51
|
Ferreira PCL, Therriault J, Tissot C, Ferrari-Souza JP, Benedet AL, Povala G, Bellaver B, Leffa DT, Brum WS, Lussier FZ, Bezgin G, Servaes S, Vermeiren M, Macedo AC, Cabrera A, Stevenson J, Triana-Baltzer G, Kolb H, Rahmouni N, Klunk WE, Lopez O, Villemagne VL, Cohen A, Tudorascu DL, Zimmer ER, Karikari TK, Ashton NJ, Zetterberg H, Blennow K, Gauthier S, Rosa-Neto P, Pascoal TA. Plasma p-tau231 and p-tau217 inform on tau tangles aggregation in cognitively impaired individuals. Alzheimers Dement 2023; 19:4463-4474. [PMID: 37534889 PMCID: PMC10592380 DOI: 10.1002/alz.13393] [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: 02/24/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 08/04/2023]
Abstract
INTRODUCTION Phosphorylated tau (p-tau) biomarkers have been recently proposed to represent brain amyloid-β (Aβ) pathology. Here, we evaluated the plasma biomarkers' contribution beyond the information provided by demographics (age and sex) to identify Aβ and tau pathologies in individuals segregated as cognitively unimpaired (CU) and impaired (CI). METHODS We assessed 138 CU and 87 CI with available plasma p-tau231, 217+ , and 181, Aβ42/40, GFAP and Aβ- and tau-PET. RESULTS In CU, only plasma p-tau231 and p-tau217+ significantly improved the performance of the demographics in detecting Aβ-PET positivity, while no plasma biomarker provided additional information to identify tau-PET positivity. In CI, p-tau217+ and GFAP significantly contributed to demographics to identify both Aβ-PET and tau-PET positivity, while p-tau231 only provided additional information to identify tau-PET positivity. DISCUSSION Our results support plasma p-tau231 and p-tau217+ as state markers of early Aβ deposition, but in later disease stages they inform on tau tangle accumulation. HIGHLIGHTS It is still unclear how much plasma biomarkers contribute to identification of AD pathology across the AD spectrum beyond the information already provided by demographics (age + sex). Plasma p-tau231 and p-tau217+ contribute to demographic information to identify brain Aβ pathology in preclinical AD. In CI individuals, plasma p-tau231 contributes to age and sex to inform on the accumulation of tau tangles, while p-tau217+ and GFAP inform on both Aβ deposition and tau pathology.
Collapse
Affiliation(s)
- Pamela C. L Ferreira
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Cécile Tissot
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - João Pedro Ferrari-Souza
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Andréa L. Benedet
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
| | - Guilherme Povala
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Bruna Bellaver
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Douglas T. Leffa
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Wagner S. Brum
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
| | - Firoza Z. Lussier
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Marie Vermeiren
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Arthur C. Macedo
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Arlec Cabrera
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Gallen Triana-Baltzer
- Neuroscience Biomarkers, Janssen Research and Development, La Jolla, CA, 92121-1126, USA
| | - Hartmuth Kolb
- Neuroscience Biomarkers, Janssen Research and Development, La Jolla, CA, 92121-1126, USA
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - William E. Klunk
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Oscar Lopez
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Victor L. Villemagne
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Ann Cohen
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Dana L. Tudorascu
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Eduardo R. Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeuctis, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
- Brain Insitute of Rio Grande do Sul, PUCRS, Porto Alegre, 90619-900, Brazil
| | - Thomas K. Karikari
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, 431 41, Sweden
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, WC1N 3BG, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1N 3BG, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, HKG, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
| | - Serge Gauthier
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Pedro Rosa-Neto
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Tharick A. Pascoal
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
52
|
Verma M, Chopra M, Kumar H. Unraveling the Potential of EphA4: A Breakthrough Target and Beacon of Hope for Neurological Diseases. Cell Mol Neurobiol 2023; 43:3375-3391. [PMID: 37477786 DOI: 10.1007/s10571-023-01390-0] [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: 04/22/2023] [Accepted: 07/13/2023] [Indexed: 07/22/2023]
Abstract
Erythropoietin-producing hepatocellular carcinoma A4 (EphA4) is a transmembrane receptor protein which is a part of the most prominent family of receptor tyrosine kinases (RTKs). It serves a crucial role in both physiological, biological, and functional states binding with their ligand like Ephrins. Its abundance in the majority of the body's systems has been reported. Moreover, it draws much attention in the CNS since it influences axonal and vascular guidance. Also, it has a widespread role at the pathological state of various CNS disorders. Reports suggest it obstructs axonal regeneration in various neurodegenerative diseases and neurological disorders. Although, neuro-regeneration is still an open challenge to the modern drug discovery community. Hence, in this review, we will provide information about the role of EphA4 in neurological diseases by which it may emerge as a therapeutic target for CNS disease. We will also provide a glance at numerous signaling pathways that activate or inhibit the EphA4-associated biological processes contributing to the course of neurodegenerative diseases. Thus, this work might serve as a basis for futuristic studies that are related to the target-based drug discovery in the field of neuro-regeneration. Pathological and physiological events associated with EphA4 and Ephrin upregulation and interaction.
Collapse
Affiliation(s)
- Meenal Verma
- National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Manjeet Chopra
- National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Hemant Kumar
- National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
53
|
Hampel H, Hu Y, Cummings J, Mattke S, Iwatsubo T, Nakamura A, Vellas B, O'Bryant S, Shaw LM, Cho M, Batrla R, Vergallo A, Blennow K, Dage J, Schindler SE. Blood-based biomarkers for Alzheimer's disease: Current state and future use in a transformed global healthcare landscape. Neuron 2023; 111:2781-2799. [PMID: 37295421 PMCID: PMC10720399 DOI: 10.1016/j.neuron.2023.05.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 03/03/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023]
Abstract
Timely detection of the pathophysiological changes and cognitive impairment caused by Alzheimer's disease (AD) is increasingly pressing because of the advent of biomarker-guided targeted therapies that may be most effective when provided early in the disease. Currently, diagnosis and management of early AD are largely guided by clinical symptoms. FDA-approved neuroimaging and cerebrospinal fluid biomarkers can aid detection and diagnosis, but the clinical implementation of these testing modalities is limited because of availability, cost, and perceived invasiveness. Blood-based biomarkers (BBBMs) may enable earlier and faster diagnoses as well as aid in risk assessment, early detection, prognosis, and management. Herein, we review data on BBBMs that are closest to clinical implementation, particularly those based on measures of amyloid-β peptides and phosphorylated tau species. We discuss key parameters and considerations for the development and potential deployment of these BBBMs under different contexts of use and highlight challenges at the methodological, clinical, and regulatory levels.
Collapse
Affiliation(s)
- Harald Hampel
- Alzheimer's Disease and Brain Health, Eisai Inc., Nutley, NJ, USA.
| | - Yan Hu
- Alzheimer's Disease and Brain Health, Eisai Inc., Nutley, NJ, USA.
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Soeren Mattke
- Center for Improving Chronic Illness Care, University of Southern California, Los Angeles, CA, USA
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akinori Nakamura
- Department of Biomarker Research, National Center for Geriatrics and Gerontology, Obu, Japan; Department of Cognition and Behavior Science, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Bruno Vellas
- University Paul Sabatier, Gérontopôle, Toulouse University Hospital, UMR INSERM 1285, Toulouse, France
| | - Sid O'Bryant
- Institute for Translational Research, Texas College of Osteopathic Medicine, Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Leslie M Shaw
- Perelman School of Medicine, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Min Cho
- Alzheimer's Disease and Brain Health, Eisai Inc., Nutley, NJ, USA
| | - Richard Batrla
- Alzheimer's Disease and Brain Health, Eisai Inc., Nutley, NJ, USA
| | - Andrea Vergallo
- Alzheimer's Disease and Brain Health, Eisai Inc., Nutley, NJ, USA
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Jeffrey Dage
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
54
|
Lopes JR, Zhang X, Mayrink J, Tatematsu BK, Guo L, LeServe DS, Abou-El-Hassan H, Rong F, Dalton MJ, Oliveira MG, Lanser TB, Liu L, Butovsky O, Rezende RM, Weiner HL. Nasal administration of anti-CD3 monoclonal antibody ameliorates disease in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 2023; 120:e2309221120. [PMID: 37669383 PMCID: PMC10500187 DOI: 10.1073/pnas.2309221120] [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: 06/04/2023] [Accepted: 07/31/2023] [Indexed: 09/07/2023] Open
Abstract
Emerging evidence suggests that dysregulation of neuroinflammation, particularly that orchestrated by microglia, plays a significant role in the pathogenesis of Alzheimer's disease (AD). Danger signals including dead neurons, dystrophic axons, phosphorylated tau, and amyloid plaques alter the functional phenotype of microglia from a homeostatic (M0) to a neurodegenerative or disease-associated phenotype, which in turn drives neuroinflammation and promotes disease. Thus, therapies that target microglia activation constitute a unique approach for treating AD. Here, we report that nasally administered anti-CD3 monoclonal antibody in the 3xTg AD mouse model reduced microglial activation and improved cognition independent of amyloid beta deposition. In addition, gene expression analysis demonstrated decreased oxidative stress, increased axogenesis and synaptic organization, and metabolic changes in the hippocampus and cortex of nasal anti-CD3 treated animals. The beneficial effect of nasal anti-CD3 was associated with the accumulation of T cells in the brain where they were in close contact with microglial cells. Taken together, our findings identify nasal anti-CD3 as a unique form of immunotherapy to treat Alzheimer's disease independent of amyloid beta targeting.
Collapse
Affiliation(s)
- Juliana R. Lopes
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Xiaoming Zhang
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Julia Mayrink
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Bruna K. Tatematsu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Lydia Guo
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Danielle S. LeServe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Hadi Abou-El-Hassan
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Felipe Rong
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Maria J. Dalton
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Marilia G. Oliveira
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Toby B. Lanser
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Lei Liu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Rafael M. Rezende
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Howard L. Weiner
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| |
Collapse
|
55
|
Self WK, Holtzman DM. Emerging diagnostics and therapeutics for Alzheimer disease. Nat Med 2023; 29:2187-2199. [PMID: 37667136 DOI: 10.1038/s41591-023-02505-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/18/2023] [Indexed: 09/06/2023]
Abstract
Alzheimer disease (AD) is the most common contributor to dementia in the world, but strategies that slow or prevent its clinical progression have largely remained elusive, until recently. This Review highlights the latest advances in biomarker technologies and therapeutic development to improve AD diagnosis and treatment. We review recent results that enable pathological staging of AD with neuroimaging and fluid-based biomarkers, with a particular emphasis on the role of amyloid, tau and neuroinflammation in disease pathogenesis. We discuss the lessons learned from randomized controlled trials, including some supporting the proposal that certain anti-amyloid antibodies slow cognitive decline during the mildly symptomatic phase of AD. In addition, we highlight evidence for newly identified therapeutic targets that may be able to modify AD pathogenesis and progression. Collectively, these recent discoveries-and the research directions that they open-have the potential to move AD clinical care toward disease-modifying treatment strategies with maximal benefits for patients.
Collapse
Affiliation(s)
- Wade K Self
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
56
|
Nadel CM, Wucherer K, Oehler A, Thwin AC, Basu K, Callahan MD, Southworth DR, Mordes DA, Craik CS, Gestwicki JE. Phosphorylation of a Cleaved Tau Proteoform at a Single Residue Inhibits Binding to the E3 Ubiquitin Ligase, CHIP. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.16.553575. [PMID: 37645969 PMCID: PMC10462110 DOI: 10.1101/2023.08.16.553575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Microtubule-associated protein tau (MAPT/tau) accumulates in a family of neurodegenerative diseases, including Alzheimer's disease (AD). In disease, tau is aberrantly modified by post-translational modifications (PTMs), including hyper-phosphorylation. However, it is often unclear which of these PTMs contribute to tau's accumulation or what mechanisms might be involved. To explore these questions, we focused on a cleaved proteoform of tau (tauC3), which selectively accumulates in AD and was recently shown to be degraded by its direct binding to the E3 ubiquitin ligase, CHIP. Here, we find that phosphorylation of tauC3 at a single residue, pS416, is sufficient to block its interaction with CHIP. A co-crystal structure of CHIP bound to the C-terminus of tauC3 revealed the mechanism of this clash and allowed design of a mutation (CHIPD134A) that partially restores binding and turnover of pS416 tauC3. We find that pS416 is produced by the known AD-associated kinase, MARK2/Par-1b, providing a potential link to disease. In further support of this idea, an antibody against pS416 co-localizes with tauC3 in degenerative neurons within the hippocampus of AD patients. Together, these studies suggest a discrete molecular mechanism for how phosphorylation at a specific site contributes to accumulation of an important tau proteoform.
Collapse
Affiliation(s)
- Cory M Nadel
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94158
| | - Kristin Wucherer
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158
| | - Abby Oehler
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94158
| | - Aye C Thwin
- Department of Biochemistry & Biophysics, University of California San Francisco, San Francisco, CA 94158
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94158
| | - Koli Basu
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158
| | - Matthew D Callahan
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94158
| | - Daniel R Southworth
- Department of Biochemistry & Biophysics, University of California San Francisco, San Francisco, CA 94158
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94158
| | - Daniel A Mordes
- Department of Pathology, University of California San Francisco, San Francisco, CA 94158
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94158
| | - Charles S Craik
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94158
| |
Collapse
|
57
|
Ondrejcak T, Klyubin I, Hu NW, O'Malley TT, Corbett GT, Winters R, Perkinton MS, Billinton A, Prenderville JA, Walsh DM, Rowan MJ. Tau and Amyloid β Protein in Patient-Derived Aqueous Brain Extracts Act Concomitantly to Disrupt Long-Term Potentiation in Vivo. J Neurosci 2023; 43:5870-5879. [PMID: 37491315 PMCID: PMC10423043 DOI: 10.1523/jneurosci.0082-23.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/07/2023] [Accepted: 07/05/2023] [Indexed: 07/27/2023] Open
Abstract
Amyloid β protein (Aβ) and tau, the two main proteins implicated in causing Alzheimer's disease (AD), are posited to trigger synaptic dysfunction long before significant synaptic loss occurs in vulnerable circuits. Whereas soluble Aβ aggregates from AD brain are well recognized potent synaptotoxins, less is known about the synaptotoxicity of soluble tau from AD or other tauopathy patient brains. Minimally manipulated patient-derived aqueous brain extracts contain the more diffusible native forms of these proteins. Here, we explore how intracerebral injection of Aβ and tau present in such aqueous extracts of patient brain contribute to disruption of synaptic plasticity in the CA1 area of the male rat hippocampus. Aqueous extracts of certain AD brains acutely inhibited long-term potentiation (LTP) of synaptic transmission in a manner that required both Aβ and tau. Tau-containing aqueous extracts of a brain from a patient with Pick's disease (PiD) also impaired LTP, and diffusible tau from either AD or PiD brain lowered the threshold for AD brain Aβ to inhibit LTP. Remarkably, the disruption of LTP persisted for at least 2 weeks after a single injection. These findings support a critical role for diffusible tau in causing rapid onset, persistent synaptic plasticity deficits, and promoting Aβ-mediated synaptic dysfunction.SIGNIFICANCE STATEMENT The microtubule-associated protein tau forms relatively insoluble fibrillar deposits in the brains of people with neurodegenerative diseases including Alzheimer's and Pick's diseases. More soluble aggregates of disease-associated tau may diffuse between cells and could cause damage to synapses in vulnerable circuits. We prepared aqueous extracts of diseased cerebral cortex and tested their ability to interfere with synaptic function in the brains of live rats. Tau in these extracts rapidly and persistently disrupted synaptic plasticity and facilitated impairments caused by amyloid β protein, the other major pathologic protein in Alzheimer's disease. These findings show that certain diffusible forms of tau can mediate synaptic dysfunction and may be a target for therapy.
