1
|
Farris F, Elhagh A, Vigorito I, Alongi N, Pisati F, Giannattasio M, Casagrande F, Veghini L, Corbo V, Tripodo C, Di Napoli A, Matafora V, Bachi A. Unveiling the mechanistic link between extracellular amyloid fibrils, mechano-signaling and YAP activation in cancer. Cell Death Dis 2024; 15:28. [PMID: 38199984 PMCID: PMC10781709 DOI: 10.1038/s41419-024-06424-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 12/20/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024]
Abstract
The tumor microenvironment is a complex ecosystem that plays a critical role in cancer progression and treatment response. Recently, extracellular amyloid fibrils have emerged as novel components of the tumor microenvironment; however, their function remains elusive. In this study, we establish a direct connection between the presence of amyloid fibrils in the secretome and the activation of YAP, a transcriptional co-activator involved in cancer proliferation and drug resistance. Furthermore, we uncover a shared mechano-signaling mechanism triggered by amyloid fibrils in both melanoma and pancreatic ductal adenocarcinoma cells. Our findings highlight the crucial role of the glycocalyx protein Agrin which binds to extracellular amyloid fibrils and acts as a necessary factor in driving amyloid-dependent YAP activation. Additionally, we reveal the involvement of the HIPPO pathway core kinase LATS1 in this signaling cascade. Finally, we demonstrate that extracellular amyloid fibrils enhance cancer cell migration and invasion. In conclusion, our research expands our knowledge of the tumor microenvironment by uncovering the role of extracellular amyloid fibrils in driving mechano-signaling and YAP activation. This knowledge opens up new avenues for developing innovative strategies to modulate YAP activation and mitigate its detrimental effects during cancer progression.
Collapse
Affiliation(s)
- Francesco Farris
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - Alice Elhagh
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - Ilaria Vigorito
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - Nicoletta Alongi
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Federica Pisati
- Histopathology Unit, Cogentech S.C.a.R.L, 20139, Milan, Italy
| | - Michele Giannattasio
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy
| | - Francesca Casagrande
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Human Technopole, Milan, Italy
| | - Lisa Veghini
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, 37134, Verona, Italy
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, 37134, Verona, Italy
- ARC-Net Centre for Applied Research on Cancer, University of Verona, 37134, Verona, Italy
| | - Claudio Tripodo
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90133, Palermo, Italy
| | - Arianna Di Napoli
- Pathology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea University Hospital, Sapienza University of Rome, 00189, Rome, Italy
| | - Vittoria Matafora
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy.
| | - Angela Bachi
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy.
| |
Collapse
|
2
|
Abstract
Experimental studies of amyloids encounter many challenges. There are many methods available for studying proteins, which can be applied to amyloids: from basic staining techniques, allowing visualization of fibers, to complex methods, e.g., AFM-IR used to their detailed biochemical and structural characterization in nanoscale. Which method is appropriate depends on the goal of an experiment: verification of aggregational properties of a peptide, distinguishing oligomers from mature fibers, or kinetic studies. Insolubility, rapid aggregation, and the need of using a high-purity peptide may be a limiting factor in studies involving amyloids. Moreover, the results obtained by various experimental methods often differ significantly, which may lead to misclassification of amyloid peptides. Due to ambiguity of experimental results, laborious and time-consuming analysis, bioinformatical methods become more widely used for amyloids.
Collapse
Affiliation(s)
| | - Natalia Szulc
- Department of Biomedical Engineering, Wroclaw University of Science and Technology, Wrocław, Poland
| | - Monika Szefczyk
- Department of Bioorganic Chemistry, Wroclaw University of Science and Technology, Wrocław, Poland
| |
Collapse
|
3
|
Kroth H, Oden F, Molette J, Schieferstein H, Gabellieri E, Mueller A, Berndt M, Sreenivasachary N, Serra AM, Capotosti F, Schmitt-Willich H, Hickman D, Pfeifer A, Dinkelborg L, Stephens A. PI-2620 Lead Optimization Highlights the Importance of Off-Target Assays to Develop a PET Tracer for the Detection of Pathological Aggregated Tau in Alzheimer's Disease and Other Tauopathies. J Med Chem 2021; 64:12808-12830. [PMID: 34455780 DOI: 10.1021/acs.jmedchem.1c00861] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The first candidate PI-2014 was tested in healthy controls and subjects with Alzheimer's disease (AD). As PI-2014 displayed off-target binding to monoamine oxidase A (MAO-A), a new lead with improved binding to Tau and decreased MAO-A binding was required. For compound optimization, Tau binding assays based on both human AD brain homogenate and Tau-paired helical filaments were employed. Furthermore, two MAO-A screening assays based on (1) human-recombinant MAO-A and (2) displacement of 2-fluoro-ethyl-harmine from mouse brain homogenate were employed. Removing the N-methyl group from the tricyclic core resulted in compounds displaying improved Tau binding. For the final round of optimization, the cyclic amine substituents were replaced by pyridine derivatives. PI-2620 (2-(2-fluoropyridin-4-yl)-9H-pyrrolo[2,3-b:4,5-c']dipyridine) emerged as a best candidate displaying high Tau binding, low MAO-A binding, high brain uptake, and fast and complete brain washout. Furthermore, PI-2620 showed Tau binding on brain sections from corticobasal degeneration, progressive supranuclear palsy, and Pick's disease.
Collapse
Affiliation(s)
- Heiko Kroth
- AC Immune SA, EPFL Innovation Park, Building B, 1015 Lausanne, Switzerland
| | - Felix Oden
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353 Berlin, Germany
| | - Jerome Molette
- AC Immune SA, EPFL Innovation Park, Building B, 1015 Lausanne, Switzerland
| | | | | | - Andre Mueller
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353 Berlin, Germany
| | - Mathias Berndt
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353 Berlin, Germany
| | | | | | | | | | - David Hickman
- AC Immune SA, EPFL Innovation Park, Building B, 1015 Lausanne, Switzerland
| | - Andrea Pfeifer
- AC Immune SA, EPFL Innovation Park, Building B, 1015 Lausanne, Switzerland
| | - Ludger Dinkelborg
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353 Berlin, Germany
| | - Andrew Stephens
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353 Berlin, Germany
| |
Collapse
|
4
|
Patten KT, Valenzuela AE, Wallis C, Berg EL, Silverman JL, Bein KJ, Wexler AS, Lein PJ. The Effects of Chronic Exposure to Ambient Traffic-Related Air Pollution on Alzheimer's Disease Phenotypes in Wildtype and Genetically Predisposed Male and Female Rats. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:57005. [PMID: 33971107 PMCID: PMC8110309 DOI: 10.1289/ehp8905] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
BACKGROUND Epidemiological data link traffic-related air pollution (TRAP) to increased risk of Alzheimer's disease (AD). Preclinical data corroborating this association are largely from studies of male animals exposed acutely or subchronically to high levels of isolated fractions of TRAP. What remains unclear is whether chronic exposure to ambient TRAP modifies AD risk and the influence of sex on this interaction. OBJECTIVES This study sought to assess effects of chronic exposure to ambient TRAP on the time to onset and severity of AD phenotypes in a preclinical model and to determine whether sex or genetic susceptibility influences outcomes. METHODS Male and female TgF344-AD rats that express human AD risk genes and wildtype littermates were housed in a vivarium adjacent to a heavily trafficked tunnel in Northern California and exposed for up to 14 months to filtered air (FA) or TRAP drawn from the tunnel and delivered to animals unchanged in real time. Refractive particles in the brain and AD phenotypes were quantified in 3-, 6-, 10-, and 15-month-old animals using hyperspectral imaging, behavioral testing, and neuropathologic measures. RESULTS Particulate matter (PM) concentrations in TRAP exposure chambers fluctuated with traffic flow but remained below 24-h PM with aerodynamic diameter less than or equal to 2.5 micrometers (PM2.5) U.S. National Ambient Air Quality Standards limits. Ultrafine PM was a predominant component of TRAP. Nano-sized refractive particles were detected in the hippocampus of TRAP animals. TRAP-exposed animals had more amyloid plaque deposition, higher hyperphosphorylated tau levels, more neuronal cell loss, and greater cognitive deficits in an age-, genotype-, and sex-dependent manner. TRAP-exposed animals also had more microglial cell activation, but not astrogliosis. DISCUSSION These data demonstrate that chronic exposure to ambient TRAP promoted AD phenotypes in wildtype and genetically susceptible rats. TRAP effects varied according to age, sex, and genotype, suggesting that AD progression depends on complex interactions between environment and genetics. These findings suggest current PM2.5 regulations are insufficient to protect the aging brain. https://doi.org/10.1289/EHP8905.
