1
|
Watamura N, Foiani MS, Bez S, Bourdenx M, Santambrogio A, Frodsham C, Camporesi E, Brinkmalm G, Zetterberg H, Patel S, Kamano N, Takahashi M, Rueda-Carrasco J, Katsouri L, Fowler S, Turkes E, Hashimoto S, Sasaguri H, Saito T, Islam AS, Benner S, Endo T, Kobayashi K, Ishida C, Vendruscolo M, Yamada M, Duff KE, Saido TC. In vivo hyperphosphorylation of tau is associated with synaptic loss and behavioral abnormalities in the absence of tau seeds. Nat Neurosci 2024:10.1038/s41593-024-01829-7. [PMID: 39719507 DOI: 10.1038/s41593-024-01829-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 10/23/2024] [Indexed: 12/26/2024]
Abstract
Tau pathology is a hallmark of several neurodegenerative diseases, including frontotemporal dementia and Alzheimer's disease. However, the sequence of events and the form of tau that confers toxicity are still unclear, due in large part to the lack of physiological models of tauopathy initiation and progression in which to test hypotheses. We have developed a series of targeted mice expressing frontotemporal-dementia-causing mutations in the humanized MAPT gene to investigate the earliest stages of tauopathy. MAPTInt10+3G>A and MAPTS305N;Int10+3G>A lines show abundant hyperphosphorylated tau in the hippocampus and entorhinal cortex, but they do not develop seed-competent fibrillar structures. Accumulation of hyperphosphorylated tau was accompanied by neurite degeneration, loss of viable synapses and indicators of behavioral abnormalities. Our results demonstrate that neuronal toxicity can occur in the absence of fibrillar, higher-order structures and that tau hyperphosphorylation is probably involved in the earliest etiological events in tauopathies showing isoform ratio imbalance.
Collapse
Affiliation(s)
- Naoto Watamura
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan.
- UK Dementia Research Institute at University College London, London, UK.
| | - Martha S Foiani
- UK Dementia Research Institute at University College London, London, UK.
| | - Sumi Bez
- UK Dementia Research Institute at University College London, London, UK
| | - Mathieu Bourdenx
- UK Dementia Research Institute at University College London, London, UK
| | - Alessia Santambrogio
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Claire Frodsham
- UK Dementia Research Institute at University College London, London, UK
| | - Elena Camporesi
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology,The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Gunnar Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology,The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- UK Dementia Research Institute at University College London, London, UK
- Queen Square Institute of Neurology, University College London, London, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology,The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Saisha Patel
- UK Dementia Research Institute at University College London, London, UK
| | - Naoko Kamano
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | - Mika Takahashi
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | | | - Loukia Katsouri
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, UK
| | - Stephanie Fowler
- UK Dementia Research Institute at University College London, London, UK
- Nuffield Department of Medicine, Oxford-GSK Institute of Molecular and Computational Medicine, Centre for Human Genetics, Oxford, UK
| | - Emir Turkes
- UK Dementia Research Institute at University College London, London, UK
| | - Shoko Hashimoto
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
- Pioneering Research Division, Medical Innovation Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
- Dementia Pathophysiology Collaboration Unit, RIKEN Center for Brain Science, Wako, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Afm Saiful Islam
- Queen Square Institute of Neurology, University College London, London, UK
| | - Seico Benner
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Japan
| | | | - Katsuji Kobayashi
- Department of Psychiatry, Awazu Neuropsychiatric Hospital, Ishikawa, Japan
| | - Chiho Ishida
- Department of Neurology, NHO Iou National Hospital, Iwade-machi, Japan
| | - Michele Vendruscolo
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Masahito Yamada
- Department of Internal Medicine, Division of Neurology, Kudanzaka Hospital, Tokyo, Japan
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Tokyo, Japan
- Kanazawa University, Kanazawa, Japan
| | - Karen E Duff
- UK Dementia Research Institute at University College London, London, UK.
- Queen Square Institute of Neurology, University College London, London, UK.
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan.
| |
Collapse
|
2
|
Jiang Y, Li W, Ma Y, Hou Y. Neuroinflammation-targeted magnetic resonance imaging nanoprobes for the early diagnosis of Alzheimer's disease. J Mater Chem B 2024. [PMID: 39686760 DOI: 10.1039/d4tb02210f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Alzheimer's disease (AD) that is an important contributor to dementia, is a chronic and irreversible neurodegenerative disease, with high rates of disability and mortality. Recently, more and more therapeutic methods have been developed to delay the progression of AD, but it remains a great challenge to achieve the early diagnosis of AD. In this work, we developed a magnetic resonance imaging (MRI) nanoprobe (NP@angiopep-2/CD137) based on angiopep-2 peptide and CD137 antibody with a NaGdF4 nanoparticle as the core and realized neuroinflammation-targeted imaging on APP/PS1 model mice using a clinical 7.0 T MRI scanner. CD137 expression was upregulated in neuroglial cells and cerebral vascular endothelial cells in inflammatory state. In the APP/PS1 mouse model, after administration, the nanoprobe-enhanced images showed specific dot-like signals in the susceptibility-weighted imaging (SWI) sequence. In summary, we designed and synthesized NP@angiopep-2/CD137 nanoprobes using the activation-dependent expression of CD137, which were applied to the pathological assessment of AD based on the hypothesis of AD neuroinflammation, and provided a reliable idea for the early molecular imaging diagnosis of AD.
