Parent
- Department of Pathology and Laboratory MedicineUniversity of VermontBurlingtonVermontUSA
| | - Russell P. Tracy
- Department of Pathology and Laboratory MedicineUniversity of VermontBurlingtonVermontUSA
| | - Izumi Maezawa
- Department of Pathology and Laboratory MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Lee‐Way Jin
- Department of Pathology and Laboratory MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Charles DeCarli
- Department of NeurologyUniversity of CaliforniaDavisCaliforniaUSA
| |
Collapse
|
28
|
Piccarducci R, Caselli MC, Zappelli E, Ulivi L, Daniele S, Siciliano G, Ceravolo R, Mancuso M, Baldacci F, Martini C. The Role of Amyloid-β, Tau, and α-Synuclein Proteins as Putative Blood Biomarkers in Patients with Cerebral Amyloid Angiopathy. J Alzheimers Dis 2022; 89:1039-1049. [PMID: 35964181 DOI: 10.3233/jad-220216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Cerebral amyloid angiopathy (CAA) is a cerebrovascular disorder characterized by the deposition of amyloid-β protein (Aβ) within brain blood vessels that develops in elderly people and Alzheimer's disease (AD) patients. Therefore, the investigation of biomarkers able to differentiate CAA patients from AD patients and healthy controls (HC) is of great interest, in particular in peripheral fluids. OBJECTIVE The current study aimed to detect the neurodegenerative disease (ND)-related protein (i.e., Aβ 1 - 40, Aβ 1 - 42, tau, and α-synuclein) levels in both red blood cells (RBCs) and plasma of CAA patients and HC, evaluating their role as putative peripheral biomarkers for CAA. METHODS For this purpose, the proteins' concentration was quantified in RBCs and plasma by homemade immunoenzymatic assays in an exploratory cohort of 20 CAA patients and 20 HC. RESULTS The results highlighted a significant increase of Aβ 1 - 40 and α-synuclein concentrations in both RBCs and plasma of CAA patients, while higher Aβ 1 - 42 and t-tau levels were detected only in RBCs of CAA individuals compared to HC. Moreover, Aβ 1 - 42/Aβ 1 - 40 ratio increased in RBCs and decreased in plasma of CAA patients. The role of these proteins as candidate peripheral biomarkers easily measurable with a blood sample in CAA needs to be confirmed in larger studies. CONCLUSION In conclusion, we provide evidence concerning the possible use of blood biomarkers for contributing to CAA diagnosis and differentiation from other NDs.
Collapse
Affiliation(s)
| | - Maria Chiara Caselli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Leonardo Ulivi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Michelangelo Mancuso
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | |
Collapse
|
29
|
Majerova P, Olesova D, Golisova G, Buralova M, Michalicova A, Vegh J, Piestansky J, Bhide M, Hanes J, Kovac A. Analog of kynurenic acid decreases tau pathology by modulating astrogliosis in rat model for tauopathy. Biomed Pharmacother 2022; 152:113257. [PMID: 35714514 DOI: 10.1016/j.biopha.2022.113257] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/30/2022] [Accepted: 06/06/2022] [Indexed: 11/17/2022] Open
Abstract
Kynurenines have immunomodulatory and neuroactive properties and can influence the central nervous system. Previous studies showed the involvement of the kynurenines in the pathogenesis and progression of neurodegenerative disease. In neurodegenerative disorders, including tauopathies, the tryptophan metabolism is shifted toward neurotoxic agents and the reduction of neuroprotectant products. Astrocyte-derived kynurenic acid serves as a neuroprotectant. However, systemic administration of kynurenic acid is not effective because of low permeability across the blood-brain barrier (BBB). We used a kynurenic acid analog with similar biological activity but higher brain permeability to overcome BBB limitations. In the present study, we used amide derivate of kynurenic acid N-(2-N, N-dimethylaminoethyl)- 4-oxo-1 H-quinoline-2-carboxamid (KYNA-1). We administered KYNA-1 for three months to tau transgenic rats SHR-24 and analyzed the effect on tau pathology and activation of glial cells. Primary glial cell cultures were applied to identify the mechanism of the KYNA-1 effect. KYNA-1 was not toxic to rats after chronic three-month administration. When chronically administered, KYNA-1 reduced hyperphosphorylation of insoluble tau in the brain of transgenic rats. Noteworthily, the plasma total tau was also reduced. We determined that the effect of KYNA-1 on tau pathology was induced through the modulation of glial activation. KYNA-1 inhibited LPS induced activation of astrocytes and induced transformation of microglia to M2 phenotype. We identified that the administration of KYNA-1 reduced tau hyperphosphorylation and neuroinflammation. KYNA-1 may serve as a promising treatment for tauopathies.
Collapse
Affiliation(s)
- Petra Majerova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia.
| | - Dominika Olesova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia; Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Kosice, Kosice, Slovakia.
| | - Greta Golisova
- Faculty of Natural Sciences, Department of Biochemistry, Comenius University in Bratislava, Mlynska dolina, Ilkovicova 6, 842 15 Bratislava, Slovakia.
| | - Martina Buralova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia.
| | - Alena Michalicova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia.
| | - Jozef Vegh
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia.
| | - Juraj Piestansky
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovakia.
| | - Mangesh Bhide
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Kosice, Kosice, Slovakia.
| | - Jozef Hanes
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia.
| | - Andrej Kovac
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia.
| |
Collapse
|
30
|
Behroozi Z, Ramezani F, Nasirinezhad F. Human umbilical cord blood-derived platelet -rich plasma: a new window for motor function recovery and axonal regeneration after spinal cord injury. Physiol Behav 2022; 252:113840. [PMID: 35525286 DOI: 10.1016/j.physbeh.2022.113840] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/01/2022] [Accepted: 05/02/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND There are complex mechanisms for reducing intrinsic repairability and neuronal regeneration following spinal cord injury (SCI). Platelet-rich plasma (PRP) is a rich source of growth factors and has been used to motivate the regeneration of peripheral nerves in neurodegenerative disorders. However, only a few studies have shown the effects of PRP on the SCI models. METHODS We investigated whether PRP derived from human umbilical cord blood (HUCB-PRP) could recover motor function in animals with spinal cord injury. Sixty adult male Wistar rats were randomly divided into 6 groups (n=60) as control, sham (laminectomy without induction of spinal cord injury), SCI, vehicle (SCI+ Platelet-Poor Plasma), PRP2day (SCI+PRP injection 2 days after SCI), and PRP14day (SCI+PRP injection 14 days after SCI). SCI was performed at the T12-T13 level. BBB test was carried out weekly after injury for six weeks. Caspase3 expression was determined using the Immunohistochemistry technique. The expression of GSK3β, CSF-tau, and MAG was determined using the Western blot technique. Data were analyzed by PRISM & SPSS software. RESULTS HUCB-PRP treated animals showed a higher locomotor function recovery than those in the SCI group (p<0.0001). The level of caspase3, GSK3β and CSF- Tau reduced and the MAG level in the spinal cord increased by the injection of HUCB-PRP in SCI animals. CONCLUSION Injection of HUCB-PRP enhanced hind limb locomotor performance by modulation of caspase3, GSK3β, CSF-tau, and MAG expression. Using HUCB-PRP could be a new therapeutic option for recovering motor function and axonal regeneration after SCI.
