1
|
Muramatsu D, Watanabe‐Nakayama T, Tsuji M, Umeda K, Hikishima S, Nakano H, Sakashita Y, Ikeda T, Konno H, Kodera N, Ando T, Noguchi‐Shinohara M, Ono K. ALZ-801 prevents amyloid β-protein assembly and reduces cytotoxicity: A preclinical experimental study. FASEB J 2025; 39:e70382. [PMID: 39921439 PMCID: PMC11806407 DOI: 10.1096/fj.202402622r] [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: 10/29/2024] [Revised: 01/20/2025] [Accepted: 01/29/2025] [Indexed: 02/10/2025]
Abstract
Alzheimer's disease (AD) is the most prevalent age-related neurodegenerative disorder, mainly characterized by amyloid β (Aβ) accumulation in the brain. Numerous new agents are currently undergoing clinical trials as disease-modifying therapies (DMTs) targeting Aβ. ALZ-801 is a promising candidate DMT for AD, with a phase 3 trial of ALZ-801 ongoing specifically for apolipoprotein E (APOE) ε4 homozygous patients with early-stage AD. This study aimed to examine the effects of ALZ-801 on Aβ assembly and explore its toxicological profile. Thioflavin T (ThT) assays and two imaging modalities-transmission electron microscopy (TEM) and high-speed atomic force microscopy (HS-AFM)-were used to evaluate ALZ-801's effects on Aβ assembly. To assess the effect of ALZ-801 on Aβ42-induced cytotoxicity, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays and lactate dehydrogenase (LDH) assays were performed. ThT assays revealed increased lag time and decreased fluorescence in the presence of ALZ-801, confirming inhibition of Aβ42 fibril formation, as confirmed by TEM. Real-time observation using HS-AFM revealed that ALZ-801 inhibited the formation of Aβ42 fibril from low-molecular-weight (LMW)-Aβ42 in the presence of Aβ42 seeds. HS-AFM also revealed that globular aggregates from LMW-Aβ42 were significantly larger with ALZ-801, with few fibrils noted. MTT and LDH assays indicated that ALZ-801 prevented LMW-Aβ42-induced cytotoxicity but did not reduce cytotoxicity induced by high-molecular-weight-Aβ42. ALZ-801 can inhibit Aβ42 aggregation by preventing both nucleus formation and fibril elongation, while promoting large globular oligomer formation, and can significantly reduce LMW-Aβ42-induced cytotoxicity. These findings underscore the potential of ALZ-801 as an effective DMT for APOE ε4 homozygous patients with AD.
Collapse
Affiliation(s)
- Daiki Muramatsu
- Department of NeurologyKanazawa University Graduate School of Medical SciencesKanazawaJapan
| | - Takahiro Watanabe‐Nakayama
- World Premier International Research Center Initiative (WPI)‐Nano Life Science InstituteKanazawa UniversityKanazawaJapan
| | - Mayumi Tsuji
- Pharmacological Research CenterShowa UniversityTokyoJapan
| | - Kenichi Umeda
- World Premier International Research Center Initiative (WPI)‐Nano Life Science InstituteKanazawa UniversityKanazawaJapan
| | - Sadao Hikishima
- Department of NeurologyKanazawa University Graduate School of Medical SciencesKanazawaJapan
| | - Hiroto Nakano
- Department of NeurologyKanazawa University Graduate School of Medical SciencesKanazawaJapan
| | - Yasuhiro Sakashita
- Department of NeurologyKanazawa University Graduate School of Medical SciencesKanazawaJapan
| | - Tokuhei Ikeda
- Department of NeurologyKanazawa University Graduate School of Medical SciencesKanazawaJapan
| | - Hiroki Konno
- World Premier International Research Center Initiative (WPI)‐Nano Life Science InstituteKanazawa UniversityKanazawaJapan
| | - Noriyuki Kodera
- World Premier International Research Center Initiative (WPI)‐Nano Life Science InstituteKanazawa UniversityKanazawaJapan
| | - Toshio Ando
- World Premier International Research Center Initiative (WPI)‐Nano Life Science InstituteKanazawa UniversityKanazawaJapan
| | | | - Kenjiro Ono
- Department of NeurologyKanazawa University Graduate School of Medical SciencesKanazawaJapan
| |
Collapse
|
2
|
Kaji S, Berghoff SA, Spieth L, Schlaphoff L, Sasmita AO, Vitale S, Büschgens L, Kedia S, Zirngibl M, Nazarenko T, Damkou A, Hosang L, Depp C, Kamp F, Scholz P, Ewers D, Giera M, Ischebeck T, Wurst W, Wefers B, Schifferer M, Willem M, Nave KA, Haass C, Arzberger T, Jäkel S, Wirths O, Saher G, Simons M. Apolipoprotein E aggregation in microglia initiates Alzheimer's disease pathology by seeding β-amyloidosis. Immunity 2024; 57:2651-2668.e12. [PMID: 39419029 DOI: 10.1016/j.immuni.2024.09.014] [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: 06/08/2023] [Revised: 02/09/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024]
Abstract
The seeded growth of pathogenic protein aggregates underlies the pathogenesis of Alzheimer's disease (AD), but how this pathological cascade is initiated is not fully understood. Sporadic AD is linked genetically to apolipoprotein E (APOE) and other genes expressed in microglia related to immune, lipid, and endocytic functions. We generated a transgenic knockin mouse expressing HaloTag-tagged APOE and optimized experimental protocols for the biochemical purification of APOE, which enabled us to identify fibrillary aggregates of APOE in mice with amyloid-β (Aβ) amyloidosis and in human AD brain autopsies. These APOE aggregates that stained positive for β sheet-binding dyes triggered Aβ amyloidosis within the endo-lysosomal system of microglia, in a process influenced by microglial lipid metabolism and the JAK/STAT signaling pathway. Taking these observations together, we propose a model for the onset of Aβ amyloidosis in AD, suggesting that the endocytic uptake and aggregation of APOE by microglia can initiate Aβ plaque formation.
Collapse
Affiliation(s)
- Seiji Kaji
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Stefan A Berghoff
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
| | - Lena Spieth
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Lennart Schlaphoff
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Andrew O Sasmita
- Max Planck Insitute for Multidisciplinary Sciences, Göttingen, Germany
| | - Simona Vitale
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Luca Büschgens
- Department of Psychiatry and Psychotherapy, University Medical Center (UMG), Georg-August-University, Göttingen, Germany
| | - Shreeya Kedia
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Martin Zirngibl
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Taisiia Nazarenko
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Alkmini Damkou
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Leon Hosang
- Institute for Neuroimmunology and Multiple Sclerosis Research, Göttingen, Germany
| | - Constanze Depp
- Max Planck Insitute for Multidisciplinary Sciences, Göttingen, Germany
| | - Frits Kamp
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Patricia Scholz
- Department of Plant Biochemistry, University of Goettingen, Albrecht-von-Haller-Institute for Plant Sciences, University of Göttingen, Göttingen, Germany
| | - David Ewers
- Max Planck Insitute for Multidisciplinary Sciences, Göttingen, Germany
| | - Martin Giera
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Albinusdreef 2, 2333ZA Leiden, the Netherlands
| | - Till Ischebeck
- Department of Plant Biochemistry, University of Goettingen, Albrecht-von-Haller-Institute for Plant Sciences, University of Göttingen, Göttingen, Germany; Institute of Plant Biology and Biotechnology (IBBP), Green Biotechnology, University of Münster, Münster, Germany
| | - Wolfgang Wurst
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany
| | - Benedikt Wefers
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Michael Willem
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Klaus-Armin Nave
- Max Planck Insitute for Multidisciplinary Sciences, Göttingen, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians University of Munich, Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Thomas Arzberger
- Center for Neuropathology and Prion Research, Ludwig-Maximilians University of Munich, Munich, Germany; Department of Psychiatry and Psychotherapy, Ludwig-Maximilians University Hospital, Munich, Germany
| | - Sarah Jäkel
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Oliver Wirths
- Department of Psychiatry and Psychotherapy, University Medical Center (UMG), Georg-August-University, Göttingen, Germany
| | - Gesine Saher
- Max Planck Insitute for Multidisciplinary Sciences, Göttingen, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.