Collapse
Affiliation(s)
- Yanjiao Jiang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Wenyue Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Yuqiang Ma
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Yi Hou
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China.
| |
Collapse
|
3
|
Bale LK, West SA, Gades NM, Baker DJ, Conover CA. Gene deletion of Pregnancy-associated Plasma Protein-A (PAPP-A) improves pathology and cognition in an Alzheimer's disease mouse model. Exp Neurol 2024; 382:114976. [PMID: 39349117 PMCID: PMC11502239 DOI: 10.1016/j.expneurol.2024.114976] [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: 04/19/2024] [Revised: 09/13/2024] [Accepted: 09/22/2024] [Indexed: 10/02/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease of age with no effective preventative or treatment approaches. Deeper understanding of the mechanisms underlying the accumulation of toxic β-amyloid oligopeptides and the formation of amyloid plaque in AD has the potential to identify new therapeutic targets. Prior research links the insulin-like growth factor (IGF) system to pathologic mechanisms underlying AD. Suppression of local IGF-I receptor (IGFIR) signaling in AD mice has been shown to reduce plaque formation in the brain and delay neurodegeneration and behavioral changes. However, direct inhibitors of IGFIR signaling are not a viable treatment option for AD due to the essentiality of the IGFIR in physiological growth and metabolism. We have previously demonstrated a more selective means to reduce local IGFIR signaling through inhibition of PAPP-A, a novel zinc metalloprotease that regulates local IGF-I bioavailability through cleavage of inhibitory IGF binding proteins. Here we tested if deletion of PAPP-A in a mouse model of AD provides protection against pathology and behavioral changes. We show that compared to AD mice, AD/PAPP-A KO mice had significantly less plaque burden, reduced astrocytic activation, decreased IGF-IR activity, and improved cognition. Human senile AD plaques showed specific immunostaining for PAPP-A. Thus, inhibition of PAPP-A expression or activity may represent a novel treatment strategy for AD.
Collapse
Affiliation(s)
- Laurie K Bale
- Department of Endocrinology, Mayo Clinic, Rochester, MN 55905, United States of America.
| | - Sally A West
- Department of Endocrinology, Mayo Clinic, Rochester, MN 55905, United States of America.
| | - Naomi M Gades
- Department of Comparative Medicine, Mayo Clinic, Scottsdale, AZ 85259, United States of America.
| | - Darren J Baker
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, United States of America; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, United States of America.
| | - Cheryl A Conover
- Department of Endocrinology, Mayo Clinic, Rochester, MN 55905, United States of America.
| |
Collapse
|
4
|
Rodriguez GA, Rothenberg EF, Shetler CO, Aoun A, Posani L, Vajram SV, Tedesco T, Fusi S, Hussaini SA. Impaired spatial coding and neuronal hyperactivity in the medial entorhinal cortex of aged App NL-G-F mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.26.624990. [PMID: 39651258 PMCID: PMC11623597 DOI: 10.1101/2024.11.26.624990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
The progressive accumulation of amyloid beta (Aβ) pathology in the brain has been associated with aberrant neuronal network activity and poor cognitive performance in preclinical mouse models of Alzheimer's disease (AD). Presently, our understanding of the mechanisms driving pathology-associated neuronal dysfunction and impaired information processing in the brain remains incomplete. Here, we assessed the impact of advanced Aβ pathology on spatial information processing in the medial entorhinal cortex (MEC) of 18-month App NL-G-F/NL- G-F knock-in (APP KI) mice as they explored contextually novel and familiar open field arenas in a two-day, four-session recording paradigm. We tracked single unit firing activity across all sessions and found that spatial information scores were decreased in MEC neurons from APP KI mice versus those in age-matched C57BL/6J controls. MEC single unit spatial representations were also impacted in APP KI mice. Border cell firing preferences were unstable across sessions and spatial periodicity in putative grid cells was disrupted. In contrast, MEC border cells and grid cells in Control mice were intact and stable across sessions. We then quantified the stability of MEC spatial maps across sessions by utilizing a metric based on the Earth Mover's Distance (EMD). We found evidence for increased instability in spatially-tuned APP KI MEC neurons versus Controls when mice were re-exposed to familiar environments and exposed to a novel environment. Additionally, spatial decoding analysis of MEC single units revealed deficits in position and speed coding in APP KI mice in all session comparisons. Finally, MEC single unit analysis revealed a mild hyperactive phenotype in APP KI mice that appeared to be driven by narrow-spiking units (putative interneurons). These findings tie Aβ-associated dysregulation in neuronal firing to disruptions in spatial information processing that may underlie certain cognitive deficits associated with AD.