Collapse
Affiliation(s)
- Tomas Ondrejcak
- Department of Pharmacology & Therapeutics, School of Medicine and Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | - Igor Klyubin
- Department of Pharmacology & Therapeutics, School of Medicine and Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | - Neng-Wei Hu
- Department of Pharmacology & Therapeutics, School of Medicine and Institute of Neuroscience, Trinity College, Dublin 2, Ireland
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Tiernan T O'Malley
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, Massachusetts 02115
| | - Grant T Corbett
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, Massachusetts 02115
| | - Róisín Winters
- Transpharmation Ireland, Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | - Michael S Perkinton
- Neuroscience, BioPharmaceuticals R&D, AstraZeneca UK, Cambridge, CB21 6GH, United Kingdom
| | - Andy Billinton
- Neuroscience, BioPharmaceuticals R&D, AstraZeneca UK, Cambridge, CB21 6GH, United Kingdom
| | - Jack A Prenderville
- Transpharmation Ireland, Institute of Neuroscience, Trinity College, Dublin 2, Ireland
- Department of Physiology, School of Medicine, Trinity College, Dublin 2, Ireland
| | - Dominic M Walsh
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, Massachusetts 02115
| | - Michael J Rowan
- Department of Pharmacology & Therapeutics, School of Medicine and Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| |
Collapse
|
58
|
Horie K, Salvadó G, Barthélemy NR, Janelidze S, Li Y, He Y, Saef B, Chen CD, Jiang H, Strandberg O, Pichet Binette A, Palmqvist S, Sato C, Sachdev P, Koyama A, Gordon BA, Benzinger TLS, Holtzman DM, Morris JC, Mattsson-Carlgren N, Stomrud E, Ossenkoppele R, Schindler SE, Hansson O, Bateman RJ. CSF MTBR-tau243 is a specific biomarker of tau tangle pathology in Alzheimer's disease. Nat Med 2023; 29:1954-1963. [PMID: 37443334 PMCID: PMC10427417 DOI: 10.1038/s41591-023-02443-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 06/05/2023] [Indexed: 07/15/2023]
Abstract
Aggregated insoluble tau is one of two defining features of Alzheimer's disease. Because clinical symptoms are strongly correlated with tau aggregates, drug development and clinical diagnosis need cost-effective and accessible specific fluid biomarkers of tau aggregates; however, recent studies suggest that the fluid biomarkers currently available cannot specifically track tau aggregates. We show that the microtubule-binding region (MTBR) of tau containing the residue 243 (MTBR-tau243) is a new cerebrospinal fluid (CSF) biomarker specific for insoluble tau aggregates and compared it to multiple other phosphorylated tau measures (p-tau181, p-tau205, p-tau217 and p-tau231) in two independent cohorts (BioFINDER-2, n = 448; and Knight Alzheimer Disease Research Center, n = 219). MTBR-tau243 was most strongly associated with tau-positron emission tomography (PET) and cognition, whereas showing the lowest association with amyloid-PET. In combination with p-tau205, MTBR-tau243 explained most of the total variance in tau-PET burden (0.58 ≤ R2 ≤ 0.75) and the performance in predicting cognitive measures (0.34 ≤ R2 ≤ 0.48) approached that of tau-PET (0.44 ≤ R2 ≤ 0.52). MTBR-tau243 levels longitudinally increased with insoluble tau aggregates, unlike CSF p-tau species. CSF MTBR-tau243 is a specific biomarker of tau aggregate pathology, which may be utilized in interventional trials and in the diagnosis of patients. Based on these findings, we propose to revise the A/T/(N) criteria to include MTBR-tau243 as representing insoluble tau aggregates ('T').
Collapse
Grants
- P30 AG066444 NIA NIH HHS
- R01 AG070941 NIA NIH HHS
- P01 AG003991 NIA NIH HHS
- P01 AG026276 NIA NIH HHS
- P30 NS048056 NINDS NIH HHS
- S10 OD025214 NIH HHS
- The Tracy Family SILQ Center established by the Tracy Family, Richard Frimel and Gary Werths, GHR Foundation, David Payne, and the Willman Family brought together by The Foundation for Barnes-Jewish Hospital.
- Eisai industry grant
- The European Union’s Horizon 2020 research and innovation program under the Marie Sklodowska-Curie action grant agreement No 101061836, from Greta och Johan Kocks research grants and, travel grants from the Strategic Research Area MultiPark (Multidisciplinary Research in Parkinson’s disease) at Lund University
- U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- The Swedish Research Council (2016-00906), the Knut and Alice Wallenberg foundation (2017-0383), the Marianne and Marcus Wallenberg foundation (2015.0125), the Strategic Research Area MultiPark (Multidisciplinary Research in Parkinson’s disease) at Lund University, the Swedish Alzheimer Foundation (AF-939932), the Swedish Brain Foundation (FO2021-0293), The Parkinson foundation of Sweden (1280/20), the Cure Alzheimer’s fund, the Konung Gustaf V:s och Drottning Victorias Frimurarestiftelse, the Skåne University Hospital Foundation (2020-O000028), Regionalt Forskningsstöd (2020-0314) and the Swedish federal government under the ALF agreement (2018-Projekt0279)
- The Knight ADRC developmental project
Collapse
Affiliation(s)
- Kanta Horie
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Eisai Inc., Nutley, NJ, USA
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Nicolas R Barthélemy
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yingxin He
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Benjamin Saef
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Charles D Chen
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hong Jiang
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Chihiro Sato
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | - Brian A Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| | - Randall J Bateman
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, USA.
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
59
|
Erickson P, Simrén J, Brum WS, Ennis GE, Kollmorgen G, Suridjan I, Langhough R, Jonaitis EM, Van Hulle CA, Betthauser TJ, Carlsson CM, Asthana S, Ashton NJ, Johnson SC, Shaw LM, Blennow K, Andreasson U, Bendlin BB, Zetterberg H. Prevalence and Clinical Implications of a β-Amyloid-Negative, Tau-Positive Cerebrospinal Fluid Biomarker Profile in Alzheimer Disease. JAMA Neurol 2023; 80:2807607. [PMID: 37523162 PMCID: PMC10391361 DOI: 10.1001/jamaneurol.2023.2338] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/05/2023] [Indexed: 08/01/2023]
Abstract
Importance Knowledge is lacking on the prevalence and prognosis of individuals with a β-amyloid-negative, tau-positive (A-T+) cerebrospinal fluid (CSF) biomarker profile. Objective To estimate the prevalence of a CSF A-T+ biomarker profile and investigate its clinical implications. Design, Setting, and Participants This was a retrospective cohort study of the cross-sectional multicenter University of Gothenburg (UGOT) cohort (November 2019-January 2021), the longitudinal multicenter Alzheimer Disease Neuroimaging Initiative (ADNI) cohort (individuals with mild cognitive impairment [MCI] and no cognitive impairment; September 2005-May 2022), and 2 Wisconsin cohorts, Wisconsin Alzheimer Disease Research Center and Wisconsin Registry for Alzheimer Prevention (WISC; individuals without cognitive impairment; February 2007-November 2020). This was a multicenter study, with data collected from referral centers in clinical routine (UGOT) and research settings (ADNI and WISC). Eligible individuals had 1 lumbar puncture (all cohorts), 2 or more cognitive assessments (ADNI and WISC), and imaging (ADNI only) performed on 2 separate occasions. Data were analyzed on August 2022 to April 2023. Exposures Baseline CSF Aβ42/40 and phosphorylated tau (p-tau)181; cognitive tests (ADNI: modified preclinical Alzheimer cognitive composite [mPACC]; WISC: modified 3-test PACC [PACC-3]). Exposures in the ADNI cohort included [18F]-florbetapir amyloid positron emission tomography (PET), magnetic resonance imaging (MRI), [18F]-fluorodeoxyglucose PET (FDG-PET), and cross-sectional tau-PET (ADNI: [18F]-flortaucipir, WISC: [18F]-MK6240). Main Outcomes and Measures Primary outcomes were the prevalence of CSF AT biomarker profiles and continuous longitudinal global cognitive outcome and imaging biomarker trajectories in A-T+ vs A-T- groups. Secondary outcomes included cross-sectional tau-PET. Results A total of 7679 individuals (mean [SD] age, 71.0 [8.4] years; 4101 male [53%]) were included in the UGOT cohort, 970 individuals (mean [SD] age, 73 [7.0] years; 526 male [54%]) were included in the ADNI cohort, and 519 individuals (mean [SD] age, 60 [7.3] years; 346 female [67%]) were included in the WISC cohort. The prevalence of an A-T+ profile in the UGOT cohort was 4.1% (95% CI, 3.7%-4.6%), being less common than the other patterns. Longitudinally, no significant differences in rates of worsening were observed between A-T+ and A-T- profiles for cognition or imaging biomarkers. Cross-sectionally, A-T+ had similar tau-PET uptake to individuals with an A-T- biomarker profile. Conclusion and Relevance Results suggest that the CSF A-T+ biomarker profile was found in approximately 5% of lumbar punctures and was not associated with a higher rate of cognitive decline or biomarker signs of disease progression compared with biomarker-negative individuals.
Collapse
Affiliation(s)
- Pontus Erickson
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joel Simrén
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Wagner S. Brum
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Gilda E. Ennis
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | | | | | - Rebecca Langhough
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Erin M. Jonaitis
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Carol A. Van Hulle
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Tobey J. Betthauser
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Cynthia M. Carlsson
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison
- Geriatric Research Education and Clinical Center of the Wm. S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Sanjay Asthana
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison
- Geriatric Research Education and Clinical Center of the Wm. S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Nicholas J. Ashton
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, King’s College London, London, England
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, England
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Sterling C. Johnson
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ulf Andreasson
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Barbara B. Bendlin
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Institute of Neurology, Department of Neurodegenerative Disease, University College London, London, England
- UK Dementia Research Institute, University College London, London, England
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| |
Collapse
|
60
|
Snellman A, Ekblad LL, Ashton NJ, Karikari TK, Lantero-Rodriguez J, Pietilä E, Koivumäki M, Helin S, Karrasch M, Zetterberg H, Blennow K, Rinne JO. Head-to-head comparison of plasma p-tau181, p-tau231 and glial fibrillary acidic protein in clinically unimpaired elderly with three levels of APOE4-related risk for Alzheimer's disease. Neurobiol Dis 2023; 183:106175. [PMID: 37268240 DOI: 10.1016/j.nbd.2023.106175] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 05/02/2023] [Accepted: 05/26/2023] [Indexed: 06/04/2023] Open
Abstract
Plasma phosphorylated tau (p-tau) and glial fibrillary acidic protein (GFAP) both reflect early changes in Alzheimer's disease (AD) pathology. Here, we compared the biomarker levels and their association with regional β-amyloid (Aβ) pathology and cognitive performance head-to-head in clinically unimpaired elderly (n = 88) at three levels of APOE4-related genetic risk for sporadic AD (APOE4/4 n = 19, APOE3/4 n = 32 or non-carriers n = 37). Concentrations of plasma p-tau181, p-tau231 and GFAP were measured using Single molecule array (Simoa), regional Aβ deposition with 11C-PiB positron emission tomography (PET), and cognitive performance with a preclinical composite. Significant differences in plasma p-tau181 and p-tau231, but not plasma GFAP concentrations were present between the APOE4 gene doses, explained solely by brain Aβ load. All plasma biomarkers correlated positively with Aβ PET in the total study population. This correlation was driven by APOE3/3 carriers for plasma p-tau markers and APOE4/4 carriers for plasma GFAP. Voxel-wise associations with amyloid-PET revealed different spatial patterns for plasma p-tau markers and plasma GFAP. Only higher plasma GFAP correlated with lower cognitive scores. Our observations suggest that plasma p-tau and plasma GFAP are both early AD markers reflecting different Aβ-related processes.
Collapse
Affiliation(s)
- Anniina Snellman
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland; Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
| | - Laura L Ekblad
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway; Department of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Elina Pietilä
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
| | - Mikko Koivumäki
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
| | - Semi Helin
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
| | - Mira Karrasch
- Department of Psychology, Åbo Akademi University, Turku, Finland
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; UK Dementia Research Institute at UCL, London, UK; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Juha O Rinne
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland; InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| |
Collapse
|
61
|
Kyalu Ngoie Zola N, Balty C, Pyr Dit Ruys S, Vanparys AAT, Huyghe NDG, Herinckx G, Johanns M, Boyer E, Kienlen-Campard P, Rider MH, Vertommen D, Hanseeuw BJ. Specific post-translational modifications of soluble tau protein distinguishes Alzheimer's disease and primary tauopathies. Nat Commun 2023; 14:3706. [PMID: 37349319 PMCID: PMC10287718 DOI: 10.1038/s41467-023-39328-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 06/07/2023] [Indexed: 06/24/2023] Open
Abstract
Tau protein aggregates in several neurodegenerative disorders, referred to as tauopathies. The tau isoforms observed in post mortem human brain aggregates is used to classify tauopathies. However, distinguishing tauopathies ante mortem remains challenging, potentially due to differences between insoluble tau in aggregates and soluble tau in body fluids. Here, we demonstrated that tau isoforms differ between tauopathies in insoluble aggregates, but not in soluble brain extracts. We therefore characterized post-translational modifications of both the aggregated and the soluble tau protein obtained from post mortem human brain tissue of patients with Alzheimer's disease, cortico-basal degeneration, Pick's disease, and frontotemporal lobe degeneration. We found specific soluble signatures for each tauopathy and its specific aggregated tau isoforms: including ubiquitination on Lysine 369 for cortico-basal degeneration and acetylation on Lysine 311 for Pick's disease. These findings provide potential targets for future development of fluid-based biomarker assays able to distinguish tauopathies in vivo.