Collapse
Affiliation(s)
- Kelley T. Patten
- Department of Molecular Biosciences, University of California Davis (UC Davis) School of Veterinary Medicine, Davis, California, USA
| | - Anthony E. Valenzuela
- Department of Molecular Biosciences, University of California Davis (UC Davis) School of Veterinary Medicine, Davis, California, USA
| | | | - Elizabeth L. Berg
- Department of Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacramento, California, USA
| | - Jill L. Silverman
- Department of Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacramento, California, USA
- The MIND Institute, UC Davis School of Medicine, Sacramento, California, USA
| | - Keith J. Bein
- Air Quality Research Center, UC Davis, Davis, California, USA
- Center for Health and the Environment, UC Davis, Davis, California, USA
| | - Anthony S. Wexler
- Air Quality Research Center, UC Davis, Davis, California, USA
- Mechanical and Aerospace Engineering, Civil and Environmental Engineering, and Land, Air and Water Resources, UC Davis, Davis, California, USA
| | - Pamela J. Lein
- Department of Molecular Biosciences, University of California Davis (UC Davis) School of Veterinary Medicine, Davis, California, USA
- The MIND Institute, UC Davis School of Medicine, Sacramento, California, USA
| |
Collapse
|
5
|
Takeda K, Uda A, Mitsubori M, Nagashima S, Iwasaki H, Ito N, Shiiba I, Ishido S, Matsuoka M, Inatome R, Yanagi S. Mitochondrial ubiquitin ligase alleviates Alzheimer's disease pathology via blocking the toxic amyloid-β oligomer generation. Commun Biol 2021; 4:192. [PMID: 33580194 PMCID: PMC7881000 DOI: 10.1038/s42003-021-01720-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 11/23/2020] [Indexed: 12/28/2022] Open
Abstract
Mitochondrial pathophysiology is implicated in the development of Alzheimer's disease (AD). An integrative database of gene dysregulation suggests that the mitochondrial ubiquitin ligase MITOL/MARCH5, a fine-tuner of mitochondrial dynamics and functions, is downregulated in patients with AD. Here, we report that the perturbation of mitochondrial dynamics by MITOL deletion triggers mitochondrial impairments and exacerbates cognitive decline in a mouse model with AD-related Aβ pathology. Notably, MITOL deletion in the brain enhanced the seeding effect of Aβ fibrils, but not the spontaneous formation of Aβ fibrils and plaques, leading to excessive secondary generation of toxic and dispersible Aβ oligomers. Consistent with this, MITOL-deficient mice with Aβ etiology exhibited worsening cognitive decline depending on Aβ oligomers rather than Aβ plaques themselves. Our findings suggest that alteration in mitochondrial morphology might be a key factor in AD due to directing the production of Aβ form, oligomers or plaques, responsible for disease development.
Collapse
Affiliation(s)
- Keisuke Takeda
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
- Department of Biology, University of Padova, Padova, Italy
- Venetian Institute of Molecular Medicine, Padova, Italy
| | - Aoi Uda
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Mikihiro Mitsubori
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Shun Nagashima
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Hiroko Iwasaki
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Naoki Ito
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo, Japan
| | - Isshin Shiiba
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo, Japan
| | - Satoshi Ishido
- Department of Microbiology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Masaaki Matsuoka
- Department of Pharmacology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Ryoko Inatome
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo, Japan
| | - Shigeru Yanagi
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan.
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo, Japan.
| |
Collapse
|
6
|
Shin J, Park S, Lee H, Kim Y. Thioflavin-positive tau aggregates complicating quantification of amyloid plaques in the brain of 5XFAD transgenic mouse model. Sci Rep 2021; 11:1617. [PMID: 33452414 PMCID: PMC7810901 DOI: 10.1038/s41598-021-81304-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 01/06/2021] [Indexed: 12/19/2022] Open
Abstract
Transgenic mouse models recapitulating Alzheimer's disease (AD) pathology are pivotal in molecular studies and drug evaluation. In transgenic models selectively expressing amyloid-β (Aβ), thioflavin S (ThS), a fluorescent dye with β-sheet binding properties, is widely employed to observe amyloid plaque accumulation. In this study, we investigated the possibility that a commonly used Aβ-expressing AD model mouse, 5XFAD, generates ThS-positive aggregates of β-sheet structures in addition to Aβ fibrils. To test this hypothesis, brain sections of male and female 5XFAD mice were double-stained with ThS and monoclonal antibodies against Aβ, tau, or α-synuclein, all of which aggregates are detected by ThS. Our results revealed that, in addition to amyloid plaques, 5XFAD mice express ThS-positive phospho-tau (p-tau) aggregates. Upon administration of a small molecule that exclusively disaggregates Aβ to 5XFAD mice for six weeks, we found that the reduction level of plaques was smaller in brain sections stained by ThS compared to an anti-Aβ antibody. Our findings implicate that the use of ThS complicates the quantification of amyloid plaques and the assessment of Aβ-targeting drugs in 5XFAD mice.
Collapse
Affiliation(s)
- Jisu Shin
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Sohui Park
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - HeeYang Lee
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - YoungSoo Kim
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea.
| |
Collapse
|
7
|
Shamir DB, Deng Y, Wu Q, Modak S, Congdon EE, Sigurdsson EM. Dynamics of Internalization and Intracellular Interaction of Tau Antibodies and Human Pathological Tau Protein in a Human Neuron-Like Model. Front Neurol 2020; 11:602292. [PMID: 33324339 PMCID: PMC7727311 DOI: 10.3389/fneur.2020.602292] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
We and others have shown in various in vivo, ex vivo and cell culture models that several tau antibodies interact with pathological tau within neurons. To further clarify this interaction in a dynamic human model, we differentiated SH-SY5Y cells with retinoic acid and BDNF to create a neuron-like model. Therein, tau antibodies were primarily taken up by receptor-mediated endocytosis, and prevented toxicity of human brain-derived paired helical filament-enriched tau (PHF). Subsequently, we monitored in real-time the interaction of antibodies and PHF within endocytic cellular compartments. Cells were pre-treated with fluorescently-tagged PHF and then incubated with tau antibodies, 4E6, 6B2, or non-specific isotype control IgG1 labeled with a pH sensitive dye. The uptake and binding of the efficacious antibody, 4E6, to PHF occurred mainly within the soma, whereas the ineffective antibody, 6B2, and ineffective control IgG1, were visualized via the processes and showed limited colocalization with PHF within this period. In summary, we have developed a neuron-like model that clarifies the early intracellular dynamics of the interaction of tau antibodies with pathological tau, and identifies features associated with efficacy. Since the model is entirely human, it is suitable to verify the therapeutic potential of humanized antibodies prior to extensive clinical trials.