Collapse
|
5
|
Keeney MT, Rocha EM, Hoffman EK, Farmer K, Di Maio R, Weir J, Wagner WG, Hu X, Clark CL, Castro SL, Scheirer A, Fazzari M, De Miranda BR, Pintchovski SA, Shrader WD, Pagano PJ, Hastings TG, Greenamyre JT. LRRK2 regulates production of reactive oxygen species in cell and animal models of Parkinson's disease. Sci Transl Med 2024; 16:eadl3438. [PMID: 39356746 DOI: 10.1126/scitranslmed.adl3438] [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: 10/13/2023] [Revised: 04/30/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024]
Abstract
Oxidative stress has long been implicated in Parkinson's disease (PD) pathogenesis, although the sources and regulation of reactive oxygen species (ROS) production are poorly defined. Pathogenic mutations in the gene encoding leucine-rich repeat kinase 2 (LRRK2) are associated with increased kinase activity and a greater risk of PD. The substrates and downstream consequences of elevated LRRK2 kinase activity are still being elucidated, but overexpression of mutant LRRK2 has been associated with oxidative stress, and antioxidants reportedly mitigate LRRK2 toxicity. Here, using CRISPR-Cas9 gene-edited HEK293 cells, RAW264.7 macrophages, rat primary ventral midbrain cultures, and PD patient-derived lymphoblastoid cells, we found that elevated LRRK2 kinase activity was associated with increased ROS production and lipid peroxidation and that this was blocked by inhibitors of either LRRK2 kinase or NADPH oxidase 2 (NOX2). Oxidative stress induced by the pesticide rotenone was ameliorated by LRRK2 kinase inhibition and was absent in cells devoid of LRRK2. In a rat model of PD induced by rotenone, a LRRK2 kinase inhibitor prevented the lipid peroxidation and NOX2 activation normally seen in nigral dopaminergic neurons in this model. Mechanistically, LRRK2 kinase activity was shown to regulate phosphorylation of serine-345 in the p47phox subunit of NOX2. This, in turn, led to translocation of p47phox from the cytosol to the membrane-associated gp91phox (NOX2) subunit, activation of the NOX2 enzyme complex, and production of ROS. Thus, LRRK2 kinase activity may drive cellular ROS production in PD through the regulation of NOX2 activity.
Collapse
Affiliation(s)
- Matthew T Keeney
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Emily M Rocha
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eric K Hoffman
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kyle Farmer
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Roberto Di Maio
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Julie Weir
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Weston G Wagner
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xiaoping Hu
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Courtney L Clark
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sandra L Castro
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Abigail Scheirer
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Marco Fazzari
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Briana R De Miranda
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Patrick J Pagano
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Teresa G Hastings
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
6
|
Gnörich J, Koehler M, Wind-Mark K, Klaus C, Zatcepin A, Palumbo G, Lalia M, Monasor LS, Beyer L, Eckenweber F, Scheifele M, Gildehaus FJ, von Ungern-Sternberg B, Barthel H, Sabri O, Bartenstein P, Herms J, Tahirovic S, Franzmeier N, Ziegler S, Brendel M. Towards multicenter β-amyloid PET imaging in mouse models: A triple scanner head-to-head comparison. Neuroimage 2024; 297:120748. [PMID: 39069223 DOI: 10.1016/j.neuroimage.2024.120748] [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: 03/29/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024] Open
Abstract
AIM β-amyloid (Aβ) small animal PET facilitates quantification of fibrillar amyloidosis in Alzheimer's disease (AD) mouse models. Thus, the methodology is receiving growing interest as a monitoring tool in preclinical drug trials. In this regard, harmonization of data from different scanners at multiple sites would allow the establishment large collaborative cohorts and may facilitate efficacy comparison of different treatments. Therefore, we objected to determine the level of agreement of Aβ-PET quantification by a head-to-head comparison of three different state-of-the-art small animal PET scanners, which could help pave the way for future multicenter studies. METHODS Within a timeframe of 5 ± 2 weeks, transgenic APPPS1 (n = 9) and wild-type (WT) (n = 8) mice (age range: 13-16 months) were examined three times by Aβ-PET ([18F]florbetaben) using a Siemens Inveon DPET, a MedisonanoScan PET/MR, and a MedisonanoScan PET/CT with harmonized reconstruction protocols. Cortex-to-white-matter 30-60 min p.i. standardized uptake value ratios (SUVRCTX/WM) were calculated to compare binding differences, effect sizes (Cohen's d) and z-score values of APPPS1 relative to WT mice. Correlation coefficients (Pearson's r) were calculated for the agreement of individual SUVR between different scanners. Voxel-wise analysis was used to determine the agreement of spatial pathology patterns. For validation of PET imaging against the histological gold standard, individual SUVR values were subject to a correlation analysis with area occupancy of methoxy‑X04 staining. RESULTS All three small animal PET scanners yielded comparable group differences between APPPS1 and WT mice (∆PET=20.4 % ± 2.9 %, ∆PET/MR=18.4 % ± 4.5 %, ∆PET/CT=18.1 % ± 3.3 %). Voxel-wise analysis confirmed a high degree of congruency of the spatial pattern (Dice coefficient (DC)PETvs.PET/MR=83.0 %, DCPETvs.PET/CT=69.3 %, DCPET/MRvs.PET/CT=81.9 %). Differences in the group level variance of the three scanners resulted in divergent z-scores (zPET=11.5 ± 1.6; zPET/MR=5.3 ± 1.3; zPET/CT=3.4 ± 0.6) and effect sizes (dPET=8.5, dPET/MR=4.5, dPET/CT=4.1). However, correlations at the individual mouse level were still strong between scanners (rPETvs.PET/MR=0.96, rPETvs.PET/CT=0.91, rPET/MRvs.PET/CT=0.87; all p ≤ 0.0001). Methoxy-X04 staining exhibited a significant correlation across all three PET machines combined (r = 0.76, p < 0.0001) but also at individual level (PET: r = 0.81, p = 0.026; PET/MR: r = 0.89, p = 0.0074; PET/CT: r = 0.93, p = 0.0028). CONCLUSIONS Our comparison of standardized small animal Aβ-PET acquired by three different scanners substantiates the possibility of moving towards a multicentric approach in preclinical AD research. The alignment of image acquisition and analysis methods achieved good overall comparability between data sets. Nevertheless, differences in variance of sensitivity and specificity of different scanners may limit data interpretation at the individual mouse level and deserves methodological optimization.
