58 results on '"Mannini, B."'
Search Results
2. The induction of α-helical structure in partially unfolded HypF-N does not affect its aggregation propensity
- Author
-
Ahmad, B., Vigliotta, I., Tatini, F., Campioni, S., Mannini, B., Winkelmann, J., Tiribilli, B., and Chiti, F.
- Published
- 2011
- Full Text
- View/download PDF
3. Role of aggregation conditions in structure, and toxicity of Yeast Hexokinase aggregates: E4.05
- Author
-
Ramshini, H., Parrini, C., Relini, A., Zampagni, M., Mannini, B., Saboury, A. A., Nemat-Gorgani, M., and Chiti, F.
- Published
- 2010
4. Multistep Inhibition of α‑Synuclein Aggregation and Toxicity in Vitro and in Vivo by Trodusquemine
- Author
-
Perni, M, Flagmeier, P, Limbocker, R, Cascella, R, Aprile, Fa, Galvagnion, C, Heller, Gt, Meisl, G, Chen, Sw, Kumita, Jr, Challa, Pk, Kirkegaard, Jb, Cohen, Sia, Mannini, B, Barbut, D, Nollen, Eaa, Cecchi, C, Cremades, N, Knowles, Tpj, Chiti, F, Zasloff, M, Vendruscolo, M, and Dobson, Cm
- Subjects
α‑Synuclein aggregation, Trodusquemine - Published
- 2018
5. The induction of α-helical structure in partially unfolded HypF-N does not affect its aggregation propensity
- Author
-
Ahmad, B., Vigliotta, I., Tatini, F., Campioni, S., Mannini, B., Winkelmann, J., Tiribilli, B., and Chiti, F.
- Abstract
The conversion of proteins into structured fibrillar aggregates is a central problem in protein chemistry, biotechnology, biology and medicine. It is generally accepted that aggregation takes place from partially structured states of proteins. However, the role of the residual structure present in such conformational states is not yet understood. In particular, it is not yet clear as to whether the α-helical structure represents a productive or counteracting structural element for protein aggregation. We have addressed this issue by studying the aggregation of pH-unfolded HypF-N. It has previously been shown that the two native α-helices of HypF-N retain a partial α-helical structure in the pH-unfolded state and that these regions are also involved in the formation of the cross-β structure of the aggregates. We have introduced mutations in such stretches of the sequence, with the aim of increasing the α-helical structure in the key regions of the pH-unfolded state, while minimizing the changes of other factors known to influence protein aggregation, such as hydrophobicity, β-Sheet propensity, etc. The resulting HypF-N mutants have higher contents of α-helical structure at the site(s) of mutation in their pH-unfolded states, but such an increase does not correlate with a change of aggregation rate. The results suggest that stabilisation of α-helical structure in amyloidogenic regions of the sequence of highly dynamic states does not have remarkable effects on the rate of protein aggregation from such conformational states. Comparison with other protein systems indicate that the effect of increasing α-helical propensity can vary if the stabilised helices are in non-amyloidogenic stretches of initially unstructured peptides (accelerating effect), in amyloidogenic stretches of initially unstructured peptides (no effect) or in amyloidogenic stretches of initially stable helices (decelerating effect)
- Published
- 2017
6. Role of aggregation conditions in structure and toxicity of Yeast Hexokinase aggregates
- Author
-
Ramshini, H., Parrini, C., Relini, Annalisa, Zampagni, M., Mannini, B., Saboury, A., Nemat Gorgani, M., and Chiti, F.
- Published
- 2010
7. Chaperones suppress the toxicity of aberrant protein aggregates. Molecular insight into the mechanism of action
- Author
-
Mannini, B, Campioni, S, Boninsegna, M, Penco, A, Cascella, R, Zampagni, M, Wilson, M, Meehan, S, Roodveldt, C, Dobson, C, Relini, A, Cecchi, C, Chiti, F, Mannini, B, Campioni, S, Boninsegna, M, Penco, A, Cascella, R, Zampagni, M, Wilson, M, Meehan, S, Roodveldt, C, Dobson, C, Relini, A, Cecchi, C, and Chiti, F
- Abstract
of poster presentations presented at 22nd IUBMB & 37th FEBS Congress, Seville, Spain, September 4-9, 2012.
- Published
- 2012
8. The induction of -helical structure in partially unfolded HypF-N does not affect its aggregation propensity
- Author
-
Ahmad, B., primary, Vigliotta, I., additional, Tatini, F., additional, Campioni, S., additional, Mannini, B., additional, Winkelmann, J., additional, Tiribilli, B., additional, and Chiti, F., additional
- Published
- 2011
- Full Text
- View/download PDF
9. The induction of α-helical structure in partially unfolded HypF-N does not affect its aggregation propensity
- Author
-
Ahmad, B., Vigliotta, I., Tatini, F., Campioni, S., Mannini, B., Winkelmann, J., Tiribilli, B., Chiti, F., Ahmad, B., Vigliotta, I., Tatini, F., Campioni, S., Mannini, B., Winkelmann, J., Tiribilli, B., and Chiti, F.