Collapse
Affiliation(s)
- Zahra Behroozi
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran; Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences. Kerman, Iran.
| | - Fatemeh Ramezani
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Farinaz Nasirinezhad
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran; Physiology Research Center, Department of Physiology, Iran University of Medical Sciences; Center for Experimental and Comparative Study, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Chen L, Niu X, Wang Y, Lv S, Zhou X, Yang Z, Peng D. Plasma tau proteins for the diagnosis of mild cognitive impairment and Alzheimer's disease: A systematic review and meta-analysis. Front Aging Neurosci 2022; 14:942629. [PMID: 35959295 PMCID: PMC9358685 DOI: 10.3389/fnagi.2022.942629] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveDetecting plasma tau biomarkers used to be impossible due to their low concentrations in blood samples. Currently, new high-sensitivity assays made it a reality. We performed a systematic review and meta-analysis in order to test the accuracy of plasma tau protein in diagnosing Alzheimer's disease (AD) or mild cognitive impairment (MCI).MethodsWe searched PubMed, Cochrane, Embase and Web of Science databases, and conducted correlation subgroup analysis, sensitivity analysis and publication bias analysis using R Programming Language.ResultsA total of 56 studies were included. Blood t-tau and p-tau levels increased from controls to MCI to AD patients, and showed significant changes in pairwise comparisons of AD, MCI and normal cognition. P-tau217 was more sensitive than p-tau181 and p-tau231 in different cognition periods. In addition, ultrasensitive analytical platforms, immunomagnetic reduction (IMR), increased the diagnostic value of tau proteins, especially the diagnostic value of t-tau.ConclusionBoth t-tau and p-tau are suitable AD blood biomarkers, and p-tau217 is more sensitive than other tau biomarkers to differentiate MCI and AD. Detection techniques also have an impact on biomarkers' results. New ultrasensitive analytical platforms of IMR increase the diagnostic value of both t-tau and p-tau biomarkers.Systematic review registrationhttps://www.crd.york.ac.uk/PROSPERO/, registration number: CRD42021264701.
Collapse
Affiliation(s)
- Leian Chen
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaoqian Niu
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yuye Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shuang Lv
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Xiao Zhou
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ziyuan Yang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Dantao Peng
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
- *Correspondence: Dantao Peng
| |
Collapse
|
32
|
Non-Invasive Nasal Discharge Fluid and Other Body Fluid Biomarkers in Alzheimer’s Disease. Pharmaceutics 2022; 14:pharmaceutics14081532. [PMID: 35893788 PMCID: PMC9330777 DOI: 10.3390/pharmaceutics14081532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/12/2022] [Accepted: 07/19/2022] [Indexed: 02/04/2023] Open
Abstract
The key to current Alzheimer’s disease (AD) therapy is the early diagnosis for prompt intervention, since available treatments only slow the disease progression. Therefore, this lack of promising therapies has called for diagnostic screening tests to identify those likely to develop full-blown AD. Recent AD diagnosis guidelines incorporated core biomarker analyses into criteria, including amyloid-β (Aβ), total-tau (T-tau), and phosphorylated tau (P-tau). Though effective, the accessibility of screening tests involving conventional cerebrospinal fluid (CSF)- and blood-based analyses is often hindered by the invasiveness and high cost. In an attempt to overcome these shortcomings, biomarker profiling research using non-invasive body fluid has shown the potential to capture the pathological changes in the patients’ bodies. These novel non-invasive body fluid biomarkers for AD have emerged as diagnostic and pathological targets. Here, we review the potential peripheral biomarkers, including non-invasive peripheral body fluids of nasal discharge, tear, saliva, and urine for AD.
Collapse
|
33
|
Cummings J, Kinney J. Biomarkers for Alzheimer's Disease: Context of Use, Qualification, and Roadmap for Clinical Implementation. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:952. [PMID: 35888671 PMCID: PMC9318582 DOI: 10.3390/medicina58070952] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 12/30/2022]
Abstract
Background and Objectives: The US Food and Drug Administration (FDA) defines a biomarker as a characteristic that is measured as an indicator of normal biological processes, pathogenic processes, or responses to an exposure or intervention. Biomarkers may be used in clinical care or as drug development tools (DDTs) in clinical trials. The goal of this review and perspective is to provide insight into the regulatory guidance for the use of biomarkers in clinical trials and clinical care. Materials and Methods: We reviewed FDA guidances relevant to biomarker use in clinical trials and their transition to use in clinical care. We identified instructive examples of these biomarkers in Alzheimer's disease (AD) drug development and their application in clinical practice. Results: For use in clinical trials, biomarkers must have a defined context of use (COU) as a risk/susceptibility, diagnostic, monitoring, predictive, prognostic, pharmacodynamic, or safety biomarker. A four-stage process defines the pathway to establish the regulatory acceptance of the COU for a biomarker including submission of a letter of intent, description of the qualification plan, submission of a full qualification package, and acceptance through a qualification recommendation. Biomarkers used in clinical care may be companion biomarkers, in vitro diagnostic devices (IVDs), or laboratory developed tests (LDTs). A five-phase biomarker development process has been proposed to structure the biomarker development process. Conclusions: Biomarkers are increasingly important in drug development and clinical care. Adherence to regulatory guidance for biomarkers used in clinical trials and patient care is required to advance these important drug development and clinical tools.