Collapse
Affiliation(s)
- Johannes Gnörich
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
| | - Mara Koehler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Karin Wind-Mark
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Carolin Klaus
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Artem Zatcepin
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Giovanna Palumbo
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Manvir Lalia
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Laura Sebastian Monasor
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Graduate School of Systemic Neuroscience, LMU Munich, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Maximilian Scheifele
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | | | | | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Center of Neuropathology and Prion Research, University of Munich, Munich Germany
| | - Sabina Tahirovic
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Nicolai Franzmeier
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Institute for Stroke and Dementia Research, LMU Munich, Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Center of Neuropathology and Prion Research, University of Munich, Munich Germany.
| |
Collapse
|
7
|
Watamura N, Kakiya N, Fujioka R, Kamano N, Takahashi M, Nilsson P, Saito T, Iwata N, Fujisawa S, Saido TC. The dopaminergic system promotes neprilysin-mediated degradation of amyloid-β in the brain. Sci Signal 2024; 17:eadk1822. [PMID: 39106321 DOI: 10.1126/scisignal.adk1822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 04/21/2024] [Accepted: 07/01/2024] [Indexed: 08/09/2024]
Abstract
Deposition of amyloid-β (Aβ) in the brain can impair neuronal function and contribute to cognitive decline in Alzheimer's disease (AD). Here, we found that dopamine and the dopamine precursor levodopa (also called l-DOPA) induced Aβ degradation in the brain. Chemogenetic approaches in mice revealed that the activation of dopamine release from ventral tegmental area (VTA) neurons increased the abundance and activity of the Aβ-degrading enzyme neprilysin and reduced the amount of Aβ deposits in the prefrontal cortex in a neprilysin-dependent manner. Aged mice had less dopamine and neprilysin in the anterior cortex, a decrease that was accentuated in AD model mice. Treating AD model mice with levodopa reduced Aβ deposition and improved cognitive function. These observations demonstrate that dopamine promotes brain region-specific, neprilysin-dependent degradation of Aβ, suggesting that dopamine-associated strategies have the potential to treat this aspect of AD pathology.
Collapse
Affiliation(s)
- Naoto Watamura
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Naomasa Kakiya
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Ryo Fujioka
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Naoko Kamano
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Mika Takahashi
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division for Neurogeriatrics, Karolinska Institutet, 171 64, Solna, Sweden
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi 464-8601, Japan
| | - Nobuhisa Iwata
- Department of Genome-based Drug Discovery & Leading Medical Research Core Unit, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8521, Japan
| | - Shigeyoshi Fujisawa
- Laboratory for Systems Neurophysiology, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
8
|
Krut' VG, Kalinichenko AL, Maltsev DI, Jappy D, Shevchenko EK, Podgorny OV, Belousov VV. Optogenetic and chemogenetic approaches for modeling neurological disorders in vivo. Prog Neurobiol 2024; 235:102600. [PMID: 38548126 DOI: 10.1016/j.pneurobio.2024.102600] [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: 08/15/2023] [Revised: 02/26/2024] [Accepted: 03/22/2024] [Indexed: 04/01/2024]
Abstract
Animal models of human neurological disorders provide valuable experimental tools which enable us to study various aspects of disorder pathogeneses, ranging from structural abnormalities and disrupted metabolism and signaling to motor and mental deficits, and allow us to test novel therapies in preclinical studies. To be valid, these animal models should recapitulate complex pathological features at the molecular, cellular, tissue, and behavioral levels as closely as possible to those observed in human subjects. Pathological states resembling known human neurological disorders can be induced in animal species by toxins, genetic factors, lesioning, or exposure to extreme conditions. In recent years, novel animal models recapitulating neuropathologies in humans have been introduced. These animal models are based on synthetic biology approaches: opto- and chemogenetics. In this paper, we review recent opto- and chemogenetics-based animal models of human neurological disorders. These models allow for the creation of pathological states by disrupting specific processes at the cellular level. The artificial pathological states mimic a range of human neurological disorders, such as aging-related dementia, Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, epilepsy, and ataxias. Opto- and chemogenetics provide new opportunities unavailable with other animal models of human neurological disorders. These techniques enable researchers to induce neuropathological states varying in severity and ranging from acute to chronic. We also discuss future directions for the development and application of synthetic biology approaches for modeling neurological disorders.