- Abstract
The conversion of proteins into structured fibrillar aggregates is a central problem in protein chemistry, biotechnology, biology and medicine. It is generally accepted that aggregation takes place from partially structured states of proteins. However, the role of the residual structure present in such conformational states is not yet understood. In particular, it is not yet clear as to whether the α-helical structure represents a productive or counteracting structural element for protein aggregation. We have addressed this issue by studying the aggregation of pH-unfolded HypF-N. It has previously been shown that the two native α-helices of HypF-N retain a partial α-helical structure in the pH-unfolded state and that these regions are also involved in the formation of the cross-β structure of the aggregates. We have introduced mutations in such stretches of the sequence, with the aim of increasing the α-helical structure in the key regions of the pH-unfolded state, while minimizing the changes of other factors known to influence protein aggregation, such as hydrophobicity, β-Sheet propensity, etc. The resulting HypF-N mutants have higher contents of α-helical structure at the site(s) of mutation in their pH-unfolded states, but such an increase does not correlate with a change of aggregation rate. The results suggest that stabilisation of α-helical structure in amyloidogenic regions of the sequence of highly dynamic states does not have remarkable effects on the rate of protein aggregation from such conformational states. Comparison with other protein systems indicate that the effect of increasing α-helical propensity can vary if the stabilised helices are in non-amyloidogenic stretches of initially unstructured peptides (accelerating effect), in amyloidogenic stretches of initially unstructured peptides (no effect) or in amyloidogenic stretches of initially stable helices (decelerating effect)
10. Small-molecule sequestration of amyloid-β as a drug discovery strategy for Alzheimer's disease
- Author
-
Benedetta Mannini, Roberta Pierattelli, Thomas Löhr, Massimiliano Bonomi, Carlo Camilloni, Thomas C. T. Michaels, Francesco Simone Ruggeri, Gabriella T. Heller, Alfonso De Simone, Michele Vendruscolo, Francesco A. Aprile, Christopher M. Dobson, Ryan Limbocker, Michele Perni, Isabella C. Felli, Tuomas P. J. Knowles, Heller, Gabriella T [0000-0002-5672-0467], Aprile, Francesco A [0000-0002-5040-4420], Perni, Michele [0000-0001-7593-8376], Ruggeri, Francesco Simone [0000-0002-1232-1907], Mannini, Benedetta [0000-0001-6812-7348], Löhr, Thomas [0000-0003-2969-810X], Bonomi, Massimiliano [0000-0002-7321-0004], Camilloni, Carlo [0000-0002-9923-8590], De Simone, Alfonso [0000-0001-8789-9546], Felli, Isabella C [0000-0002-6018-9090], Pierattelli, Roberta [0000-0001-7755-0885], Knowles, Tuomas PJ [0000-0002-7879-0140], Dobson, Christopher M [0000-0002-5445-680X], Vendruscolo, Michele [0000-0002-3616-1610], Apollo - University of Cambridge Repository, Heller, G. T., Aprile, F. A., Michaels, T. C. T., Limbocker, R., Perni, M., Ruggeri, F. S., Mannini, B., Lohr, T., Bonomi, M., Camilloni, C., de Simone, A., Felli, I. C., Pierattelli, R., Knowles, T. P. J., Dobson, C. M., Vendruscolo, M., University of Cambridge [UK] (CAM), Imperial College London, Harvard University, Bioinformatique structurale - Structural Bioinformatics, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Università degli Studi di Milano = University of Milan (UNIMI), University of Naples Federico II = Università degli studi di Napoli Federico II, Università degli Studi di Firenze = University of Florence (UniFI), Funding: G.T.H. is supported by the Gates Cambridge Trust and the Rosalind Franklin Research Fellowship at Newnham College, Cambridge, F.A.A. is supported by UK Research and Innovation (Future Leaders Fellowship MR/S033947/1) and the Alzheimer’s Society, UK (317, 511), R.L. is supported by the Gates Cambridge Trust, TCTM by Peterhouse, Cambridge and the Swiss National Science Foundation, and F.S.R. is supported by Darwin College and the Swiss National Foundation (grant numbers P300P2_171219 and P2ELP2_162116, respectively). We acknowledge ARCHER UK National Supercomputing Service under ARCHER Leadership project (grant number e510) and PRACE for awarding us access to MareNostrum at Barcelona Supercomputing Center (BSC), Spain for metadynamic metainference simulations. Parameterization of 10074-G5 was performed using resources provided by the Cambridge Service for Data Driven Discovery (CSD3) operated by the University of Cambridge Research Computing Service (www.csd3.cam.ac.uk), provided by Dell EMC and Intel using Tier-2 funding from the Engineering and Physical Sciences Research Council (capital grant EP/P020259/1), and DiRAC funding from the Science and Technology Facilities Council (www.dirac.ac.uk). MALDI mass spectrometry measurements were performed by L. Packman at the Protein and Nucleic Acid Chemistry Facility (PNAC) at the Department of Biochemistry, University of Cambridge. The NMR measurements were supported by the iNEXT H2020 Programme (EC contract no. 653706). OW450 C. elegans were donated by E. Nollen. BLI measurements were performed in the Biophysics facility at the Department of Biochemistry, University of Cambridge. The work was also supported by the Centre for Misfolding Diseases and the INCEPTION project ANR-16-CONV-0005., ANR-16-CONV-0005,INCEPTION,Institut Convergences pour l'étude de l'Emergence des Pathologies au Travers des Individus et des populatiONs(2016), Harvard University [Cambridge], Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Università degli Studi di Milano [Milano] (UNIMI), University of Naples Federico II, and Università degli Studi di Firenze = University of Florence [Firenze] (UNIFI)
- Subjects
Amyloid beta ,In silico ,Biophysics ,Intrinsically disordered proteins ,03 medical and health sciences ,0302 clinical medicine ,Alzheimer Disease ,Drug Discovery ,medicine ,Humans ,Life Science ,[SDV.BBM.BC]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biochemistry [q-bio.BM] ,Molecular Biology ,Research Articles ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Amyloid beta-Peptides ,biology ,Drug discovery ,Chemistry ,SciAdv r-articles ,Conformational entropy ,Small molecule ,Peptide Fragments ,3. Good health ,Mechanism of action ,biology.protein ,Small molecule binding ,medicine.symptom ,Hydrophobic and Hydrophilic Interactions ,030217 neurology & neurosurgery ,Research Article - Abstract
A small molecule binds to a disordered protein in its monomeric form, preventing its aggregation linked to Alzheimer’s disease., Disordered proteins are challenging therapeutic targets, and no drug is currently in clinical use that modifies the properties of their monomeric states. Here, we identify a small molecule (10074-G5) capable of binding and sequestering the intrinsically disordered amyloid-β (Aβ) peptide in its monomeric, soluble state. Our analysis reveals that this compound interacts with Aβ and inhibits both the primary and secondary nucleation pathways in its aggregation process. We characterize this interaction using biophysical experiments and integrative structural ensemble determination methods. We observe that this molecule increases the conformational entropy of monomeric Aβ while decreasing its hydrophobic surface area. We also show that it rescues a Caenorhabditis elegans model of Aβ-associated toxicity, consistent with the mechanism of action identified from the in silico and in vitro studies. These results illustrate the strategy of stabilizing the monomeric states of disordered proteins with small molecules to alter their behavior for therapeutic purposes.