| |
Collapse
|
3
|
Muir RT, Callum JL, Yu AYX, Kapral MK, Swartz RH, Black SE, MacIntosh BJ, Fergusson DA, Kleinman S, Demchuk AD, Stys PK, Smith EE, Hill MD. Beta-Amyloid Related Neurodegenerative and Neurovascular Diseases: Potential Implications for Transfusion Medicine. Transfus Med Rev 2024; 38:150858. [PMID: 39413667 DOI: 10.1016/j.tmrv.2024.150858] [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: 03/18/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 10/18/2024]
Abstract
Cerebral amyloid angiopathy (CAA) is a progressive cerebrovascular and neurodegenerative disorder that is caused by the aberrant accumulation of soluble beta-amyloid isoforms in the small vessel walls of the cerebral and cerebellar cortices and the leptomeninges. Vascular beta-amyloid deposition increases vulnerability to intracerebral hemorrhage (ICH). Clinically, CAA can be the underlying cause of up to half of spontaneous lobar ICHs and can also present with convexity subarachnoid hemorrhage, transient focal neurologic episodes and progressive cognitive decline leading to dementia. The majority of CAA is sporadic, with increasing prevalence with age and often coexists with Alzheimer's Disease (AD). Genetic and iatrogenic etiologies are rare. Cases of CAA and AD have been linked to the use of cadaveric pituitary hormone and later life iatrogenic CAA has also been described following early-life neurosurgical procedures with cadaveric dura grafts. Together these data suggest a capacity of beta-amyloid transmissibility. A recent study found that in over 1 million transfusion recipients from donors who later developed (i) >1 ICH or (ii) one ICH event and dementia, had an elevated risk of developing future ICH. Considering prior reports of transfusion associated variant-Creutzfeldt Jakob Disease in humans and in vivo evidence in sheep, coupled with emerging data supporting beta-amyloid's prion-like properties, raises the question of whether CAA could be transmissible by blood transfusion. This would also have implications for screening, especially in an era of emerging plasma biomarkers of cerebral amyloidosis. Given the public health concerns raised by this biologically plausible question, there is a need for future studies with well-characterized definitions - and temporal ascertainment - of CAA exposure and outcomes to examine whether CAA is transfusion-transmissible, and, if so, with what frequency and timing of onset.
Collapse
Affiliation(s)
- Ryan T Muir
- Calgary Stroke Program, Department of Clinical Neurosciences, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Calgary, Alberta, Canada; Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Jeannie L Callum
- Department of Pathology and Molecular Medicine, Queen's University, Ontario, Canada; Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Research Program Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Amy Y X Yu
- Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Research Program Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada; Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada; ICES (formerly Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
| | - Moira K Kapral
- ICES (formerly Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada; Department of Medicine, General Internal Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Richard H Swartz
- Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Research Program Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada; Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada; ICES (formerly Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
| | - Sandra E Black
- Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Research Program Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada; Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Bradley J MacIntosh
- Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Research Program Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Dean A Fergusson
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Steven Kleinman
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrew D Demchuk
- Calgary Stroke Program, Department of Clinical Neurosciences, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Calgary, Alberta, Canada
| | - Peter K Stys
- Calgary Stroke Program, Department of Clinical Neurosciences, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Calgary, Alberta, Canada
| | - Eric E Smith
- Calgary Stroke Program, Department of Clinical Neurosciences, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Calgary, Alberta, Canada
| | - Michael D Hill
- Calgary Stroke Program, Department of Clinical Neurosciences, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Calgary, Alberta, Canada; Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
4
|
Zhong MZ, Peng T, Duarte ML, Wang M, Cai D. Updates on mouse models of Alzheimer's disease. Mol Neurodegener 2024; 19:23. [PMID: 38462606 PMCID: PMC10926682 DOI: 10.1186/s13024-024-00712-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 02/14/2024] [Indexed: 03/12/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease in the United States (US). Animal models, specifically mouse models have been developed to better elucidate disease mechanisms and test therapeutic strategies for AD. A large portion of effort in the field was focused on developing transgenic (Tg) mouse models through over-expression of genetic mutations associated with familial AD (FAD) patients. Newer generations of mouse models through knock-in (KI)/knock-out (KO) or CRISPR gene editing technologies, have been developed for both familial and sporadic AD risk genes with the hope to more accurately model proteinopathies without over-expression of human AD genes in mouse brains. In this review, we summarized the phenotypes of a few commonly used as well as newly developed mouse models in translational research laboratories including the presence or absence of key pathological features of AD such as amyloid and tau pathology, synaptic and neuronal degeneration as well as cognitive and behavior deficits. In addition, advantages and limitations of these AD mouse models have been elaborated along with discussions of any sex-specific features. More importantly, the omics data from available AD mouse models have been analyzed to categorize molecular signatures of each model reminiscent of human AD brain changes, with the hope to guide future selection of most suitable models for specific research questions to be addressed in the AD field.
Collapse
Affiliation(s)
- Michael Z Zhong
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Biology, College of Arts and Science, Boston University, Boston, MA, 02215, USA
| | - Thomas Peng
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Science Research Program, Scarsdale High School, New York, NY, 10583, USA
| | - Mariana Lemos Duarte
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Research & Development, James J Peters VA Medical Center, Bronx, NY, 10468, USA.
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
| | - Dongming Cai
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Research & Development, James J Peters VA Medical Center, Bronx, NY, 10468, USA.
- Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Neurology, N. Bud Grossman Center for Memory Research and Care, The University of Minnesota, Minneapolis, MN, 55455, USA.
- Geriatric Research Education & Clinical Center (GRECC), The Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA.
| |
Collapse
|
5
|
Stepanenko OV, Sulatskaya AI, Sulatsky MI, Mikhailova EV, Kuznetsova IM, Turoverov KK, Stepanenko OV. Mammalian odorant-binding proteins are prone to form amorphous aggregates and amyloid fibrils. Int J Biol Macromol 2023; 253:126872. [PMID: 37722633 DOI: 10.1016/j.ijbiomac.2023.126872] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/06/2023] [Accepted: 09/10/2023] [Indexed: 09/20/2023]
Abstract
Odorant-binding proteins are involved in perceiving smell by capturing odorants within the protein's β-barrel. On the example of bovine odorant-binding protein (bOBP), the structural organization of such proteins and their ability to bind ligands under various conditions in vitro were examined. We found a tendency of bOBP to form oligomers and small amorphous aggregates without disturbing the integrity of protein monomers at physiological conditions. Changes in environmental parameters (increased temperature and pH) favored the formation of larger and dense supramolecular complexes that significantly reduce the binding of ligands by bOBP. The ability of bOBP to form fibrillar aggregates with the properties of amyloids, including high cytotoxicity, was revealed at sample stirring (even at physiological temperature and pH), at medium acidification or pre-solubilization with hexafluoroisopropanol. Fibrillogenesis of bOBP was initiated by the dissociation of the protein's supramolecular complexes into monomers and the destabilization of the protein's β-barrels without a significant destruction of its native β-strands.
Collapse
Affiliation(s)
- Olga V Stepanenko
- Laboratory of Structural Dynamics, Stability and folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave, 194064 St. Petersburg, Russia.
| | - Anna I Sulatskaya
- Laboratory of Structural Dynamics, Stability and folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave, 194064 St. Petersburg, Russia.
| | - Maksim I Sulatsky
- Laboratory of Cell Morphology, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave., 194064 St. Petersburg, Russia.
| | - Ekaterina V Mikhailova
- Laboratory of Structural Dynamics, Stability and folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave, 194064 St. Petersburg, Russia.
| | - Irina M Kuznetsova
- Laboratory of Structural Dynamics, Stability and folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave, 194064 St. Petersburg, Russia.
| | - Konstantin K Turoverov
- Laboratory of Structural Dynamics, Stability and folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave, 194064 St. Petersburg, Russia.
| | - Olesya V Stepanenko
- Laboratory of Structural Dynamics, Stability and folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave, 194064 St. Petersburg, Russia.
| |
Collapse
|
6
|
Kozin SA, Kechko OI, Adzhubei AA, Makarov AA, Mitkevich VA. Switching On/Off Amyloid Plaque Formation in Transgenic Animal Models of Alzheimer's Disease. Int J Mol Sci 2023; 25:72. [PMID: 38203242 PMCID: PMC10778642 DOI: 10.3390/ijms25010072] [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: 11/24/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
A hallmark of Alzheimer's disease (AD) are the proteinaceous aggregates formed by the amyloid-beta peptide (Aβ) that is deposited inside the brain as amyloid plaques. The accumulation of aggregated Aβ may initiate or enhance pathologic processes in AD. According to the amyloid hypothesis, any agent that has the capability to inhibit Aβ aggregation and/or destroy amyloid plaques represents a potential disease-modifying drug. In 2023, a humanized IgG1 monoclonal antibody (lecanemab) against the Aβ-soluble protofibrils was approved by the US FDA for AD therapy, thus providing compelling support to the amyloid hypothesis. To acquire a deeper insight on the in vivo Aβ aggregation, various animal models, including aged herbivores and carnivores, non-human primates, transgenic rodents, fish and worms were widely exploited. This review is based on the recent data obtained using transgenic animal AD models and presents experimental verification of the critical role in Aβ aggregation seeding of the interactions between zinc ions, Aβ with the isomerized Asp7 (isoD7-Aβ) and the α4β2 nicotinic acetylcholine receptor.