Collapse
Affiliation(s)
- Dov B Shamir
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
| | - Yan Deng
- Microscopy Core, New York University Grossman School of Medicine, New York, NY, United States
| | - Qian Wu
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
| | - Swananda Modak
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
| | - Erin E Congdon
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States.,Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
8
|
Gabellieri E, Capotosti F, Molette J, Sreenivasachary N, Mueller A, Berndt M, Schieferstein H, Juergens T, Varisco Y, Oden F, Schmitt-Willich H, Hickman D, Dinkelborg L, Stephens A, Pfeifer A, Kroth H. Discovery of 2-(4-(2-fluoroethoxy)piperidin-1-yl)-9-methyl-9H-pyrrolo[2,3-b:4,5-c']dipyridine ([18F]PI-2014) as PET tracer for the detection of pathological aggregated tau in Alzheimer's disease and other tauopathies. Eur J Med Chem 2020; 204:112615. [PMID: 32771872 DOI: 10.1016/j.ejmech.2020.112615] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/04/2020] [Accepted: 06/21/2020] [Indexed: 12/29/2022]
Abstract
The compound screening was initiated with a direct staining assay to identify compounds binding to Tau aggregates and not Abeta plaques using human brain sections derived from late stage Alzheimer's disease donors. The binding of Tau aggregate selective compounds was then quantitatively assessed with human brain derived paired helical filaments utilizing the label-free Back Scattering Interferometry assay. In vivo biodistribution experiments of selected fluorine-18 labeled compounds were performed in mice to assess brain uptake, brain washout, and defluorination. Compound 11 emerged as the most promising candidate, displaying high in vitro binding affinity and selectivity to neurofibrillary tangles. Fluorine-18 labeled compound 11 showed high brain uptake and rapid washout from the mouse brain with no observed bone uptake. Furthermore, compound 11 was able to detect Tau aggregates in tauopathy brain sections from corticobasal degeneration, progressive supranuclear palsy, and Pick's disease donors. Thus, 2-(4-(2-fluoroethoxy)piperidin-1-yl)-9-methyl-9H-pyrrolo[2,3-b:4,5-c']dipyridine (PI-2014, compound 11) was selected for characterization in a first-in-human study.
Collapse
Affiliation(s)
| | | | - Jerome Molette
- AC Immune SA, EPFL Innovation Park, Building B, 1015, Lausanne, Switzerland
| | | | - Andre Mueller
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353, Berlin, Germany
| | - Mathias Berndt
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353, Berlin, Germany
| | - Hanno Schieferstein
- Formerly Piramal Imaging GmbH, Tegeler Strasse 6-7, 13353, Berlin, Germany; Merck KGaA, Frankfurter Strasse 250, 64293, Darmstadt, Germany
| | - Tanja Juergens
- AC Immune SA, EPFL Innovation Park, Building B, 1015, Lausanne, Switzerland
| | - Yvan Varisco
- AC Immune SA, EPFL Innovation Park, Building B, 1015, Lausanne, Switzerland
| | - Felix Oden
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353, Berlin, Germany
| | | | - David Hickman
- AC Immune SA, EPFL Innovation Park, Building B, 1015, Lausanne, Switzerland
| | - Ludger Dinkelborg
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353, Berlin, Germany
| | - Andrew Stephens
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353, Berlin, Germany
| | - Andrea Pfeifer
- AC Immune SA, EPFL Innovation Park, Building B, 1015, Lausanne, Switzerland
| | - Heiko Kroth
- AC Immune SA, EPFL Innovation Park, Building B, 1015, Lausanne, Switzerland.
| |
Collapse
|
9
|
Subchronic administration of auranofin reduced amyloid-β plaque pathology in a transgenic APP NL-G-F/NL-G-F mouse model. Brain Res 2020; 1746:147022. [PMID: 32707043 DOI: 10.1016/j.brainres.2020.147022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/13/2020] [Accepted: 07/18/2020] [Indexed: 01/26/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Neuropathological processes, including the accumulation of amyloid-β (Aβ) plaques and neurofibrillary tangles, and neuroinflammation, lead to cognitive impairment at middle and eventually later stages of AD progression. Over the last decade, focused efforts have explored repurposed drug approaches for AD pathophysiological mechanisms. Recently, auranofin, an anti-inflammatory drug, was shown to have therapeutic potential in a number of diseases in addition to rheumatoid arthritis. Surprisingly, no data regarding the effects of auranofin on cognitive deficits in AD mice or the influence of auranofin on Aβ pathology and neuroinflammatory processes are available. In the present study, we used 14-month-old transgenic male APPNL-G-F/NL-G-F mice to assess the effects of subchronic administration of auranofin at low doses (1 and 5 mg/kg, intraperitoneal) on spatial memory, Aβ pathology and the expression of cortical and hippocampal proteins (glial fibrillary acidic protein (GFAP), ionized calcium binding adaptor molecule-1 (Iba-1)) and proteins related to synaptic plasticity (glutamic acid decarboxylase 67 (GAD67), homer proteins homologue-1 (Homer-1)). The data demonstrated that auranofin significantly decreased Aβ deposition in the hippocampus and the number of Aβ plaques in the cingulate cortex, but it did not have memory-enhancing effects or induce changes in the expression of the studied proteins. Our current results highlight the importance of considering further pre-clinical research to investigate the possible beneficial effects of auranofin on the other pathological aspects of AD.
Collapse
|
10
|
Sahoo BR, Cox SJ, Ramamoorthy A. High-resolution probing of early events in amyloid-β aggregation related to Alzheimer's disease. Chem Commun (Camb) 2020; 56:4627-4639. [PMID: 32300761 PMCID: PMC7254607 DOI: 10.1039/d0cc01551b] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In Alzheimer's disease (AD), soluble oligomers of amyloid-β (Aβ) are emerging as a crucial entity in driving disease progression as compared to insoluble amyloid deposits. The lacuna in establishing the structure to function relationship for Aβ oligomers prevents the development of an effective treatment for AD. While the transient and heterogeneous properties of Aβ oligomers impose many challenges for structural investigation, an effective use of a combination of NMR techniques has successfully identified and characterized them at atomic-resolution. Here, we review the successful utilization of solution and solid-state NMR techniques to probe the aggregation and structures of small and large oligomers of Aβ. Biophysical studies utilizing the commonly used solution and 19F based NMR experiments to identify the formation of small size early intermediates and to obtain their structures, and dock-lock mechanism of fiber growth at atomic-resolution are discussed. In addition, the use of proton-detected magic angle spinning (MAS) solid-state NMR experiments to obtain high-resolution insights into the aggregation pathways and structures of large oligomers and other aggregates is also presented. We expect these NMR based studies to be valuable for real-time monitoring of the depletion of monomers and the formation of toxic oligomers and high-order aggregates under a variety of conditions, and to solve the high-resolution structures of small and large size oligomers for most amyloid proteins, and therefore to develop inhibitors and drugs.
Collapse
Affiliation(s)
- Bikash R Sahoo
- Biophysics Program, Department of Chemistry, University of Michigan, Ann Arbor, MI 48109-1055, USA.
| | | | | |
Collapse
|
11
|
Yan T, Liang J, Gao J, Wang L, Fujioka H, Zhu X, Wang X. FAM222A encodes a protein which accumulates in plaques in Alzheimer's disease. Nat Commun 2020; 11:411. [PMID: 31964863 PMCID: PMC6972869 DOI: 10.1038/s41467-019-13962-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/10/2019] [Indexed: 01/05/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by amyloid plaques and progressive cerebral atrophy. Here, we report FAM222A as a putative brain atrophy susceptibility gene. Our cross-phenotype association analysis of imaging genetics indicates a potential link between FAM222A and AD-related regional brain atrophy. The protein encoded by FAM222A is predominantly expressed in the CNS and is increased in brains of patients with AD and in an AD mouse model. It accumulates within amyloid deposits, physically interacts with amyloid-β (Aβ) via its N-terminal Aβ binding domain, and facilitates Aβ aggregation. Intracerebroventricular infusion or forced expression of this protein exacerbates neuroinflammation and cognitive dysfunction in an AD mouse model whereas ablation of this protein suppresses the formation of amyloid deposits, neuroinflammation and cognitive deficits in the AD mouse model. Our data support the pathological relevance of protein encoded by FAM222A in AD.