Collapse
Affiliation(s)
- Nathalie Kyalu Ngoie Zola
- Universite catholique de Louvain (UCLouvain) and Institute of Neuroscience (IONS), 1200, Brussels, Belgium
- Universite catholique de Louvain (UCLouvain) and de Duve Institute (DDUV), Protein Phosphorylation (PHOS), 1200, Brussels, Belgium
| | - Clémence Balty
- Universite catholique de Louvain (UCLouvain) and de Duve Institute (DDUV), Protein Phosphorylation (PHOS), 1200, Brussels, Belgium
| | - Sébastien Pyr Dit Ruys
- Universite catholique de Louvain (UClouvain) and Louvain Drug Research Institute (LDRI), Integrated Pharmacometrics, Pharmacogenomics and Pharmacokinetics Group (PMGK), 1200, Brussels, Belgium
| | - Axelle A T Vanparys
- Universite catholique de Louvain (UCLouvain) and Institute of Neuroscience (IONS), 1200, Brussels, Belgium
| | - Nicolas D G Huyghe
- Université catholique de Louvain (UCLouvain) and Institut de Recherche Expérimentale et Clinique (IREC), 1200, Brussels, Belgium
| | - Gaëtan Herinckx
- Universite catholique de Louvain (UCLouvain), de Duve Institute (DDUV), and MASSPROT Platform, 1200, Brussels, Belgium
| | - Manuel Johanns
- Universite catholique de Louvain (UCLouvain) and de Duve Institute (DDUV), Protein Phosphorylation (PHOS), 1200, Brussels, Belgium
| | - Emilien Boyer
- Universite catholique de Louvain (UCLouvain) and Institute of Neuroscience (IONS), 1200, Brussels, Belgium
- Cliniques universitaires Saint-Luc, Neurology Department, 1200, Brussels, Belgium
| | - Pascal Kienlen-Campard
- Universite catholique de Louvain (UCLouvain) and Institute of Neuroscience (IONS), 1200, Brussels, Belgium
| | - Mark H Rider
- Universite catholique de Louvain (UCLouvain) and de Duve Institute (DDUV), Protein Phosphorylation (PHOS), 1200, Brussels, Belgium
| | - Didier Vertommen
- Universite catholique de Louvain (UCLouvain), de Duve Institute (DDUV), and MASSPROT Platform, 1200, Brussels, Belgium
| | - Bernard J Hanseeuw
- Universite catholique de Louvain (UCLouvain) and Institute of Neuroscience (IONS), 1200, Brussels, Belgium.
- Cliniques universitaires Saint-Luc, Neurology Department, 1200, Brussels, Belgium.
- Universite catholique de Louvain (UCLouvain), WELBIO department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium.
- Harvard Medical School, Massachusetts General Hospital, Department of Radiology, Gordon Center for Medical Imaging, Boston, MA, USA.
| |
Collapse
|
62
|
Li B, Shi K, Ren C, Kong M, Ba M. Detection of Tau-PET Positivity in Clinically Diagnosed Mild Cognitive Impairment with Multidimensional Features. J Alzheimers Dis 2023:JAD230180. [PMID: 37334600 DOI: 10.3233/jad-230180] [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: 06/20/2023]
Abstract
BACKGROUND The way to evaluate brain tau pathology in vivo is tau positron emission tomography (tau-PET) or cerebrospinal fluid (CSF) analysis. In the clinically diagnosed mild cognitive impairment (MCI), a significant proportion of tau-PET are negative. Interest in less expensive and convenient ways to detect tau pathology in Alzheimer's disease has increased due to the high cost of tau-PET and the invasiveness of lumbar puncture, which typically slows down the cost and enrollment of clinical trials. OBJECTIVE We aimed to investigate one simple and effective method in predicting tau-PET status in MCI individuals. METHODS The sample included 154 individuals which were dichotomized into tau-PET (+) and tau-PET (-) using a cut-off of >1.33. We used stepwise regression to select the unitary or combination of variables that best predicted tau-PET. The receiver operating characteristic curve was used to assess the accuracy of single and multiple clinical markers. RESULTS The combined performance of three variables [Alzheimer's Disease Assessment Scale-Cognitive Subscale 13 (ADAS-Cog13), Mini-Mental State Examination (MMSE), ADNI-Memory summary score (ADNI-MEM)] in neurocognitive measures demonstrated good predictive accuracy of tau-PET status [accuracy = 85.7%, area under the curve (AUC) = 0.879]. The combination of clinical markers model (APOEɛ4, neurocognitive measures and structural MRI imaging of middle temporal) had the best discriminative power (AUC = 0.946). CONCLUSION As a noninvasive test, the combination of APOEɛ4, neurocognitive measures and structural MRI imaging of middle temporal accurately predicts tau-PET status. The finding may provide a non-invasive, cost-effective tool for clinical application in predicting tau pathology among MCI individuals.
Collapse
Affiliation(s)
- Bingyu Li
- Department of Neurology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Kening Shi
- Department of Neurology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Chao Ren
- Department of Neurology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Min Kong
- Department of Neurology, Yantaishan Hospital, Yantai, Shandong, China
| | - Maowen Ba
- Department of Neurology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
- Yantai Regional Sub Center of National Center for Clinical Medical Research of Neurological Diseases, Shandong, China
| |
Collapse
|
63
|
Zhang L, Xia Y, Gui Y. Neuronal ApoE4 in Alzheimer's disease and potential therapeutic targets. Front Aging Neurosci 2023; 15:1199434. [PMID: 37333457 PMCID: PMC10272394 DOI: 10.3389/fnagi.2023.1199434] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/16/2023] [Indexed: 06/20/2023] Open
Abstract
The most prevalent genetic risk factor for Alzheimer's disease (AD) is Apolipoprotein E (ApoE), a gene located on chromosome 19 that encodes three alleles (e2, e3, and e4) that give rise to the ApoE subtypes E2, E3, and E4, respectively. E2 and E4 have been linked to increased plasma triglyceride concentrations and are known to play a critical role in lipoprotein metabolism. The prominent pathological features of AD mainly include senile plaques formed by amyloid β (Aβ42) aggregation and neuronal fibrous tangles (NFTs), and the deposited plaques are mainly composed of Aβ hyperphosphorylation and truncated head. In the central nervous system, the ApoE protein is primarily derived from astrocytes, but ApoE is also produced when neurons are stressed or affected by certain stress, injury, and aging conditions. ApoE4 in neurons induces Aβ and tau protein pathologies, leading to neuroinflammation and neuronal damage, impairing learning and memory functions. However, how neuronal ApoE4 mediates AD pathology remains unclear. Recent studies have shown that neuronal ApoE4 may lead to greater neurotoxicity, which increases the risk of AD development. This review focuses on the pathophysiology of neuronal ApoE4 and explains how neuronal ApoE4 mediates Aβ deposition, pathological mechanisms of tau protein hyperphosphorylation, and potential therapeutic targets.
Collapse
|
64
|
Younes K, Sha SJ. The most valuable player or the tombstone: is tau the correct target to treat Alzheimer's disease? Brain 2023; 146:2211-2213. [PMID: 37150887 DOI: 10.1093/brain/awad151] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 05/02/2023] [Indexed: 05/09/2023] Open
Abstract
This scientific commentary refers to ‘Tilavonemab in early Alzheimer’s disease: results from a phase 2, randomized, double-blind study’ by Florian et al. (https://doi.org/10.1093/brain/awad024).
Collapse
Affiliation(s)
- Kyan Younes
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Sharon J Sha
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
65
|
Horie K, Li Y, Barthélemy NR, Gordon BA, Hassenstab J, Benzinger TL, Fagan AM, Morris JC, Karch CM, Xiong C, Allegri R, Mendez PC, Ikeuchi T, Kasuga K, Noble J, Farlow M, Chhatwal J, Day GS, Schofield PR, Masters CL, Levin J, Jucker M, Lee JH, Hoon Roh J, Sato C, Sachdev P, Koyama A, Reyderman L, Bateman RJ, McDade E. Change in Cerebrospinal Fluid Tau Microtubule Binding Region Detects Symptom Onset, Cognitive Decline, Tangles, and Atrophy in Dominantly Inherited Alzheimer's Disease. Ann Neurol 2023; 93:1158-1172. [PMID: 36843330 PMCID: PMC10238659 DOI: 10.1002/ana.26620] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/28/2023]
Abstract
OBJECTIVE Identifying cerebrospinal fluid measures of the microtubule binding region of tau (MTBR-tau) species that reflect tau aggregation could provide fluid biomarkers that track Alzheimer's disease related neurofibrillary tau pathological changes. We examined the cerebrospinal fluid (CSF) MTBR-tau species in dominantly inherited Alzheimer's disease (DIAD) mutation carriers to assess the association with Alzheimer's disease (AD) biomarkers and clinical symptoms. METHODS Cross-sectional and longitudinal CSF from 229 DIAD mutation carriers and 130 mutation non-carriers had sequential characterization of N-terminal/mid-domain phosphorylated tau (p-tau) followed by MTBR-tau species and tau positron emission tomography (tau PET), other soluble tau and amyloid biomarkers, comprehensive clinical and cognitive assessments, and brain magnetic resonance imaging of atrophy. RESULTS CSF MTBR-tau species located within the putative "border" region and one species corresponding to the "core" region of aggregates in neurofibrillary tangles (NFTs) increased during the presymptomatic stage and decreased during the symptomatic stage. The "border" MTBR-tau species were associated with amyloid pathology and CSF p-tau; whereas the "core" MTBR-tau species were associated stronger with tau PET and CSF measures of neurodegeneration. The ratio of the border to the core species provided a continuous measure of increasing amounts that tracked clinical progression and NFTs. INTERPRETATION Changes in CSF soluble MTBR-tau species preceded the onset of dementia, tau tangle increase, and atrophy in DIAD. The ratio of 4R-specific MTBR-tau (border) to the NFT (core) MTBR-tau species corresponds to the pathology of NFTs in DIAD and change with disease progression. The dynamics between different MTBR-tau species in the CSF may serve as a marker of tau-related disease progression and target engagement of anti-tau therapeutics. ANN NEUROL 2023;93:1158-1172.
Collapse
Affiliation(s)
- Kanta Horie
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Eisai Inc., Nutley, NJ, 07110, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Nicolas R. Barthélemy
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Brian A. Gordon
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Tammie. L.S. Benzinger
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Anne M. Fagan
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Celeste M. Karch
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Ricardo Allegri
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI) Instituto de Investigaciones Neurológicas Raúl Correa, Buenos Aires, Argentina
| | - Patricio Chrem Mendez
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI) Instituto de Investigaciones Neurológicas Raúl Correa, Buenos Aires, Argentina
| | | | | | - James Noble
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, G.H. Sergievsky Center, Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032 USA
| | - Martin Farlow
- Department of Neurology, Indiana University, Indianapolis, IN 46202, USA
| | - Jasmeer Chhatwal
- Massachusetts General Hospital, Harvard Medical School Boston, MA 02114, USA
| | - Gregory S. Day
- Department of Neurology, Mayo Clinic in Florida, Jacksonville, FL 32224, USA
| | - Peter R. Schofield
- Neuroscience Research Australia, Sydney, 2031 NSW, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, 2052 NSW, Australia
| | - Colin L. Masters
- The Florey Institute and the University of Melbourne, Parkville, Victoria 3010, Australia
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) Munich, Marchioninistr 15, D-83177 Munchen, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology, Ludwig-Maximilians Universität München, Marchioninistr 15, 83177 Munich, Germany
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE) Tübingen; and Hertie-Institute for Clinical Brain Research, University of Tübingen, D-72076 Tübingen, Germany
| | - Jae-Hong Lee
- Department of Neurology, Asan Medical Center, Seoul 05505, Korea
| | - Jee Hoon Roh
- Departments of Biomedical Sciences, Physiology, and Neurology, Korea University College of Medicine, Seoul 02841, Korea
| | - Chihiro Sato
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | | | | | | | - Randall J. Bateman
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Eric McDade
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | | |
Collapse
|
66
|
Sun Z, Kwon JS, Ren Y, Chen S, Cates K, Lu X, Walker CK, Karahan H, Sviben S, Fitzpatrick JAJ, Valdez C, Houlden H, Karch CM, Bateman RJ, Sato C, Mennerick SJ, Diamond MI, Kim J, Tanzi RE, Holtzman DM, Yoo AS. Endogenous recapitulation of Alzheimer's disease neuropathology through human 3D direct neuronal reprogramming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.542155. [PMID: 37292658 PMCID: PMC10245934 DOI: 10.1101/2023.05.24.542155] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that primarily affects elderly individuals, and is characterized by hallmark neuronal pathologies including extracellular amyloid-β (Aβ) plaque deposition, intracellular tau tangles, and neuronal death. However, recapitulating these age-associated neuronal pathologies in patient-derived neurons has remained a significant challenge, especially for late-onset AD (LOAD), the most common form of the disorder. Here, we applied the high efficiency microRNA-mediated direct neuronal reprogramming of fibroblasts from AD patients to generate cortical neurons in three-dimensional (3D) Matrigel and self-assembled neuronal spheroids. Our findings indicate that neurons and spheroids reprogrammed from both autosomal dominant AD (ADAD) and LOAD patients exhibited AD-like phenotypes linked to neurons, including extracellular Aβ deposition, dystrophic neurites with hyperphosphorylated, K63-ubiquitin-positive, seed-competent tau, and spontaneous neuronal death in culture. Moreover, treatment with β- or γ-secretase inhibitors in LOAD patient-derived neurons and spheroids before Aβ deposit formation significantly lowered Aβ deposition, as well as tauopathy and neurodegeneration. However, the same treatment after the cells already formed Aβ deposits only had a mild effect. Additionally, inhibiting the synthesis of age-associated retrotransposable elements (RTEs) by treating LOAD neurons and spheroids with the reverse transcriptase inhibitor, lamivudine, alleviated AD neuropathology. Overall, our results demonstrate that direct neuronal reprogramming of AD patient fibroblasts in a 3D environment can capture age-related neuropathology and reflect the interplay between Aβ accumulation, tau dysregulation, and neuronal death. Moreover, miRNA-based 3D neuronal conversion provides a human-relevant AD model that can be used to identify compounds that can potentially ameliorate AD-associated pathologies and neurodegeneration.
Collapse
|
67
|
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:JAD221202. [PMID: 37125550 DOI: 10.3233/jad-221202] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [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
| | - Chia-Chi Ku
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan, ROC
| | - Po-Sheng Hu
- College of Photonics, National Yang Ming Chiao Tung University, Tainan City, Taiwan
| | - Wei-Hung Chiang
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan, R.O.C
| |
Collapse
|
68
|
Brown J, Camporesi E, Lantero-Rodriguez J, Olsson M, Wang A, Medem B, Zetterberg H, Blennow K, Karikari TK, Wall M, Hill E. Tau in cerebrospinal fluid induces neuronal hyperexcitability and alters hippocampal theta oscillations. Acta Neuropathol Commun 2023; 11:67. [PMID: 37095572 PMCID: PMC10127378 DOI: 10.1186/s40478-023-01562-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/03/2023] [Indexed: 04/26/2023] Open
Abstract
Alzheimer's disease (AD) and other tauopathies are characterized by the aggregation of tau into soluble and insoluble forms (including tangles and neuropil threads). In humans, a fraction of both phosphorylated and non-phosphorylated N-terminal to mid-domain tau species, are secreted into cerebrospinal fluid (CSF). Some of these CSF tau species can be measured as diagnostic and prognostic biomarkers, starting from early stages of disease. While in animal models of AD pathology, soluble tau aggregates have been shown to disrupt neuronal function, it is unclear whether the tau species present in CSF will modulate neural activity. Here, we have developed and applied a novel approach to examine the electrophysiological effects of CSF from patients with a tau-positive biomarker profile. The method involves incubation of acutely-isolated wild-type mouse hippocampal brain slices with small volumes of diluted human CSF, followed by a suite of electrophysiological recording methods to evaluate their effects on neuronal function, from single cells through to the network level. Comparison of the toxicity profiles of the same CSF samples, with and without immuno-depletion for tau, has enabled a pioneering demonstration that CSF-tau potently modulates neuronal function. We demonstrate that CSF-tau mediates an increase in neuronal excitability in single cells. We then observed, at the network level, increased input-output responses and enhanced paired-pulse facilitation as well as an increase in long-term potentiation. Finally, we show that CSF-tau modifies the generation and maintenance of hippocampal theta oscillations, which have important roles in learning and memory and are known to be altered in AD patients. Together, we describe a novel method for screening human CSF-tau to understand functional effects on neuron and network activity, which could have far-reaching benefits in understanding tau pathology, thus allowing for the development of better targeted treatments for tauopathies in the future.