- Published
- 2020
11. Targeting Protein Aggregation in ALS.
- Author
-
Perni M and Mannini B
- Subjects
- Humans, Protein Aggregates, Animals, Amyotrophic Lateral Sclerosis metabolism, Amyotrophic Lateral Sclerosis genetics, Amyotrophic Lateral Sclerosis drug therapy, Protein Aggregation, Pathological metabolism
- Abstract
Proteinopathies involve the abnormal accumulation of specific proteins. Maintaining the balance of the proteome is a finely regulated process managed by a complex network of cellular machinery responsible for protein synthesis, folding, and degradation. However, stress and ageing can disrupt this balance, leading to widespread protein aggregation. Currently, several therapies targeting protein aggregation are in clinical trials for ALS. These approaches mainly focus on two strategies: addressing proteins that are prone to aggregation due to mutations and targeting the cellular mechanisms that maintain protein homeostasis to prevent aggregation. This review will cover these emerging drugs. Advances in ALS research not only offer hope for better outcomes for ALS patients but also provide valuable insights and methodologies that can benefit the broader field of neurodegenerative disease drug discovery.
- Published
- 2024
- Full Text
- View/download PDF
12. Preparation and Characterization of Zn(II)-Stabilized Aβ 42 Oligomers.
- Author
-
González Díaz A, Cataldi R, Mannini B, and Vendruscolo M
- Subjects
- Humans, Cell Line, Tumor, Alzheimer Disease metabolism, Cell Survival drug effects, Amyloid beta-Peptides chemistry, Amyloid beta-Peptides metabolism, Zinc chemistry, Peptide Fragments chemistry, Peptide Fragments metabolism
- Abstract
Aβ oligomers are being investigated as cytotoxic agents in Alzheimer's disease (AD). Because of their transient nature and conformational heterogeneity, the relationship between the structure and activity of these oligomers is still poorly understood. Hence, methods for stabilizing Aβ oligomeric species relevant to AD are needed to uncover the structural determinants of their cytotoxicity. Here, we build on the observation that metal ions and metabolites have been shown to interact with Aβ, influencing its aggregation and stabilizing its oligomeric species. We thus developed a method that uses zinc ions, Zn(II), to stabilize oligomers produced by the 42-residue form of Aβ (Aβ
42 ), which is dysregulated in AD. These Aβ42 -Zn(II) oligomers are small in size, spanning the 10-30 nm range, stable at physiological temperature, and with a broad toxic profile in human neuroblastoma cells. These oligomers offer a tool to study the mechanisms of toxicity of Aβ oligomers in cellular and animal AD models.- Published
- 2024
- Full Text
- View/download PDF
13. A Relationship between the Structures and Neurotoxic Effects of Aβ Oligomers Stabilized by Different Metal Ions.
- Author
-
Chia S, Cataldi RL, Ruggeri FS, Limbocker R, Condado-Morales I, Pisani K, Possenti A, Linse S, Knowles TPJ, Habchi J, Mannini B, and Vendruscolo M
- Subjects
- Humans, Metals, Ions, Peptide Fragments chemistry, Amyloid beta-Peptides chemistry, Alzheimer Disease
- Abstract
Oligomeric assemblies of the amyloid β peptide (Aβ) have been investigated for over two decades as possible neurotoxic agents in Alzheimer's disease. However, due to their heterogeneous and transient nature, it is not yet fully established which of the structural features of these oligomers may generate cellular damage. Here, we study distinct oligomer species formed by Aβ40 (the 40-residue form of Aβ) in the presence of four different metal ions (Al
3+ , Cu2+ , Fe2+ , and Zn2+ ) and show that they differ in their structure and toxicity in human neuroblastoma cells. We then describe a correlation between the size of the oligomers and their neurotoxic activity, which provides a type of structure-toxicity relationship for these Aβ40 oligomer species. These results provide insight into the possible role of metal ions in Alzheimer's disease by the stabilization of Aβ oligomers.- Published
- 2024
- Full Text
- View/download PDF
14. Structure-Based Discovery of Small-Molecule Inhibitors of the Autocatalytic Proliferation of α-Synuclein Aggregates.
- Author
-
Chia S, Faidon Brotzakis Z, Horne RI, Possenti A, Mannini B, Cataldi R, Nowinska M, Staats R, Linse S, Knowles TPJ, Habchi J, and Vendruscolo M
- Subjects
- Humans, Amyloid metabolism, Kinetics, Cell Proliferation, Protein Aggregates, alpha-Synuclein metabolism, Parkinson Disease metabolism
- Abstract
The presence of amyloid fibrils of α-synuclein is closely associated with Parkinson's disease and related synucleinopathies. It is still very challenging, however, to systematically discover small molecules that prevent the formation of these aberrant aggregates. Here, we describe a structure-based approach to identify small molecules that specifically inhibit the surface-catalyzed secondary nucleation step in the aggregation of α-synuclein by binding to the surface of the amyloid fibrils. The resulting small molecules are screened using a range of kinetic and thermodynamic assays for their ability to bind α-synuclein fibrils and prevent the further generation of α-synuclein oligomers. This study demonstrates that the combination of structure-based and kinetic-based drug discovery methods can lead to the identification of small molecules that selectively inhibit the autocatalytic proliferation of α-synuclein aggregates.