Collapse
Affiliation(s)
- Viktoriya G Krut'
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia
| | - Andrei L Kalinichenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Dmitry I Maltsev
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - David Jappy
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia
| | - Evgeny K Shevchenko
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia
| | - Oleg V Podgorny
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia.
| | - Vsevolod V Belousov
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia; Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow 143025, Russia.
| |
Collapse
|
9
|
Amartumur S, Nguyen H, Huynh T, Kim TS, Woo RS, Oh E, Kim KK, Lee LP, Heo C. Neuropathogenesis-on-chips for neurodegenerative diseases. Nat Commun 2024; 15:2219. [PMID: 38472255 PMCID: PMC10933492 DOI: 10.1038/s41467-024-46554-8] [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: 10/04/2023] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Developing diagnostics and treatments for neurodegenerative diseases (NDs) is challenging due to multifactorial pathogenesis that progresses gradually. Advanced in vitro systems that recapitulate patient-like pathophysiology are emerging as alternatives to conventional animal-based models. In this review, we explore the interconnected pathogenic features of different types of ND, discuss the general strategy to modelling NDs using a microfluidic chip, and introduce the organoid-on-a-chip as the next advanced relevant model. Lastly, we overview how these models are being applied in academic and industrial drug development. The integration of microfluidic chips, stem cells, and biotechnological devices promises to provide valuable insights for biomedical research and developing diagnostic and therapeutic solutions for NDs.
Collapse
Affiliation(s)
- Sarnai Amartumur
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Huong Nguyen
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Thuy Huynh
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Testaverde S Kim
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, 34824, Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon, 35015, Korea
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Anti-microbial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
| | - Luke P Lee
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Harvard Medical School, Division of Engineering in Medicine and Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA.
| | - Chaejeong Heo
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea.
| |
Collapse
|
10
|
Feng Y, Murphy MC, Hojo E, Li F, Roberts N. Magnetic Resonance Elastography in the Study of Neurodegenerative Diseases. J Magn Reson Imaging 2024; 59:82-96. [PMID: 37084171 DOI: 10.1002/jmri.28747] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/22/2023] Open
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD) present a major health burden to society. Changes in brain structure and cognition are generally only observed at the late stage of the disease. Although advanced magnetic resonance imaging (MRI) techniques such as diffusion imaging may allow identification of biomarkers at earlier stages of neurodegeneration, early diagnosis is still challenging. Magnetic resonance elastography (MRE) is a noninvasive MRI technique for studying the mechanical properties of tissues by measuring the wave propagation induced in the tissues using a purpose-built actuator. Here, we present a systematic review of preclinical and clinical studies in which MRE has been applied to study neurodegenerative diseases. Actuator systems for data acquisition, inversion algorithms for data analysis, and sample demographics are described and tissue stiffness measures obtained for the whole brain and internal structures are summarized. A total of six animal studies and eight human studies have been published. The animal studies refer to 123 experimental animals (68 AD and 55 PD) and 121 wild-type animals, while the human studies refer to 142 patients with neurodegenerative disease (including 56 AD and 17 PD) and 166 controls. The animal studies are consistent in the reporting of decreased stiffness of the hippocampal region in AD mice. However, in terms of disease progression, although consistent decreases in either storage modulus or shear modulus magnitude are reported for whole brain, there is variation in the results reported for the hippocampal region. The clinical studies are consistent in reports of a significant decrease in either whole brain storage modulus or shear modulus magnitude, in both AD and PD and with different brain structures affected in different neurodegenerative diseases. MRE studies of neurodegenerative diseases are still in their infancy, and in future it will be interesting to investigate potential relationships between brain mechanical properties and clinical measures, which may help elucidate the mechanisms underlying onset and progression of neurodegenerative diseases. EVIDENCE LEVEL: 1. TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Yuan Feng
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, China
- National Engineering Research Center of Advanced Magnetic Resonance Technologies for Diagnosis and Therapy (NERC-AMRT), Shanghai Jiao Tong University, Shanghai, China
| | - Matthew C Murphy
- Department of Radiology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Emi Hojo
- Centre for Reproductive Health (CRH), School of Clinical Sciences, University of Edinburgh, Edinburgh, UK
| | - Fei Li
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Neil Roberts
- Centre for Reproductive Health (CRH), School of Clinical Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
11
|