Collapse
Affiliation(s)
- Jeffrey Cummings
- Pam Quirk Brain Health and Biomarker Laboratory, Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV 89154, USA;
| | | |
Collapse
|
34
|
Faldu KG, Shah JS. Alzheimer's disease: a scoping review of biomarker research and development for effective disease diagnosis. Expert Rev Mol Diagn 2022; 22:681-703. [PMID: 35855631 DOI: 10.1080/14737159.2022.2104639] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 07/19/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is regarded as the foremost reason for neurodegeneration that prominently affects the geriatric population. Characterized by extracellular accumulation of amyloid-beta (Aβ), intracellular aggregation of hyperphosphorylated tau (p-tau), and neuronal degeneration that causes impairment of memory and cognition. Amyloid/tau/neurodegeneration (ATN) classification is utilized for research purposes and involves amyloid, tau, and neuronal injury staging through MRI, PET scanning, and CSF protein concentration estimations. CSF sampling is invasive, and MRI and PET scanning requires sophisticated radiological facilities which limit its widespread diagnostic use. ATN classification lacks effectiveness in preclinical AD. AREAS COVERED This publication intends to collate and review the existing biomarker profile and the current research and development of a new arsenal of biomarkers for AD pathology from different biological samples, microRNA (miRNA), proteomics, metabolomics, artificial intelligence, and machine learning for AD screening, diagnosis, prognosis, and monitoring of AD treatments. EXPERT OPINION It is an accepted observation that AD-related pathological changes occur over a long period of time before the first symptoms are observed providing ample opportunity for detection of biological alterations in various biological samples that can aid in early diagnosis and modify treatment outcomes.
Collapse
Affiliation(s)
- Khushboo Govind Faldu
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Jigna Samir Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| |
Collapse
|
35
|
Hawksworth J, Fernández E, Gevaert K. A new generation of AD biomarkers: 2019 to 2021. Ageing Res Rev 2022; 79:101654. [PMID: 35636691 DOI: 10.1016/j.arr.2022.101654] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 05/17/2022] [Accepted: 05/25/2022] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia and cases are rising worldwide. The effort to fight this disease is hampered by a lack of disease-modifying treatments and the absence of an early, accurate diagnostic tool. Neuropathology begins years or decades before symptoms occur and, upon onset of symptoms, diagnosis can take a year or more. Such delays postpone treatment and make research into the early stages of the disease difficult. Ideally, clinicians require a minimally invasive test that can detect AD in its early stages, before cognitive symptoms occur. Advances in proteomic technologies have facilitated the study of promising biomarkers of AD. Over the last two years (2019-2021) studies have identified and validated many species which can be measured in cerebrospinal fluid (CSF), plasma, or in both fluids, and which have a high predictive value for AD. We herein discuss proteins which have been highlighted as promising biomarkers of AD in the last two years, and consider implications for future research within the research framework of the amyloid (A), tau (T), neurodegeneration (N) scoring system. We review recently identified species of amyloid and tau which may improve diagnosis when used in combination with current measures such as amyloid-beta-42 (Aβ42), total tau (t-tau) and phosphorylated tau (p-tau). In addition, several proteins have been identified as likely proxies for neurodegeneration, including neurofilament light (NfL), synaptosomal-associated protein 25 (SNAP-25) and neurogranin (NRGN). Finally, proteins originating from diverse processes such as neuroinflammation, lipid transport and mitochondrial dysfunction could aid in both AD diagnosis and patient stratification.
Collapse
|
36
|
Fernandes M, Mari L, Chiaravalloti A, Paoli B, Nuccetelli M, Izzi F, Giambrone MP, Camedda R, Bernardini S, Schillaci O, Mercuri NB, Placidi F, Liguori C. 18F-FDG PET, cognitive functioning, and CSF biomarkers in patients with obstructive sleep apnoea before and after continuous positive airway pressure treatment. J Neurol 2022; 269:5356-5367. [PMID: 35608659 PMCID: PMC9468130 DOI: 10.1007/s00415-022-11182-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/07/2022] [Accepted: 05/10/2022] [Indexed: 11/23/2022]
Abstract
Introduction Dysregulation of cerebral glucose consumption, alterations in cerebrospinal fluid (CSF) biomarkers, and cognitive impairment have been reported in patients with obstructive sleep apnoea (OSA). On these bases, OSA has been considered a risk factor for Alzheimer’s disease (AD). This study aimed to measure cognitive performance, CSF biomarkers, and cerebral glucose consumption in OSA patients and to evaluate the effects of continuous positive airway pressure (CPAP) treatment on these biomarkers over a 12-month period. Methods Thirty-four OSA patients and 34 controls underwent 18F-fluoro-2-deoxy-d-glucose positron emission tomography (18F-FDG PET), cognitive evaluation, and CSF analysis. A subgroup of 12 OSA patients treated with beneficial CPAP and performing the 12-month follow-up was included in the longitudinal analysis, and cognitive evaluation and 18F-FDG PET were repeated. Results Significantly reduced glucose consumption was observed in the bilateral praecuneus, posterior cingulate cortex, and frontal areas in OSA patients than controls. At baseline, OSA patients also showed lower β-amyloid42 and higher phosphorylated-tau CSF levels than controls. Increased total tau and phosphorylated tau levels correlated with a reduction in brain glucose consumption in a cluster of different brain areas. In the longitudinal analysis, OSA patients showed an improvement in cognition and a global increase in cerebral 18F-FDG uptake. Conclusions Cognitive impairment, reduced cerebral glucose consumption, and alterations in CSF biomarkers were observed in OSA patients, which may reinforce the hypothesis of AD neurodegenerative processes triggered by OSA. Notably, cognition and brain glucose consumption improved after beneficial CPAP treatment. Further studies are needed to evaluate the long-term effects of CPAP treatment on these AD biomarkers.
Collapse
Affiliation(s)
- Mariana Fernandes
- Department of Systems Medicine, Sleep Medicine Centre, University of Rome "Tor Vergata", Rome, Italy
| | - Luisa Mari
- Neurology Unit, University Hospital of Rome "Tor Vergata", Rome, Italy
| | - Agostino Chiaravalloti
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - Barbara Paoli
- Department of Systems Medicine, Sleep Medicine Centre, University of Rome "Tor Vergata", Rome, Italy
| | - Marzia Nuccetelli
- Department of Clinical Biochemistry and Molecular Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Francesca Izzi
- Neurology Unit, University Hospital of Rome "Tor Vergata", Rome, Italy
| | | | - Riccardo Camedda
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Sergio Bernardini
- Department of Clinical Biochemistry and Molecular Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - Nicola Biagio Mercuri
- Neurology Unit, University Hospital of Rome "Tor Vergata", Rome, Italy.,IRCSS Santa Lucia Foundation, Rome, Italy
| | - Fabio Placidi
- Department of Systems Medicine, Sleep Medicine Centre, University of Rome "Tor Vergata", Rome, Italy.,Neurology Unit, University Hospital of Rome "Tor Vergata", Rome, Italy
| | - Claudio Liguori
- Department of Systems Medicine, Sleep Medicine Centre, University of Rome "Tor Vergata", Rome, Italy. .,Neurology Unit, University Hospital of Rome "Tor Vergata", Rome, Italy.
| |
Collapse
|
37
|
Pomilio AB, Vitale AA, Lazarowski AJ. Neuroproteomics Chip-Based Mass Spectrometry and Other Techniques for Alzheimer´S Disease Biomarkers – Update. Curr Pharm Des 2022; 28:1124-1151. [DOI: 10.2174/1381612828666220413094918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/25/2022] [Indexed: 11/22/2022]
Abstract
Background:
Alzheimer's disease (AD) is a progressive neurodegenerative disease of growing interest given that there is cognitive damage and symptom onset acceleration. Therefore, it is important to find AD biomarkers for early diagnosis, disease progression, and discrimination of AD and other diseases.