Collapse
Affiliation(s)
- Sergey A. Kozin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.I.K.); (A.A.A.); (A.A.M.)
| | | | | | | | - Vladimir A. Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.I.K.); (A.A.A.); (A.A.M.)
| |
Collapse
|
7
|
Zhao J, Rostgaard K, Lauwers E, Dahlén T, Ostrowski SR, Erikstrup C, Pedersen OB, de Strooper B, Lemmens R, Hjalgrim H, Edgren G. Intracerebral Hemorrhage Among Blood Donors and Their Transfusion Recipients. JAMA 2023; 330:941-950. [PMID: 37698562 PMCID: PMC10498336 DOI: 10.1001/jama.2023.14445] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 07/13/2023] [Indexed: 09/13/2023]
Abstract
Importance Recent reports have suggested that cerebral amyloid angiopathy, a common cause of multiple spontaneous intracerebral hemorrhages (ICHs), may be transmissible through parenteral injection of contaminated cadaveric pituitary hormone in humans. Objective To determine whether spontaneous ICH in blood donors after blood donation is associated with development of spontaneous ICH in transfusion recipients. Design, Setting, and Participants Exploratory retrospective cohort study using nationwide blood bank and health register data from Sweden (main cohort) and Denmark (validation cohort) and including all 1 089 370 patients aged 5 to 80 years recorded to have received a red blood cell transfusion from January 1, 1970 (Sweden), or January 1, 1980 (Denmark), until December 31, 2017. Exposures Receipt of red blood cell transfusions from blood donors who subsequently developed (1) a single spontaneous ICH, (2) multiple spontaneous ICHs, or (3) no spontaneous ICH. Main Outcomes and Measures Spontaneous ICH in transfusion recipients; ischemic stroke was a negative control outcome. Results A total of 759 858 patients from Sweden (median age, 65 [IQR, 48-73] years; 59% female) and 329 512 from Denmark (median age, 64 [IQR, 50-73] years; 58% female) were included, with a median follow-up of 5.8 (IQR, 1.4-12.5) years and 6.1 (IQR, 1.5-11.6) years, respectively. Patients who underwent transfusion with red blood cell units from donors who developed multiple spontaneous ICHs had a significantly higher risk of a single spontaneous ICH themselves, compared with patients receiving transfusions from donors who did not develop spontaneous ICH, in both the Swedish cohort (unadjusted incidence rate [IR], 3.16 vs 1.12 per 1000 person-years; adjusted hazard ratio [HR], 2.73; 95% CI, 1.72-4.35; P < .001) and the Danish cohort (unadjusted IR, 2.82 vs 1.09 per 1000 person-years; adjusted HR, 2.32; 95% CI, 1.04-5.19; P = .04). No significant difference was found for patients receiving transfusions from donors who developed a single spontaneous ICH in the Swedish cohort (unadjusted IR, 1.35 vs 1.12 per 1000 person-years; adjusted HR, 1.06; 95% CI, 0.84-1.36; P = .62) nor the Danish cohort (unadjusted IR, 1.36 vs 1.09 per 1000 person-years; adjusted HR, 1.06; 95% CI, 0.70-1.60; P = .73), nor for ischemic stroke as a negative control outcome. Conclusions and Relevance In an exploratory analysis of patients who received red blood cell transfusions, patients who underwent transfusion with red blood cells from donors who later developed multiple spontaneous ICHs were at significantly increased risk of spontaneous ICH themselves. This may suggest a transfusion-transmissible agent associated with some types of spontaneous ICH, although the findings may be susceptible to selection bias and residual confounding, and further research is needed to investigate if transfusion transmission of cerebral amyloid angiopathy might explain this association.
Collapse
Affiliation(s)
- Jingcheng Zhao
- Department of Medicine, Solna, Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
| | - Klaus Rostgaard
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Elsa Lauwers
- VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Torsten Dahlén
- Department of Medicine, Solna, Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
- Hematology Department, Karolinska University Hospital, Stockholdm, Sweden
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Ole Birger Pedersen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
| | - Bart de Strooper
- VIB Center for Brain and Disease Research, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute, KU Leuven (University of Leuven), Leuven, Belgium
- Dementia Research Institute, University College London, London, England
| | - Robin Lemmens
- VIB Center for Brain and Disease Research, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute, KU Leuven (University of Leuven), Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Henrik Hjalgrim
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| | - Gustaf Edgren
- Department of Medicine, Solna, Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
- Department of Cardiology, Södersjukhuset, Stockholm, Sweden
| |
Collapse
|
8
|
Sulatsky MI, Belousov MV, Kosolapova AO, Mikhailova EV, Romanenko MN, Antonets KS, Kuznetsova IM, Turoverov KK, Nizhnikov AA, Sulatskaya AI. Amyloid Fibrils of Pisum sativum L. Vicilin Inhibit Pathological Aggregation of Mammalian Proteins. Int J Mol Sci 2023; 24:12932. [PMID: 37629113 PMCID: PMC10454621 DOI: 10.3390/ijms241612932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Although incurable pathologies associated with the formation of highly ordered fibrillar protein aggregates called amyloids have been known for about two centuries, functional roles of amyloids have been studied for only two decades. Recently, we identified functional amyloids in plants. These amyloids formed using garden pea Pisum sativum L. storage globulin and vicilin, accumulated during the seed maturation and resisted treatment with gastric enzymes and canning. Thus, vicilin amyloids ingested with food could interact with mammalian proteins. In this work, we analyzed the effects of vicilin amyloids on the fibril formation of proteins that form pathological amyloids. We found that vicilin amyloids inhibit the fibrillogenesis of these proteins. In particular, vicilin amyloids decrease the number and length of lysozyme amyloid fibrils; the length and width of β-2-microglobulin fibrils; the number, length and the degree of clustering of β-amyloid fibrils; and, finally, they change the structure and decrease the length of insulin fibrils. Such drastic influences of vicilin amyloids on the pathological amyloids' formation cause the alteration of their toxicity for mammalian cells, which decreases for all tested amyloids with the exception of insulin. Taken together, our study, for the first time, demonstrates the anti-amyloid effect of vicilin fibrils and suggests the mechanisms underlying this phenomenon.
Collapse
Affiliation(s)
- Maksim I. Sulatsky
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (M.I.S.); (E.V.M.); (I.M.K.); (K.K.T.)
| | - Mikhail V. Belousov
- All-Russia Research Institute for Agricultural Microbiology, 196608 St. Petersburg, Russia; (M.V.B.); (A.O.K.); (M.N.R.); (K.S.A.)
- Faculty of Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Anastasiia O. Kosolapova
- All-Russia Research Institute for Agricultural Microbiology, 196608 St. Petersburg, Russia; (M.V.B.); (A.O.K.); (M.N.R.); (K.S.A.)
- Faculty of Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Ekaterina V. Mikhailova
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (M.I.S.); (E.V.M.); (I.M.K.); (K.K.T.)
| | - Maria N. Romanenko
- All-Russia Research Institute for Agricultural Microbiology, 196608 St. Petersburg, Russia; (M.V.B.); (A.O.K.); (M.N.R.); (K.S.A.)
- Faculty of Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Kirill S. Antonets
- All-Russia Research Institute for Agricultural Microbiology, 196608 St. Petersburg, Russia; (M.V.B.); (A.O.K.); (M.N.R.); (K.S.A.)
- Faculty of Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Irina M. Kuznetsova
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (M.I.S.); (E.V.M.); (I.M.K.); (K.K.T.)
| | - Konstantin K. Turoverov
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (M.I.S.); (E.V.M.); (I.M.K.); (K.K.T.)
| | - Anton A. Nizhnikov
- All-Russia Research Institute for Agricultural Microbiology, 196608 St. Petersburg, Russia; (M.V.B.); (A.O.K.); (M.N.R.); (K.S.A.)
- Faculty of Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Anna I. Sulatskaya
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (M.I.S.); (E.V.M.); (I.M.K.); (K.K.T.)
| |
Collapse
|
9
|
Johansson B, Oasa S, Muntsant Soria A, Tiiman A, Söderberg L, Amandius E, Möller C, Lannfelt L, Terenius L, Giménez-Llort L, Vukojević V. The interwoven fibril-like structure of amyloid-beta plaques in mouse brain tissue visualized using super-resolution STED microscopy. Cell Biosci 2023; 13:142. [PMID: 37542303 PMCID: PMC10403925 DOI: 10.1186/s13578-023-01086-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/14/2023] [Indexed: 08/06/2023] Open
Abstract
BACKGROUND Standard neuropathologic analysis of Alzheimer's brain relies on traditional fluorescence microscopy, which suffers from limited spatial resolution due to light diffraction. As a result, it fails to reveal intricate details of amyloid plaques. While electron microscopy (EM) offers higher resolution, its extensive sample preparation, involving fixation, dehydration, embedding, and sectioning, can introduce artifacts and distortions in the complex brain tissue. Moreover, EM lacks molecular specificity and has limited field of view and imaging depth. RESULTS In our study, we employed super-resolution Stimulated Emission Depletion (STED) microscopy in conjunction with the anti-human APP recombinant antibody 1C3 fluorescently labelled with DyLightTM633 (1C3-DyLight633). This combination allowed us to visualize amyloidogenic aggregates in vitro and in brain sections from a 17-month-old 3×Tg-AD mouse with sub-diffraction limited spatial resolution. Remarkably, we achieved a spatial resolution of 29 nm in vitro and 62 nm in brain tissue sections, surpassing the capabilities of conventional confocal microscopy by 5-10 times. Consequently, we could discern individual fibrils within plaques, an achievement previously only possible with EM. CONCLUSIONS The utilization of STED microscopy represents a groundbreaking advancement in the field, enabling researchers to delve into the characterization of local mechanisms that underlie Amyloid (Aβ) deposition into plaques and their subsequent clearance. This unprecedented level of detail is especially crucial for comprehending the etiology of Alzheimer's disease and developing the next generation of anti-amyloid treatments. By facilitating the evaluation of drug candidates and non-pharmacological interventions aiming to reduce amyloid burden, STED microscopy emerges as an indispensable tool for driving scientific progress in Alzheimer's research.