Ramsden CE, Zamora D, Horowitz MS, Jahanipour J, Calzada E, Li X, Keyes GS, Murray HC, Curtis MA, Faull RM, Sedlock A, Maric D. ApoER2-Dab1 disruption as the origin of pTau-associated neurodegeneration in sporadic Alzheimer's disease. Acta Neuropathol Commun 2023; 11:197. [PMID: 38093390 PMCID: PMC10720169 DOI: 10.1186/s40478-023-01693-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/16/2023] [Indexed: 12/17/2023] Open
Abstract
In sporadic Alzheimer's disease (sAD) specific regions, layers and neurons accumulate hyperphosphorylated Tau (pTau) and degenerate early while others remain unaffected even in advanced disease. ApoER2-Dab1 signaling suppresses Tau phosphorylation as part of a four-arm pathway that regulates lipoprotein internalization and the integrity of actin, microtubules, and synapses; however, the role of this pathway in sAD pathogenesis is not fully understood. We previously showed that multiple ApoER2-Dab1 pathway components including ApoE, Reelin, ApoER2, Dab1, pP85αTyr607, pLIMK1Thr508, pTauSer202/Thr205 and pPSD95Thr19 accumulate together within entorhinal-hippocampal terminal zones in sAD, and proposed a unifying hypothesis wherein disruption of this pathway underlies multiple aspects of sAD pathogenesis. However, it is not yet known whether ApoER2-Dab1 disruption can help explain the origin(s) and early progression of pTau pathology in sAD. In the present study, we applied in situ hybridization and immunohistochemistry (IHC) to characterize ApoER2 expression and accumulation of ApoER2-Dab1 pathway components in five regions known to develop early pTau pathology in 64 rapidly autopsied cases spanning the clinicopathological spectrum of sAD. We found that (1) these selectively vulnerable neuron populations strongly express ApoER2; and (2) multiple ApoER2-Dab1 components representing all four arms of this pathway accumulate in abnormal neurons and neuritic plaques in mild cognitive impairment (MCI) and sAD cases and correlate with histological progression and cognitive deficits. Multiplex-IHC revealed that Dab1, pP85αTyr607, pLIMK1Thr508, pTauSer202/Thr205 and pPSD95Thr19 accumulate together within many of the same ApoER2-expressing neurons and in the immediate vicinity of ApoE/ApoJ-enriched extracellular plaques. Collective findings reveal that pTau is only one of many ApoER2-Dab1 pathway components that accumulate in multiple neuroanatomical sites in the earliest stages of sAD and provide support for the concept that ApoER2-Dab1 disruption drives pTau-associated neurodegeneration in human sAD.
Collapse
Affiliation(s)
- Christopher E Ramsden
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH (NIA/NIH), 251 Bayview Blvd., Baltimore, MD, 21224, USA.
- Intramural Program of the National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA.
| | - Daisy Zamora
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH (NIA/NIH), 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Mark S Horowitz
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH (NIA/NIH), 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Jahandar Jahanipour
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892, USA
| | - Elizabeth Calzada
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH (NIA/NIH), 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Xiufeng Li
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH (NIA/NIH), 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Gregory S Keyes
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH (NIA/NIH), 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Helen C Murray
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, Private Bag, Auckland, 92019, New Zealand
- Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892, USA
| | - Maurice A Curtis
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, Private Bag, Auckland, 92019, New Zealand
| | - Richard M Faull
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, Private Bag, Auckland, 92019, New Zealand
| | - Andrea Sedlock
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892, USA
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892, USA
| |
Collapse
|
12
|
Li X, Quan M, Wei Y, Wang W, Xu L, Wang Q, Jia J. Critical thinking of Alzheimer's transgenic mouse model: current research and future perspective. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2711-2754. [PMID: 37480469 DOI: 10.1007/s11427-022-2357-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/23/2023] [Indexed: 07/24/2023]
Abstract
Transgenic models are useful tools for studying the pathogenesis of and drug development for Alzheimer's Disease (AD). AD models are constructed usually using overexpression or knock-in of multiple pathogenic gene mutations from familial AD. Each transgenic model has its unique behavioral and pathological features. This review summarizes the research progress of transgenic mouse models, and their progress in the unique mechanism of amyloid-β oligomers, including the first transgenic mouse model built in China based on a single gene mutation (PSEN1 V97L) found in Chinese familial AD. We further summarized the preclinical findings of drugs using the models, and their future application in exploring the upstream mechanisms and multitarget drug development in AD.
Collapse
Affiliation(s)
- Xinyue Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Meina Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Yiping Wei
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wei Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Lingzhi Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Qi Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China.
- Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, 100053, China.
- Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, 100053, China.