Objective:
To update the relevance of mass spectrometry for the identification of peptides and proteins involved in AD useful as discriminating biomarkers.
Methods:
Proteomics and peptidomics technologies that show the highest possible specificity and selectivity for AD biomarkers are analyzed, together with the biological fluids used. In addition to positron emission tomography and magnetic resonance imaging, MALDI-TOF mass spectrometry is widely used to identify proteins and peptides involved in AD. The use of protein chips in SELDI technology and electroblotting chips for peptides makes feasible small amounts (L) of samples for analysis.
Results:
Suitable biomarkers are related to AD pathology, such as intracellular neurofibrillary tangles; extraneuronal senile plaques; neuronal and axonal degeneration; inflammation and oxidative stress. Recently, peptides were added to the candidate list, which are not amyloid-b or tau fragments, but are related to coagulation, brain plasticity, and complement/neuroinflammation systems involving the neurovascular unit.
Conclusion:
The progress made in the application of mass spectrometry and recent chip techniques is promising for discriminating between AD, mild cognitive impairment, and matched healthy controls. The application of this technique to blood samples from patients with AD has shown to be less invasive and fast enough to determine the diagnosis, stage of the disease, prognosis, and follow-up of the therapeutic response.
Collapse
Affiliation(s)
- Alicia B. Pomilio
- Departamento de Bioquímica Clínica, Área Hematología, Hospital de Clínicas “José de San Martín”, Universidad de Buenos Aires, Av. Córdoba 2351, C1120AAF Buenos Aires, Argentina
| | - Arturo A. Vitale
- Departamento de Bioquímica Clínica, Área Hematología, Hospital de Clínicas “José de San Martín”, Universidad de Buenos Aires, Av. Córdoba 2351, C1120AAF Buenos Aires, Argentina
| | - Alberto J. Lazarowski
- Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Córdoba 2351, C1120AAF Buenos Aires, Argentina
| |
Collapse
|
38
|
Sarnowski C, Ghanbari M, Bis JC, Logue M, Fornage M, Mishra A, Ahmad S, Beiser AS, Boerwinkle E, Bouteloup V, Chouraki V, Cupples LA, Damotte V, DeCarli CS, DeStefano AL, Djoussé L, Fohner AE, Franz CE, Kautz TF, Lambert JC, Lyons MJ, Mosley TH, Mukamal KJ, Pase MP, Portilla Fernandez EC, Rissman RA, Satizabal CL, Vasan RS, Yaqub A, Debette S, Dufouil C, Launer LJ, Kremen WS, Longstreth WT, Ikram MA, Seshadri S. Meta-analysis of genome-wide association studies identifies ancestry-specific associations underlying circulating total tau levels. Commun Biol 2022; 5:336. [PMID: 35396452 PMCID: PMC8993877 DOI: 10.1038/s42003-022-03287-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 03/17/2022] [Indexed: 01/27/2023] Open
Abstract
Circulating total-tau levels can be used as an endophenotype to identify genetic risk factors for tauopathies and related neurological disorders. Here, we confirmed and better characterized the association of the 17q21 MAPT locus with circulating total-tau in 14,721 European participants and identified three novel loci in 953 African American participants (4q31, 5p13, and 6q25) at P < 5 × 10-8. We additionally detected 14 novel loci at P < 5 × 10-7, specific to either Europeans or African Americans. Using whole-exome sequence data in 2,279 European participants, we identified ten genes associated with circulating total-tau when aggregating rare variants. Our genetic study sheds light on genes reported to be associated with neurological diseases including stroke, Alzheimer's, and Parkinson's (F5, MAP1B, and BCAS3), with Alzheimer's pathological hallmarks (ADAMTS12, IL15, and FHIT), or with an important function in the brain (PARD3, ELFN2, UBASH3B, SLIT3, and NSD3), and suggests that the genetic architecture of circulating total-tau may differ according to ancestry.
Collapse
Affiliation(s)
- Chloé Sarnowski
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mark Logue
- National Center for PTSD, Behavioral Sciences Division, VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry and Biomedical Genetics, Boston University School of Medicine, Boston, MA, USA
| | - Myriam Fornage
- University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Aniket Mishra
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, F-33000, Bordeaux, France
| | - Shahzad Ahmad
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Alexa S Beiser
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Eric Boerwinkle
- University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Vincent Bouteloup
- Centre Inserm U1219 Bordeaux Population Health, CIC1401-EC, Institut de Santé Publique, d'Epidémiologie et de Développement, Université de Bordeaux, CHU de Bordeaux, Pôle Santé Publique, Bordeaux, France
| | - Vincent Chouraki
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE- LabEx DISTALZ - Risk factors and molecular determinants of aging diseases, F-59000, Lille, France
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
| | - Vincent Damotte
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE- LabEx DISTALZ - Risk factors and molecular determinants of aging diseases, F-59000, Lille, France
| | - Charles S DeCarli
- Department of Neurology and Center for Neuroscience, University of California at Davis, Davis, CA, USA
| | - Anita L DeStefano
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Luc Djoussé
- Department of Medicine, Division of Aging, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alison E Fohner
- Institute of Public Health Genetics and Department of Epidemiology and Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
| | - Carol E Franz
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, USA
| | - Tiffany F Kautz
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Jean-Charles Lambert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE- LabEx DISTALZ - Risk factors and molecular determinants of aging diseases, F-59000, Lille, France
| | - Michael J Lyons
- Department of Psychology and Brain Sciences, Boston University, Boston, MA, USA
| | | | - Kenneth J Mukamal
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Matthew P Pase
- Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | | | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Claudia L Satizabal
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Ramachandran S Vasan
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
- Preventive Medicine & Epidemiology, Boston University School of Medicine, Boston, MA, USA
| | - Amber Yaqub
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Stephanie Debette
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, F-33000, Bordeaux, France
- Bordeaux University Hospital, Department of Neurology, Institute for Neurodegenerative Diseases, Bordeaux, France
| | - Carole Dufouil
- Bordeaux University Hospital, Department of Neurology, Institute for Neurodegenerative Diseases, Bordeaux, France
| | | | - William S Kremen
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, USA
| | - William T Longstreth
- Departments of Neurology and Epidemiology, University of Washington, Seattle, WA, USA
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sudha Seshadri
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| |
Collapse
|
39
|
Mankhong S, Kim S, Lee S, Kwak HB, Park DH, Joa KL, Kang JH. Development of Alzheimer’s Disease Biomarkers: From CSF- to Blood-Based Biomarkers. Biomedicines 2022; 10:biomedicines10040850. [PMID: 35453600 PMCID: PMC9025524 DOI: 10.3390/biomedicines10040850] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/25/2022] [Accepted: 03/31/2022] [Indexed: 02/05/2023] Open
Abstract
In the 115 years since the discovery of Alzheimer’s disease (AD), our knowledge, diagnosis, and therapeutics have significantly improved. Biomarkers are the primary tools for clinical research, diagnostics, and therapeutic monitoring in clinical trials. They provide much insightful information, and while they are not clinically used routinely, they help us to understand the mechanisms of this disease. This review charts the journey of AD biomarker discovery and development from cerebrospinal fluid (CSF) amyloid-beta 1-42 (Aβ42), total tau (T-tau), and phosphorylated tau (p-tau) biomarkers and imaging technologies to the next generation of biomarkers. We also discuss advanced high-sensitivity assay platforms for CSF Aβ42, T-tau, p-tau, and blood analysis. The recently proposed Aβ deposition/tau biomarker/neurodegeneration or neuronal injury (ATN) scheme might facilitate the definition of the biological status underpinning AD and offer a common language among researchers across biochemical biomarkers and imaging. Moreover, we highlight blood-based biomarkers for AD that offer a scalable alternative to CSF biomarkers through cost-saving and reduced invasiveness, and may provide an understanding of disease initiation and development. We discuss different groups of blood-based biomarker candidates, their advantages and limitations, and paths forward, from identification and analysis to clinical validation. The development of valid blood-based biomarkers may facilitate the implementation of future AD therapeutics and diagnostics.