Collapse
Affiliation(s)
- Björn Johansson
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17176, Stockholm, Sweden
- Theme Aging, Karolinska University Hospital, Karolinska Institutet, SE-17176, Stockholm, Sweden
| | - Sho Oasa
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17176, Stockholm, Sweden
| | - Aida Muntsant Soria
- Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
| | - Ann Tiiman
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17176, Stockholm, Sweden
| | | | | | | | | | - Lars Terenius
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17176, Stockholm, Sweden
| | - Lydia Giménez-Llort
- Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
| | - Vladana Vukojević
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17176, Stockholm, Sweden.
| |
Collapse
|
10
|
Greenberg SM, Charidimou A. Seed to Bleed: Iatrogenic Cerebral Amyloid Angiopathy. Stroke 2023; 54:1224-1226. [PMID: 37035915 PMCID: PMC10473030 DOI: 10.1161/strokeaha.123.042583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Affiliation(s)
- Steven M. Greenberg
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston (S.M.G.)
| | - Andreas Charidimou
- Department of Neurology, Boston University Medical Center and Boston University School of Medicine, MA (A.C.)
| |
Collapse
|
11
|
Lacoursiere SG, Safar J, Westaway D, Mohajerani MH, Sutherland RJ. The effect of Aβ seeding is dependent on the presence of knock-in genes in the App NL-G-F mice. FRONTIERS IN DEMENTIA 2022; 1:941879. [PMID: 39081481 PMCID: PMC11285652 DOI: 10.3389/frdem.2022.941879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/08/2022] [Indexed: 08/02/2024]
Abstract
Alzheimer's disease (AD) is characterized by the prion-like propagation of amyloid-β (Aβ). However, the role of Aβ in cognitive impairment is still unclear. To determine the causal role of Aβ in AD, we intracerebrally seeded the entorhinal cortex of a 2-month-old App NL-G-F mouse model with an Aβ peptide derived from patients who died from rapidly progressing AD. When the mice were 3 months of age or 1 month following seeding, spatial learning and memory were tested using the Morris water task. Immunohistochemical labeling showed seeding with the Aβ was found accelerate Aβ plaque deposition and microgliosis in the App NL-G-F mice, but this was dependent on the presence of the knocked-in genes. However, we found no correlation between pathology and spatial performance. The results of the present study show the seeding effects in the App NL-G-F knock-in model, and how these are dependent on the presence of a humanized App gene. But these pathological changes were not initially causal in memory impairment.
Collapse
Affiliation(s)
- Sean G. Lacoursiere
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Jiri Safar
- Departments of Pathology, Neurology, Psychiatry, and National Prion Disease Pathology Surveillance Center, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - David Westaway
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
| | - Majid H. Mohajerani
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Robert J. Sutherland
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| |
Collapse
|
12
|
Aires V, Ziegler-Waldkirch S, Friesen M, Reichardt W, Erny D, Loreth D, Harborne A, Kretz O, von Elverfeldt D, Meyer-Luehmann M. Seed-induced Aβ deposits in the corpus callosum disrupt white matter integrity in a mouse model of Alzheimer’s disease. Front Cell Neurosci 2022; 16:862918. [PMID: 36003141 PMCID: PMC9393256 DOI: 10.3389/fncel.2022.862918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Neuropathologically, Alzheimer’s disease (AD) is characterized by the accumulation of amyloid-beta peptide (Aβ) and subsequent formation of the so-called Aβ plaques. Along with neuronal loss, previous studies report white matter anomalies and corpus callosum (CC) atrophy in AD patients. Notably, perturbations in the white matter can be observed years before expected disease onset, suggesting that early stages of disease progression play a role in AD-associated loss of myelin integrity. Through seed-induced deposition of Aβ, we are able to examine alterations of central nervous system (CNS) integrity during the initial stages of plaque formation. In this study, we investigate the impact of Aβ seeding in the CC utilizing various imaging techniques as well as quantitative gene expression analysis and demonstrate that Aβ deposits result in an imbalance of glial cells in the CC. We found increased amounts of phagocytic microglia and reactive astrocytes, while oligodendrocyte progenitor cell (OPC) numbers were reduced. Moreover, white matter aberrations adjacent to the Aβ seeding were observed together with an overall decline in callosal myelination. This data indicate that the initial stages of plaque formation induce oligodendrocyte dysfunction, which might ultimately lead to myelin loss.
Collapse
Affiliation(s)
- Vanessa Aires
- Department of Neurology, Medical Center – University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Stephanie Ziegler-Waldkirch
- Department of Neurology, Medical Center – University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marina Friesen
- Department of Neurology, Medical Center – University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Wilfried Reichardt
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Radiology, Medical Physics, Medical Center – University of Freiburg, Freiburg, Germany
- German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Erny
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute of Neuropathology, University of Freiburg, Freiburg, Germany
- Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Desiree Loreth
- Department of Neurology, Medical Center – University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andrew Harborne
- Department of Neurology, Medical Center – University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Kretz
- Department of Internal Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dominik von Elverfeldt
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Radiology, Medical Physics, Medical Center – University of Freiburg, Freiburg, Germany
| | - Melanie Meyer-Luehmann
- Department of Neurology, Medical Center – University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- *Correspondence: Melanie Meyer-Luehmann,
| |
Collapse
|
13
|
Inhibition of PLK2 activity affects APP and tau pathology and improves synaptic content in a sex-dependent manner in a 3xTg mouse model of Alzheimer's disease. Neurobiol Dis 2022; 172:105833. [PMID: 35905928 DOI: 10.1016/j.nbd.2022.105833] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 07/14/2022] [Accepted: 07/22/2022] [Indexed: 11/23/2022] Open
Abstract
Converging lines of evidence suggest that abnormal accumulation of the kinase Polo-like kinase 2 (PLK2) might play a role in the pathogenesis of Alzheimer's disease (AD), possibly through its role in regulating the amyloid β (Aβ) cascade. In the present study, we investigated the effect of inhibiting PLK2 kinase activity in in vitro and in vivo models of AD neuropathology. First, we confirmed that PLK2 overexpression modulated APP and Tau protein levels and phosphorylation in cell culture, in a kinase activity dependent manner. Furthermore, a transient treatment of triple transgenic mouse model of AD (3xTg-AD) with a potent and specific PLK2 pharmacological inhibitor (PLK2i #37) reduced some neuropathological aspects in a sex-dependent manner. In 3xTg-AD males, treatment with PLK2i #37 led to lower Tau burden, higher synaptic protein content, and prevented learning and memory deficits. In contrast, treated females showed an exacerbation of Tau pathology, associated with a reduction in amyloid plaque accumulation. Overall, our findings suggest that PLK2 inhibition alters key components of AD neuropathology in a sex-dependent manner and might display a therapeutic potential for the treatment for AD and related dementia.