- Published
- 2023
- Full Text
- View/download PDF
15. The Pathological G51D Mutation in Alpha-Synuclein Oligomers Confers Distinct Structural Attributes and Cellular Toxicity.
- Author
-
Xu CK, Castellana-Cruz M, Chen SW, Du Z, Meisl G, Levin A, Mannini B, Itzhaki LS, Knowles TPJ, Dobson CM, Cremades N, and Kumita JR
- Subjects
- Humans, Neurodegenerative Diseases, Protein Aggregates, Protein Binding, Spectrum Analysis, alpha-Synuclein metabolism, Mutation, Protein Aggregation, Pathological genetics, Protein Aggregation, Pathological metabolism, Protein Multimerization genetics, alpha-Synuclein chemistry, alpha-Synuclein genetics
- Abstract
A wide variety of oligomeric structures are formed during the aggregation of proteins associated with neurodegenerative diseases. Such soluble oligomers are believed to be key toxic species in the related disorders; therefore, identification of the structural determinants of toxicity is of upmost importance. Here, we analysed toxic oligomers of α-synuclein and its pathological variants in order to identify structural features that could be related to toxicity and found a novel structural polymorphism within G51D oligomers. These G51D oligomers can adopt a variety of β-sheet-rich structures with differing degrees of α-helical content, and the helical structural content of these oligomers correlates with the level of induced cellular dysfunction in SH-SY5Y cells. This structure-function relationship observed in α-synuclein oligomers thus presents the α-helical structure as another potential structural determinant that may be linked with cellular toxicity in amyloid-related proteins.
- Published
- 2022
- Full Text
- View/download PDF
16. Surface-Catalyzed Secondary Nucleation Dominates the Generation of Toxic IAPP Aggregates.
- Author
-
Rodriguez Camargo DC, Chia S, Menzies J, Mannini B, Meisl G, Lundqvist M, Pohl C, Bernfur K, Lattanzi V, Habchi J, Cohen SI, Knowles TPJ, Vendruscolo M, and Linse S
- Abstract
The aggregation of the human islet amyloid polypeptide (IAPP) is associated with diabetes type II. A quantitative understanding of this connection at the molecular level requires that the aggregation mechanism of IAPP is resolved in terms of the underlying microscopic steps. Here we have systematically studied recombinant IAPP, with amidated C-terminus in oxidised form with a disulphide bond between residues 3 and 7, using thioflavin T fluorescence to monitor the formation of amyloid fibrils as a function of time and IAPP concentration. We used global kinetic analyses to connect the macroscopic measurements of aggregation to the microscopic mechanisms, and show that the generation of new aggregates is dominated by the secondary nucleation of monomers on the fibril surface. We then exposed insulinoma cells to aliquots extracted from different time points of the aggregation process, finding the highest toxicity at the midpoint of the reaction, when the secondary nucleation rate reaches its maximum. These results identify IAPP oligomers as the most cytotoxic species generated during IAPP aggregation, and suggest that compounds that target secondary nucleation of IAPP could be most effective as therapeutic candidates for diabetes type II., Competing Interests: Authors DC, SKRC, JM, BM, JH and SC were employed by Wren Therapeutics Limited. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Rodriguez Camargo, Chia, Menzies, Mannini, Meisl, Lundqvist, Pohl, Bernfur, Lattanzi, Habchi, Cohen, Knowles, Vendruscolo and Linse.)
- Published
- 2021
- Full Text
- View/download PDF
17. Distinct responses of human peripheral blood cells to different misfolded protein oligomers.
- Author
-
Leal-Lasarte M, Mannini B, Chiti F, Vendruscolo M, Dobson CM, Roodveldt C, and Pozo D
- Subjects
- Adult, CD4-Positive T-Lymphocytes metabolism, Cell Differentiation physiology, Cells, Cultured, Cytokines metabolism, Humans, Lymphocyte Activation physiology, Middle Aged, Protein Folding, T-Lymphocytes, Regulatory metabolism, Th1 Cells metabolism, Th17 Cells metabolism, Leukocytes, Mononuclear metabolism, Proteostasis Deficiencies metabolism
- Abstract
Increasing evidence indicates that peripheral immune cells play a prominent role in neurodegeneration connected to protein misfolding, which are associated with formation of aberrant aggregates, including soluble protein misfolded oligomers. The precise links, however, between the physicochemical features of diverse oligomers and their effects on the immune system, particularly on adaptive immunity, remain currently unexplored, due partly to the transient and heterogeneous nature of the oligomers themselves. To overcome these limitations, we took advantage of two stable and well-characterized types of model oligomers (A and B), formed by HypF-N bacterial protein, type B oligomers displaying lower solvent-exposed hydrophobicity. Exposure to oligomers of human peripheral blood mononuclear cells (PBMCs) revealed differential effects, with type B, but not type A, oligomers leading to a reduction in CD4
+ cells. Type A oligomers promoted enhanced differentiation towards CD4+ CD25High FoxP3+ Tregs and displayed a higher suppressive effect on lymphocyte proliferation than Tregs treated with oligomers B or untreated cells. Moreover, our results reveal Th1 and Th17 lymphocyte differentiation mediated by type A oligomers and a differential balance of TGF-β, IL-6, IL-23, IFN-γ and IL-10 mediators. These results indicate that type B oligomers recapitulate some of the biological responses associated with Parkinson's disease in peripheral immunocompetent cells, while type A oligomers resemble responses associated with Alzheimer's disease. We anticipate that further studies characterizing the differential effects of protein misfolded oligomers on the peripheral immune system may lead to the development of blood-based diagnostics, which could report on the type and properties of oligomers present in patients., (© 2021 The Authors. Immunology published by John Wiley & Sons Ltd.)- Published
- 2021
- Full Text
- View/download PDF
18. Publisher Correction: Two human metabolites rescue a C. elegans model of Alzheimer's disease via a cytosolic unfolded protein response.