Collapse
Affiliation(s)
- Sakulrat Mankhong
- Department of Pharmacology, Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.)
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Korea; (S.L.); (H.-B.K.); (D.-H.P.)
| | - Sujin Kim
- Department of Pharmacology, Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.)
| | - Seongju Lee
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Korea; (S.L.); (H.-B.K.); (D.-H.P.)
- Department of Anatomy, College of Medicine, Inha University, Incheon 22212, Korea
| | - Hyo-Bum Kwak
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Korea; (S.L.); (H.-B.K.); (D.-H.P.)
- Department of Kinesiology, Inha University, Incheon 22212, Korea
| | - Dong-Ho Park
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Korea; (S.L.); (H.-B.K.); (D.-H.P.)
- Department of Kinesiology, Inha University, Incheon 22212, Korea
| | - Kyung-Lim Joa
- Department of Physical & Rehabilitation Medicine, College of Medicine, Inha University, Incheon 22212, Korea;
| | - Ju-Hee Kang
- Department of Pharmacology, Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.)
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Korea; (S.L.); (H.-B.K.); (D.-H.P.)
- Correspondence: ; Tel.: +82-32-860-9872
| |
Collapse
|
40
|
Heshmatollah A, Fani L, Koudstaal PJ, Ghanbari M, Ikram MA, Ikram MK. Plasma Amyloid Beta, Total-Tau and Neurofilament Light Chain Levels and the Risk of Stroke: A Prospective Population-Based Study. Neurology 2022; 98:e1729-e1737. [PMID: 35232820 DOI: 10.1212/wnl.0000000000200004] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 01/03/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES To unravel whether Alzheimer's disease-related pathology or neurodegeneration play a role in stroke etiology, we determined the effect of plasma levels amyloid β (Aβ), total-tau and neurofilament light chain (NfL) on risk of stroke and its subtypes. METHODS Between 2002 and 2005, we measured plasma Aβ40, Aβ42, total-tau, and NfL in 4,661 stroke-free participants from the population-based Rotterdam Study. We used Cox proportional-hazards models to determine the association between these markers with incident stroke for the entire cohort, per stroke subtype, and by median age, sex, Apolipoprotein E (APOE) ε4 carriership, and education. RESULTS After a mean follow-up of 10.8 ± 3.3 years, 379 participants suffered a first-ever stroke. Log2 total-tau at baseline showed a non-linear association with risk of any stroke and ischemic stroke: compared to the first (lowest) quartile the adjusted hazard ratio for the highest quartile total-tau was 1.68, 95% CI: 1.18-2.40 for any stroke. Log2 NfL was associated with an increased risk of any stroke (HR per SD increase 1.27, 95% CI: 1.12-1.44), ischemic stroke, and hemorrhagic stroke (HR 1.56, 95% CI: 1.14-2.12). Log2 Aβ40, Aβ42, and Aβ42/40 ratio levels were not associated with stroke risk.Discussion Participants with higher total-tau and NfL at baseline had a higher risk of stroke and several stroke subtypes. These findings support the role of markers of neurodegeneration in the etiology of stroke. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that higher plasma levels of total-tau and NfL are associated with an increased risk of subsequent stroke.
Collapse
Affiliation(s)
- Alis Heshmatollah
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands.,Department of Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Lana Fani
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Peter J Koudstaal
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - M Kamran Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands.,Department of Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
41
|
Schirinzi T, Zenuni H, Grillo P, Bovenzi R, Guerrera G, Gargano F, Pieri M, Bernardini S, Biagio Mercuri N, Battistini L, Sancesario GM. Tau and Amyloid-β Peptides in Serum of Patients With Parkinson's Disease: Correlations With CSF Levels and Clinical Parameters. Front Neurol 2022; 13:748599. [PMID: 35280296 PMCID: PMC8914101 DOI: 10.3389/fneur.2022.748599] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 01/18/2022] [Indexed: 12/13/2022] Open
Abstract
Relevance of blood-based biomarkers is increasing into the neurodegenerative diseases field, but data on Parkinson's disease (PD) remain still scarce. In this study, we used the SiMoA technique to measure serum content of total tau protein and amyloid-β peptides (Aβ-42, Aβ-40) in 22 PD patients and ten control subjects. Serum levels of each biomarker were correlated with the respective CSF levels in both the groups; in PD patients, also the correlations between serum biomarkers and main clinical parameters were tested (motor, non-motor, cognitive scores and levodopa equivalent daily dose). Serum biomarkers did not exhibit quantitative differences between patients and controls; however, only PD patients had inter-fluids (serum-CSF) associations in tau and amyloid-β-42 levels. Moreover, serum content of tau protein was inversely correlated with cognitive performances (MoCA score). These findings, albeit preliminary, indicate that brain-derived peptides may change in parallel in both peripheral blood and CSF of PD patients, eventually even in association with some clinical features. Further studies are now needed to validate the use of blood-based biomarkers in PD.