Collapse
|
14
|
Wakeman DR, Weed MR, Perez SE, Cline EN, Viola KL, Wilcox KC, Moddrelle DS, Nisbett EZ, Kurian AM, Bell AF, Pike R, Jacobson PB, Klein WL, Mufson EJ, Lawrence MS, Elsworth JD. Intrathecal amyloid-beta oligomer administration increases tau phosphorylation in the medial temporal lobe in the African green monkey: A nonhuman primate model of Alzheimer's disease. Neuropathol Appl Neurobiol 2022; 48:e12800. [PMID: 35156715 PMCID: PMC10902791 DOI: 10.1111/nan.12800] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/31/2022] [Accepted: 02/05/2022] [Indexed: 11/26/2022]
Abstract
AIMS An obstacle to developing new treatment strategies for Alzheimer's disease (AD) has been the inadequate translation of findings in current AD transgenic rodent models to the prediction of clinical outcomes. By contrast, nonhuman primates (NHPs) share a close neurobiology with humans in virtually all aspects relevant to developing a translational AD model. The present investigation used African green monkeys (AGMs) to refine an inducible NHP model of AD based on the administration of amyloid-beta oligomers (AβOs), a key upstream initiator of AD pathology. METHODS AβOs or vehicle were repeatedly delivered over 4 weeks to age-matched young adult AGMs by intracerebroventricular (ICV) or intrathecal (IT) injections. Induction of AD-like pathology was assessed in subregions of the medial temporal lobe (MTL) by quantitative immunohistochemistry (IHC) using the AT8 antibody to detect hyperphosphorylated tau. Hippocampal volume was measured by magnetic resonance imaging (MRI) scans prior to, and after, intrathecal injections. RESULTS IT administration of AβOs in young adult AGMs revealed an elevation of tau phosphorylation in the MTL cortical memory circuit compared with controls. The largest increases were detected in the entorhinal cortex that persisted for at least 12 weeks after dosing. MRI scans showed a reduction in hippocampal volume following AβO injections. CONCLUSIONS Repeated IT delivery of AβOs in young adult AGMs led to an accelerated AD-like neuropathology in MTL, similar to human AD, supporting the value of this translational model to de-risk the clinical trial of diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Sylvia E Perez
- Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Erika N Cline
- Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - Kirsten L Viola
- Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - Kyle C Wilcox
- Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - David S Moddrelle
- Virscio Inc., St. Kitts Biomedical Research Foundation, St. Kitts, West Indies
| | - Ernell Z Nisbett
- Virscio Inc., St. Kitts Biomedical Research Foundation, St. Kitts, West Indies
| | | | - Amanda F Bell
- Virscio Inc., St. Kitts Biomedical Research Foundation, St. Kitts, West Indies
| | - Ricaldo Pike
- Virscio Inc., St. Kitts Biomedical Research Foundation, St. Kitts, West Indies
| | | | - William L Klein
- Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - Elliott J Mufson
- Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | | | | |
Collapse
|
15
|
Moretto E, Stuart S, Surana S, Vargas JNS, Schiavo G. The Role of Extracellular Matrix Components in the Spreading of Pathological Protein Aggregates. Front Cell Neurosci 2022; 16:844211. [PMID: 35573838 PMCID: PMC9100790 DOI: 10.3389/fncel.2022.844211] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/08/2022] [Indexed: 11/23/2022] Open
Abstract
Several neurodegenerative diseases are characterized by the accumulation of aggregated misfolded proteins. These pathological agents have been suggested to propagate in the brain via mechanisms similar to that observed for the prion protein, where a misfolded variant is transferred from an affected brain region to a healthy one, thereby inducing the misfolding and/or aggregation of correctly folded copies. This process has been characterized for several proteins, such as α-synuclein, tau, amyloid beta (Aβ) and less extensively for huntingtin and TDP-43. α-synuclein, tau, TDP-43 and huntingtin are intracellular proteins, and their aggregates are located in the cytosol or nucleus of neurons. They have been shown to spread between cells and this event occurs, at least partially, via secretion of these protein aggregates in the extracellular space followed by re-uptake. Conversely, Aβ aggregates are found mainly extracellularly, and their spreading occurs in the extracellular space between brain regions. Due to the inherent nature of their spreading modalities, these proteins are exposed to components of the extracellular matrix (ECM), including glycans, proteases and core matrix proteins. These ECM components can interact with or process pathological misfolded proteins, potentially changing their properties and thus regulating their spreading capabilities. Here, we present an overview of the documented roles of ECM components in the spreading of pathological protein aggregates in neurodegenerative diseases with the objective of identifying the current gaps in knowledge and stimulating further research in the field. This could potentially lead to the identification of druggable targets to slow down the spreading and/or progression of these pathologies.
Collapse
Affiliation(s)
- Edoardo Moretto
- Institute of Neuroscience, National Research Council, CNR, Milan, Italy
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- *Correspondence: Edoardo Moretto,
| | - Skye Stuart
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Sunaina Surana
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
| | - Jose Norberto S. Vargas
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
| | - Giampietro Schiavo
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
- Giampietro Schiavo,
| |
Collapse
|
16
|
Chourrout M, Roux M, Boisvert C, Gislard C, Legland D, Arganda-Carreras I, Olivier C, Peyrin F, Boutin H, Rama N, Baron T, Meyronet D, Brun E, Rositi H, Wiart M, Chauveau F. Brain virtual histology with X-ray phase-contrast tomography Part II:3D morphologies of amyloid- β plaques in Alzheimer's disease models. BIOMEDICAL OPTICS EXPRESS 2022; 13:1640-1653. [PMID: 35414980 PMCID: PMC8973161 DOI: 10.1364/boe.438890] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 05/15/2023]
Abstract
While numerous transgenic mouse strains have been produced to model the formation of amyloid-β (Aβ) plaques in the brain, efficient methods for whole-brain 3D analysis of Aβ deposits have to be validated and standardized. Moreover, routine immunohistochemistry performed on brain slices precludes any shape analysis of Aβ plaques, or require complex procedures for serial acquisition and reconstruction. The present study shows how in-line (propagation-based) X-ray phase-contrast tomography (XPCT) combined with ethanol-induced brain sample dehydration enables hippocampus-wide detection and morphometric analysis of Aβ plaques. Performed in three distinct Alzheimer mouse strains, the proposed workflow identified differences in signal intensity and 3D shape parameters: 3xTg displayed a different type of Aβ plaques, with a larger volume and area, greater elongation, flatness and mean breadth, and more intense average signal than J20 and APP/PS1. As a label-free non-destructive technique, XPCT can be combined with standard immunohistochemistry. XPCT virtual histology could thus become instrumental in quantifying the 3D spreading and the morphological impact of seeding when studying prion-like properties of Aβ aggregates in animal models of Alzheimer's disease. This is Part II of a series of two articles reporting the value of in-line XPCT for virtual histology of the brain; Part I shows how in-line XPCT enables 3D myelin mapping in the whole rodent brain and in human autopsy brain tissue.
Collapse
Affiliation(s)
- Matthieu Chourrout
- Univ. Lyon, Lyon Neuroscience Research Center, CNRS UMR5292, Inserm U1028, Université Claude Bernard Lyon 1, Lyon, France
| | - Margaux Roux
- Univ. Lyon, Lyon Neuroscience Research Center, CNRS UMR5292, Inserm U1028, Université Claude Bernard Lyon 1, Lyon, France
| | - Carlie Boisvert
- Univ. Lyon, Lyon Neuroscience Research Center, CNRS UMR5292, Inserm U1028, Université Claude Bernard Lyon 1, Lyon, France
- Current affiliation: Faculty of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ontario, Canada
| | - Coralie Gislard
- Univ. Lyon, Lyon Neuroscience Research Center, CNRS UMR5292, Inserm U1028, Université Claude Bernard Lyon 1, Lyon, France
| | | | - Ignacio Arganda-Carreras
- University of the Basque Country (UPV/EHU), San Sebastian, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- Donostia International Physics Center (DIPC), San Sebastian, Spain
| | - Cécile Olivier
- Univ. Lyon, CREATIS; CNRS UMR5220; INSERM U1044; INSA-Lyon; Univ. Lyon 1, Lyon, France
| | - Françoise Peyrin
- Univ. Lyon, CREATIS; CNRS UMR5220; INSERM U1044; INSA-Lyon; Univ. Lyon 1, Lyon, France
| | - Hervé Boutin
- Univ. Manchester, Faculty of Biology Medicine and Health, Wolfson Molecular Imaging Centre, Manchester, UK
| | - Nicolas Rama
- Univ. Lyon, CRCL; INSERM U1052; CNRS UMR5286; Univ. Lyon 1; Centre Léon Bérard, Lyon, France
| | | | | | - Emmanuel Brun
- Univ. Grenoble Alpes, Inserm UA07 Strobe Grenoble, France
| | - Hugo Rositi
- Univ. Clermont Auvergne, Institut Pascal; CNRS UMR 6602; SIGMA Clermont, Clermont-Ferrand, France
| | - Marlène Wiart
- Univ. Lyon, CarMeN Laboratory; INSERM U1060; INRA U1397; Hospices Civils de Lyon, Lyon, France
- CNRS, Lyon, France
- These authors contributed equally to this work
| | - Fabien Chauveau
- Univ. Lyon, Lyon Neuroscience Research Center, CNRS UMR5292, Inserm U1028, Université Claude Bernard Lyon 1, Lyon, France
- CNRS, Lyon, France
- These authors contributed equally to this work
| |
Collapse
|
17
|
Ziegler-Waldkirch S, Friesen M, Loreth D, Sauer JF, Kemna S, Hilse A, Erny D, Helm C, d´Errico P, Prinz M, Bartos M, Meyer-Luehmann M. Seed-induced Aβ deposition alters neuronal function and impairs olfaction in a mouse model of Alzheimer's disease. Mol Psychiatry 2022; 27:4274-4284. [PMID: 35869271 PMCID: PMC9718674 DOI: 10.1038/s41380-022-01686-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of amyloid-β (Aβ) which ultimately forms plaques. These Aβ deposits can be induced in APP transgenic mouse models by prion-like seeding. It has been widely accepted that anosmia and hyposmia occur during the early stages of AD, even before cognitive deficits are present. In order to determine the impact of seed-induced Aβ deposits on olfaction, we performed intracerebral injections of seed-competent brain homogenate into the olfactory bulb of young pre-depositing APP transgenic mice. Remarkably, we observed a dramatic olfactory impairment in those mice. Furthermore, the number of newborn neurons as well as the activity of cells in the mitral cell layer was decreased. Notably, exposure to an enriched environment reduced Aβ seeding, vivified neurogenesis and most importantly reversed olfactory deficits. Based on our findings, we conclude that altered neuronal function as a result of induced Aβ pathology might contribute to olfactory dysfunction in AD.