Collapse
Affiliation(s)
- Tingxiang Yan
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Jingjing Liang
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA.
| | - Ju Gao
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Luwen Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Hisashi Fujioka
- Electron Microscopy Core Facility, Case Western Reserve University, Cleveland, OH, USA
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA.
| | - Xinglong Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
12
|
Pedrero-Prieto CM, Flores-Cuadrado A, Saiz-Sánchez D, Úbeda-Bañón I, Frontiñán-Rubio J, Alcaín FJ, Mateos-Hernández L, de la Fuente J, Durán-Prado M, Villar M, Martínez-Marcos A, Peinado JR. Human amyloid-β enriched extracts: evaluation of in vitro and in vivo internalization and molecular characterization. Alzheimers Res Ther 2019; 11:56. [PMID: 31253170 PMCID: PMC6599264 DOI: 10.1186/s13195-019-0513-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/12/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Intracerebral inoculation of extracts from post-mortem human Alzheimer's disease brains into mice produces a prion-like spreading effect of amyloid-β. The differences observed between these extracts and the synthetic peptide, in terms of amyloid-β internalization and seed and cell-to-cell transmission of cytosolic protein aggregates, suggest that brain extracts contain key contributors that enhance the prion-like effect of amyloid-β. Nevertheless, these potential partners are still unknown due to the complexity of whole brain extracts. METHODS Herein, we established a method based on sequential detergent solubilization of post-mortem samples of human brains affected by Alzheimer's disease that strongly enrich amyloid-β aggregates by eliminating 92% of the remaining proteins. Internalization of Aβ1-42 from the enriched AD extracts was evaluated in vitro, and internalization of fluorescent-labeled AD extracts was also investigated in vivo. Furthermore, we carried out a molecular characterization of the Aβ-enriched fraction using label-free proteomics, studying the distribution of representative components in the amygdala and the olfactory cortex of additional human AD brain samples by immunohistochemistry. RESULTS Aβ1-42 from the enriched AD extracts are internalized into endothelial cells in vitro after 48 h. Furthermore, accumulation of fluorescent-labeled Aβ-enriched extracts into mouse microglia was observed in vivo after 4 months of intracerebral inoculation. Label-free proteomics (FDR < 0.01) characterization of the amyloid-β-enriched fraction from different post-mortem samples allowed for the identification of more than 130 proteins, several of which were significantly overrepresented (i.e., ANXA5 and HIST1H2BK; p < 0.05) and underrepresented (i.e., COL6A or FN1; p < 0.05) in the samples with Alzheimer's disease. We were also able to identify proteins exclusively observed in Alzheimer's disease (i.e., RNF213) or only detected in samples not affected by the disease (i.e., CNTN1) after the enrichment process. Immunohistochemistry against these proteins in additional tissues revealed their particular distribution in the amygdala and the olfactory cortex in relation to the amyloid-β plaque. CONCLUSIONS Identification and characterization of the unique features of these extracts, in terms of amyloid-β enrichment, identification of the components, in vitro and in vivo cell internalization, and tissue distribution, constitute the best initial tool to further investigate the seeding and transmissibility proposed in the prion-like hypothesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Cristina M. Pedrero-Prieto
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Alicia Flores-Cuadrado
- Department of Medical Sciences, Ciudad Real Medical School; Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Daniel Saiz-Sánchez
- Department of Medical Sciences, Ciudad Real Medical School; Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Isabel Úbeda-Bañón
- Department of Medical Sciences, Ciudad Real Medical School; Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Javier Frontiñán-Rubio
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Francisco J. Alcaín
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Lourdes Mateos-Hernández
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC, CSIC-UCLM-JCCM, Ciudad Real, Spain
- UMR BIPAR, INRA, ANSES, Ecole Nationale Vétérinaire d’Alfort, Université Paris-Est, 94700 Maisons-Alfort, France
| | - José de la Fuente
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC, CSIC-UCLM-JCCM, Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK USA
| | - Mario Durán-Prado
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Margarita Villar
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC, CSIC-UCLM-JCCM, Ciudad Real, Spain
| | - Alino Martínez-Marcos
- Department of Medical Sciences, Ciudad Real Medical School; Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Juan R. Peinado
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| |
Collapse
|
13
|
Jamieson-Lucy A, Mullins MC. Isolation of Zebrafish Balbiani Bodies for Proteomic Analysis. Methods Mol Biol 2019; 1920:295-302. [PMID: 30737698 DOI: 10.1007/978-1-4939-9009-2_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Proteomic characterization of isolated organelles can provide insight into the functional components of the structure and novel targets for further testing. Germplasm in developing oocytes is difficult to isolate for protein identification because not all types of germplasm are stable outside of the cytoplasm. In zebrafish, the Balbiani body forms a proteinaceous aggregate that contains the germplasm and we found is stable outside of the oocyte. Here we present a manual isolation protocol that collects intact Balbiani bodies from stage I zebrafish oocytes. We lysed oocytes by passing them through a syringe, and then used a fine injection needle to wick up Balbiani bodies by capillary action with minimal buffer solution. Using this protocol we collected sufficient material for proteomic analysis of the zebrafish Balbiani body.
Collapse
Affiliation(s)
- Allison Jamieson-Lucy
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mary C Mullins
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
14
|
Modeling amyloid beta and tau pathology in human cerebral organoids. Mol Psychiatry 2018; 23:2363-2374. [PMID: 30171212 PMCID: PMC6594704 DOI: 10.1038/s41380-018-0229-8] [Citation(s) in RCA: 234] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 04/24/2018] [Accepted: 06/05/2018] [Indexed: 12/15/2022]
Abstract
The typical abnormalities observed in the brain of Alzheimer's disease (AD) patients include synaptic alterations, neuronal death, brain inflammation, and the accumulation of protein aggregates in the form of amyloid plaques and neurofibrillary tangles. Despite the development of many animal and in vitro models for AD, there is a lack of an experimental approach that fully recapitulates essential aspects of the disease in human cells. Here, we report the generation of a new model to study AD, consisting of cerebral organoids (COs) produced from human-induced pluripotent stem cells (iPSCs). Under our experimental conditions, COs grow to form three-dimensional (3D) structures containing neural areas with cortical-like organization. Analysis of COs by histological and biochemical methods revealed that organoids produced from iPSCs derived from patients affected by familial AD or Down syndrome (DS) spontaneously develop over time pathological features of AD, including accumulation of structures highly reminiscent to amyloid plaques and neurofibrillary tangles. These pathological abnormalities were not observed in COs generated from various controls, including human iPSCs from healthy individuals, human iPSCs from patients affected by Creutzfeldt-Jakob disease, mouse embryonic stem cells (ESCs), or mouse iPSCs. These findings enable modeling genetic AD in a human cellular context in a 3D cortical-like tissue developed in vitro from patient-specific stem cells. This system provides a more relevant disease model compared to pre-existing methods and offers a new platform for discovery of novel targets and screening of drugs for therapeutic intervention.
Collapse
|
15
|
Koss DJ, Dubini M, Buchanan H, Hull C, Platt B. Distinctive temporal profiles of detergent-soluble and -insoluble tau and Aβ species in human Alzheimer's disease. Brain Res 2018; 1699:121-134. [PMID: 30102892 DOI: 10.1016/j.brainres.2018.08.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/13/2018] [Accepted: 08/09/2018] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) pathology relevant proteins tau and beta-amyloid (Aβ) exist as an array of post-translationally modified and conformationally altered species with varying abundance, solubility and toxicity. Insoluble neurofibrillary tau tangles and Aβ plaques are end-stage AD hallmarks, yet may carry less disease significance compared to soluble species. At present, it is unclear how soluble and insoluble tau and Aβ relate to each other as well as to disease progression. Here, detergent soluble and insoluble fractions generated from post-mortem human temporal lobe samples (Brodmann area 21) were probed for tau and Aβ markers in immuno-dot assays. Measures were quantified according to diagnosis (AD cf. Non-AD), neuropathological severity, and correlated with disease progression (Braak stages). All markers were elevated within AD cases cf. non-AD controls (p < 0.05) independent of solubility. However, when considered according to neuropathological severity, phospho-tau (detected via CP13 and AT8 antibodies) was elevated early within the soluble fraction (p < 0.05 intermediate cf. low severity) and emerged only later within the insoluble fraction (p < 0.05 high cf. low severity). In contrast, PHF1 phospho-tau, TOC1 reactive tau oligomers and amyloid markers rose within the two fractions simultaneously. Independent of solubility, cognitive correlations were observed for tau makers and for fibrillary amyloid (OC), however only soluble total Aβ was significantly correlated with intellectual impairment. Following the exclusion of end-stage cases, only soluble total Aβ remained correlated with cognition. The data indicate differential rates of protein aggregation during AD progression and confirm the disease relevance of early emerging soluble Aβ species.