Collapse
Affiliation(s)
- Jessica Brown
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Elena Camporesi
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
| | - Maria Olsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180, Mölndal, Sweden
| | - Alice Wang
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Blanca Medem
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin, Madison, WI, 53792, USA
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180, Mölndal, Sweden
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180, Mölndal, Sweden
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Mark Wall
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Emily Hill
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK.
| |
Collapse
|
69
|
Quinn J, Ethier EC, Novielli A, Malone A, Ramirez CE, Salloum L, Trombetta BA, Kivisäkk P, Bremang M, Selzer S, Fournier M, Das S, Xing Y, Arnold SE, Carlyle BC. Cerebrospinal Fluid and Brain Proteoforms of the Granin Neuropeptide Family in Alzheimer's Disease. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:649-667. [PMID: 36912488 PMCID: PMC10080684 DOI: 10.1021/jasms.2c00341] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/11/2023] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
The granin neuropeptide family is composed of acidic secretory signaling molecules that act throughout the nervous system to help modulate synaptic signaling and neural activity. Granin neuropeptides have been shown to be dysregulated in different forms of dementia, including Alzheimer's disease (AD). Recent studies have suggested that the granin neuropeptides and their protease-cleaved bioactive peptides (proteoforms) may act as both powerful drivers of gene expression and as a biomarker of synaptic health in AD. The complexity of granin proteoforms in human cerebrospinal fluid (CSF) and brain tissue has not been directly addressed. We developed a reliable nontryptic mass spectrometry assay to comprehensively map and quantify endogenous neuropeptide proteoforms in the brain and CSF of individuals diagnosed with mild cognitive impairment and dementia due to AD compared to healthy controls, individuals with preserved cognition despite AD pathology ("Resilient"), and those with impaired cognition but no AD or other discernible pathology ("Frail"). We drew associations between neuropeptide proteoforms, cognitive status, and AD pathology values. Decreased levels of VGF proteoforms were observed in CSF and brain tissue from individuals with AD compared to controls, while select proteoforms from chromogranin A showed the opposite effect. To address mechanisms of neuropeptide proteoform regulation, we showed that the proteases Calpain-1 and Cathepsin S can cleave chromogranin A, secretogranin-1, and VGF into proteoforms found in both the brain and CSF. We were unable to demonstrate differences in protease abundance in protein extracts from matched brains, suggesting that regulation may occur at the level of transcription.
Collapse
Affiliation(s)
- James
P. Quinn
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Elizabeth C. Ethier
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Angelo Novielli
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Aygul Malone
- Advanced
Proteomics Facility, Department of Biochemistry, University of Oxford, Oxford, Oxfordshire OX1 3QU, United Kingdom
| | - Christopher E. Ramirez
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Lauren Salloum
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Bianca A. Trombetta
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Pia Kivisäkk
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Michael Bremang
- Proteome
Sciences LLC, Frankfurt am Main, Hessen 60438, Germany
| | - Stefan Selzer
- Proteome
Sciences LLC, Frankfurt am Main, Hessen 60438, Germany
| | - Marjorie Fournier
- Advanced
Proteomics Facility, Department of Biochemistry, University of Oxford, Oxford, Oxfordshire OX1 3QU, United Kingdom
| | - Sudeshna Das
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Yaoyi Xing
- Department
of Physiology, Anatomy & Genetics, University
of Oxford, Oxford, Oxfordshire OX1 3QU, United Kingdom
- Kavli
Institute for Nanoscience Discovery, University
of Oxford, Oxford OX1 3QU, United
Kingdom
| | - Steven E. Arnold
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Becky C. Carlyle
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
- Department
of Physiology, Anatomy & Genetics, University
of Oxford, Oxford, Oxfordshire OX1 3QU, United Kingdom
- Kavli
Institute for Nanoscience Discovery, University
of Oxford, Oxford OX1 3QU, United
Kingdom
| |
Collapse
|
70
|
Yamada K. Multifaceted Roles of Aquaporins in the Pathogenesis of Alzheimer’s Disease. Int J Mol Sci 2023; 24:ijms24076528. [PMID: 37047501 PMCID: PMC10095057 DOI: 10.3390/ijms24076528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/29/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
The central nervous system is highly dependent on water, and disturbances in water homeostasis can have a significant impact on its normal functions. The regulation of water balance is, at least in part, carried out via specialized water channels called aquaporins. In the central nervous system, two major aquaporins (AQPs), AQP1 and AQP4, and their potential involvements have been long implicated in the pathophysiology of many brain disorders such as brain edema and Neuromyelitis optica. In addition to these diseases, there is growing attention to the involvement of AQPs in the removal of waste products in Alzheimer’s disease (AD). This indicates that targeting fluid homeostasis is a novel and attractive approach for AD. This review article aims to summarize recent knowledge on the pathological implications of AQPs in AD, discussing unsolved questions and future prospects.
Collapse
Affiliation(s)
- Kaoru Yamada
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| |
Collapse
|
71
|
Ennis GE, Betthauser TJ, Koscik RL, Chin NA, Christian BT, Asthana S, Johnson SC, Bendlin BB. The relationship of insulin resistance and diabetes to tau PET SUVR in middle-aged to older adults. Alzheimers Res Ther 2023; 15:55. [PMID: 36932429 PMCID: PMC10022314 DOI: 10.1186/s13195-023-01180-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/31/2023] [Indexed: 03/19/2023]
Abstract
BACKGROUND Insulin resistance (IR) and type 2 diabetes have been found to increase the risk for Alzheimer's clinical syndrome in epidemiologic studies but have not been associated with tau tangles in neuropathological research and have been inconsistently associated with cerebrospinal fluid P-tau181. IR and type 2 diabetes are well-recognized vascular risk factors. Some studies suggest that cardiovascular risk may act synergistically with cortical amyloid to increase tau measured using tau PET. Utilizing data from largely nondemented middle-aged and older adult cohorts enriched for AD risk, we investigated the association of IR and diabetes to tau PET and whether amyloid moderated those relationships. METHODS Participants were enrolled in either the Wisconsin Registry for Alzheimer's Prevention (WRAP) or Wisconsin Alzheimer's Disease Research Center (WI-ADRC) Clinical Core. Two partially overlapping samples were studied: a sample characterized using HOMA-IR (n=280 WRAP participants) and a sample characterized on diabetic status (n=285 WRAP and n=109 WI-ADRC). IR was measured using the homeostasis model assessment of insulin resistance (HOMA-IR). Tau PET employing the radioligand 18F-MK-6240 was used to detect AD-specific aggregated tau. Linear regression tested the relationship of IR and diabetic status to tau PET standardized uptake value ratio (SUVR) within the entorhinal cortex and whether relationships were moderated by amyloid assessed by amyloid PET distribution volume ratio (DVR) and amyloid PET positivity status. RESULTS Neither HOMA-IR nor diabetic status was significantly associated with tau PET SUVR. The relationship between IR and tau PET SUVR was not moderated by amyloid PET DVR or positivity status. The association between diabetic status and tau PET SUVR was not significantly moderated by amyloid PET DVR but was significantly moderated by amyloid PET positivity status. Among the amyloid PET-positive participants, the estimated marginal tau PET SUVR mean was higher in the diabetic (n=6) relative to the nondiabetic group (n=88). CONCLUSION Findings indicate that IR may not be related to tau in generally healthy middle-aged and older adults who are in the early stages of the AD clinicopathologic continuum but suggest the need for additional research to investigate whether a synergistic relationship between type 2 diabetes and amyloid is associated with increased tau levels.
Collapse
Affiliation(s)
- Gilda E Ennis
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
| | - Tobey J Betthauser
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Rebecca Langhough Koscik
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Nathaniel A Chin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Bradley T Christian
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Hospital Department of Veterans Affairs, Madison, WI, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Hospital Department of Veterans Affairs, Madison, WI, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Hospital Department of Veterans Affairs, Madison, WI, USA
| |
Collapse
|
72
|
Lou T, Tao B, Chen M. Relationship of Apolipoprotein E with Alzheimer's Disease and Other Neurological Disorders: An Updated Review. Neuroscience 2023; 514:123-140. [PMID: 36736614 DOI: 10.1016/j.neuroscience.2023.01.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023]
Abstract
Alzheimer's disease (AD) and other neurodegenerative diseases, for which there is no effective cure, cause great social burden. Apolipoprotein E (APOE) is an important lipid transporter, which has been shown to have a close relationship with AD and other neurological disorders in an increasing number of studies, suggesting its potential as a therapeutic target. In this review, we summarize the recent advances in clinical and basic research on the role of APOE in the pathogenesis of multiple neurological diseases, with an emphasis on the new associations between APOE and AD, and between APOE and depression. The progress of APOE research in Parkinson's disease (PD) and some other neurological diseases is briefly discussed.
Collapse
Affiliation(s)
- Tianwen Lou
- The First Clinical Medical College, Anhui Medical University, Hefei, China; Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Borui Tao
- The First Clinical Medical College, Anhui Medical University, Hefei, China; Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Ming Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
73
|
Bijttebier S, Rodrigues Martins D, Mertens L, Grauwen K, Bruinzeel W, Willems R, Bartolomé-Nebreda JM, Theunis C, Bretteville A, Ebneth A, Dillen L. IP-LC-MSMS Enables Identification of Three Tau O-GlcNAcylation Sites as O-GlcNAcase Inhibition Pharmacodynamic Readout in Transgenic Mice Overexpressing Human Tau. J Proteome Res 2023; 22:1309-1321. [PMID: 36888912 DOI: 10.1021/acs.jproteome.2c00822] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
O-β-linked N-acetylglucosaminylation (O-GlcNAcylation) modulates tau phosphorylation and aggregation: the pharmacological increase of tau O-GlcNAcylation upon treatment with inhibitors of O-GlcNAc hydrolase (OGA) constitutes a potential strategy to tackle neurodegenerative diseases. Analysis of tau O-GlcNAcylation could potentially be used as a pharmacodynamic biomarker both in preclinical and clinical studies. The goal of the current study was to confirm tau O-GlcNAcylation at S400 as a pharmacodynamic readout of OGA inhibition in P301S transgenic mice overexpressing human tau and treated with the OGA inhibitor Thiamet G and to explore if additional O-GlcNAcylation sites on tau could be identified. As a first step, an immunoprecipitation-liquid chromatography-mass spectrometry (IP-LC-MS) methodology was developed to monitor changes in O-GlcNAcylation around S400 of tau in mouse brain homogenate (BH) extracts. Second, additional O-GlcNAc sites were identified in in-house produced recombinant O-GlcNAcylated human tau at relatively high concentrations, thereby facilitating collection of informative LC-MS data for identification of low-concentration O-GlcNAc-tryptic tau peptides in human transgenic mouse BH extracts. This strategy enabled, for the first time, identification of three low abundant N-terminal and mid-domain O-GlcNAc sites of tau (at S208, S191, and S184 or S185) in human transgenic mouse BH. Data are openly available at data.mendeley.com (doi: 10.17632/jp57yk9469.1; doi: 10.17632/8n5j45dnd8.1; doi: 10.17632/h5vdrx4n3d.1).
Collapse
Affiliation(s)
- Sebastiaan Bijttebier
- Bioanalytical Discovery & Development Sciences, Janssen R&D, Turnhoutseweg 30, 2340 Beerse, Belgium
| | | | - Liesbeth Mertens
- R&D Neurosciences, Janssen R&D, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Karolien Grauwen
- R&D Neurosciences, Janssen R&D, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Wouter Bruinzeel
- R&D Structural & Protein Sciences, Janssen R&D, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Roland Willems
- R&D Neurosciences, Janssen R&D, Turnhoutseweg 30, 2340 Beerse, Belgium
| | | | - Clara Theunis
- R&D Neurosciences, Janssen R&D, Turnhoutseweg 30, 2340 Beerse, Belgium
| | | | - Andreas Ebneth
- R&D Neurosciences, Janssen R&D, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Lieve Dillen
- Bioanalytical Discovery & Development Sciences, Janssen R&D, Turnhoutseweg 30, 2340 Beerse, Belgium
| |
Collapse
|
74
|
Gouveia Roque C, Chung KM, McCurdy EP, Jagannathan R, Randolph LK, Herline-Killian K, Baleriola J, Hengst U. CREB3L2-ATF4 heterodimerization defines a transcriptional hub of Alzheimer's disease gene expression linked to neuropathology. SCIENCE ADVANCES 2023; 9:eadd2671. [PMID: 36867706 PMCID: PMC9984184 DOI: 10.1126/sciadv.add2671] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 01/30/2023] [Indexed: 06/18/2023]
Abstract
Gene expression is changed by disease, but how these molecular responses arise and contribute to pathophysiology remains less understood. We discover that β-amyloid, a trigger of Alzheimer's disease (AD), promotes the formation of pathological CREB3L2-ATF4 transcription factor heterodimers in neurons. Through a multilevel approach based on AD datasets and a novel chemogenetic method that resolves the genomic binding profile of dimeric transcription factors (ChIPmera), we find that CREB3L2-ATF4 activates a transcription network that interacts with roughly half of the genes differentially expressed in AD, including subsets associated with β-amyloid and tau neuropathologies. CREB3L2-ATF4 activation drives tau hyperphosphorylation and secretion in neurons, in addition to misregulating the retromer, an endosomal complex linked to AD pathogenesis. We further provide evidence for increased heterodimer signaling in AD brain and identify dovitinib as a candidate molecule for normalizing β-amyloid-mediated transcriptional responses. The findings overall reveal differential transcription factor dimerization as a mechanism linking disease stimuli to the development of pathogenic cellular states.