Collapse
Affiliation(s)
- Tommaso Schirinzi
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
- *Correspondence: Tommaso Schirinzi
| | - Henri Zenuni
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Piergiorgio Grillo
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Roberta Bovenzi
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Gisella Guerrera
- European Centre for Brain Research, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Francesca Gargano
- European Centre for Brain Research, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Massimo Pieri
- Department of Experimental Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
- European Centre for Brain Research, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Luca Battistini
- European Centre for Brain Research, IRCCS Fondazione Santa Lucia, Rome, Italy
| | | |
Collapse
|
42
|
Bhuniya S, Goyal M, Chowdhury N, Mishra P. Intermittent hypoxia and sleep disruption in obstructive sleep apnea increase serum tau and amyloid-beta levels. J Sleep Res 2022; 31:e13566. [PMID: 35165967 DOI: 10.1111/jsr.13566] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 01/16/2023]
Abstract
Obstructive sleep apnea is characterized by intermittent hypoxia and sleep disruption, leading to accelerated neurodegenerative changes and cognitive decline. Serum amyloid-beta and tau proteins, which are markers for Alzheimer's disease, have been reported to increase in patients with obstructive sleep apnea. This study compared the serum levels of amyloid-beta proteins and tau proteins in 46 cognitively normal obstructive sleep apnea patients and 30 healthy controls. Sleep parameters and severity of obstructive sleep apnea were determined using overnight polysomnography. Serum levels of Aβ40, Aβ42, total tau and phosphorylated-tau were determined by enzyme-linked immunosorbent assay. Patients with obstructive sleep apnea had significantly higher median serum levels of Aβ40 (121.0 versus 78.3 pg ml-1 ), Aβ42 (105.6 versus 18.6 pg ml-1 ) and total tau (168.5 versus 10.9 pg ml-1 ) than controls. Serum levels of phosphorylated-tau did not differ significantly between the two groups. Serum levels of amyloid and tau proteins correlated with parameters of nocturnal oxygen saturation. Rapid eye movement sleep was negatively correlated with total amyloid-beta proteins. We conclude that serum levels of amyloid-beta and total tau are higher in patients with obstructive sleep apnea and hypoxia as well as changes in sleep architecture associated with their increased levels. Patients with obstructive sleep apnea should be closely monitored for the signs of cognitive impairment. Obstructive sleep apnea is a modifiable risk factor, and its treatment may reverse neurodegenerative changes and prevent cognitive impairment.
Collapse
Affiliation(s)
- Sourin Bhuniya
- Department of Pulmonary Medicine, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Manish Goyal
- Department of Physiology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Nilotpal Chowdhury
- Department of Pathology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Priyadarshini Mishra
- Department of Physiology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| |
Collapse
|
43
|
Phan LMT, Cho S. Fluorescent Aptasensor and Colorimetric Aptablot for p-tau231 Detection: Toward Early Diagnosis of Alzheimer's Disease. Biomedicines 2022; 10:93. [PMID: 35052773 PMCID: PMC8772959 DOI: 10.3390/biomedicines10010093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/27/2021] [Accepted: 12/30/2021] [Indexed: 02/05/2023] Open
Abstract
The pathology of Alzheimer's disease (AD), the most common cause of dementia, is considered to be mainly driven by two major hallmarks (tau and amyloid beta). It is highly desirable to develop an affordable medicinal diagnostic that can be utilized worldwide for the early diagnosis of AD. Hence, p-tau231 was selected as a specific target, which appears both in AD serum and cerebrospinal fluid, for the development of a sensing platform for the diagnosis of AD. To the best of our knowledge, these are the first aptamer-mediated biosensors that rely on sensitive fluorescent and colorimetric aptasensors for the rapid monitoring of p-tau231. The nitrogen-doped carbon dot-based turn-on fluorescent aptasensor could rapidly analyze p-tau231 down to 3.64 ng/mL within 40 min, and the colorimetric Cu-enhanced-Au aptablot displayed high sensitivity at 4.71 pg/mL through a digital camera, with visibility to the naked eye down to 8 ng/mL p-tau231 within 140 min. Owing to their advantages, which include affordability, rapidity, high sensitivity, and dependence on complicated instruments, these aptamer-based biosensors offer significant potential for the early diagnosis of AD worldwide.
Collapse
Affiliation(s)
- Le Minh Tu Phan
- Department of Electronic Engineering, Gachon University, Seongnam-si 13120, Korea
- School of Medicine and Pharmacy, The University of Danang, Danang 550000, Vietnam
| | - Sungbo Cho
- Department of Electronic Engineering, Gachon University, Seongnam-si 13120, Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| |
Collapse
|
44
|
Sun HL, Zhou FY, Chen DW, Tan CR, Zeng GH, Liu YH, Wang J, Bu XL, Wang YJ, Li HY, Jin WS. The Correlation of Tau Levels with Blood Monocyte Count in Patients with Alzheimer’s Disease. J Alzheimers Dis 2021; 85:1321-1328. [PMID: 34924377 DOI: 10.3233/jad-210692] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background: Recent studies have shown that monocytes can phagocytize the tau protein, which may ameliorate tau-type pathology in Alzheimer’s disease (AD). However, there are few clinical studies on the relationship between monocytes and tau-type pathology in AD patients. Objective: We aimed to explore changes in peripheral monocytes and their association with tau protein in AD patients. Methods: A total of 127 clinically diagnosed AD patients and 100 age- and sex-matched cognitively normal controls were recruited for analysis of the correlation of plasma tau levels with the blood monocyte count. Cerebrospinal fluid (CSF) samples from 46 AD patients and 88 controls were further collected to analyze the correlation of CSF tau and amyloid-β (Aβ) levels with the blood monocyte count. 105 clinically diagnosed mild cognitive impairment (MCI) patients and 149 age- and sex-matched cognitively normal controls were recruited from another cohort for verification. Results: Compared to normal controls, AD patients showed a significant reduction in the blood monocyte count. In addition, the monocyte count of AD patients was negatively correlated with CSF t-tau and p-tau levels but not with plasma tau levels. In normal people, monocyte count lack correlation with tau levels both in plasma and CSF. Monocyte count were not correlated with CSF Aβ levels in either group but were negatively correlated with CSF tau/Aβ 42 levels in the AD group. We had further verified the correlations of monocyte count with CSF tau levels in another cohort. Conclusion: This study suggests that monocytes may play an important role in the clearance of tau protein in the brain.