Collapse
Affiliation(s)
- David J Koss
- School of Medicine, Medical Sciences & Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| | - Marina Dubini
- School of Medicine, Medical Sciences & Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Heather Buchanan
- School of Medicine, Medical Sciences & Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Claire Hull
- School of Medicine, Medical Sciences & Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Bettina Platt
- School of Medicine, Medical Sciences & Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
16
|
ApoE isoforms and carboxyl-terminal-truncated apoE4 forms affect neuronal BACE1 levels and Aβ production independently of their cholesterol efflux capacity. Biochem J 2018; 475:1839-1859. [DOI: 10.1042/bcj20180068] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/25/2018] [Accepted: 05/02/2018] [Indexed: 01/14/2023]
Abstract
The β-site amyloid precursor protein-cleaving enzyme 1 (BACE1) initiates the production of amyloid-β peptide (Aβ), which is central to the pathogenesis of Alzheimer's disease (AD). Changes in brain cholesterol homeostasis have been suggested to affect Aβ metabolism. Cholesterol homeostasis is maintained in the brain by apolipoprotein E (apoE). The apoE4 isoform constitutes the major risk factor for AD. Here, we investigated the effect of apoE forms on Aβ generation and on BACE1 levels. We also examined the potential involvement in these processes of cholesterol transporters ABCG1 and ABCG4 or the lipoprotein receptor SR-BI, which are implicated in cholesterol efflux to apoE. It was found that reconstituted lipoprotein-associated apoE isoforms promoted the increase of Aβ production and oligomerization and of BACE1 levels in human neuroblastoma SK-N-SH cells, with an apoE4 ≥ apoE3 > apoE2 potency rank order. Progressive carboxyl-terminal apoE4 deletions between residues 230–299 decreased the protein's ability to increase BACE1, while further truncations up to residue 166 prevented apoE4 from increasing BACE1 and Aβ levels in SK-N-SH and primary mouse neuronal cells. ABCG1, but not ABCG4 or SR-BI, moderately increased Aβ production and BACE1 levels in SK-N-SH cells. All apoE forms affected Aβ production/oligomerization and BACE1 levels in a pattern that did not follow that of their capacity to promote ABCG1, ABCG4 or SR-BI-mediated cholesterol efflux. Overall, our data indicate that apoE-containing lipoprotein particles can have a direct effect on BACE1 levels and Aβ secretion and possibly contribute to AD pathogenetic processes, independently of their capacity to promote cholesterol efflux.
Collapse
|
17
|
Marwarha G, Schommer J, Lund J, Schommer T, Ghribi O. Palmitate-induced C/EBP homologous protein activation leads to NF-κB-mediated increase in BACE1 activity and amyloid beta genesis. J Neurochem 2018; 144:761-779. [PMID: 29315574 PMCID: PMC6371812 DOI: 10.1111/jnc.14292] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/29/2017] [Accepted: 12/23/2017] [Indexed: 12/12/2022]
Abstract
The etiology of Alzheimer's disease (AD) is egregiously comprehended, but epidemiological studies have posited that diets rich in the saturated fatty acid palmitic acid (palmitate) are a significant risk factor. The production and accumulation of amyloid beta peptide (Aβ) is considered the core pathological molecular event in the pathogenesis of AD. The rate-limiting step in Aβ genesis from amyloid-β precursor protein (AβPP) is catalyzed by the enzyme β-site amyloid precursor protein cleaving enzyme 1 (BACE1), the expression and enzymatic activity of which is significantly up-regulated in the AD brain. In this study, we determined the molecular mechanisms that potentially underlie the palmitate-induced up-regulation in BACE1 expression and augmented Aβ production. We demonstrate that a palmitate-enriched diet and exogenous palmitate treatment evoke an increase in BACE1 expression and activity leading to enhanced Aβ genesis in the mouse brain and SH-SY5Y-APPSwe cells, respectively, through the activation of the transcription factor NF-κB. Chromatin immunoprecipitation (ChIP) assays and luciferase reporter assays revealed that palmitate enhances BACE1 expression by increasing the binding of NF-κB in the BACE1 promoter followed by an enhancement in the transactivation of the BACE1 promoter. Elucidation and delineation of upstream molecular events unveiled a critical role of the endoplasmic reticulum stress-associated transcription factor, C/EBP homologous protein (CHOP) in the palmitate-induced NF-κB activation, as CHOP knock-down cells and Chop-/- mice do not exhibit the same degree of NF-κB activation in response to the palmitate challenge. Our study delineates a novel CHOP-NF-κB signaling pathway that mediates palmitate-induced up-regulation of BACE1 expression and Aβ genesis.
Collapse
Affiliation(s)
- Gurdeep Marwarha
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Jared Schommer
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Jonah Lund
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Trevor Schommer
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Othman Ghribi
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203
| |
Collapse
|
18
|
Shamir DB, Deng Y, Sigurdsson EM. Live Imaging of Pathological Tau Protein and Tau Antibodies in a Neuron-Like Cellular Model. Methods Mol Biol 2018; 1779:371-379. [PMID: 29886544 DOI: 10.1007/978-1-4939-7816-8_22] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Several tau antibody therapies are now in clinical trials and numerous other tau antibodies are in various stages of preclinical development to treat Alzheimer's disease and related tauopathies. This involves long-term studies in mouse models that are necessary but time consuming and typically provide only a limited mechanistic understanding of how the antibodies work and why some are not effective. Live cellular imaging with fluorescently tagged pathological tau proteins and tau antibodies provides a valuable insight into their dynamic interaction outside or within the cell. Furthermore, this acute technique may have predictive validity to assess the potential efficacy of different tau antibodies in neutralizing and/or clearing tau aggregates, and can likely be applied to other amyloid diseases. Overall, it should facilitate identifying candidate antibodies for more detailed long-term validation. Due to the human origin of the model, it may be particularly useful to characterize humanized antibodies that utilize receptor-mediated uptake to reach their intracellular target.
Collapse
Affiliation(s)
- Dov B Shamir
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Yan Deng
- Microscopy Core, New York University School of Medicine, New York, NY, USA
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA.
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
19
|
Abstract
Amyloid β (Aβ) is the major constituent of the brain deposits found in parenchymal plaques and cerebral blood vessels of patients with Alzheimer's disease (AD). Besides classic full-length peptides, biochemical analyses of brain deposits have revealed high degree of Aβ heterogeneity likely resulting from the action of multiple proteolytic enzymes. This chapter describes a sequential extraction protocol allowing the differential fractionation of soluble and deposited Aβ species taking advantage of their differential solubility properties. Soluble Aβ is extracted by water-based buffers like phosphate-buffered saline-PBS-whereas pre-fibrillar and fibrillar deposits, usually poorly soluble in PBS, are extractable in detergent containing solutions or more stringent conditions as formic acid. The extraction procedure is followed by the biochemical identification of the extracted Aβ species using Western blot and a targeted proteomic analysis which combines immunoprecipitation with MALDI-ToF mass spectrometry. This approach revealed the presence of numerous C- and N-terminal truncated Aβ species in addition to Aβ1-40/42. Notably, the more soluble C-terminal cleaved fragments constitute a main part of PBS homogenates. On the contrary, N-terminal truncated species typically require more stringent conditions for the extraction in agreement with their lower solubility and enhanced aggregability. Detailed assessment of the molecular diversity of Aβ species composing interstitial fluid and amyloid deposits at different disease stages, as well as the evaluation of the truncation profile during various pharmacologic approaches will provide a comprehensive understanding of the still undefined contribution of Aβ truncations to AD pathogenesis and their potential as novel therapeutic targets.