Collapse
Affiliation(s)
- Cláudio Gouveia Roque
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Kyung Min Chung
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Ethan P. McCurdy
- Integrated Program in Cellular, Molecular, and Biomedical Studies, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Radhika Jagannathan
- Division of Aging and Dementia, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Lisa K. Randolph
- Doctoral Program in Neurobiology and Behavior, Columbia University, New York, NY, USA
| | - Krystal Herline-Killian
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Jimena Baleriola
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- IKERBASQUE Basque Foundation for Science, Bilbao, Spain
- Department of Cell Biology and Histology, University of the Basque Country, Leioa, Spain
| | - Ulrich Hengst
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| |
Collapse
|
75
|
Madsen LS, Parbo P, Ismail R, Gottrup H, Østergaard L, Brooks DJ, Eskildsen SF. Capillary dysfunction correlates with cortical amyloid load in early Alzheimer's disease. Neurobiol Aging 2023; 123:1-9. [PMID: 36610198 DOI: 10.1016/j.neurobiolaging.2022.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Alterations in cerebral perfusion is increasingly considered to play a crucial role in Alzheimer's disease (AD) and together with accumulated amyloid-β, deficiencies in the brain microvascular circulation may result in local hypoxia. Here, we studied alterations in cerebral circulation and the correlation between amyloid-β load and cerebral perfusion in prodromal AD (pAD). Using dynamic susceptibility contrast MRI and PET, we evaluated cerebral perfusion and amyloid-β levels in 19 individuals with mild cognitive impairment (MCI) and high amyloid-β load (pAD-MCI), 13 MCI individuals without AD pathology and 21 healthy controls. The pAD-MCI group showed significantly lower microvascular blood flow and significantly higher heterogeneity of microvascular blood transit times (p < 0.01) compared with the other 2 groups. Additionally, in the pAD-MCI group raised amyloid-β levels correlated with decreased microvascular blood flow and increased heterogeneity of microvascular blood flow in frontal and temporal areas (p < 0.01). These results indicate a close connection between levels of amyloid-β deposition and brain microvascular perfusion in pAD. A vicious cycle may be established where amyloid-β load and deficiencies in brain perfusion may reinforce each other.
Collapse
Affiliation(s)
- Lasse S Madsen
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark; Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Peter Parbo
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | - Rola Ismail
- Department of Nuclear Medicine, Sygehus Lillebaelt, Kolding, Denmark
| | - Hanne Gottrup
- Dementia Clinic, Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark
| | - David J Brooks
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark; Institute of Translational and Clinical Research, University of Newcastle upon Tyne, Newcastle, UK
| | - Simon F Eskildsen
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
76
|
Sato H, Kasuga K, Isoo N, Hayashi T, Ikeuchi T, Hori Y, Tomita T. Soluble form of the APP fragment, sAPPβ, positively regulates tau secretion. Neurosci Res 2023:S0168-0102(23)00068-8. [PMID: 36967088 DOI: 10.1016/j.neures.2023.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 04/05/2023]
Abstract
Extracellular tau has been highlighted in the pathogenesis of Alzheimer disease (AD), which is the most common neurodegenerative disease. Pathological analyses as well as model animal studies suggest that amyloid-β peptide (Aβ) deposition facilitates the spreading of tau aggregation pathology via extracellular tau. However, the precise mechanism of tau secretion remains unknown. Here, we show that the overexpression of amyloid precursor protein (APP) enhances the secretion of tau phosphorylated at threonine 181 in mouse neuroblastoma Neuro2a cells. Moreover, we found that soluble amyloid precursor protein β (sAPPβ), which is generated by β-site APP cleaving enzyme 1 (BACE1), mediates tau secretion. Our results demonstrate that BACE1-mediated cleavage of APP plays pathological roles in AD pathogenesis by not only Aβ production, but by the spreading of tau aggregation pathology via sAPPβ in AD patients.
Collapse
Affiliation(s)
- Haruaki Sato
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Noriko Isoo
- Department of Physiology, School of Medicine, Teikyo University, Tokyo, Japan
| | - Toshihiro Hayashi
- Department of Physiology, School of Medicine, Teikyo University, Tokyo, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yukiko Hori
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
77
|
Rahimi B, Behroozi Z, Motamednezhad A, Jafarpour M, Hamblin MR, Moshiri A, Janzadeh A, Ramezani F. Study of nerve cell regeneration on nanofibers containing cerium oxide nanoparticles in a spinal cord injury model in rats. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2023; 34:9. [PMID: 36809518 PMCID: PMC9944598 DOI: 10.1007/s10856-023-06711-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 01/15/2023] [Indexed: 05/17/2023]
Abstract
Since the CNS is unable to repair itself via neuronal regeneration in adult mammals, alternative therapies need to be found. The use of cerium oxide nanoparticles to repair nerve damage could be a promising approach for spinal cord reconstruction. In this study, we constructed a scaffold containing cerium oxide nanoparticles (Scaffold-CeO2) and investigated the rate of nerve cell regeneration in a rat model of spinal cord injury. The scaffold of gelatin and polycaprolactone was synthesized, and a gelatin solution containing cerium oxide nanoparticles was attached to the scaffold. For the animal study, 40 male Wistar rats were randomly divided into 4 groups (n = 10): (a) Control; (b) Spinal cord injury (SCI); (c) Scaffold (SCI + scaffold without CeO2 nanoparticles); (d) Scaffold-CeO2 (SCI + scaffold containing CeO2 nanoparticles). After creation of a hemisection SCI, scaffolds were placed at the site of injury in groups c and d, and after 7 weeks the rats were subjected to behavioral tests and then sacrificed for preparation of the spinal cord tissue to measure the expression of G-CSF, Tau and Mag proteins by Western blotting and Iba-1 protein by immunohistochemistry. The result of behavioral tests confirmed motor improvement and pain reduction in the Scaffold-CeO2 group compared to the SCI group. Decreased expression of Iba-1 and higher expression of Tau and Mag in the Scaffold-CeO2 group compared to the SCI group could be the result of nerve regeneration caused by the scaffold containing CeONPs as well as relief of pain symptoms.
Collapse
Affiliation(s)
- Behnaz Rahimi
- Department of basic sciences, Saveh University of Medical Sciences, Saveh, Iran
| | - Zahra Behroozi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran
| | - Ali Motamednezhad
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maral Jafarpour
- International Campus, Iran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | | | - Atousa Janzadeh
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Fatemeh Ramezani
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
78
|
Fornasiero EF, Savas JN. Determining and interpreting protein lifetimes in mammalian tissues. Trends Biochem Sci 2023; 48:106-118. [PMID: 36163144 PMCID: PMC9868050 DOI: 10.1016/j.tibs.2022.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 01/26/2023]
Abstract
The orchestration of protein production and degradation, and the regulation of protein lifetimes, play a central role in the majority of biological processes. Recent advances in proteomics have enabled the estimation of protein half-lives for thousands of proteins in vivo. What is the utility of these measurements, and how can they be leveraged to interpret the proteome changes occurring during development, aging, and disease? This opinion article summarizes leading technical approaches and highlights their strengths and weaknesses. We also disambiguate frequently used terminology, illustrate recent mechanistic insights, and provide guidance for interpreting and validating protein turnover measurements. Overall, protein lifetimes, coupled to estimates of protein levels, are essential for obtaining a deep understanding of mammalian biology and the basic processes defining life itself.
Collapse
Affiliation(s)
- Eugenio F Fornasiero
- Department of Neuro-Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany.
| | - Jeffrey N Savas
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
79
|
Therriault J, Vermeiren M, Servaes S, Tissot C, Ashton NJ, Benedet AL, Karikari TK, Lantero-Rodriguez J, Brum WS, Lussier FZ, Bezgin G, Stevenson J, Rahmouni N, Kunach P, Wang YT, Fernandez-Arias J, Socualaya KQ, Macedo AC, Ferrari-Souza JP, Ferreira PCL, Bellaver B, Leffa DT, Zimmer ER, Vitali P, Soucy JP, Triana-Baltzer G, Kolb HC, Pascoal TA, Saha-Chaudhuri P, Gauthier S, Zetterberg H, Blennow K, Rosa-Neto P. Association of Phosphorylated Tau Biomarkers With Amyloid Positron Emission Tomography vs Tau Positron Emission Tomography. JAMA Neurol 2023; 80:188-199. [PMID: 36508198 PMCID: PMC9856704 DOI: 10.1001/jamaneurol.2022.4485] [Citation(s) in RCA: 81] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/13/2022] [Indexed: 12/14/2022]
Abstract
Importance The recent proliferation of phosphorylated tau (p-tau) biomarkers has raised questions about their preferential association with the hallmark pathologies of Alzheimer disease (AD): amyloid-β plaques and tau neurofibrillary tangles. Objective To determine whether cerebrospinal fluid (CSF) and plasma p-tau biomarkers preferentially reflect cerebral β-amyloidosis or neurofibrillary tangle aggregation measured with positron emission tomography (PET). Design, Setting, and Participants This was a cross-sectional study of 2 observational cohorts: the Translational Biomarkers in Aging and Dementia (TRIAD) study, with data collected between October 2017 and August 2021, and the Alzheimer's Disease Neuroimaging Initiative (ADNI), with data collected between September 2015 and November 2019. TRIAD was a single-center study, and ADNI was a multicenter study. Two independent subsamples were derived from TRIAD. The first TRIAD subsample comprised individuals assessed with CSF p-tau (p-tau181, p-tau217, p-tau231, p-tau235), [18F]AZD4694 amyloid PET, and [18F]MK6240 tau PET. The second TRIAD subsample included individuals assessed with plasma p-tau (p-tau181, p-tau217, p-tau231), [18F]AZD4694 amyloid PET, and [18F]MK6240 tau PET. An independent cohort from ADNI comprised individuals assessed with CSF p-tau181, [18F]florbetapir PET, and [18F]flortaucipir PET. Participants were included based on the availability of p-tau and PET biomarker assessments collected within 9 months of each other. Exclusion criteria were a history of head trauma or magnetic resonance imaging/PET safety contraindications. No participants who met eligibility criteria were excluded. Exposures Amyloid PET, tau PET, and CSF and plasma assessments of p-tau measured with single molecule array (Simoa) assay or enzyme-linked immunosorbent assay. Main Outcomes and Measures Associations between p-tau biomarkers with amyloid PET and tau PET. Results A total of 609 participants (mean [SD] age, 66.9 [13.6] years; 347 female [57%]; 262 male [43%]) were included in the study. For all 4 phosphorylation sites assessed in CSF, p-tau was significantly more closely associated with amyloid-PET values than tau-PET values (p-tau181 difference, 13%; 95% CI, 3%-22%; P = .006; p-tau217 difference, 11%; 95% CI, 3%-20%; P = .003; p-tau231 difference, 15%; 95% CI, 5%-22%; P < .001; p-tau235 difference, 9%; 95% CI, 1%-19%; P = .02) . These results were replicated with plasma p-tau181 (difference, 11%; 95% CI, 1%-22%; P = .02), p-tau217 (difference, 9%; 95% CI, 1%-19%; P = .02), p-tau231 (difference, 13%; 95% CI, 3%-24%; P = .009), and CSF p-tau181 (difference, 9%; 95% CI, 1%-21%; P = .02) in independent cohorts. Conclusions and Relevance Results of this cross-sectional study of 2 observational cohorts suggest that the p-tau abnormality as an early event in AD pathogenesis was associated with amyloid-β accumulation and highlights the need for careful interpretation of p-tau biomarkers in the context of the amyloid/tau/neurodegeneration, or A/T/(N), framework.
Collapse
Affiliation(s)
- Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Marie Vermeiren
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Erasmus Medical Center, Erasmus University Rotterdam, Rotterdam, the Netherlands
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular Medicine, University of Gothenburg, Gothenburg, Sweden
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, United Kingdom
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, United Kingdom
| | - Andréa Lessa Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Thomas K. Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Wagner S. Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Department of Pharmacology, Graduate Program in Biological Sciences: Biochemistry and Pharmacology and Therapeutics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Firoza Z. Lussier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
| | - Peter Kunach
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Jaime Fernandez-Arias
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Kely Quispialaya Socualaya
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Arthur C. Macedo
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - João Pedro Ferrari-Souza
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Pâmela C. L. Ferreira
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Bruna Bellaver
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Douglas T. Leffa
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Eduardo R. Zimmer
- Department of Pharmacology, Graduate Program in Biological Sciences: Biochemistry and Pharmacology and Therapeutics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Paolo Vitali
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Jean-Paul Soucy
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | | | - Hartmuth C. Kolb
- Neuroscience Biomarkers, Janssen Research & Development, La Jolla, California
| | - Tharick A. Pascoal
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
80
|
Sahlgren Bendtsen KM, Hall VJ. The Breakthroughs and Caveats of Using Human Pluripotent Stem Cells in Modeling Alzheimer's Disease. Cells 2023; 12:cells12030420. [PMID: 36766763 PMCID: PMC9913971 DOI: 10.3390/cells12030420] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Modeling Alzheimer's disease (AD) using human-induced pluripotent stem cells (iPSCs) is a field now spanning 15 years. Developments in the field have shown a shift in using simple 2D cortical neuron models to more advanced tri-cultures and 3D cerebral organoids that recapitulate more features of the disease. This is largely due to development and optimization of new cell protocols. In this review, we highlight recent major breakthroughs in the AD field and the implications this has in modeling AD using iPSCs (AD-iPSCs). To date, AD-iPSCs have been largely used to recapitulate and study impaired amyloid precursor protein (APP) processing and tau phosphorylation in both familial and sporadic AD. AD-iPSCs have also been studied for varying neuronal and glial dysfunctions. Moreover, they have been useful for discovering new molecular mechanisms, such as identifying proteins that bridge APP processing with tau phosphorylation and for identifying molecular pathways that bridge APP processing dysfunction with impaired cholesterol biosynthesis. Perhaps the greatest use of AD-iPSCs has been in discovering compounds via drug screening, that reduce amyloid beta (Aβ) in neurons, such as the anti-inflammatory compound, cromolyn, and antiparasitic drugs, avermectins. In addition, high content screening using AD-iPSCs has led to the identification of statins that can reduce levels of phosphorylated tau (p-Tau) in neurons. Some of these compounds have made it through to testing in human clinical trials. Improvements in omic technologies including single cell RNA sequencing and proteomics as well as advances in production of iPSC-cerebral organoids and tri-cultures is likely to result in the further discovery of new drugs and treatments for AD. Some caveats remain in the field, including, long experimental conditions to create mature neurons, high costs of media that limit research capabilities, and a lack of reproducibility using current iPSC-cerebral organoid protocols. Despite these current limitations, AD-iPSCs remain an excellent cellular model for studying AD mechanisms and for drug discovery.