Collapse
Affiliation(s)
- Stephanie Ziegler-Waldkirch
- grid.7708.80000 0000 9428 7911Department of Neurology, Medical Center – University of Freiburg, 79106 Freiburg, Germany ,grid.5963.9Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Marina Friesen
- grid.7708.80000 0000 9428 7911Department of Neurology, Medical Center – University of Freiburg, 79106 Freiburg, Germany ,grid.5963.9Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany ,grid.5963.9Faculty of Biology, University of Freiburg, 79110 Freiburg, Germany
| | - Desirée Loreth
- grid.7708.80000 0000 9428 7911Department of Neurology, Medical Center – University of Freiburg, 79106 Freiburg, Germany ,grid.5963.9Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany ,grid.13648.380000 0001 2180 3484Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Jonas-Frederic Sauer
- grid.5963.9Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany ,grid.5963.9Institute for Physiology I, Systemic and Cellular Neurophysiology, University of Freiburg, 79104 Freiburg, Germany
| | - Solveig Kemna
- grid.7708.80000 0000 9428 7911Department of Neurology, Medical Center – University of Freiburg, 79106 Freiburg, Germany ,grid.5963.9Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Alexandra Hilse
- grid.7708.80000 0000 9428 7911Department of Neurology, Medical Center – University of Freiburg, 79106 Freiburg, Germany ,grid.5963.9Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany ,grid.5963.9Faculty of Biology, University of Freiburg, 79110 Freiburg, Germany
| | - Daniel Erny
- grid.5963.9Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany ,grid.5963.9Institute of Neuropathology, University of Freiburg, 79106 Freiburg, Germany ,grid.5963.9Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Christina Helm
- grid.7708.80000 0000 9428 7911Department of Neurology, Medical Center – University of Freiburg, 79106 Freiburg, Germany ,grid.5963.9Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Paolo d´Errico
- grid.7708.80000 0000 9428 7911Department of Neurology, Medical Center – University of Freiburg, 79106 Freiburg, Germany ,grid.5963.9Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Marco Prinz
- grid.5963.9Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany ,grid.5963.9Institute of Neuropathology, University of Freiburg, 79106 Freiburg, Germany ,grid.5963.9Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine University of Freiburg, 79110 Freiburg, Germany ,grid.5963.9Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Marlene Bartos
- grid.5963.9Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany ,grid.5963.9Institute for Physiology I, Systemic and Cellular Neurophysiology, University of Freiburg, 79104 Freiburg, Germany ,grid.5963.9Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine University of Freiburg, 79110 Freiburg, Germany
| | - Melanie Meyer-Luehmann
- Department of Neurology, Medical Center - University of Freiburg, 79106, Freiburg, Germany. .,Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany. .,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine University of Freiburg, 79110, Freiburg, Germany.
| |
Collapse
|
18
|
Nakano H, Hamaguchi T, Ikeda T, Watanabe‐Nakayama T, Ono K, Yamada M. Inactivation of seeding activity of amyloid β‐protein aggregates in vitro. J Neurochem 2021; 160:499-516. [DOI: 10.1111/jnc.15563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/25/2021] [Accepted: 12/14/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Hiroto Nakano
- Department of Neurology and Neurobiology of Aging Kanazawa University Graduate School of Medical Sciences Kanazawa Japan
| | - Tsuyoshi Hamaguchi
- Department of Neurology and Neurobiology of Aging Kanazawa University Graduate School of Medical Sciences Kanazawa Japan
| | - Tokuhei Ikeda
- Department of Neurology and Neurobiology of Aging Kanazawa University Graduate School of Medical Sciences Kanazawa Japan
- Department of Neurology Ishikawa Prefectural Central Hospital Kanazawa Japan
| | - Takahiro Watanabe‐Nakayama
- World Premier International Research Center Initiative (WPI)‐Nano Life Science Institute Kanazawa University Kanazawa Japan
| | - Kenjiro Ono
- Division of Neurology Department of Internal Medicine Showa University Tokyo Japan
| | - Masahito Yamada
- Department of Neurology and Neurobiology of Aging Kanazawa University Graduate School of Medical Sciences Kanazawa Japan
- Department of Internal Medicine Department of Neurology Kudanzaka Hospital Tokyo Japan
| |
Collapse
|
19
|
Kaur S, Verma H, Dhiman M, Tell G, Gigli GL, Janes F, Mantha AK. Brain Exosomes: Friend or Foe in Alzheimer's Disease? Mol Neurobiol 2021; 58:6610-6624. [PMID: 34595669 DOI: 10.1007/s12035-021-02547-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 08/23/2021] [Indexed: 01/18/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. It is known to be a multifactorial disease and several causes are associated with its occurrence as well as progression. However, the accumulation of amyloid beta (Aβ) is widely considered its major pathogenic hallmark. Additionally, neurofibrillary tangles (NFT), mitochondrial dysfunction, oxidative stress, and aging (cellular senescence) are considered as additional hits affecting the disease pathology. Several studies are now suggesting important role of inflammation in AD, which shifts our thought towards the brain's resident immune cells, microglia, and astrocytes; how they interact with neurons; and how these interactions are affected by intra and extracellular stressful factors. These interactions can be modulated by different mechanisms and pathways, in which exosomes could play an important role. Exosomes are multivesicular bodies secreted by nearly all types of cells. The exosomes secreted by glial cells or neurons affect the interactions and thus the physiology of these cells by transmitting miRNAs, proteins, and lipids. Exosomes can serve as a friend or foe to the neuron function, depending upon the carried signals. Exosomes, from the healthy microenvironment, may assist neuron function and health, whereas, from the stressed microenvironment, they carry oxidative and inflammatory signals to the neurons and thus prove detrimental to the neuronal function. Furthermore, exosomes can cross the blood-brain barrier (BBB), and from the blood plasma they can enter the brain cells and activate microglia and astrocytes. Exosomes can transport Aβ or Tau, cytokines, miRNAs between the cells, and alter the physiology of recipient cells. They can also assist in Aβ clearance and regulation of synaptic activity. The exosomes derived from different cells play different roles, and this field is still in its infancy stage. This review advocates exosomes' role as a friend or foe in neurodegenerative diseases, especially in the case of Alzheimer's disease.
Collapse
Affiliation(s)
- Sharanjot Kaur
- Department of Microbiology, School of Biological Sciences , Central University of Punjab, Bathinda, Punjab, India
| | - Harkomal Verma
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Village Ghudda151 401, Punjab, Bathinda, India
| | - Monisha Dhiman
- Department of Microbiology, School of Biological Sciences , Central University of Punjab, Bathinda, Punjab, India
| | - Gianluca Tell
- Department of Medicine, University of Udine, Udine, Italy
| | - Gian Luigi Gigli
- Department of Medicine, University of Udine, Udine, Italy
- Clinical Neurology, Udine University Hospital, Udine, Italy
| | | | - Anil K Mantha
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Village Ghudda151 401, Punjab, Bathinda, India.
| |
Collapse
|
20
|
Friesen M, Ziegler-Waldkirch S, Egenolf M, d'Errico P, Helm C, Mezö C, Dokalis N, Erny D, Katzmarski N, Coelho R, Loreth D, Prinz M, Meyer-Luehmann M. Distinct Aβ pathology in the olfactory bulb and olfactory deficits in a mouse model of Aβ and α-syn co-pathology. Brain Pathol 2021; 32:e13032. [PMID: 34713522 PMCID: PMC9048518 DOI: 10.1111/bpa.13032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 01/05/2023] Open
Abstract
Several degenerative brain disorders such as Alzheimer's disease (AD), Parkinson's disease (PD) and Dementia with Lewy bodies (DLB) are characterized by the simultaneous appearance of amyloid‐β (Aβ) and α‐synuclein (α‐syn) pathologies and symptoms that are similar, making it difficult to differentiate between these diseases. Until now, an accurate diagnosis can only be made by postmortem analysis. Furthermore, the role of α‐syn in Aβ aggregation and the arising characteristic olfactory impairments observed during the progression of these diseases is still not well understood. Therefore, we assessed Aβ load in olfactory bulbs of APP‐transgenic mice expressing APP695KM670/671NL and PSEN1L166P under the control of the neuron‐specific Thy‐1 promoter (referred to here as APPPS1) and APPPS1 mice co‐expressing SNCAA30P (referred to here as APPPS1 × [A30P]aSYN). Furthermore, the olfactory capacity of these mice was evaluated in the buried food and olfactory avoidance test. Our results demonstrate an age‐dependent increase in Aβ load in the olfactory bulb of APP‐transgenic mice that go along with exacerbated olfactory performance. Our study provides clear evidence that the presence of α‐syn significantly diminished the endogenous and seed‐induced Aβ deposits and significantly ameliorated olfactory dysfunction in APPPS1 × [A30P]aSYN mice.