Collapse
Affiliation(s)
- Agueda Rostagno
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Thomas A Neubert
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY, USA
| | - Jorge Ghiso
- Department of Pathology, New York University School of Medicine, New York, NY, USA.
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
20
|
Lomont JP, Rich KL, Maj M, Ho JJ, Ostrander JS, Zanni MT. Spectroscopic Signature for Stable β-Amyloid Fibrils versus β-Sheet-Rich Oligomers. J Phys Chem B 2017; 122:144-153. [PMID: 29220175 DOI: 10.1021/acs.jpcb.7b10765] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
We use two-dimensional IR (2D IR) spectroscopy to explore fibril formation for the two predominant isoforms of the β-amyloid (Aβ1-40 and Aβ1-42) protein associated with Alzheimer's disease. Two-dimensional IR spectra resolve a transition at 1610 cm-1 in Aβ fibrils that does not appear in other Aβ aggregates, even those with predominantly β-sheet-structure-like oligomers. This transition is not resolved in linear IR spectroscopy because it lies under the broad band centered at 1625 cm-1, which is the traditional infrared signature for amyloid fibrils. The feature is prominent in 2D IR spectra because 2D lineshapes are narrower and scale nonlinearly with transition dipole strengths. Transmission electron microscopy measurements demonstrate that the 1610 cm-1 band is a positive identification of amyloid fibrils. Sodium dodecyl sulfate micelles that solubilize and disaggregate preaggregated Aβ samples deplete the 1625 cm-1 band but do not affect the 1610 cm-1 band, demonstrating that the 1610 cm-1 band is due to very stable fibrils. We demonstrate that the 1610 cm-1 transition arises from amide I modes by mutating out the only side-chain residue that could give rise to this transition, and we explore the potential structural origins of the transition by simulating 2D IR spectra based on Aβ crystal structures. It was not previously possible to distinguish stable Aβ fibrils from the less stable β-sheet-rich oligomers with infrared light. This 2D IR signature will be useful for Alzheimer's research on Aβ aggregation, fibril formation, and toxicity.
Collapse
Affiliation(s)
- Justin P Lomont
- Department of Chemistry, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States
| | - Kacie L Rich
- Department of Chemistry, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States
| | - Michał Maj
- Department of Chemistry, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States
| | - Jia-Jung Ho
- Department of Chemistry, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States
| | - Joshua S Ostrander
- Department of Chemistry, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States
| | - Martin T Zanni
- Department of Chemistry, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States
| |
Collapse
|
21
|
Microglia-derived ASC specks cross-seed amyloid-β in Alzheimer’s disease. Nature 2017; 552:355-361. [DOI: 10.1038/nature25158] [Citation(s) in RCA: 470] [Impact Index Per Article: 67.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 11/22/2017] [Indexed: 12/28/2022]
|
22
|
Marwarha G, Rostad S, Lilek J, Kleinjan M, Schommer J, Ghribi O. Palmitate Increases β-site AβPP-Cleavage Enzyme 1 Activity and Amyloid-β Genesis by Evoking Endoplasmic Reticulum Stress and Subsequent C/EBP Homologous Protein Activation. J Alzheimers Dis 2017; 57:907-925. [PMID: 28304295 PMCID: PMC5389045 DOI: 10.3233/jad-161130] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Epidemiological studies implicate diets rich in saturated free fatty acids (sFFA) as a potential risk factor for developing Alzheimer's disease (AD). In particular, high plasma levels of the sFFA palmitic acid (palmitate) were shown to inversely correlate with cognitive function. However, the cellular mechanisms by which sFFA may increase the risk for AD are not well known. Endoplasmic reticulum (ER) stress has emerged as one of the signaling pathways initiating and fostering the neurodegenerative changes in AD by increasing the aspartyl protease β-site AβPP cleaving enzyme 1 (BACE1) and amyloid-β (Aβ) genesis. In this study, we determined the extent to which palmitate increases BACE1 and Aβ levels in vitro and in vivo as well as the potential role of ER stress as cellular mechanism underlying palmitate effects. We demonstrate, in palmitate-treated SH-SY5Y neuroblastoma cells and in the hippocampi of palmitate-enriched diet-fed mice, that palmitate evokes the activation of the C/EBP Homologous Protein (CHOP), a transcription factor that is specifically responsive to ER stress. Induction of CHOP expression is associated with increased BACE1 mRNA, protein and activity levels, and subsequent enhanced amyloidogenic processing of amyloid-β protein precursor (AβPP) that culminates in a substantial increase in Aβ genesis. We further show that CHOP is an indispensable molecular mediator of palmitate-induced upregulation in BACE1 activity and Aβ genesis. Indeed, we show that Chop-/- mice and CHOP knocked-down SH-SY5Y neuroblastoma cells do not exhibit the same commensurate degree of palmitate-induced increase in BACE1 expression levels and Aβ genesis.
Collapse
Affiliation(s)
| | | | | | | | | | - Othman Ghribi
- Correspondence to: Dr. Othman Ghribi, Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, North Dakota 58202, USA. Tel.: +1 701 777 2522; Fax: +1 701 777 4490; E-mail:
| |
Collapse
|
23
|
Aβ truncated species: Implications for brain clearance mechanisms and amyloid plaque deposition. Biochim Biophys Acta Mol Basis Dis 2017; 1864:208-225. [PMID: 28711595 DOI: 10.1016/j.bbadis.2017.07.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/24/2017] [Accepted: 07/11/2017] [Indexed: 12/14/2022]
Abstract
Extensive parenchymal and vascular Aβ deposits are pathological hallmarks of Alzheimer's disease (AD). Besides classic full-length peptides, biochemical analyses of brain deposits have revealed high degree of Aβ heterogeneity likely resulting from the action of multiple proteolytic enzymes. In spite of the numerous studies focusing in Aβ, the relevance of N- and C-terminal truncated species for AD pathogenesis remains largely understudied. In the present work, using novel antibodies specifically recognizing Aβ species N-terminally truncated at position 4 or C-terminally truncated at position 34, we provide a clear assessment of the differential topographic localization of these species in AD brains and transgenic models. Based on their distinct solubility, brain N- and C-terminal truncated species were extracted by differential fractionation and identified via immunoprecipitation coupled to mass spectrometry analysis. Biochemical/biophysical studies with synthetic homologues further confirmed the different solubility properties and contrasting fibrillogenic characteristics of the truncated species composing the brain Aβ peptidome. Aβ C-terminal degradation leads to the production of more soluble fragments likely to be more easily eliminated from the brain. On the contrary, N-terminal truncation at position 4 favors the formation of poorly soluble, aggregation prone peptides with high amyloidogenic propensity and the potential to exacerbate the fibrillar deposits, self-perpetuating the amyloidogenic loop. Detailed assessment of the molecular diversity of Aβ species composing interstitial fluid and amyloid deposits at different disease stages, as well as the evaluation of the truncation profile during various pharmacologic approaches will provide a comprehensive understanding of the still undefined contribution of Aβ truncations to the disease pathogenesis and their potential as novel therapeutic targets.