- Author
-
Joshi P, Perni M, Limbocker R, Mannini B, Casford S, Chia S, Habchi J, Labbadia J, Dobson CM, and Vendruscolo M
- Published
- 2021
- Full Text
- View/download PDF
19. Exogenous misfolded protein oligomers can cross the intestinal barrier and cause a disease phenotype in C. elegans.
- Author
-
Perni M, Mannini B, Xu CK, Kumita JR, Dobson CM, Chiti F, and Vendruscolo M
- Subjects
- Animals, Caenorhabditis elegans Proteins metabolism, Caenorhabditis elegans Proteins genetics, Oxidative Stress drug effects, Intestinal Mucosa metabolism, Protein Aggregates, Protein Multimerization, Caenorhabditis elegans metabolism, Caenorhabditis elegans drug effects, Phenotype, Protein Folding
- Abstract
Misfolded protein oligomers are increasingly recognized as highly cytotoxic agents in a wide range of human disorders associated with protein aggregation. In this study, we assessed the possible uptake and resulting toxic effects of model protein oligomers administered to C. elegans through the culture medium. We used an automated machine-vision, high-throughput screening procedure to monitor the phenotypic changes in the worms, in combination with confocal microscopy to monitor the diffusion of the oligomers, and oxidative stress assays to detect their toxic effects. Our results suggest that the oligomers can diffuse from the intestinal lumen to other tissues, resulting in a disease phenotype. We also observed that pre-incubation of the oligomers with a molecular chaperone (αB-crystallin) or a small molecule inhibitor of protein aggregation (squalamine), reduced the oligomer absorption. These results indicate that exogenous misfolded protein oligomers can be taken up by the worms from their environment and spread across tissues, giving rise to pathological effects in regions distant from their place of absorbance., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
20. Two human metabolites rescue a C. elegans model of Alzheimer's disease via a cytosolic unfolded protein response.
- Author
-
Joshi P, Perni M, Limbocker R, Mannini B, Casford S, Chia S, Habchi J, Labbadia J, Dobson CM, and Vendruscolo M
- Subjects
- Alzheimer Disease prevention & control, Amyloid beta-Peptides chemistry, Amyloid beta-Peptides metabolism, Animals, Caenorhabditis elegans Proteins metabolism, Carnosine pharmacology, Cytosol metabolism, HSP40 Heat-Shock Proteins metabolism, Humans, Kynurenic Acid pharmacology, Protein Aggregates, Protein Aggregation, Pathological prevention & control, Transcription Factors metabolism, Unfolded Protein Response drug effects, Alzheimer Disease metabolism, Caenorhabditis elegans metabolism, Carnosine metabolism, Disease Models, Animal, Kynurenic Acid metabolism, Unfolded Protein Response physiology
- Abstract
Age-related changes in cellular metabolism can affect brain homeostasis, creating conditions that are permissive to the onset and progression of neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. Although the roles of metabolites have been extensively studied with regard to cellular signaling pathways, their effects on protein aggregation remain relatively unexplored. By computationally analysing the Human Metabolome Database, we identified two endogenous metabolites, carnosine and kynurenic acid, that inhibit the aggregation of the amyloid beta peptide (Aβ) and rescue a C. elegans model of Alzheimer's disease. We found that these metabolites act by triggering a cytosolic unfolded protein response through the transcription factor HSF-1 and downstream chaperones HSP40/J-proteins DNJ-12 and DNJ-19. These results help rationalise previous observations regarding the possible anti-ageing benefits of these metabolites by providing a mechanism for their action. Taken together, our findings provide a link between metabolite homeostasis and protein homeostasis, which could inspire preventative interventions against neurodegenerative disorders.
- Published
- 2021
- Full Text
- View/download PDF
21. Squalamine and Its Derivatives Modulate the Aggregation of Amyloid-β and α-Synuclein and Suppress the Toxicity of Their Oligomers.
- Author
-
Limbocker R, Staats R, Chia S, Ruggeri FS, Mannini B, Xu CK, Perni M, Cascella R, Bigi A, Sasser LR, Block NR, Wright AK, Kreiser RP, Custy ET, Meisl G, Errico S, Habchi J, Flagmeier P, Kartanas T, Hollows JE, Nguyen LT, LeForte K, Barbut D, Kumita JR, Cecchi C, Zasloff M, Knowles TPJ, Dobson CM, Chiti F, and Vendruscolo M
- Abstract
The aberrant aggregation of proteins is a key molecular event in the development and progression of a wide range of neurodegenerative disorders. We have shown previously that squalamine and trodusquemine, two natural products in the aminosterol class, can modulate the aggregation of the amyloid-β peptide (Aβ) and of α-synuclein (αS), which are associated with Alzheimer's and Parkinson's diseases. In this work, we expand our previous analyses to two squalamine derivatives, des-squalamine and α-squalamine, obtaining further insights into the mechanism by which aminosterols modulate Aβ and αS aggregation. We then characterize the ability of these small molecules to alter the physicochemical properties of stabilized oligomeric species in vitro and to suppress the toxicity of these aggregates to varying degrees toward human neuroblastoma cells. We found that, despite the fact that these aminosterols exert opposing effects on Aβ and αS aggregation under the conditions that we tested, the modifications that they induced to the toxicity of oligomers were similar. Our results indicate that the suppression of toxicity is mediated by the displacement of toxic oligomeric species from cellular membranes by the aminosterols. This study, thus, provides evidence that aminosterols could be rationally optimized in drug discovery programs to target oligomer toxicity in Alzheimer's and Parkinson's diseases., Competing Interests: DB and MZ are inventors in a patent for the use of aminosterols in the treatment of Parkinson’s disease. DB and MZ are co-founders of Enterin Inc. and serve as the President and CSO, respectively, of the company. MV, TPJK, and JH are co-founders, and BM and MP are employees of Wren Therapeutics Ltd., which is independently pursuing inhibitors of protein misfolding and aggregation. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Limbocker, Staats, Chia, Ruggeri, Mannini, Xu, Perni, Cascella, Bigi, Sasser, Block, Wright, Kreiser, Custy, Meisl, Errico, Habchi, Flagmeier, Kartanas, Hollows, Nguyen, LeForte, Barbut, Kumita, Cecchi, Zasloff, Knowles, Dobson, Chiti and Vendruscolo.)