Collapse
Affiliation(s)
- Hao-Lun Sun
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Shigatse Branch, Xinqiao Hospital, Third Military Medical University, Shigatse, China
| | - Fa-Ying Zhou
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Dong-Wan Chen
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Cheng-Rong Tan
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Gui-Hua Zeng
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yu-Hui Liu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Jun Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xian-Le Bu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yan-Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Hui-Yun Li
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Wang-Sheng Jin
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
45
|
Milanesi E, Dobre M, Cucos CA, Rojo AI, Jiménez-Villegas J, Capetillo-Zarate E, Matute C, Piñol-Ripoll G, Manda G, Cuadrado A. Whole Blood Expression Pattern of Inflammation and Redox Genes in Mild Alzheimer's Disease. J Inflamm Res 2021; 14:6085-6102. [PMID: 34848989 PMCID: PMC8612672 DOI: 10.2147/jir.s334337] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 09/29/2021] [Indexed: 12/21/2022] Open
Abstract
Background Although Alzheimer’s disease (AD) is associated with alterations of the central nervous system, this disease has an echo in blood that might represent a valuable source of biomarkers for improved diagnosis, prognosis and for monitoring drug response. Methods We performed a targeted transcriptomics study on 38 mild Alzheimer’s disease (AD) patients and 38 matched controls for evaluating the expression levels of 136 inflammation and 84 redox genes in whole blood. Patients were diagnosed as mild AD based on altered levels of total TAU, phospho-TAU and Abeta(1–42) in cerebrospinal fluid, and Abeta(1–40), Abeta(1–42) and total TAU levels in plasma. Whenever possible, blood and brain comparisons were made using public datasets. Results We found 48 inflammation and 34 redox genes differentially expressed in the blood of AD patients vs controls (FC >1.5, p < 0.01), out of which 22 pro-inflammatory and 12 redox genes exhibited FC >2 and p < 0.001. Receiver operating characteristic (ROC) analysis identified nine inflammation and seven redox genes that discriminated between AD patients and controls (area under the curve >0.9). Correlations of the dysregulated inflammation and redox transcripts indicated that RELA may regulate several redox genes including DUOX1 and GSR. Based on the gene expression profile, we have found that the master regulators of inflammation and redox homeostasis, NFκB and NRF2, were significantly disturbed in the blood of AD patients, as well as several zinc finger and helix-loop-helix transcription factors. Conclusion The selected inflammation and redox genes might be useful biomarkers for monitoring anti-inflammatory therapy in mild AD.
Collapse
Affiliation(s)
- Elena Milanesi
- "Victor Babes" National Institute of Pathology, Bucharest, 050096, Romania
| | - Maria Dobre
- "Victor Babes" National Institute of Pathology, Bucharest, 050096, Romania
| | | | - Ana I Rojo
- Department of Endocrine Physiology and Nervous System, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Madrid, 28029, Spain.,Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, 28049, Spain.,Neuroscience Section, Instituto de Investigación Sanitaria La Paz (IDIPAZ), Madrid, 28046, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, 28031, Spain
| | - José Jiménez-Villegas
- Department of Endocrine Physiology and Nervous System, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Madrid, 28029, Spain.,Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, 28049, Spain
| | - Estibaliz Capetillo-Zarate
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, 28031, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, 48009, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Carlos Matute
- IKERBASQUE, Basque Foundation for Science, Bilbao, 48009, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Gerard Piñol-Ripoll
- Unitat Trastons Cognitius, Hospital Universitari Santa Maria-IRB Leida, Lleida, 25198, Spain
| | - Gina Manda
- "Victor Babes" National Institute of Pathology, Bucharest, 050096, Romania
| | - Antonio Cuadrado
- "Victor Babes" National Institute of Pathology, Bucharest, 050096, Romania.,Department of Endocrine Physiology and Nervous System, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Madrid, 28029, Spain.,Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, 28049, Spain.,Neuroscience Section, Instituto de Investigación Sanitaria La Paz (IDIPAZ), Madrid, 28046, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, 28031, Spain
| |
Collapse
|
46
|
Andreou D, Jørgensen KN, Nerland S, Smelror RE, Wedervang-Resell K, Johannessen CH, Myhre AM, Andreassen OA, Blennow K, Zetterberg H, Agartz I. Lower plasma total tau in adolescent psychosis: Involvement of the orbitofrontal cortex. J Psychiatr Res 2021; 144:255-261. [PMID: 34700214 DOI: 10.1016/j.jpsychires.2021.10.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/16/2021] [Accepted: 10/19/2021] [Indexed: 10/20/2022]
Abstract
Schizophrenia is thought to be a neurodevelopmental disorder with neuronal migration, differentiation and maturation disturbances. Tau is a microtubule-associated protein with a crucial role in these processes. Lower circulating tau levels have been reported in adults with schizophrenia, but this association has not been investigated in adolescent psychosis. We aimed to test the hypotheses that a) adolescents with early-onset psychosis (EOP; age of onset <18 years) display lower plasma tau concentrations compared to healthy controls, and b) among patients with psychosis, tau levels are linked to structural brain measures associated with the microtubule-associated tau (MAPT) gene and psychosis. We included 37 adolescent patients with EOP (mean age 16.4 years) and 59 adolescent healthy controls (mean age 16.2 years). We investigated putative patient-control differences in plasma total tau concentrations measured by a Single molecule array (Simoa) immunoassay. We explored the correlations between tau and selected structural brain measures based on T1-weighted MRI scans processed in FreeSurfer v6.0. We found significantly lower plasma tau concentrations in patients compared to healthy controls (p = 0.017, partial eta-squared = 0.061). Tau was not associated with antipsychotic use or the antipsychotic dosage. Among patients but not healthy controls, tau levels were positively correlated with the cortical orbitofrontal surface area (p = 0.013, R-squared = 0.24). The results are suggestive of a tau-related neurodevelopmental disturbance in adolescent psychosis.