Collapse
|
24
|
Drew SC. The Case for Abandoning Therapeutic Chelation of Copper Ions in Alzheimer's Disease. Front Neurosci 2017; 11:317. [PMID: 28626387 PMCID: PMC5455140 DOI: 10.3389/fnins.2017.00317] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 05/18/2017] [Indexed: 12/26/2022] Open
Abstract
The "therapeutic chelation" approach to treating Alzheimer's disease (AD) evolved from the metals hypothesis, with the premise that small molecules can be designed to prevent transition metal-induced amyloid deposition and oxidative stress within the AD brain. Over more than 20 years, countless in vitro studies have been devoted to characterizing metal binding, its effect on Aβ aggregation, ROS production, and in vitro toxicity. Despite a lack of evidence for any clinical benefit, the conjecture that therapeutic chelation is an effective approach for treating AD remains widespread. Here, the author plays the devil's advocate, questioning the experimental evidence, the dogma, and the value of therapeutic chelation, with a major focus on copper ions.
Collapse
Affiliation(s)
- Simon C. Drew
- Department of Medicine, Royal Melbourne Hospital, University of MelbourneMelbourne, VIC, Australia
| |
Collapse
|
25
|
Zhang TO, Alperstein AM, Zanni MT. Amyloid β-Sheet Secondary Structure Identified in UV-Induced Cataracts of Porcine Lenses using 2D IR Spectroscopy. J Mol Biol 2017; 429:1705-1721. [PMID: 28454743 PMCID: PMC5493149 DOI: 10.1016/j.jmb.2017.04.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 04/08/2017] [Accepted: 04/24/2017] [Indexed: 02/04/2023]
Abstract
Cataracts are formed by the aggregation of crystallin proteins in the eye lens. Many in vitro studies have established that crystallin proteins precipitate into aggregates that contain amyloid fibers when denatured, but there is little evidence that ex vivo cataracts contain amyloid. In this study, we collect two-dimensional infrared (2D IR) spectra on tissue slices of porcine eye lenses. As shown in control experiments on in vitro αB- and γD-crystallin, 2D IR spectroscopy can identify the highly ordered β-sheets typical of amyloid secondary structure even if the fibers themselves are too short to be resolved with TEM. In ex vivo experiments of acid-treated tissues, characteristic 2D IR features are observed and fibers >50nm in length are resolved by transmission electron microscopy (TEM), consistent with amyloid fibers. In UV-irradiated lens tissues, fibers are not observed with TEM, but highly ordered β-sheets of amyloid secondary structure is identified from the 2D IR spectra. The characteristic 2D IR features of amyloid β-sheet secondary structure are created by as few as four or five strands and so identify amyloid secondary structure even if the aggregates themselves are too small to be resolved with TEM. We discuss these findings in the context of the chaperone system of the lens, which we hypothesize sequesters small aggregates, thereby preventing long fibers from forming. This study expands the scope of heterodyned 2D IR spectroscopy to tissues. The results provide a link between in vitro and ex vivo studies and support the hypothesis that cataracts are an amyloid disease.
Collapse
Affiliation(s)
- Tianqi O Zhang
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI 53706, USA.
| | - Ariel M Alperstein
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI 53706, USA.
| | - Martin T Zanni
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI 53706, USA.
| |
Collapse
|
26
|
The chaperonin CCT inhibits assembly of α-synuclein amyloid fibrils by a specific, conformation-dependent interaction. Sci Rep 2017; 7:40859. [PMID: 28102321 PMCID: PMC5244355 DOI: 10.1038/srep40859] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 12/12/2016] [Indexed: 12/11/2022] Open
Abstract
The eukaryotic chaperonin CCT (chaperonin containing TCP-1) uses cavities built into its double-ring structure to encapsulate and to assist folding of a large subset of proteins. CCT can inhibit amyloid fibre assembly and toxicity of the polyQ extended mutant of huntingtin, the protein responsible for Huntington’s disease. This raises the possibility that CCT modulates other amyloidopathies, a still-unaddressed question. We show here that CCT inhibits amyloid fibre assembly of α-synuclein A53T, one of the mutants responsible for Parkinson’s disease. We evaluated fibrillation blockade in α-synuclein A53T deletion mutants and CCT interactions of full-length A53T in distinct oligomeric states to define an inhibition mechanism specific for α-synuclein. CCT interferes with fibre assembly by interaction of its CCTζ and CCTγ subunits with the A53T central hydrophobic region (NAC). This interaction is specific to NAC conformation, as it is produced once soluble α-synuclein A53T oligomers form and blocks the reaction before fibres begin to grow. Finally, we show that this association inhibits α-synuclein A53T oligomer toxicity in neuroblastoma cells. In summary, our results and those for huntingtin suggest that CCT is a general modulator of amyloidogenesis via a specific mechanism.
Collapse
|
27
|
High-resolution analytical imaging and electron holography of magnetite particles in amyloid cores of Alzheimer's disease. Sci Rep 2016; 6:24873. [PMID: 27121137 PMCID: PMC4848473 DOI: 10.1038/srep24873] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 03/31/2016] [Indexed: 12/20/2022] Open
Abstract
Abnormal accumulation of brain metals is a key feature of Alzheimer’s disease (AD). Formation of amyloid-β plaque cores (APC) is related to interactions with biometals, especially Fe, Cu and Zn, but their particular structural associations and roles remain unclear. Using an integrative set of advanced transmission electron microscopy (TEM) techniques, including spherical aberration-corrected scanning transmission electron microscopy (Cs-STEM), nano-beam electron diffraction, electron holography and analytical spectroscopy techniques (EDX and EELS), we demonstrate that Fe in APC is present as iron oxide (Fe3O4) magnetite nanoparticles. Here we show that Fe was accumulated primarily as nanostructured particles within APC, whereas Cu and Zn were distributed through the amyloid fibers. Remarkably, these highly organized crystalline magnetite nanostructures directly bound into fibrillar Aβ showed characteristic superparamagnetic responses with saturated magnetization with circular contours, as observed for the first time by off-axis electron holography of nanometer scale particles.
Collapse
|
28
|
Hernandez-Guillamon M, Mawhirt S, Blais S, Montaner J, Neubert TA, Rostagno A, Ghiso J. Sequential Amyloid-β Degradation by the Matrix Metalloproteases MMP-2 and MMP-9. J Biol Chem 2015; 290:15078-91. [PMID: 25897080 DOI: 10.1074/jbc.m114.610931] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Indexed: 01/11/2023] Open
Abstract
Matrix metalloproteases (MMPs) MMP-2 and MMP-9 have been implicated in the physiological catabolism of Alzheimer's amyloid-β (Aβ). Conversely, their association with vascular amyloid deposits, blood-brain barrier disruption, and hemorrhagic transformations after ischemic stroke also highlights their involvement in pathological processes. To better understand this dichotomy, recombinant human (rh) MMP-2 and MMP-9 were incubated with Aβ40 and Aβ42, and the resulting proteolytic fragments were assessed via immunoprecipitation and quantitative mass spectrometry. Both MMPs generated Aβ fragments truncated only at the C terminus, ending at positions 34, 30, and 16. Using deuterated homologues as internal standards, we observed limited and relatively slow degradation of Aβ42 by rhMMP-2, although the enzyme cleaved >80% of Aβ40 during the 1st h of incubation. rhMMP-9 was significantly less effective, particularly in degrading Aβ(1-42), although the targeted peptide bonds were identical. Using Aβ(1-34) and Aβ(1-30), we demonstrated that these peptides are also substrates for both MMPs, cleaving Aβ(1-34) to produce Aβ(1-30) first and Aβ(1-16) subsequently. Consistent with the kinetics observed with full-length Aβ, rhMMP-9 degraded only a minute fraction of Aβ(1-34) and was even less effective in producing Aβ(1-16). Further degradation of Aβ(1-16) by either MMP-2 or MMP-9 was not observed even after prolonged incubation times. Notably, all MMP-generated C-terminally truncated Aβ fragments were highly soluble and did not exhibit fibrillogenic properties or induce cytotoxicity in human cerebral microvascular endothelial or neuronal cells supporting the notion that these truncated Aβ species are associated with clearance mechanisms rather than being key elements in the fibrillogenesis process.