- Published
- 2021
- Full Text
- View/download PDF
22. Aβ Oligomers Dysregulate Calcium Homeostasis by Mechanosensitive Activation of AMPA and NMDA Receptors.
- Author
-
Fani G, Mannini B, Vecchi G, Cascella R, Cecchi C, Dobson CM, Vendruscolo M, and Chiti F
- Subjects
- Amyloid beta-Peptides metabolism, Calcium metabolism, Homeostasis, Humans, Neurons metabolism, Peptide Fragments metabolism, alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid, Alzheimer Disease, Receptors, N-Methyl-D-Aspartate
- Abstract
Alzheimer's disease, which is the most common form of dementia, is characterized by the aggregation of the amyloid β peptide (Aβ) and by an impairment of calcium homeostasis caused by excessive activation of glutamatergic receptors (excitotoxicity). Here, we studied the effects on calcium homeostasis caused by the formation of Aβ oligomeric assemblies. We found that Aβ oligomers cause a rapid influx of calcium ions (Ca
2+ ) across the cell membrane by rapidly activating extrasynaptic N -methyl-d-aspartate (NMDA) receptors and, to a lower extent, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. We also observed, however, that misfolded oligomers do not interact directly with these receptors. Further experiments with lysophosphatidylcholine and arachidonic acid, which cause membrane compression and stretch, respectively, indicated that these receptors are activated through a change in membrane tension induced by the oligomers and transmitted mechanically to the receptors via the lipid bilayer. Indeed, lysophosphatidylcholine is able to neutralize the oligomer-induced activation of the NMDA receptors, whereas arachidonic acid activates the receptors similarly to the oligomers with no additive effects. An increased rotational freedom observed for a fluorescent probe embedded within the membrane in the presence of the oligomers also indicates a membrane stretch. These results reveal a mechanism of toxicity of Aβ oligomers in Alzheimer's disease through the perturbation of the mechanical properties of lipid membranes sensed by NMDA and AMPA receptors.- Published
- 2021
- Full Text
- View/download PDF
23. Publisher Correction: A dopamine metabolite stabilizes neurotoxic amyloid-β oligomers.
- Author
-
Cataldi R, Chia S, Pisani K, Ruggeri FS, Xu CK, Šneideris T, Perni M, Sarwat S, Joshi P, Kumita JR, Linse S, Habchi J, Knowles TPJ, Mannini B, Dobson CM, and Vendruscolo M
- Published
- 2021
- Full Text
- View/download PDF
24. A dopamine metabolite stabilizes neurotoxic amyloid-β oligomers.
- Author
-
Cataldi R, Chia S, Pisani K, Ruggeri FS, Xu CK, Šneideris T, Perni M, Sarwat S, Joshi P, Kumita JR, Linse S, Habchi J, Knowles TPJ, Mannini B, Dobson CM, and Vendruscolo M
- Subjects
- Alzheimer Disease metabolism, Cell Line, Tumor, Escherichia coli, Humans, Alzheimer Disease etiology, Amyloid beta-Peptides metabolism, Dopamine metabolism
- Abstract
Aberrant soluble oligomers formed by the amyloid-β peptide (Aβ) are major pathogenic agents in the onset and progression of Alzheimer's disease. A variety of biomolecules can influence the formation of these oligomers in the brain, although their mechanisms of action are still largely unknown. Here, we studied the effects on Aβ aggregation of DOPAL, a reactive catecholaldehyde intermediate of dopamine metabolism. We found that DOPAL is able to stabilize Aβ oligomeric species, including dimers and trimers, that exert toxic effects on human neuroblastoma cells, in particular increasing cytosolic calcium levels and promoting the generation of reactive oxygen species. These results reveal an interplay between Aβ aggregation and key biochemical processes regulating cellular homeostasis in the brain.
- Published
- 2021
- Full Text
- View/download PDF
25. Therapeutic Strategies to Reduce the Toxicity of Misfolded Protein Oligomers.
- Author
-
Kreiser RP, Wright AK, Block NR, Hollows JE, Nguyen LT, LeForte K, Mannini B, Vendruscolo M, and Limbocker R
- Subjects
- Animals, Humans, Protein Aggregation, Pathological metabolism, Protein Aggregation, Pathological pathology, Proteostasis Deficiencies metabolism, Proteostasis Deficiencies pathology, Protein Aggregation, Pathological therapy, Protein Multimerization, Proteostasis Deficiencies therapy
- Abstract
The aberrant aggregation of proteins is implicated in the onset and pathogenesis of a wide range of neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. Mounting evidence indicates that misfolded protein oligomers produced as intermediates in the aggregation process are potent neurotoxic agents in these diseases. Because of the transient and heterogeneous nature of these elusive aggregates, however, it has proven challenging to develop therapeutics that can effectively target them. Here, we review approaches aimed at reducing oligomer toxicity, including (1) modulating the oligomer populations (e.g., by altering the kinetics of aggregation by inhibiting, enhancing, or redirecting the process), (2) modulating the oligomer properties (e.g., through the size-hydrophobicity-toxicity relationship), (3) modulating the oligomer interactions (e.g., by protecting cell membranes by displacing oligomers), and (4) reducing oligomer toxicity by potentiating the protein homeostasis system. We analyze examples of these complementary approaches, which may lead to the development of compounds capable of preventing or treating neurodegenerative disorders associated with protein aggregation.