Collapse
|
81
|
Kawarabayashi T, Nakamura T, Miyashita K, Segawa T, Fukamachi I, Sugawara T, Oka H, Ishizawa K, Amari M, Kasahara H, Makioka K, Ikeda Y, Takatama M, Shoji M. Clinical Evaluation of Cerebrospinal Fluid p217tau and Neurofilament Light Chain Levels in Patients with Alzheimer's Disease or Other Neurological Diseases. J Alzheimers Dis 2023; 96:1623-1638. [PMID: 38007650 PMCID: PMC10741340 DOI: 10.3233/jad-230419] [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] [Accepted: 09/29/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUND The cerebrospinal fluid (CSF) levels of tau phosphorylated at threonine 217 (p217tau) or 181 (p181tau), and neurofilament light chain (NfL) are definite biomarkers of tauopathy and neurodegeneration in Alzheimer's disease (AD). OBJECTIVE To validate their utility in excluding other neurological diseases and age-related changes in clinical settings. METHODS We developed monoclonal antibodies against p217tau and NfL, established novel ELISAs, and analyzed 170 CSF samples from patients with AD or other neurological diseases. RESULTS In AD, p217tau is a more specific and abundant CSF component than p181tau. However, CSF NfL levels increase age-dependently and to a greater extent in central and peripheral nervous diseases than in AD. CONCLUSIONS CSF p217tau correlates better with AD neurodegeneration than other tau-related biomarkers and the major phosphorylated tau species. The clinical usage of NfL as a neurodegeneration biomarker in AD requires exclusion of various central and peripheral neurological diseases.
Collapse
Affiliation(s)
- Takeshi Kawarabayashi
- Department of Neurology, Dementia Research Center, Geriatrics Research Institute and Hospital, Maebashi, Japan
- Department of Neurology, Gunma University Hospital, Maebashi, Japan
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Takumi Nakamura
- Department of Neurology, Gunma University Hospital, Maebashi, Japan
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | | | | | | | - Takashi Sugawara
- Department of Neurology, Dementia Research Center, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Hironori Oka
- Department of Neurology, Dementia Research Center, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Kunihiko Ishizawa
- Department of Neurology, Dementia Research Center, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Masakuni Amari
- Department of Neurology, Dementia Research Center, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Hiroo Kasahara
- Department of Neurology, Gunma University Hospital, Maebashi, Japan
| | - Kouki Makioka
- Department of Neurology, Gunma University Hospital, Maebashi, Japan
| | - Yoshio Ikeda
- Department of Neurology, Gunma University Hospital, Maebashi, Japan
| | - Masamitsu Takatama
- Department of Neurology, Dementia Research Center, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Mikio Shoji
- Department of Neurology, Dementia Research Center, Geriatrics Research Institute and Hospital, Maebashi, Japan
- Department of Neurology, Gunma University Hospital, Maebashi, Japan
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|
82
|
Dong H, Guo L, Yang H, Zhu W, Liu F, Xie Y, Zhang Y, Xue K, Li Q, Liang M, Zhang N, Qin W. Association between gray matter atrophy, cerebral hypoperfusion, and cognitive impairment in Alzheimer's disease. Front Aging Neurosci 2023; 15:1129051. [PMID: 37091519 PMCID: PMC10117777 DOI: 10.3389/fnagi.2023.1129051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/15/2023] [Indexed: 04/25/2023] Open
Abstract
Background Alzheimer's disease (AD) is one of the most severe neurodegenerative diseases leading to dementia in the elderly. Cerebral atrophy and hypoperfusion are two important pathophysiological characteristics. However, it is still unknown about the area-specific causal pathways between regional gray matter atrophy, cerebral hypoperfusion, and cognitive impairment in AD patients. Method Forty-two qualified AD patients and 49 healthy controls (HC) were recruited in this study. First, we explored voxel-wise inter-group differences in gray matter volume (GMV) and arterial spin labeling (ASL) -derived cerebral blood flow (CBF). Then we explored the voxel-wise associations between GMV and Mini-Mental State Examination (MMSE) score, GMV and CBF, and CBF and MMSE to identify brain targets contributing to cognitive impairment in AD patients. Finally, a mediation analysis was applied to test the causal pathways among atrophied GMV, hypoperfusion, and cognitive impairment in AD. Results Voxel-wise permutation test identified that the left middle temporal gyrus (MTG) had both decreased GMV and CBF in the AD. Moreover, the GMV of this region was positively correlated with MMSE and its CBF, and CBF of this region was also positively correlated with MMSE in AD (p < 0.05, corrected). Finally, mediation analysis revealed that gray matter atrophy of left MTG drives cognitive impairment of AD via the mediation of CBF (proportion of mediation = 55.82%, β = 0.242, 95% confidence interval by bias-corrected and accelerated bootstrap: 0.082 to 0.530). Conclusion Our findings indicated suggested that left MTG is an important hub linking gray matter atrophy, hypoperfusion, and cognitive impairment for AD, and might be a potential treatment target for AD.
Collapse
Affiliation(s)
- Haoyang Dong
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Lining Guo
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Hailei Yang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenshuang Zhu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Fang Liu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yingying Xie
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Yu Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Kaizhong Xue
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiang Li
- Technical College for the Deaf, Tianjin University of Technology, Tianjin, China
| | - Meng Liang
- School of Medical Imaging, Tianjin Medical University, Tianjin, China
| | - Nan Zhang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Nan Zhang,
| | - Wen Qin
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
- Wen Qin,
| |
Collapse
|
83
|
Murray ME, Moloney CM, Kouri N, Syrjanen JA, Matchett BJ, Rothberg DM, Tranovich JF, Sirmans TNH, Wiste HJ, Boon BDC, Nguyen AT, Reichard RR, Dickson DW, Lowe VJ, Dage JL, Petersen RC, Jack CR, Knopman DS, Vemuri P, Graff-Radford J, Mielke MM. Global neuropathologic severity of Alzheimer's disease and locus coeruleus vulnerability influences plasma phosphorylated tau levels. Mol Neurodegener 2022; 17:85. [PMID: 36575455 PMCID: PMC9795667 DOI: 10.1186/s13024-022-00578-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 10/26/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Advances in ultrasensitive detection of phosphorylated tau (p-tau) in plasma has enabled the use of blood tests to measure Alzheimer's disease (AD) biomarker changes. Examination of postmortem brains of participants with antemortem plasma p-tau levels remains critical to understanding comorbid and AD-specific contribution to these biomarker changes. METHODS We analyzed 35 population-based Mayo Clinic Study of Aging participants with plasma p-tau at threonine 181 and threonine 217 (p-tau181, p-tau217) available within 3 years of death. Autopsied participants included cognitively unimpaired, mild cognitive impairment, AD dementia, and non-AD neurodegenerative disorders. Global neuropathologic scales of tau, amyloid-β, TDP-43, and cerebrovascular disease were examined. Regional digital pathology measures of tau (phosphorylated threonine 181 and 217 [pT181, pT217]) and amyloid-β (6F/3D) were quantified in hippocampus and parietal cortex. Neurotransmitter hubs reported to influence development of tangles (nucleus basalis of Meynert) and amyloid-β plaques (locus coeruleus) were evaluated. RESULTS The strongest regional associations were with parietal cortex for tau burden (p-tau181 R = 0.55, p = 0.003; p-tau217 R = 0.66, p < 0.001) and amyloid-β burden (p-tau181 R = 0.59, p < 0.001; p-tau217 R = 0.71, p < 0.001). Linear regression analysis of global neuropathologic scales explained 31% of variability in plasma p-tau181 (Adj. R2 = 0.31) and 59% in plasma p-tau217 (Adj. R2 = 0.59). Neither TDP-43 nor cerebrovascular disease global scales independently contributed to variability. Global scales of tau pathology (β-coefficient = 0.060, p = 0.016) and amyloid-β pathology (β-coefficient = 0.080, p < 0.001) independently predicted plasma p-tau217 when modeled together with co-pathologies, but only amyloid-β (β-coefficient = 0.33, p = 0.021) significantly predicted plasma p-tau181. While nucleus basalis of Meynert neuron count/mm2 was not associated with plasma p-tau levels, a lower locus coeruleus neuron count/mm2 was associated with higher plasma p-tau181 (R = -0.50, p = 0.007) and higher plasma p-tau217 (R = -0.55, p = 0.002). Cognitive scores (Adj. R2 = 0.25-0.32) were predicted by the global tau scale, but not by the global amyloid-β scale or plasma p-tau when modeled simultaneously. CONCLUSIONS Higher soluble plasma p-tau levels may be the result of an intersection between insoluble deposits of amyloid-β and tau accumulation in brain, and may be associated with locus coeruleus degeneration.
Collapse
Affiliation(s)
- Melissa E. Murray
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Christina M. Moloney
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Naomi Kouri
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Jeremy A. Syrjanen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN USA
| | - Billie J. Matchett
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Darren M. Rothberg
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Jessica F. Tranovich
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Tiffany N. Hicks Sirmans
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Heather J. Wiste
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN USA
| | - Baayla D. C. Boon
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Aivi T. Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
| | - R. Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
| | - Dennis W. Dickson
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Val J. Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN USA
| | - Jeffrey L. Dage
- Department of Neurology, Indiana University, Indianapolis, IN USA
| | | | | | | | | | | | - Michelle M. Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN USA
- Wake Forest University School of Medicine, Winston-Salem, NC USA
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest University School of Medicine, 525 Vine, 5th floor, Winston-Salem, NC 27157 USA
| |
Collapse
|
84
|
Simrén J, Brum WS, Ashton NJ, Benedet AL, Karikari TK, Kvartsberg H, Sjons E, Lussier FZ, Chamoun M, Stevenson J, Hopewell R, Pallen V, Ye K, Pascoal TA, Zetterberg H, Rosa-Neto P, Blennow K. CSF tau368/total-tau ratio reflects cognitive performance and neocortical tau better compared to p-tau181 and p-tau217 in cognitively impaired individuals. Alzheimers Res Ther 2022; 14:192. [PMID: 36544221 PMCID: PMC9773470 DOI: 10.1186/s13195-022-01142-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Cerebrospinal fluid (CSF) tau biomarkers are reliable diagnostic markers for Alzheimer's disease (AD). However, their strong association with amyloid pathology may limit their reliability as specific markers of tau neurofibrillary tangles. A recent study showed evidence that a ratio of CSF C-terminally truncated tau (tau368, a tangle-enriched tau species), especially in ratio with total tau (t-tau), correlates strongly with tau PET tracer uptake. In this study, we set to evaluate the performance of the tau368/t-tau ratio in capturing tangle pathology, as indexed by a high-affinity tau PET tracer, as well as its association with severity of clinical symptoms. METHODS In total, 125 participants were evaluated cross-sectionally from the Translational Biomarkers of Aging and Dementia (TRIAD) cohort (21 young, 60 cognitively unimpaired [CU] elderly [15 Aβ+], 10 Aβ+ with mild cognitive impairment [MCI], 14 AD dementia patients, and 20 Aβ- individuals with non-AD cognitive disorders). All participants underwent amyloid and tau PET scanning, with [18F]-AZD4694 and [18F]-MK6240, respectively, and had CSF measurements of p-tau181, p-tau217, and t-tau. CSF concentrations of tau368 were quantified in all individuals with an in-house single molecule array assay. RESULTS CSF tau368 concentration was not significantly different across the diagnostic groups, although a modest increase was observed in all groups as compared with healthy young individuals (all P < 0.01). In contrast, the CSF tau368/t-tau ratio was the lowest in AD dementia, being significantly lower than in CU individuals (Aβ-, P < 0.001; Aβ+, P < 0.01), as well as compared to those with non-AD cognitive disorders (P < 0.001). Notably, in individuals with symptomatic AD, tau368/t-tau correlated more strongly with [18F]-MK6240 PET SUVR as compared to the other CSF tau biomarkers, with increasing associations being seen in brain regions associated with more advanced disease (isocortical regions > limbic regions > transentorhinal regions). Importantly, linear regression models indicated that these associations were not confounded by Aβ PET SUVr. CSF tau368/t-tau also tended to continue to become more abnormal with higher tau burden, whereas the other biomarkers plateaued after the limbic stage. Finally, the tau368/t-tau ratio correlated more strongly with cognitive performance in individuals with symptomatic AD as compared to t-tau, p-tau217 and p-tau181. CONCLUSION The tau368/t-tau ratio captures novel aspects of AD pathophysiology and disease severity in comparison to established CSF tau biomarkers, as it is more closely related to tau PET SUVR and cognitive performance in the symptomatic phase of the disease.
Collapse
Affiliation(s)
- Joel Simrén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Wagner S Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, London, UK
| | - Andrea L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hlin Kvartsberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Emma Sjons
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Firoza Z Lussier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- Montreal Neurological Institute, Montreal, QC, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- Montreal Neurological Institute, Montreal, QC, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- Montreal Neurological Institute, Montreal, QC, Canada
| | - Robert Hopewell
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- Montreal Neurological Institute, Montreal, QC, Canada
| | - Vanessa Pallen
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- Montreal Neurological Institute, Montreal, QC, Canada
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Tharick A Pascoal
- Department of Neurology and Psychiatry, University of Pittsburgh, Pittsburgh, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- Montreal Neurological Institute, Montreal, QC, Canada
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
85
|
Mahali S, Martinez R, King M, Verbeck A, Harari O, Benitez BA, Horie K, Sato C, Temple S, Karch CM. Defective proteostasis in induced pluripotent stem cell models of frontotemporal lobar degeneration. Transl Psychiatry 2022; 12:508. [PMID: 36494352 PMCID: PMC9734180 DOI: 10.1038/s41398-022-02274-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
Impaired proteostasis is associated with normal aging and is accelerated in neurodegeneration. This impairment may lead to the accumulation of protein, which can be toxic to cells and tissue. In a subset of frontotemporal lobar degeneration with tau pathology (FTLD-tau) cases, pathogenic mutations in the microtubule-associated protein tau (MAPT) gene are sufficient to cause tau accumulation and neurodegeneration. However, the pathogenic events triggered by the expression of the mutant tau protein remain poorly understood. Here, we show that molecular networks associated with lysosomal biogenesis and autophagic function are disrupted in brains from FTLD-tau patients carrying a MAPT p.R406W mutation. We then used human induced pluripotent stem cell (iPSC)-derived neurons and 3D cerebral organoids from patients carrying the MAPT p.R406W mutation and CRISPR/Cas9, corrected controls to evaluate proteostasis. MAPT p.R406W was sufficient to induce morphological and functional deficits in the lysosomal pathway in iPSC-neurons. These phenotypes were reversed upon correction of the mutant allele with CRISPR/Cas9. Treatment with mTOR inhibitors led to tau degradation specifically in MAPT p.R406W neurons. Together, our findings suggest that MAPT p.R406W is sufficient to cause impaired lysosomal function, which may contribute to disease pathogenesis and serve as a cellular phenotype for drug screening.
Collapse
Affiliation(s)
- Sidhartha Mahali
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Rita Martinez
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Melvin King
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Anthony Verbeck
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Oscar Harari
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University in St Louis, St Louis, MO, USA
| | - Bruno A Benitez
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University in St Louis, St Louis, MO, USA
| | - Kanta Horie
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Chihiro Sato
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | | | - Celeste M Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA.