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100053, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China.
| |
Collapse
|
13
|
Maniv I, Sarji M, Bdarneh A, Feldman A, Ankawa R, Koren E, Magid-Gold I, Reis N, Soteriou D, Salomon-Zimri S, Lavy T, Kesselman E, Koifman N, Kurz T, Kleifeld O, Michaelson D, van Leeuwen FW, Verheijen BM, Fuchs Y, Glickman MH. Altered ubiquitin signaling induces Alzheimer's disease-like hallmarks in a three-dimensional human neural cell culture model. Nat Commun 2023; 14:5922. [PMID: 37739965 PMCID: PMC10516951 DOI: 10.1038/s41467-023-41545-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 09/08/2023] [Indexed: 09/24/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by toxic protein accumulation in the brain. Ubiquitination is essential for protein clearance in cells, making altered ubiquitin signaling crucial in AD development. A defective variant, ubiquitin B + 1 (UBB+1), created by a non-hereditary RNA frameshift mutation, is found in all AD patient brains post-mortem. We now detect UBB+1 in human brains during early AD stages. Our study employs a 3D neural culture platform derived from human neural progenitors, demonstrating that UBB+1 alone induces extracellular amyloid-β (Aβ) deposits and insoluble hyperphosphorylated tau aggregates. UBB+1 competes with ubiquitin for binding to the deubiquitinating enzyme UCHL1, leading to elevated levels of amyloid precursor protein (APP), secreted Aβ peptides, and Aβ build-up. Crucially, silencing UBB+1 expression impedes the emergence of AD hallmarks in this model system. Our findings highlight the significance of ubiquitin signalling as a variable contributing to AD pathology and present a nonclinical platform for testing potential therapeutics.
Collapse
Affiliation(s)
- Inbal Maniv
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Mahasen Sarji
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Anwar Bdarneh
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Alona Feldman
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Roi Ankawa
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Elle Koren
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Inbar Magid-Gold
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Noa Reis
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Despina Soteriou
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Shiran Salomon-Zimri
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences, The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Tali Lavy
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Ellina Kesselman
- The Wolfson Department of Chemical Engineering, The Technion Center for Electron Microscopy of Soft Matter, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Naama Koifman
- The Wolfson Department of Chemical Engineering, The Technion Center for Electron Microscopy of Soft Matter, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Thimo Kurz
- School of Molecular Biosciences, University of Glasgow, Glasgow, G12 8QQ, Scotland, UK
| | - Oded Kleifeld
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Daniel Michaelson
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences, The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Fred W van Leeuwen
- Department of Neuroscience, Maastricht University, 6229 ER, Maastricht, the Netherlands
| | - Bert M Verheijen
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
- Department of Neuroscience, Maastricht University, 6229 ER, Maastricht, the Netherlands
| | - Yaron Fuchs
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel.
- Augmanity, Rehovot, 7670308, Israel.
| | - Michael H Glickman
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel.
| |
Collapse
|
14
|
Murayama MA. The past and present of therapeutic strategy for Alzheimer's diseases: potential for stem cell therapy. Exp Anim 2023; 72:285-293. [PMID: 36878603 PMCID: PMC10435354 DOI: 10.1538/expanim.22-0164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disease characterized by cognitive dysfunction and neuropsychiatric symptoms, is the most prevalent form of dementia among the elderly. Amyloid aggregation, tau hyperphosphorylation, and neural cell loss are the main pathological features. Various hypotheses have been proposed to explain the development of AD. Some therapeutic agents have shown clinical benefits in patients with AD; however, many of these agents have failed. The degree of neural cell loss is associated with the severity of AD. Adult neurogenesis, which governs cognitive and emotional behaviors, occurs in the hippocampus, and some research groups have reported that neural cell transplantation into the hippocampus improves cognitive dysfunction in AD model mice. Based on these clinical findings, stem cell therapy for patients with AD has recently attracted attention. This review provides past and present therapeutic strategies for the management and treatment of AD.
Collapse
Affiliation(s)
- Masanori A Murayama
- Department of Animal Models for Human Diseases, Institute of Biomedical Science, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
15
|
Perrotta M, Carnevale D, Carnevale L. Mouse models of cerebral injury and cognitive impairment in hypertension. Front Aging Neurosci 2023; 15:1199612. [PMID: 37539342 PMCID: PMC10394515 DOI: 10.3389/fnagi.2023.1199612] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/05/2023] [Indexed: 08/05/2023] Open
Abstract
Hypertension is a major risk factor for dementia, including both vascular and neurodegenerative etiologies. With the original aim of studying the effect of blood pressure elevation on canonical target organs of hypertension as the heart, the vasculature or the kidneys, several experimental models of hypertension have sprouted during the years. With the more recent interest of understanding the cerebral injury burden caused by hypertension, it is worth understanding how the main models of hypertension or localized cerebral hypertension stand in the field of hypertension-induced cerebral injury and cognitive impairment. With this review we will report main genetic, pharmacological and surgical models of cognitive impairment induced by hypertension, summarizing how each specific category and model can improve our understanding of the complex phenomenon of cognitive loss of vascular etiology.