- Published
- 2020
- Full Text
- View/download PDF
26. Small-molecule sequestration of amyloid-β as a drug discovery strategy for Alzheimer's disease.
- Author
-
Heller GT, Aprile FA, Michaels TCT, Limbocker R, Perni M, Ruggeri FS, Mannini B, Löhr T, Bonomi M, Camilloni C, De Simone A, Felli IC, Pierattelli R, Knowles TPJ, Dobson CM, and Vendruscolo M
- Subjects
- Amyloid beta-Peptides metabolism, Drug Discovery, Humans, Hydrophobic and Hydrophilic Interactions, Peptide Fragments metabolism, Alzheimer Disease drug therapy, Alzheimer Disease metabolism
- Abstract
Disordered proteins are challenging therapeutic targets, and no drug is currently in clinical use that modifies the properties of their monomeric states. Here, we identify a small molecule (10074-G5) capable of binding and sequestering the intrinsically disordered amyloid-β (Aβ) peptide in its monomeric, soluble state. Our analysis reveals that this compound interacts with Aβ and inhibits both the primary and secondary nucleation pathways in its aggregation process. We characterize this interaction using biophysical experiments and integrative structural ensemble determination methods. We observe that this molecule increases the conformational entropy of monomeric Aβ while decreasing its hydrophobic surface area. We also show that it rescues a Caenorhabditis elegans model of Aβ-associated toxicity, consistent with the mechanism of action identified from the in silico and in vitro studies. These results illustrate the strategy of stabilizing the monomeric states of disordered proteins with small molecules to alter their behavior for therapeutic purposes., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution License 4.0 (CC BY).)
- Published
- 2020
- Full Text
- View/download PDF
27. A rationally designed bicyclic peptide remodels Aβ42 aggregation in vitro and reduces its toxicity in a worm model of Alzheimer's disease.
- Author
-
Ikenoue T, Aprile FA, Sormanni P, Ruggeri FS, Perni M, Heller GT, Haas CP, Middel C, Limbocker R, Mannini B, Michaels TCT, Knowles TPJ, Dobson CM, and Vendruscolo M
- Subjects
- Amyloid metabolism, Animals, Disease Models, Animal, Peptide Fragments, Plaque, Amyloid metabolism, Alzheimer Disease metabolism, Amyloid beta-Peptides metabolism, Caenorhabditis elegans metabolism, Protein Aggregation, Pathological metabolism
- Abstract
Bicyclic peptides have great therapeutic potential since they can bridge the gap between small molecules and antibodies by combining a low molecular weight of about 2 kDa with an antibody-like binding specificity. Here we apply a recently developed in silico rational design strategy to produce a bicyclic peptide to target the C-terminal region (residues 31-42) of the 42-residue form of the amyloid β peptide (Aβ42), a protein fragment whose aggregation into amyloid plaques is linked with Alzheimer's disease. We show that this bicyclic peptide is able to remodel the aggregation process of Aβ42 in vitro and to reduce its associated toxicity in vivo in a C. elegans worm model expressing Aβ42. These results provide an initial example of a computational approach to design bicyclic peptides to target specific epitopes on disordered proteins.
- Published
- 2020
- Full Text
- View/download PDF
28. Trodusquemine displaces protein misfolded oligomers from cell membranes and abrogates their cytotoxicity through a generic mechanism.
- Author
-
Limbocker R, Mannini B, Ruggeri FS, Cascella R, Xu CK, Perni M, Chia S, Chen SW, Habchi J, Bigi A, Kreiser RP, Wright AK, Albright JA, Kartanas T, Kumita JR, Cremades N, Zasloff M, Cecchi C, Knowles TPJ, Chiti F, Vendruscolo M, and Dobson CM
- Subjects
- Amyloid beta-Peptides chemistry, Amyloid beta-Peptides toxicity, Biophysical Phenomena drug effects, Carboxyl and Carbamoyl Transferases chemistry, Carboxyl and Carbamoyl Transferases toxicity, Cell Death drug effects, Cell Line, Tumor, Cell Membrane drug effects, Escherichia coli Proteins chemistry, Escherichia coli Proteins toxicity, Humans, Spermine pharmacology, alpha-Synuclein chemistry, alpha-Synuclein toxicity, Cell Membrane metabolism, Cholestanes pharmacology, Protein Folding drug effects, Protein Multimerization drug effects, Spermine analogs & derivatives
- Abstract
The onset and progression of numerous protein misfolding diseases are associated with the presence of oligomers formed during the aberrant aggregation of several different proteins, including amyloid-β (Aβ) in Alzheimer's disease and α-synuclein (αS) in Parkinson's disease. These small, soluble aggregates are currently major targets for drug discovery. In this study, we show that trodusquemine, a naturally-occurring aminosterol, markedly reduces the cytotoxicity of αS, Aβ and HypF-N oligomers to human neuroblastoma cells by displacing the oligomers from cell membranes in the absence of any substantial morphological and structural changes to the oligomers. These results indicate that the reduced toxicity results from a mechanism that is common to oligomers from different proteins, shed light on the origin of the toxicity of the most deleterious species associated with protein aggregation and suggest that aminosterols have the therapeutically-relevant potential to protect cells from the oligomer-induced cytotoxicity associated with numerous protein misfolding diseases.