- Hope Center for Neurological Disorders, Washington University in St Louis, St Louis, MO, USA.
| |
Collapse
|
86
|
Dai CL, Liu F, Iqbal K, Gong CX. Gut Microbiota and Immunotherapy for Alzheimer's Disease. Int J Mol Sci 2022; 23:15230. [PMID: 36499564 PMCID: PMC9741026 DOI: 10.3390/ijms232315230] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/08/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that eventually leads to dementia and death of the patient. Currently, no effective treatment is available that can slow or halt the progression of the disease. The gut microbiota can modulate the host immune system in the peripheral and central nervous system through the microbiota-gut-brain axis. Growing evidence indicates that gut microbiota dysbiosis plays an important role in the pathogenesis of AD, and modulation of the gut microbiota may represent a new avenue for treating AD. Immunotherapy targeting Aβ and tau has emerged as the most promising disease-modifying therapy for the treatment of AD. However, the underlying mechanism of AD immunotherapy is not known. Importantly, preclinical and clinical studies have highlighted that the gut microbiota exerts a major influence on the efficacy of cancer immunotherapy. However, the role of the gut microbiota in AD immunotherapy has not been explored. We found that immunotherapy targeting tau can modulate the gut microbiota in an AD mouse model. In this article, we focused on the crosstalk between the gut microbiota, immunity, and AD immunotherapy. We speculate that modulation of the gut microbiota induced by AD immunotherapy may partially underlie the efficacy of the treatment.
Collapse
Affiliation(s)
| | | | | | - Cheng-Xin Gong
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, NY 10314, USA
| |
Collapse
|
87
|
Horie K, Barthélemy NR, Spina S, VandeVrede L, He Y, Paterson RW, Wright BA, Day GS, Davis AA, Karch CM, Seeley WW, Perrin RJ, Koppisetti RK, Shaikh F, Lago AL, Heuer HW, Ghoshal N, Gabelle A, Miller BL, Boxer AL, Bateman RJ, Sato C. CSF tau microtubule-binding region identifies pathological changes in primary tauopathies. Nat Med 2022; 28:2547-2554. [PMID: 36424467 PMCID: PMC9800273 DOI: 10.1038/s41591-022-02075-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 10/05/2022] [Indexed: 11/27/2022]
Abstract
Despite recent advances in fluid biomarker research in Alzheimer's disease (AD), there are no fluid biomarkers or imaging tracers with utility for diagnosis and/or theragnosis available for other tauopathies. Using immunoprecipitation and mass spectrometry, we show that 4 repeat (4R) isoform-specific tau species from microtubule-binding region (MTBR-tau275 and MTBR-tau282) increase in the brains of corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), frontotemporal lobar degeneration (FTLD)-MAPT and AD but decrease inversely in the cerebrospinal fluid (CSF) of CBD, FTLD-MAPT and AD compared to control and other FTLD-tau (for example, Pick's disease). CSF MTBR-tau measures are reproducible in repeated lumbar punctures and can be used to distinguish CBD from control (receiver operating characteristic area under the curve (AUC) = 0.889) and other FTLD-tau, such as PSP (AUC = 0.886). CSF MTBR-tau275 and MTBR-tau282 may represent the first affirmative biomarkers to aid in the diagnosis of primary tauopathies and facilitate clinical trial designs.
Collapse
Affiliation(s)
- Kanta Horie
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- The Tracy Family Stable Isotope Labeling Quantitation Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicolas R Barthélemy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- The Tracy Family Stable Isotope Labeling Quantitation Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Salvatore Spina
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Lawren VandeVrede
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Yingxin He
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- The Tracy Family Stable Isotope Labeling Quantitation Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Ross W Paterson
- Department of Neurology, University College London Queen Square Institute of Neurology, University College London, London, UK
| | - Brenton A Wright
- Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Gregory S Day
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Albert A Davis
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, St. Louis, MO, USA
| | - Celeste M Karch
- Hope Center for Neurological Disorders, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - William W Seeley
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Richard J Perrin
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rama K Koppisetti
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Faris Shaikh
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Argentina Lario Lago
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Hilary W Heuer
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Nupur Ghoshal
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Audrey Gabelle
- Memory Research and Resources Center, Department of Neurology, University Hospital of Montpellier, Neurosciences Institute of Montpellier, University of Montpellier, Montpellier, France
| | - Bruce L Miller
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Adam L Boxer
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- The Tracy Family Stable Isotope Labeling Quantitation Center, Washington University School of Medicine, St. Louis, MO, USA.
- Hope Center for Neurological Disorders, St. Louis, MO, USA.
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
| | - Chihiro Sato
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- The Tracy Family Stable Isotope Labeling Quantitation Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
88
|
Mori H, Yoshino Y, Ueno M, Funahashi Y, Kumon H, Ozaki Y, Yamazaki K, Ochi S, Iga J, Ueno S. Blood MAPT expression and methylation status in Alzheimer's disease. PCN REPORTS : PSYCHIATRY AND CLINICAL NEUROSCIENCES 2022; 1:e65. [PMID: 38868661 PMCID: PMC11114303 DOI: 10.1002/pcn5.65] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/20/2022] [Accepted: 11/25/2022] [Indexed: 06/14/2024]
Abstract
Aim This study aimed to investigate the expression levels and methylation status of microtubule-associated protein tau (MAPT) in the blood of Alzheimer's disease (AD) patients and age- and sex-matched healthy controls. Methods Fifty AD outpatients and 50 healthy contorls were enrolled. Blood samples were collected for processing of complementary DNA and genomic DNA. MAPT messenger ribonucleic acid (mRNA) expression was analyzed by real-time quantitative polymerase chain reaction. The methylation rates of four cytosine-phosphate-guanine (CpG) sites in the upstream region of MAPT exon1 were evaluated by the pyrosequencing method. Results No significant differences in MAPT mRNA expression levels were found between AD and control subjects (AD 0.97 ± 0.49 vs. control 1.0 ± 0.64, p = 0.62). MAPT mRNA expression levels were not correlated with any other clinical characteristics or results of psychological tests. MAPT mRNA expression levels were significantly higher in AD subjects treated with acetylcholinesterase inhibitors (AchEIs) (n = 25) than in subjects not treated with AChEIs (n = 25) (unmedicated 0.83 ± 0.33 vs. medicated 1.12 ± 0.59, p = 0.049). The AD subjects did not differ from the control subjects in methylation rates at selected CpG sites. MAPT methylation status were not correlated with clinical characteristics, the results of psychological tests, or MAPT mRNA expression. Conclusion MAPT mRNA expression levels and methylation status in blood do not appear useful as biomarkers for AD or the examined CpG sites were not genetically significant for MAPT gene expression or AD pathology. However, AChEIs may alter MAPT mRNA expression. Further studies are needed to explore blood biomarkers that can discriminate AD patients from controls.
Collapse
Affiliation(s)
- Hiroaki Mori
- Department of Neuropsychiatry, Molecules and FunctionEhime University Graduate School of Medicine, ShitsukawaToonEhimeJapan
| | - Yuta Yoshino
- Department of Neuropsychiatry, Molecules and FunctionEhime University Graduate School of Medicine, ShitsukawaToonEhimeJapan
| | - Mariko Ueno
- Department of Neuropsychiatry, Molecules and FunctionEhime University Graduate School of Medicine, ShitsukawaToonEhimeJapan
| | - Yu Funahashi
- Department of Neuropsychiatry, Molecules and FunctionEhime University Graduate School of Medicine, ShitsukawaToonEhimeJapan
| | - Hiroshi Kumon
- Department of Neuropsychiatry, Molecules and FunctionEhime University Graduate School of Medicine, ShitsukawaToonEhimeJapan
| | - Yuki Ozaki
- Department of Neuropsychiatry, Molecules and FunctionEhime University Graduate School of Medicine, ShitsukawaToonEhimeJapan
| | - Kiyohiro Yamazaki
- Department of Neuropsychiatry, Molecules and FunctionEhime University Graduate School of Medicine, ShitsukawaToonEhimeJapan
| | - Shinichiro Ochi
- Department of Neuropsychiatry, Molecules and FunctionEhime University Graduate School of Medicine, ShitsukawaToonEhimeJapan
| | - Jun‐ichi Iga
- Department of Neuropsychiatry, Molecules and FunctionEhime University Graduate School of Medicine, ShitsukawaToonEhimeJapan
| | - Shu‐ichi Ueno
- Department of Neuropsychiatry, Molecules and FunctionEhime University Graduate School of Medicine, ShitsukawaToonEhimeJapan
| |
Collapse
|
89
|
Gao L, Shuai Y, Wen L, Zhang H, Zhang Y, Zhang X. Benefit and safety of antibiotics for Alzheimer's disease: Protocol for a systematic review and meta-analysis. Medicine (Baltimore) 2022; 101:e31637. [PMID: 36451430 PMCID: PMC9704868 DOI: 10.1097/md.0000000000031637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is an age-related degenerative change of the central nervous system, the cause of which remains unclear. Recent studies have found that brain inflammation caused by microbial infections may be one of the etiologies of AD, and antibiotics as novel treatments may be beneficial for delaying the development of AD. Several prospective studies have investigated the effects of different antibiotics on Alzheimer's disease. However, no systematic review or meta-analysis has evaluated the benefits and safety of antibiotics in AD patients. METHODS This study will analyze randomized controlled trials and observational studies published from database inception to December 31, 2022, and included direct or indirect evidence. Studies will be retrieved by searching PubMed, Scopus, Web of Science, Cochrane Central Register of Controlled Clinical Trials, CNKI, and Wan Fang databases. The outcomes of this study included the Alzheimer's Disease Assessment Scale cognitive subscale (ADAS-cog), Montreal Cognitive Assessment (MoCA), Standardized Mini-Mental State Examination (SMMSE), Clinical Dementia Rating (CDR), Frontal Functioning Scale (FAB), Dysfunctional Behavior Rating Instrument (DBRI), Activities of Daily Living (ADLs) Index, and Geriatric Depression Scale (GDS). The risk of bias will be assessed using the Cochrane risk-of-bias assessment instrument for randomized controlled trials. A random-effect/fixed-effects model will be used to summarize the estimates of the mean difference/risk ratio using a 95% confidence interval. RESULTS This study will analyze the benefits and safety of antibiotics in patients with AD. CONCLUSION The results of this analysis will provide evidence to evaluate the benefits and safety of antibiotics in the treatment of AD.
Collapse
Affiliation(s)
- Lin Gao
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Yinqi Shuai
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Lihong Wen
- The Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan Province, China
| | - Hong Zhang
- Chengdu University of Traditional Chinese Medicine Affiliated Hospital, Chengdu, Sichuan Province, China
| | - Yi Zhang
- Chengdu University of Traditional Chinese Medicine Affiliated Hospital, Chengdu, Sichuan Province, China
| | - Xiaoyun Zhang
- Chengdu University of Traditional Chinese Medicine Affiliated Hospital, Chengdu, Sichuan Province, China
- * Correspondence: Xiaoyun Zhang, Chengdu University of Traditional Chinese Medicine Affiliated Hospital, Chengdu, Sichuan Province 610000, China (e-mail: )
| |
Collapse
|
90
|
Zhou X, Ma G, Wan Z, Wang S. Label-Free Multimetric Measurement of Molecular Binding Kinetics by Electrical Modulation of a Flexible Nanobiolayer. ACS Sens 2022; 7:3461-3469. [PMID: 36273329 PMCID: PMC10358282 DOI: 10.1021/acssensors.2c01804] [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] [Indexed: 01/31/2023]
Abstract
Most label-free techniques rely on measuring refractive index or mass change on the sensor surface. Thus, it is challenging for them to measure small molecules or enzymatic processes that only induce a minor mass change on the analyte molecules. Here, we have developed a technique by combining Surface Plasmon Resonance sensing with an Oscillating Biomolecule Layer approach (SPR-OBL) to enhance the sensitivity of traditional SPR. In addition to the inherent mass sensitivity, SPR-OBL is also sensitive to the charge and conformational change of the analyte; hence it overcomes the mass limit and is able to detect small molecules. We show that the multimetric SPR-OBL measurement allows for sensing any changes regarding mass, charge, and conformation, which expands the detection capability of SPR.
Collapse
Affiliation(s)
- Xiaoyan Zhou
- Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, AZ 85287, USA
- School of Electrical, Computer and Energy Engineering, Arizona State University, Tempe, AZ 85287, USA
| | - Guangzhong Ma
- Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, AZ 85287, USA
| | - Zijian Wan
- Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, AZ 85287, USA
- School of Electrical, Computer and Energy Engineering, Arizona State University, Tempe, AZ 85287, USA
| | - Shaopeng Wang
- Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, AZ 85287, USA
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
91
|
Kandiah N, Choi SH, Hu CJ, Ishii K, Kasuga K, Mok VC. Current and Future Trends in Biomarkers for the Early Detection of Alzheimer's Disease in Asia: Expert Opinion. J Alzheimers Dis Rep 2022; 6:699-710. [PMID: 36606209 PMCID: PMC9741748 DOI: 10.3233/adr-220059] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/23/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) poses a substantial healthcare burden in the rapidly aging Asian population. Early diagnosis of AD, by means of biomarkers, can lead to interventions that might alter the course of the disease. The amyloid, tau, and neurodegeneration (AT[N]) framework, which classifies biomarkers by their core pathophysiological features, is a biomarker measure of amyloid plaques and neurofibrillary tangles. Our current AD biomarker armamentarium, comprising neuroimaging biomarkers and cerebrospinal fluid biomarkers, while clinically useful, may be invasive and expensive and hence not readily available to patients. Several studies have also investigated the use of blood-based measures of established core markers for detection of AD, such as amyloid-β and phosphorylated tau. Furthermore, novel non-invasive peripheral biomarkers and digital biomarkers could potentially expand access to early AD diagnosis to patients in Asia. Despite the multiplicity of established and potential biomarkers in AD, a regional framework for their optimal use to guide early AD diagnosis remains lacking. A group of experts from five regions in Asia gathered at a meeting in March 2021 to review the current evidence on biomarkers in AD diagnosis and discuss best practice around their use, with the goal of developing practical guidance that can be implemented easily by clinicians in Asia to support the early diagnosis of AD. This article summarizes recent key evidence on AD biomarkers and consolidates the experts' insights into the current and future use of these biomarkers for the screening and early diagnosis of AD in Asia.