Collapse
Affiliation(s)
- Dimitrios Andreou
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden.
| | - Kjetil Nordbø Jørgensen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Stener Nerland
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Runar Elle Smelror
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Kirsten Wedervang-Resell
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Child and Adolescent Mental Health Research Unit, Department of Research and Innovation, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Cecilie Haggag Johannessen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anne Margrethe Myhre
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Research and Innovation, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Kaj Blennow
- 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
| | - 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
| | - Ingrid Agartz
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
47
|
Hall JR, Petersen M, Johnson L, O'Bryant SE. Plasma Total Tau and Neurobehavioral Symptoms of Cognitive Decline in Cognitively Normal Older Adults. Front Psychol 2021; 12:774049. [PMID: 34803857 PMCID: PMC8603823 DOI: 10.3389/fpsyg.2021.774049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Depression and related neurobehavioral symptoms are common features of Alzheimer’s disease and other dementias. The presence of these potentially modifiable neurobehavioral symptoms in cognitively intact older adults may represent an early indication of pathophysiological processes in the brain. Tau pathology is a key feature of a number of dementias. A number of studies have found an association between tau and neurobehavioral symptoms. The current study investigated the relationship of a blood-based biomarker of tau and symptoms of depression, anxiety, worry, and sleep disturbances in 538 community based, cognitively normal older adults. Logistic regression revealed no significant relationship between plasma total tau and any measures of neurobehavioral symptoms. To assess the impact of level of tau on these relationships, participants were divided into those in the highest quintile of tau and those in the lower four quintiles. Regression analyses showed a significant relationship between level of plasma total tau and measures of depression, apathy, anxiety, worry and sleep. The presence of higher levels of plasma tau and elevated neurobehavioral symptoms may be an early indicator of cognitive decline and prodromal Alzheimer’s disease. Longitudinal research is needed to evaluate the impact of these factors on the development of dementia and may suggest areas for early intervention.
Collapse
Affiliation(s)
- James R Hall
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, United States.,Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Melissa Petersen
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, United States.,Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Leigh Johnson
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, United States.,Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Sid E O'Bryant
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, United States.,Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
| |
Collapse
|
48
|
Sodhi K, Pratt R, Wang X, Lakhani HV, Pillai SS, Zehra M, Wang J, Grover L, Henderson B, Denvir J, Liu J, Pierre S, Nelson T, Shapiro JI. Role of adipocyte Na,K-ATPase oxidant amplification loop in cognitive decline and neurodegeneration. iScience 2021; 24:103262. [PMID: 34755095 PMCID: PMC8564125 DOI: 10.1016/j.isci.2021.103262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 08/10/2021] [Accepted: 10/11/2021] [Indexed: 11/21/2022] Open
Abstract
Recent studies suggest that a western diet may contribute to clinical neurodegeneration and dementia. Adipocyte-specific expression of the Na,K-ATPase signaling antagonist, NaKtide, ameliorates the pathophysiological consequences of murine experimental obesity and renal failure. In this study, we found that a western diet produced systemic oxidant stress along with evidence of activation of Na,K-ATPase signaling within both murine brain and peripheral tissues. We also noted this diet caused increases in circulating inflammatory cytokines as well as behavioral, and brain biochemical changes consistent with neurodegeneration. Adipocyte specific NaKtide affected by a doxycycline on/off expression system ameliorated all of these diet effects. These data suggest that a western diet produces cognitive decline and neurodegeneration through augmented Na,K-ATPase signaling and that antagonism of this pathway in adipocytes ameliorates the pathophysiology. If this observation is confirmed in humans, the adipocyte Na,K-ATPase may serve as a clinical target in the therapy of neurodegenerative disorders.
Collapse
Affiliation(s)
- Komal Sodhi
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Rebecca Pratt
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Xiaoliang Wang
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Hari Vishal Lakhani
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Sneha S. Pillai
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Mishghan Zehra
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Jiayan Wang
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Lawrence Grover
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Brandon Henderson
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - James Denvir
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Jiang Liu
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Sandrine Pierre
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Thomas Nelson
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Joseph I. Shapiro
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| |
Collapse
|
49
|
Plasma phosphorylated-tau181 as a predictive biomarker for Alzheimer's amyloid, tau and FDG PET status. Transl Psychiatry 2021; 11:585. [PMID: 34775468 PMCID: PMC8590691 DOI: 10.1038/s41398-021-01709-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 10/17/2021] [Accepted: 10/26/2021] [Indexed: 12/13/2022] Open
Abstract
Plasma phosphorylated-tau181 (p-tau181) showed the potential for Alzheimer's diagnosis and prognosis, but its role in detecting cerebral pathologies is unclear. We aimed to evaluate whether it could serve as a marker for Alzheimer's pathology in the brain. A total of 1189 participants with plasma p-tau181 and PET data of amyloid, tau or FDG PET were included from ADNI. Cross-sectional relationships of plasma p-tau181 with PET biomarkers were tested. Longitudinally, we further investigated whether different p-tau181 levels at baseline predicted different progression of Alzheimer's pathological changes in the brain. We found plasma p-tau181 significantly correlated with brain amyloid (Spearman ρ = 0.45, P < 0.0001), tau (0.25, P = 0.0003), and FDG PET uptakes (-0.37, P < 0.0001), and increased along the Alzheimer's continuum. Individually, plasma p-tau181 could detect abnormal amyloid, tau pathologies and hypometabolism in the brain, similar with or even better than clinical indicators. The diagnostic accuracy of plasma p-tau181 elevated significantly when combined with clinical information (AUC = 0.814 for amyloid PET, 0.773 for tau PET, and 0.708 for FDG PET). Relationships of plasma p-tau181 with brain pathologies were partly or entirely mediated by the corresponding CSF biomarkers. Besides, individuals with abnormal plasma p-tau181 level (>18.85 pg/ml) at baseline had a higher risk of pathological progression in brain amyloid (HR: 2.32, 95%CI 1.32-4.08) and FDG PET (3.21, 95%CI 2.06-5.01) status. Plasma p-tau181 may be a sensitive screening test for detecting brain pathologies, and serve as a predictive biomarker for Alzheimer's pathophysiology.
Collapse
|
50
|
Singh K, Cheung BM, Xu A. Ultrasensitive detection of blood biomarkers of Alzheimer's and Parkinson's diseases: a systematic review. Biomark Med 2021; 15:1693-1708. [PMID: 34743546 DOI: 10.2217/bmm-2021-0219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose: Neurodegenerative disorders are a global health burden with costly and invasive diagnoses relying on brain imaging technology or CSF-based biomarkers. Therefore, considerable efforts to identify blood-biomarkers for Alzheimer's (AD) and Parkinson's diseases (PD) are ongoing. Objectives: This review evaluates the blood biomarkers for AD and PD for their diagnostic value. Methods: This study systematically reviewed articles published between July 1984 and February 2021. Among 1266 papers, we selected 42 studies for a systematic review and 23 studies for meta-analysis. Results & conclusion: Our analysis highlights P-tau181, T-tau and Nfl as promising blood biomarkers for AD diagnosis. Nfl levels were consistently raised in 16 AD and three PD cohorts. P-tau181 and T-tau were also significantly increased in 12 and eight AD cohorts, respectively.
Collapse
Affiliation(s)
- Kailash Singh
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| | - Bernard My Cheung
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China.,Department of Pharmacy & Pharmacology, The University of Hong Kong, Hong Kong, China
| |
Collapse
|