Collapse
Affiliation(s)
- Marina Friesen
- Department of Neurology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Stephanie Ziegler-Waldkirch
- Department of Neurology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Milena Egenolf
- Department of Neurology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Paolo d'Errico
- Department of Neurology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christina Helm
- Department of Neurology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Charlotte Mezö
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany.,Institute of Neuropathology, University of Freiburg, Freiburg, Germany
| | - Nikolaos Dokalis
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany.,Institute of Neuropathology, University of Freiburg, Freiburg, Germany
| | - Daniel Erny
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Institute of Neuropathology, University of Freiburg, Freiburg, Germany.,Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Natalie Katzmarski
- Department of Neurology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Romina Coelho
- Department of Neurology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Departamento de Química e Bioquímica, Universidade de Lisboa, Lisbon, Portugal
| | - Desirée Loreth
- Department of Neurology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marco Prinz
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Institute of Neuropathology, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Melanie Meyer-Luehmann
- Department of Neurology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
21
|
Joshi P, Riffel F, Kumar S, Villacampa N, Theil S, Parhizkar S, Haass C, Colonna M, Heneka MT, Arzberger T, Herms J, Walter J. TREM2 modulates differential deposition of modified and non-modified Aβ species in extracellular plaques and intraneuronal deposits. Acta Neuropathol Commun 2021; 9:168. [PMID: 34663480 PMCID: PMC8522217 DOI: 10.1186/s40478-021-01263-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 12/29/2022] Open
Abstract
Progressive accumulation of Amyloid-β (Aβ) deposits in the brain is a characteristic neuropathological hallmark of Alzheimer’s disease (AD). During disease progression, extracellular Aβ plaques undergo specific changes in their composition by the sequential deposition of different modified Aβ species. Microglia are implicated in the restriction of amyloid deposits and play a major role in internalization and degradation of Aβ. Recent studies showed that rare variants of the Triggering Receptor Expressed on Myeloid cells 2 (TREM2) are associated with an increased risk for AD. Post-translational modifications of Aβ could modulate the interaction with TREM2, and the uptake by microglia. Here, we demonstrate that genetic deletion of TREM2 or expression of a disease associated TREM2 variant in mice lead to differential accumulation of modified and non-modified Aβ species in extracellular plaques and intraneuronal deposits. Human brains with rare TREM2 AD risk variants also showed altered deposition of modified Aβ species in the different brain lesions as compared to cases with the common variant of TREM2. These findings indicate that TREM2 plays a critical role in the development and the composition of Aβ deposits, not only in extracellular plaques, but also intraneuronally, that both could contribute to the pathogenesis of AD.
Collapse
|
22
|
IKK2/NF-κB Activation in Astrocytes Reduces amyloid β Deposition: A Process Associated with Specific Microglia Polarization. Cells 2021; 10:cells10102669. [PMID: 34685649 PMCID: PMC8534251 DOI: 10.3390/cells10102669] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease that is accompanied by pronounced neuroinflammatory responses mainly characterized by marked microgliosis and astrogliosis. However, it remains open as to how different aspects of astrocytic and microglial activation affect disease progression. Previously, we found that microglia expansion in the spinal cord, initiated by IKK2/NF-κB activation in astrocytes, exhibits stage-dependent beneficial effects on the progression of amyotrophic lateral sclerosis. Here, we investigated the impact of NF-κB-initiated neuroinflammation on AD pathogenesis using the APP23 mouse model of AD in combination with conditional activation of IKK2/NF-κB signaling in astrocytes. We show that NF-κB activation in astrocytes triggers a distinct neuroinflammatory response characterized by striking astrogliosis as well as prominent microglial reactivity. Immunohistochemistry and Congo red staining revealed an overall reduction in the size and number of amyloid plaques in the cerebral cortex and hippocampus. Interestingly, isolated primary astrocytes and microglia cells exhibit specific marker gene profiles which, in the case of microglia, point to an enhanced plaque clearance capacity. In contrast, direct IKK2/NF-κB activation in microglia results in a pro-inflammatory polarization program. Our findings suggest that IKK2/NF-κB signaling in astrocytes may activate paracrine mechanisms acting on microglia function but also on APP processing in neurons.
Collapse
|
23
|
Gour N, Gazit E. Metabolite assemblies: A surprising extension to the amyloid hypothesis. Curr Opin Chem Biol 2021; 64:154-164. [PMID: 34482124 DOI: 10.1016/j.cbpa.2021.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/10/2021] [Accepted: 07/25/2021] [Indexed: 12/15/2022]
Abstract
The realization of the ability of metabolites to form self-assembled amyloid-like nanostructures was a surprising phenomenon. This discovery paved the way towards understanding the pathophysiology of the inborn error of metabolism disorders from a new perspective, relating them to amyloid-associated diseases that are characterized by the aggregation of proteins and polypeptides. Hence, a 'generic amyloid hypothesis' can be proposed. This theory implies that the formation of amyloid-like structures is a general phenomenon not limited to proteins and reflects a common etiology for both age-related amyloid-associated diseases and inborn error of metabolism disorders. Here, we present a comprehensive survey of the recent research related to metabolite amyloids including their structure formation through self-association, propagation, interactions, transmission, and their role in metabolic disorders and neurodegenerative diseases and their applications for the fabrication of novel materials which implicate metabolite assemblies as a surprising extension to the amyloid scheme.
Collapse
Affiliation(s)
- Nidhi Gour
- School of Science, Department of Chemistry, Indrashil University, Mehsana, Gujarat, 382740 India
| | - Ehud Gazit
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, 6997801, Israel; BLAVATNIK CENTER for Drug Discovery, Tel Aviv University, Tel Aviv, 6997801, Israel.
| |
Collapse
|
24
|
Willbold D, Strodel B, Schröder GF, Hoyer W, Heise H. Amyloid-type Protein Aggregation and Prion-like Properties of Amyloids. Chem Rev 2021; 121:8285-8307. [PMID: 34137605 DOI: 10.1021/acs.chemrev.1c00196] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This review will focus on the process of amyloid-type protein aggregation. Amyloid fibrils are an important hallmark of protein misfolding diseases and therefore have been investigated for decades. Only recently, however, atomic or near-atomic resolution structures have been elucidated from various in vitro and ex vivo obtained fibrils. In parallel, the process of fibril formation has been studied in vitro under highly artificial but comparatively reproducible conditions. The review starts with a summary of what is known and speculated from artificial in vitro amyloid-type protein aggregation experiments. A partially hypothetic fibril selection model will be described that may be suitable to explain why amyloid fibrils look the way they do, in particular, why at least all so far reported high resolution cryo-electron microscopy obtained fibril structures are in register, parallel, cross-β-sheet fibrils that mostly consist of two protofilaments twisted around each other. An intrinsic feature of the model is the prion-like nature of all amyloid assemblies. Transferring the model from the in vitro point of view to the in vivo situation is not straightforward, highly hypothetic, and leaves many open questions that need to be addressed in the future.
Collapse
Affiliation(s)
- Dieter Willbold
- Institute of Biological Information Processing, Structural Biochemistry, IBI-7, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.,Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany.,Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology (State University), 141700 Dolgoprudny, Russia
| | - Birgit Strodel
- Institute of Biological Information Processing, Structural Biochemistry, IBI-7, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.,Institute of Theoretical and Computational Chemistry, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Gunnar F Schröder
- Institute of Biological Information Processing, Structural Biochemistry, IBI-7, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.,Physics Department, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Wolfgang Hoyer
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Henrike Heise
- Institute of Biological Information Processing, Structural Biochemistry, IBI-7, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.,Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
25
|
Bacterial Extracellular DNA Promotes β-Amyloid Aggregation. Microorganisms 2021; 9:microorganisms9061301. [PMID: 34203755 PMCID: PMC8232312 DOI: 10.3390/microorganisms9061301] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/05/2021] [Accepted: 06/11/2021] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease is associated with prion-like aggregation of the amyloid β (Aβ) peptide and the subsequent accumulation of misfolded neurotoxic aggregates in the brain. Therefore, it is critical to clearly identify the factors that trigger the cascade of Aβ misfolding and aggregation. Numerous studies have pointed out the association between microorganisms and their virulence factors and Alzheimer’s disease; however, their exact mechanisms of action remain unclear. Recently, we discovered a new pathogenic role of bacterial extracellular DNA, triggering the formation of misfolded Tau aggregates. In this study, we investigated the possible role of DNA extracted from different bacterial and eukaryotic cells in triggering Aβ aggregation in vitro. Interestingly, we found that the extracellular DNA of some, but not all, bacteria is an effective trigger of Aβ aggregation. Furthermore, the acceleration of Aβ nucleation and elongation can vary based on the concentration of the bacterial DNA and the bacterial strain from which this DNA had originated. Our findings suggest that bacterial extracellular DNA might play a previously overlooked role in the Aβ protein misfolding associated with Alzheimer’s disease pathogenesis. Moreover, it highlights a new mechanism of how distantly localized bacteria can remotely contribute to protein misfolding and diseases associated with this process. These findings might lead to the use of bacterial DNA as a novel therapeutic target for the prevention and treatment of Alzheimer’s disease.