Collapse
Affiliation(s)
- Nagaendran Kandiah
- Dementia Research Centre (Singapore), Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore,Correspondence to: Nagaendran Kandiah, Dementia Research Centre, Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore 308232. Tel.: +65 6592 2653; Fax: +65 6339 2889; E-mail: ; ORCID: 0000-0001-9244-4298
| | - Seong Hye Choi
- Department of Neurology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Chaur-Jong Hu
- Department of Neurology, Dementia Center, Shuang Ho Hospital, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kenji Ishii
- Team for Neuroimaging Research, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Center for Bioresources, Brain Research Institute, Niigata University, Niigata, Japan
| | - Vincent C.T. Mok
- Division of Neurology, Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China,Li Ka Shing Institute of Health Sciences, Gerald Choa Neuroscience Institute, Lui Che Woo Institute of Innovative Medicine, Therese Pei Fong Chow Research Centre for Prevention of Dementia, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
92
|
Wang Y, Zhang Z, Li B, He B, Li L, Nice EC, Zhang W, Xu J. New Insights into the Gut Microbiota in Neurodegenerative Diseases from the Perspective of Redox Homeostasis. Antioxidants (Basel) 2022; 11:2287. [PMID: 36421473 PMCID: PMC9687622 DOI: 10.3390/antiox11112287] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/08/2022] [Accepted: 11/16/2022] [Indexed: 08/27/2023] Open
Abstract
An imbalance between oxidants and antioxidants in the body can lead to oxidative stress, which is one of the major causes of neurodegenerative diseases. The gut microbiota contains trillions of beneficial bacteria that play an important role in maintaining redox homeostasis. In the last decade, the microbiota-gut-brain axis has emerged as a new field that has revolutionized the study of the pathology, diagnosis, and treatment of neurodegenerative diseases. Indeed, a growing number of studies have found that communication between the brain and the gut microbiota can be accomplished through the endocrine, immune, and nervous systems. Importantly, dysregulation of the gut microbiota has been strongly associated with the development of oxidative stress-mediated neurodegenerative diseases. Therefore, a deeper understanding of the relationship between the gut microbiota and redox homeostasis will help explain the pathogenesis of neurodegenerative diseases from a new perspective and provide a theoretical basis for proposing new therapeutic strategies for neurodegenerative diseases. In this review, we will describe the role of oxidative stress and the gut microbiota in neurodegenerative diseases and the underlying mechanisms by which the gut microbiota affects redox homeostasis in the brain, leading to neurodegenerative diseases. In addition, we will discuss the potential applications of maintaining redox homeostasis by modulating the gut microbiota to treat neurodegenerative diseases, which could open the door for new therapeutic approaches to combat neurodegenerative diseases.
Collapse
Affiliation(s)
- Yu Wang
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Zhe Zhang
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Bowen Li
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Bo He
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lei Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Wei Zhang
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, China
| | - Jia Xu
- School of Medicine, Ningbo University, Ningbo 315211, China
| |
Collapse
|
93
|
Hu Z, Ondrejcak T, Yu P, Zhang Y, Yang Y, Klyubin I, Kennelly SP, Rowan MJ, Hu NW. Do tau-synaptic long-term depression interactions in the hippocampus play a pivotal role in the progression of Alzheimer's disease? Neural Regen Res 2022; 18:1213-1219. [PMID: 36453396 PMCID: PMC9838152 DOI: 10.4103/1673-5374.360166] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Cognitive decline in Alzheimer's disease correlates with the extent of tau pathology, in particular tau hyperphosphorylation that initially appears in the transentorhinal and related regions of the brain including the hippocampus. Recent evidence indicates that tau hyperphosphorylation caused by either amyloid-β or long-term depression, a form of synaptic weakening involved in learning and memory, share similar mechanisms. Studies from our group and others demonstrate that long-term depression-inducing low-frequency stimulation triggers tau phosphorylation at different residues in the hippocampus under different experimental conditions including aging. Conversely, certain forms of long-term depression at hippocampal glutamatergic synapses require endogenous tau, in particular, phosphorylation at residue Ser396. Elucidating the exact mechanisms of interaction between tau and long-term depression may help our understanding of the physiological and pathological functions of tau/tau (hyper)phosphorylation. We first summarize experimental evidence regarding tau-long-term depression interactions, followed by a discussion of possible mechanisms by which this interplay may influence the pathogenesis of Alzheimer's disease. Finally, we conclude with some thoughts and perspectives on future research about these interactions.
Collapse
Affiliation(s)
- Zhengtao Hu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China,Department of Gerontology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Tomas Ondrejcak
- Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Pengpeng Yu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yangyang Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yin Yang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China,Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Igor Klyubin
- Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Sean P. Kennelly
- Department of Age-Related Healthcare, Tallaght University Hospital, Dublin, Ireland,Department of Medical Gerontology, Trinity College, Dublin, Ireland
| | - Michael J. Rowan
- Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Neng-Wei Hu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China,Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin, Ireland,Correspondence to: Neng-Wei Hu, .
| |
Collapse
|
94
|
Fišar Z. Linking the Amyloid, Tau, and Mitochondrial Hypotheses of Alzheimer's Disease and Identifying Promising Drug Targets. Biomolecules 2022; 12:1676. [PMID: 36421690 PMCID: PMC9687482 DOI: 10.3390/biom12111676] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/23/2022] [Accepted: 11/09/2022] [Indexed: 08/27/2023] Open
Abstract
Damage or loss of brain cells and impaired neurochemistry, neurogenesis, and synaptic and nonsynaptic plasticity of the brain lead to dementia in neurodegenerative diseases, such as Alzheimer's disease (AD). Injury to synapses and neurons and accumulation of extracellular amyloid plaques and intracellular neurofibrillary tangles are considered the main morphological and neuropathological features of AD. Age, genetic and epigenetic factors, environmental stressors, and lifestyle contribute to the risk of AD onset and progression. These risk factors are associated with structural and functional changes in the brain, leading to cognitive decline. Biomarkers of AD reflect or cause specific changes in brain function, especially changes in pathways associated with neurotransmission, neuroinflammation, bioenergetics, apoptosis, and oxidative and nitrosative stress. Even in the initial stages, AD is associated with Aβ neurotoxicity, mitochondrial dysfunction, and tau neurotoxicity. The integrative amyloid-tau-mitochondrial hypothesis assumes that the primary cause of AD is the neurotoxicity of Aβ oligomers and tau oligomers, mitochondrial dysfunction, and their mutual synergy. For the development of new efficient AD drugs, targeting the elimination of neurotoxicity, mutual potentiation of effects, and unwanted protein interactions of risk factors and biomarkers (mainly Aβ oligomers, tau oligomers, and mitochondrial dysfunction) in the early stage of the disease seems promising.
Collapse
Affiliation(s)
- Zdeněk Fišar
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague, Czech Republic
| |
Collapse
|
95
|
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: 4.0] [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
|
96
|
Pichet Binette A, Franzmeier N, Spotorno N, Ewers M, Brendel M, Biel D, Strandberg O, Janelidze S, Palmqvist S, Mattsson-Carlgren N, Smith R, Stomrud E, Ossenkoppele R, Hansson O. Amyloid-associated increases in soluble tau relate to tau aggregation rates and cognitive decline in early Alzheimer's disease. Nat Commun 2022; 13:6635. [PMID: 36333294 PMCID: PMC9636262 DOI: 10.1038/s41467-022-34129-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
For optimal design of anti-amyloid-β (Aβ) and anti-tau clinical trials, we need to better understand the pathophysiological cascade of Aβ- and tau-related processes. Therefore, we set out to investigate how Aβ and soluble phosphorylated tau (p-tau) relate to the accumulation of tau aggregates assessed with PET and subsequent cognitive decline across the Alzheimer's disease (AD) continuum. Using human cross-sectional and longitudinal neuroimaging and cognitive assessment data, we show that in early stages of AD, increased concentration of soluble CSF p-tau is strongly associated with accumulation of insoluble tau aggregates across the brain, and CSF p-tau levels mediate the effect of Aβ on tau aggregation. Further, higher soluble p-tau concentrations are mainly related to faster accumulation of tau aggregates in the regions with strong functional connectivity to individual tau epicenters. In this early stage, higher soluble p-tau concentrations is associated with cognitive decline, which is mediated by faster increase of tau aggregates. In contrast, in AD dementia, when Aβ fibrils and soluble p-tau levels have plateaued, cognitive decline is related to the accumulation rate of insoluble tau aggregates. Our data suggest that therapeutic approaches reducing soluble p-tau levels might be most favorable in early AD, before widespread insoluble tau aggregates.
Collapse
Affiliation(s)
- Alexa Pichet Binette
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden.
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Nicola Spotorno
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
| | - Michael Ewers
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Davina Biel
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Olof Strandberg
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
- Department of Neurology, Skåne University Hospital, Lund, 205 02, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
- Department of Neurology, Skåne University Hospital, Lund, 205 02, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Oskar Hansson
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
97
|
Abstract
Alzheimer's disease (AD) characterization has progressed from being indexed using clinical symptomatology followed by neuropathological examination at autopsy to in vivo signatures using cerebrospinal fluid (CSF) biomarkers and positron emission tomography. The core AD biomarkers reflect amyloid-β plaques (A), tau pathology (T) and neurodegeneration (N), following the ATN schedule, and are now being introduced into clinical routine practice. This is an important development, as disease-modifying treatments are now emerging. Further, there are now reproducible data on CSF biomarkers which reflect synaptic pathology, neuroinflammation and common co-pathologies. In addition, the development of ultrasensitive techniques has enabled the core CSF biomarkers of AD pathophysiology to be translated to blood (e.g., phosphorylated tau, amyloid-β and neurofilament light). In this chapter, we review where we stand with both core and novel CSF biomarkers, as well as the explosion of data on blood biomarkers. Also, we discuss potential applications in research aiming to better understand the disease, as well as possible use in routine clinical practice and therapeutic trials.
Collapse
Affiliation(s)
- Joel Simrén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Anders Elmgren
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, United Kingdom; UK Dementia Research Institute, University College London, London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| |
Collapse
|
98
|
Zhao L, Tannenbaum A, Bakker ENTP, Benveniste H. Physiology of Glymphatic Solute Transport and Waste Clearance from the Brain. Physiology (Bethesda) 2022; 37:0. [PMID: 35881783 PMCID: PMC9550574 DOI: 10.1152/physiol.00015.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/12/2022] [Accepted: 07/20/2022] [Indexed: 12/25/2022] Open
Abstract
This review focuses on the physiology of glymphatic solute transport and waste clearance, using evidence from experimental animal models as well as from human studies. Specific topics addressed include the biophysical characteristics of fluid and solute transport in the central nervous system, glymphatic-lymphatic coupling, as well as the role of cerebrospinal fluid movement for brain waste clearance. We also discuss the current understanding of mechanisms underlying increased waste clearance during sleep.
Collapse
Affiliation(s)
- Lucy Zhao
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut
| | - Allen Tannenbaum
- Departments of Computer Science and Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York City, New York
| | - Erik N T P Bakker
- Department of Biomedical Engineering and Physics, Amsterdam UMC, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut
- Department of Biomedical Engineering, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
99
|
Hicks C, Dhiman A, Barrymore C, Goswami T. Traumatic Brain Injury Biomarkers, Simulations and Kinetics. Bioengineering (Basel) 2022; 9:612. [PMID: 36354523 PMCID: PMC9687153 DOI: 10.3390/bioengineering9110612] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/02/2022] [Accepted: 10/20/2022] [Indexed: 10/21/2023] Open
Abstract
This paper reviews the predictive capabilities of blood-based biomarkers to quantify traumatic brain injury (TBI). Biomarkers for concussive conditions also known as mild, to moderate and severe TBI identified along with post-traumatic stress disorder (PTSD) and chronic traumatic encephalopathy (CTE) that occur due to repeated blows to the head during one's lifetime. Since the pathways of these biomarkers into the blood are not fully understood whether there is disruption in the blood-brain barrier (BBB) and the time it takes after injury for the expression of the biomarkers to be able to predict the injury effectively, there is a need to understand the protein biomarker structure and other physical properties. The injury events in terms of brain and mechanics are a result of external force with or without the shrapnel, in the wake of a wave result in local tissue damage. Thus, these mechanisms express specific biomarkers kinetics of which reaches half-life within a few hours after injury to few days. Therefore, there is a need to determine the concentration levels that follow injury. Even though current diagnostics linking biomarkers with TBI severity are not fully developed, there is a need to quantify protein structures and their viability after injury. This research was conducted to fully understand the structures of 12 biomarkers by performing molecular dynamics simulations involving atomic movement and energies of forming hydrogen bonds. Molecular dynamics software, NAMD and VMD were used to determine and compare the approximate thermodynamic stabilities of the biomarkers and their bonding energies. Five biomarkers used clinically were S100B, GFAP, UCHL1, NF-L and tau, the kinetics obtained from literature show that the concentration values abruptly change with time after injury. For a given protein length, associated number of hydrogen bonds and bond energy describe a lower bound region where proteins self-dissolve and do not have long enough half-life to be detected in the fluids. However, above this lower bound, involving higher number of bonds and energy, we hypothesize that biomarkers will be viable to disrupt the BBB and stay longer to be modeled for kinetics for diagnosis and therefore may help in the discoveries of new biomarkers.
Collapse
Affiliation(s)
- Celeste Hicks
- Biomedical, Industrial and Human Factors Engineering, Wright State University, 3640 Col. Glen Hwy, Dayton, OH 45435, USA
| | - Akshima Dhiman
- Boonshoft School of Medicine, Wright State University, 3640 Col. Glen Hwy, Dayton, OH 45435, USA
| | - Chauntel Barrymore
- Boonshoft School of Medicine, Wright State University, 3640 Col. Glen Hwy, Dayton, OH 45435, USA
| | - Tarun Goswami
- Biomedical, Industrial and Human Factors Engineering, Wright State University, 3640 Col. Glen Hwy, Dayton, OH 45435, USA
| |
Collapse
|
100
|
Shiravandi A, Yari F, Tofigh N, Kazemi Ashtiani M, Shahpasand K, Ghanian MH, Shekari F, Faridbod F. Earlier Detection of Alzheimer's Disease Based on a Novel Biomarker cis P-tau by a Label-Free Electrochemical Immunosensor. BIOSENSORS 2022; 12:879. [PMID: 36291017 PMCID: PMC9599477 DOI: 10.3390/bios12100879] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/01/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
Early detection of cis phosphorylated tau (cis P-tau) may help as an effective treatment to control the progression of Alzheimer's disease (AD). Recently, we introduced for the first time a monoclonal antibody (mAb) with high affinity against cis P-tau. In this study, the cis P-tau mAb was utilized to develop a label-free immunosensor. The antibody was immobilized onto a gold electrode and the electrochemical responses to the analyte were acquired by electrochemical impedance spectroscopy (EIS), cyclic voltammetry (CV), and differential pulse voltammetry (DPV). The immunosensor was capable of selective detection of cis P-tau among non-specific targets like trans P-tau and major plasma proteins. A wide concentration range (10 × 10-14 M-3.0 × 10-9 M) of cis P-tau was measured in PBS and human serum matrices with a limit of detection of 0.02 and 0.05 pM, respectively. Clinical applicability of the immunosensor was suggested by its long-term storage stability and successful detection of cis P-tau in real samples of cerebrospinal fluid (CSF) and blood serum collected from human patients at different stages of AD. These results suggest that this simple immunosensor may find great application in clinical settings for early detection of AD which is an unmet urgent need in today's healthcare services.
Collapse
Affiliation(s)
- Ayoub Shiravandi
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran
| | - Farzaneh Yari
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran P.O. Box 14155-6455, Iran
| | - Nahid Tofigh
- Laboratory of Neuro-Organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 1417935840, Iran
| | - Mohammad Kazemi Ashtiani
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran
| | - Koorosh Shahpasand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran
| | - Mohammad-Hossein Ghanian
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran
| | - Faezeh Shekari
- Advanced Therapy Medicinal Product Technology Development Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran
| | - Farnoush Faridbod
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran P.O. Box 14155-6455, Iran
| |
Collapse
|