Collapse
Affiliation(s)
- Marialuisa Perrotta
- Department of Molecular Medicine, “Sapienza” University of Rome, Rome, Italy
- Research Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Department of Angiocardioneurology and Translational Medicine, Pozzilli, Italy
| | - Daniela Carnevale
- Department of Molecular Medicine, “Sapienza” University of Rome, Rome, Italy
- Research Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Department of Angiocardioneurology and Translational Medicine, Pozzilli, Italy
| | - Lorenzo Carnevale
- Research Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Department of Angiocardioneurology and Translational Medicine, Pozzilli, Italy
| |
Collapse
|
16
|
Fertan E, Gendron WH, Wong AA, Hanson GM, Brown RE, Weaver ICG. Noncanonical regulation of imprinted gene Igf2 by amyloid-beta 1-42 in Alzheimer's disease. Sci Rep 2023; 13:2043. [PMID: 36739453 PMCID: PMC9899226 DOI: 10.1038/s41598-023-29248-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Reduced insulin-like growth factor 2 (IGF2) levels in Alzheimer's disease (AD) may be the mechanism relating age-related metabolic disorders to dementia. Since Igf2 is an imprinted gene, we examined age and sex differences in the relationship between amyloid-beta 1-42 (Aβ42) accumulation and epigenetic regulation of the Igf2/H19 gene cluster in cerebrum, liver, and plasma of young and old male and female 5xFAD mice, in frontal cortex of male and female AD and non-AD patients, and in HEK293 cell cultures. We show IGF2 levels, Igf2 expression, histone acetylation, and H19 ICR methylation are lower in females than males. However, elevated Aβ42 levels are associated with Aβ42 binding to Igf2 DMR2, increased DNA and histone methylation, and a reduction in Igf2 expression and IGF2 levels in 5xFAD mice and AD patients, independent of H19 ICR methylation. Cell culture results confirmed the binding of Aβ42 to Igf2 DMR2 increased DNA and histone methylation, and reduced Igf2 expression. These results indicate an age- and sex-related causal relationship among Aβ42 levels, epigenomic state, and Igf2 expression in AD and provide a potential mechanism for Igf2 regulation in normal and pathological conditions, suggesting IGF2 levels may be a useful diagnostic biomarker for Aβ42 targeted AD therapies.
Collapse
Affiliation(s)
- Emre Fertan
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - William H Gendron
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Aimée A Wong
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Gabrielle M Hanson
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada.,Brain Repair Centre, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Ian C G Weaver
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada. .,Department of Psychiatry, Dalhousie University, Halifax, NS, B3H 4R2, Canada. .,Department of Pathology, Dalhousie University, Halifax, NS, B3H 4R2, Canada. .,Brain Repair Centre, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
| |
Collapse
|
17
|
Akhtar A, Gupta SM, Dwivedi S, Kumar D, Shaikh MF, Negi A. Preclinical Models for Alzheimer's Disease: Past, Present, and Future Approaches. ACS OMEGA 2022; 7:47504-47517. [PMID: 36591205 PMCID: PMC9798399 DOI: 10.1021/acsomega.2c05609] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/22/2022] [Indexed: 05/13/2023]
Abstract
A robust preclinical disease model is a primary requirement to understand the underlying mechanisms, signaling pathways, and drug screening for human diseases. Although various preclinical models are available for several diseases, clinical models for Alzheimer's disease (AD) remain underdeveloped and inaccurate. The pathophysiology of AD mainly includes the presence of amyloid plaques and neurofibrillary tangles (NFT). Furthermore, neuroinflammation and free radical generation also contribute to AD. Currently, there is a wide gap in scientific approaches to preventing AD progression. Most of the available drugs are limited to symptomatic relief and improve deteriorating cognitive functions. To mimic the pathogenesis of human AD, animal models like 3XTg-AD and 5XFAD are the primarily used mice models in AD therapeutics. Animal models for AD include intracerebroventricular-streptozotocin (ICV-STZ), amyloid beta-induced, colchicine-induced, etc., focusing on parameters such as cognitive decline and dementia. Unfortunately, the translational rate of the potential drug candidates in clinical trials is poor due to limitations in imitating human AD pathology in animal models. Therefore, the available preclinical models possess a gap in AD modeling. This paper presents an outline that critically assesses the applicability and limitations of the current approaches in disease modeling for AD. Also, we attempted to provide key suggestions for the best-fit model to evaluate potential therapies, which might improve therapy translation from preclinical studies to patients with AD.
Collapse
Affiliation(s)
- Ansab Akhtar
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Shraddha M. Gupta
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Shubham Dwivedi
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Devendra Kumar
- Faculty
of Pharmacy, DIT University, Uttarakhand, Dehradun 248009, India
| | - Mohd. Farooq Shaikh
- Neuropharmacology
Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 47500, Malaysia
| | - Arvind Negi
- Department
of Bioproducts and Biosystems, Aalto University, FI-00076 Espoo, Finland
- E-mail:
| |
Collapse
|