Collapse
|
26
|
Simons ES, Smith MA, Dengler-Crish CM, Crish SD. Retinal ganglion cell loss and gliosis in the retinofugal projection following intravitreal exposure to amyloid-beta. Neurobiol Dis 2021; 147:105146. [PMID: 33122075 DOI: 10.1016/j.nbd.2020.105146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 10/09/2020] [Accepted: 10/23/2020] [Indexed: 01/07/2023] Open
Abstract
Pathological accumulations of amyloid-beta (Aβ) peptide are found in retina early in Alzheimer's disease, yet its effects on retinal neuronal structure remain unknown. To investigate this, we injected fibrillized Aβ1-42 protein into the eye of adult C57BL/6 J mice and analyzed the retina, optic nerve (ON), and the superior colliculus (SC), the primary retinal target in mice. We found that retinal Aβ exposure stimulated microglial activation and retinal ganglion cell (RGC) loss as early as 1-week post-injection. Pathology was not limited to the retina, but propagated into other areas of the central nervous system. Microgliosis spread throughout the retinal projection (retina, ON, and SC), with multiplex protein quantitation demonstrating an increase in endogenously produced Aβ in the ON and SC corresponding to the injected retinas. Surprisingly, this pathology spread to the opposite side, with unilateral Aβ eye injections driving increased Aβ levels, neuroinflammation, and RGC death in the opposite, un-injected retinal projection. As Aβ-mediated microglial activation has been shown to propagate Aβ pathology, we also investigated the role of the Aβ-binding microglial scavenger receptor CD36 in this pathology. Transgenic mice lacking the CD36 receptor were resistant to Aβ-induced inflammation and RGC death up to 2 weeks following exposure. These results indicate that Aβ pathology drives regional neuropathology in the retina and does not remain isolated to the affected eye, but spreads throughout the nervous system. Further, CD36 may serve as a promising target to prevent Aβ-mediated inflammatory damage.
Collapse
Affiliation(s)
- E S Simons
- Northeast Ohio Medical University, Rootstown, OH 44272, United States; Kent State Biomedical Sciences Graduate Program, Kent, OH 44240, United States
| | - M A Smith
- Northeast Ohio Medical University, Rootstown, OH 44272, United States; Kent State Biomedical Sciences Graduate Program, Kent, OH 44240, United States; Akron Children's Hospital, Rebecca D. Considine Research Institute, Akron, OH 44302, United States
| | - C M Dengler-Crish
- Northeast Ohio Medical University, Rootstown, OH 44272, United States; Kent State Biomedical Sciences Graduate Program, Kent, OH 44240, United States
| | - S D Crish
- Northeast Ohio Medical University, Rootstown, OH 44272, United States; Kent State Biomedical Sciences Graduate Program, Kent, OH 44240, United States.
| |
Collapse
|
27
|
Abstract
Aβ plaques are one of the two lesions in the brain that define the neuropathological diagnosis of Alzheimer's disease. Plaques are highly diverse structures; many of them include massed, fibrillar polymers of the Aβ protein referred to as Aβ-amyloid, but some lack the defining features of amyloid. Cellular elements in 'classical' plaques include abnormal neuronal processes and reactive glial cells, but these are not present in all plaques. Plaques have been given various names since their discovery in 1892, including senile plaques, amyloid plaques, and neuritic plaques. However, with the identification in the 1980s of Aβ as the obligatory and universal component of plaques, the term 'Aβ plaques' has become a unifying term for these heterogeneous formations. Tauopathy, the second essential lesion of the Alzheimer's disease diagnostic dyad, is downstream of Aβ-proteopathy, but it is critically important for the manifestation of dementia. The etiologic link between Aβ-proteopathy and tauopathy in Alzheimer's disease remains largely undefined. Aβ plaques develop and propagate via the misfolding, self-assembly and spread of Aβ by the prion-like mechanism of seeded protein aggregation. Partially overlapping sets of risk factors and sequelae, including inflammation, genetic variations, and various environmental triggers have been linked to plaque development and idiopathic Alzheimer's disease, but no single factor has emerged as a requisite cause. The value of Aβ plaques per se as therapeutic targets is uncertain; although some plaques are sites of focal gliosis and inflammation, the complexity of inflammatory biology presents challenges to glia-directed intervention. Small, soluble, oligomeric assemblies of Aβ are enriched in the vicinity of plaques, and these probably contribute to the toxic impact of Aβ aggregation on the brain. Measures designed to reduce the production or seeded self-assembly of Aβ can impede the formation of Aβ plaques and oligomers, along with their accompanying abnormalities; given the apparent long timecourse of the emergence, maturation and proliferation of Aβ plaques in humans, such therapies are likely to be most effective when begun early in the pathogenic process, before significant damage has been done to the brain. Since their discovery in the late 19th century, Aβ plaques have, time and again, illuminated fundamental mechanisms driving neurodegeneration, and they should remain at the forefront of efforts to understand, and therefore treat, Alzheimer's disease.
Collapse
Affiliation(s)
- Lary C. Walker
- Department of Neurology and Yerkes National Primate Research Center, Emory University
| |
Collapse
|
28
|
Morgan GJ, Wall JS. The Process of Amyloid Formation due to Monoclonal Immunoglobulins. Hematol Oncol Clin North Am 2020; 34:1041-1054. [PMID: 33099422 DOI: 10.1016/j.hoc.2020.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Monoclonal antibodies secreted by clonally expanded plasma cells can form a range of pathologic aggregates including amyloid fibrils. The enormous diversity in the sequences of the involved light chains may be responsible for complexity of the disease. Nevertheless, important common features have been recognized. Two recent high-resolution structures of light chain fibrils show related but distinct conformations. The native structure of the light chains is lost when they are incorporated into the amyloid fibrils. The authors discuss the processes that lead to aggregation and describe how existing and emerging therapies aim to prevent aggregation or remove amyloid fibrils from tissues.
Collapse
Affiliation(s)
- Gareth J Morgan
- Amyloidosis Center and Section of Hematology and Medical Oncology, Department of Medicine, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA.
| | - Jonathan S Wall
- Amyloidosis and Cancer Theranostics Program, Preclinical and Diagnostic Molecular Imaging Laboratory, The University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway, Knoxville, TN 37920, USA
| |
Collapse
|
29
|
Catania M, Di Fede G. One or more β-amyloid(s)? New insights into the prion-like nature of Alzheimer's disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 175:213-237. [PMID: 32958234 DOI: 10.1016/bs.pmbts.2020.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Misfolding and aggregation of proteins play a central role in the pathogenesis of several neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's and Lewy Body diseases, Frontotemporal Lobar Degeneration and prion diseases. Increasing evidence supports the view that Aβ and tau, which are the two main molecular players in AD, share with the prion protein several "prion-like" features that can be relevant for disease pathogenesis. These features essentially include structural/conformational/biochemical variations, resistance to degradation by endogenous proteases, seeding ability, attitude to form neurotoxic assemblies, spreading and propagation of toxic aggregates, transmissibility of tau- and Aβ-related pathology to animal models. Following this view, part of the recent scientific literature has generated a new reading frame for AD pathophysiology, based on the application of the prion paradigm to the amyloid cascade hypothesis in an attempt to definitely explain the key events causing the disease and inducing its occurrence under different clinical phenotypes.
Collapse
Affiliation(s)
- Marcella Catania
- Neurology 5 / Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giuseppe Di Fede
- Neurology 5 / Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| |
Collapse
|
30
|
d'Errico P, Meyer-Luehmann M. Mechanisms of Pathogenic Tau and Aβ Protein Spreading in Alzheimer's Disease. Front Aging Neurosci 2020; 12:265. [PMID: 33061903 PMCID: PMC7481386 DOI: 10.3389/fnagi.2020.00265] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/03/2020] [Indexed: 01/01/2023] Open
Abstract
Alzheimer’s disease (AD) is pathologically defined by extracellular accumulation of amyloid-β (Aβ) peptides generated by the cleavage of amyloid precursor protein (APP), strings of hyperphosphorylated Tau proteins accumulating inside neurons known as neurofibrillary tangles (NFTs) and neuronal loss. The association between the two hallmarks and cognitive decline has been known since the beginning of the 20th century when the first description of the disease was carried out by Alois Alzheimer. Today, more than 40 million people worldwide are affected by AD that represents the most common cause of dementia and there is still no effective treatment available to cure the disease. In general, the aggregation of Aβ is considered an essential trigger in AD pathogenesis that gives rise to NFTs, neuronal dysfunction and dementia. During the process leading to AD, tau and Aβ first misfold and form aggregates in one brain region, from where they spread to interconnected areas of the brain thereby inducing its gradual morphological and functional deterioration. In this mini-review article, we present an overview of the current literature on the spreading mechanisms of Aβ and tau pathology in AD since a more profound understanding is necessary to design therapeutic approaches aimed at preventing or halting disease progression.
Collapse
Affiliation(s)
- Paolo d'Errico
- Department of Neurology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Meyer-Luehmann
- Department of Neurology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|