Collapse
Affiliation(s)
- Mar Hernandez-Guillamon
- From the Departments of Pathology, the Neurovascular Research Laboratory, Institut de Recerca, 08035 Barcelona, Spain
| | | | - Steven Blais
- Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York 10016, the Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, New York 10016, and
| | - Joan Montaner
- the Neurovascular Research Laboratory, Institut de Recerca, 08035 Barcelona, Spain, the Neurovascular Unit, Neurology and Medicine Departments, Universitat Autònoma de Barcelona, Vall d'Hebron Hospital, 08035 Barcelona, Spain
| | - Thomas A Neubert
- Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York 10016, the Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, New York 10016, and
| | | | - Jorge Ghiso
- From the Departments of Pathology, Psychiatry, and
| |
Collapse
|
29
|
Jimenez S, Navarro V, Moyano J, Sanchez-Mico M, Torres M, Davila JC, Vizuete M, Gutierrez A, Vitorica J. Disruption of amyloid plaques integrity affects the soluble oligomers content from Alzheimer disease brains. PLoS One 2014; 9:e114041. [PMID: 25485545 PMCID: PMC4259387 DOI: 10.1371/journal.pone.0114041] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 11/03/2014] [Indexed: 11/18/2022] Open
Abstract
The implication of soluble Abeta in the Alzheimer’s disease (AD) pathology is currently accepted. In fact, the content of soluble extracellular Abeta species, such as monomeric and/or oligomeric Abeta, seems to correlate with the clinico-pathological dysfunction observed in AD patients. However, the nature (monomeric, dimeric or other oligomers), the relative abundance, and the origin (extra-/intraneuronal or plaque-associated), of these soluble species are actually under debate. In this work we have characterized the soluble (defined as soluble in Tris-buffered saline after ultracentrifugation) Abeta, obtained from hippocampal samples of Braak II, Braak III–IV and Braak V–VI patients. Although the content of both Abeta40 and Abeta42 peptides displayed significant increase with pathology progression, our results demonstrated the presence of low, pg/µg protein, amount of both peptides. This low content could explain the absence (or below detection limits) of soluble Abeta peptides detected by western blots or by immunoprecipitation-western blot analysis. These data were in clear contrast to those published recently by different groups. Aiming to explain the reasons that determine these substantial differences, we also investigated whether the initial homogenization could mobilize Abeta from plaques, using 12-month-old PS1xAPP cortical samples. Our data demonstrated that manual homogenization (using Dounce) preserved the integrity of Abeta plaques whereas strong homogenization procedures (such as sonication) produced a vast redistribution of the Abeta species in all soluble and insoluble fractions. This artifact could explain the dissimilar and somehow controversial data between different groups analyzing human AD samples.
Collapse
Affiliation(s)
- Sebastian Jimenez
- Departamento Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Victoria Navarro
- Departamento Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Javier Moyano
- Departamento Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - María Sanchez-Mico
- Departamento Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Manuel Torres
- Departamento Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Jose Carlos Davila
- Departamento Biología Celular, Genética y Fisiología, Facultad de Ciencias, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Marisa Vizuete
- Departamento Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Antonia Gutierrez
- Departamento Biología Celular, Genética y Fisiología, Facultad de Ciencias, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- * E-mail: (JV); (AG)
| | - Javier Vitorica
- Departamento Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- * E-mail: (JV); (AG)
| |
Collapse
|
30
|
Alzheimer brain-derived tau oligomers propagate pathology from endogenous tau. Sci Rep 2012; 2:700. [PMID: 23050084 PMCID: PMC3463004 DOI: 10.1038/srep00700] [Citation(s) in RCA: 358] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 09/05/2012] [Indexed: 11/12/2022] Open
Abstract
Intracerebral injection of brain extracts containing amyloid or tau aggregates in transgenic animals can induce cerebral amyloidosis and tau pathology. We extracted pure populations of tau oligomers directly from the cerebral cortex of Alzheimer disease (AD) brain. These oligomers are potent inhibitors of long term potentiation (LTP) in hippocampal brain slices and disrupt memory in wild type mice. We observed for the first time that these authentic brain-derived tau oligomers propagate abnormal tau conformation of endogenous murine tau after prolonged incubation. The conformation and hydrophobicity of tau oligomers play a critical role in the initiation and spread of tau pathology in the naïve host in a manner reminiscent of sporadic AD.
Collapse
|
31
|
Lasagna-Reeves CA, Castillo-Carranza DL, Sengupta U, Sarmiento J, Troncoso J, Jackson GR, Kayed R. Identification of oligomers at early stages of tau aggregation in Alzheimer's disease. FASEB J 2012; 26:1946-59. [PMID: 22253473 DOI: 10.1096/fj.11-199851] [Citation(s) in RCA: 367] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Neurofibrillary tangles (NFTs) are a pathological hallmark of Alzheimer's disease (AD); however, the relationship between NFTs and disease progression remains controversial. Analyses of tau animal models suggest that phenotypes coincide with accumulation of soluble aggregated tau species but not the accumulation of NFTs. The pathological role of prefilamentous tau aggregates, e.g., tau oligomeric intermediates, is poorly understood, in part because of methodological challenges. Here, we engineered a novel tau oligomer-specific antibody, T22, and used it to elucidate the temporal course and biochemical features of oligomers during NFT development in AD brain. We found that tau oligomers in human AD brain samples were 4-fold higher than those in the controls. We also revealed the role of oligomeric tau conformers in pretangles, neuritic plaques, and neuropil threads in the frontal cortex tissue from AD brains; this analysis uncovers a consistent code that governs tau oligomerization with regard to degree of neuronal cytopathology. These data are the first to characterize the role of tau oligomers in the natural history of NFTs, and they highlight the suitability of tau oligomers as therapeutic targets in AD and related tauopathies.
Collapse
Affiliation(s)
- Cristian A Lasagna-Reeves
- University of Texas Medical Branch, 301 University Blvd., Medical Research Building, Room 10.138C, Galveston, TX, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Tomidokoro Y, Rostagno A, Neubert TA, Lu Y, Rebeck GW, Frangione B, Greenberg SM, Ghiso J. Iowa variant of familial Alzheimer's disease: accumulation of posttranslationally modified AbetaD23N in parenchymal and cerebrovascular amyloid deposits. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:1841-54. [PMID: 20228223 PMCID: PMC2843474 DOI: 10.2353/ajpath.2010.090636] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/04/2009] [Indexed: 12/30/2022]
Abstract
Mutations within the amyloid-beta (Abeta) sequence, especially those clustered at residues 21-23, which are linked to early onset familial Alzheimer's disease (AD), are primarily associated with cerebral amyloid angiopathy (CAA). The basis for this predominant vascular amyloid burden and the differential clinical phenotypes of cerebral hemorrhage/stroke in some patients and dementia in others remain unknown. The AbetaD23N Iowa mutation is associated with progressive AD-like dementia, often without clinically manifested intracerebral hemorrhage. Neuropathologically, the disease is characterized by predominant preamyloid deposits, severe CAA, and abundant neurofibrillary tangles in the presence of remarkably few mature plaques. Biochemical analyses using a combination of immunoprecipitation, mass spectrometry, amino acid sequence, and Western blot analysis performed after sequential tissue extractions to separately isolate soluble components, preamyloid, and fibrillar amyloid species indicated that the Iowa deposits are complex mixtures of mutated and nonmutated Abeta molecules. These molecules exhibited various degrees of solubility, were highly heterogeneous at both the N- and C-termini, and showed partial aspartate isomerization at positions 1, 7, and 23. This collection of Abeta species-the Iowa brain Abeta peptidome-contained clear imprints of amyloid clearance mechanisms yet highlighted the unique neuropathological features shared by a non-Abeta cerebral amyloidosis, familial Danish dementia, in which neurofibrillary tangles coexist with extensive pre-amyloid deposition in the virtual absence of fibrillar lesions. These data therefore challenge the importance of neuritic plaques as the sole contributors for the development of dementia.
Collapse
Affiliation(s)
- Yasushi Tomidokoro
- Department of Pathology and Psychiatry, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | | | |
Collapse
|