- Published
- 2020
- Full Text
- View/download PDF
29. Rationally Designed Antibodies as Research Tools to Study the Structure-Toxicity Relationship of Amyloid-β Oligomers.
- Author
-
Limbocker R, Mannini B, Cataldi R, Chhangur S, Wright AK, Kreiser RP, Albright JA, Chia S, Habchi J, Sormanni P, Kumita JR, Ruggeri FS, Dobson CM, Chiti F, Aprile FA, and Vendruscolo M
- Subjects
- Alzheimer Disease metabolism, Amyloid beta-Peptides metabolism, Amyloid beta-Protein Precursor metabolism, Antibody Formation immunology, Brain metabolism, Drug Design, Humans, Neurons metabolism, Peptide Fragments metabolism, Plaque, Amyloid metabolism, Protein Aggregates physiology, Protein Engineering methods, Structure-Activity Relationship, Amyloid beta-Peptides immunology, Antibodies immunology, Antibodies metabolism
- Abstract
Alzheimer's disease is associated with the aggregation of the amyloid-β peptide (Aβ), resulting in the deposition of amyloid plaques in brain tissue. Recent scrutiny of the mechanisms by which Aβ aggregates induce neuronal dysfunction has highlighted the importance of the Aβ oligomers of this protein fragment. Because of the transient and heterogeneous nature of these oligomers, however, it has been challenging to investigate the detailed mechanisms by which these species exert cytotoxicity. To address this problem, we demonstrate here the use of rationally designed single-domain antibodies (DesAbs) to characterize the structure-toxicity relationship of Aβ oligomers. For this purpose, we use Zn
2+ -stabilized oligomers of the 40-residue form of Aβ (Aβ40 ) as models of brain Aβ oligomers and two single-domain antibodies (DesAb18-24 and DesAb34-40 ), designed to bind to epitopes at residues 18-24 and 34-40 of Aβ40 , respectively. We found that the DesAbs induce a change in structure of the Zn2+ -stabilized Aβ40 oligomers, generating a simultaneous increase in their size and solvent-exposed hydrophobicity. We then observed that these increments in both the size and hydrophobicity of the oligomers neutralize each other in terms of their effects on cytotoxicity, as predicted by a recently proposed general structure-toxicity relationship, and observed experimentally. These results illustrate the use of the DesAbs as research tools to investigate the biophysical and cytotoxicity properties of Aβ oligomers.- Published
- 2020
- Full Text
- View/download PDF
30. Rational design of a conformation-specific antibody for the quantification of Aβ oligomers.
- Author
-
Aprile FA, Sormanni P, Podpolny M, Chhangur S, Needham LM, Ruggeri FS, Perni M, Limbocker R, Heller GT, Sneideris T, Scheidt T, Mannini B, Habchi J, Lee SF, Salinas PC, Knowles TPJ, Dobson CM, and Vendruscolo M
- Subjects
- Alzheimer Disease diagnosis, Alzheimer Disease metabolism, Amyloid beta-Peptides chemistry, Animals, Antibodies chemistry, Antibodies metabolism, Antibody Specificity, Caenorhabditis elegans, Disease Models, Animal, Epitopes, Hippocampus metabolism, Mice, Protein Binding, Protein Conformation, Single-Domain Antibodies, Amyloid beta-Peptides metabolism, Antibodies immunology, Protein Aggregates
- Abstract
Protein misfolding and aggregation is the hallmark of numerous human disorders, including Alzheimer's disease. This process involves the formation of transient and heterogeneous soluble oligomers, some of which are highly cytotoxic. A major challenge for the development of effective diagnostic and therapeutic tools is thus the detection and quantification of these elusive oligomers. Here, to address this problem, we develop a two-step rational design method for the discovery of oligomer-specific antibodies. The first step consists of an "antigen scanning" phase in which an initial panel of antibodies is designed to bind different epitopes covering the entire sequence of a target protein. This procedure enables the determination through in vitro assays of the regions exposed in the oligomers but not in the fibrillar deposits. The second step involves an "epitope mining" phase, in which a second panel of antibodies is designed to specifically target the regions identified during the scanning step. We illustrate this method in the case of the amyloid β (Aβ) peptide, whose oligomers are associated with Alzheimer's disease. Our results show that this approach enables the accurate detection and quantification of Aβ oligomers in vitro, and in Caenorhabditis elegans and mouse hippocampal tissues., Competing Interests: The authors declare no competing interest., (Copyright © 2020 the Author(s). Published by PNAS.)
- Published
- 2020
- Full Text
- View/download PDF
31. Single molecule secondary structure determination of proteins through infrared absorption nanospectroscopy.
- Author
-
Ruggeri FS, Mannini B, Schmid R, Vendruscolo M, and Knowles TPJ
- Subjects
- Biophysics, Molecular Conformation, Nanotechnology methods, Spectrophotometry, Infrared methods
- Abstract
The chemical and structural properties of biomolecules determine their interactions, and thus their functions, in a wide variety of biochemical processes. Innovative imaging methods have been developed to characterise biomolecular structures down to the angstrom level. However, acquiring vibrational absorption spectra at the single molecule level, a benchmark for bulk sample characterization, has remained elusive. Here, we introduce off-resonance, low power and short pulse infrared nanospectroscopy (ORS-nanoIR) to allow the acquisition of infrared absorption spectra and chemical maps at the single molecule level, at high throughput on a second timescale and with a high signal-to-noise ratio (~10-20). This high sensitivity enables the accurate determination of the secondary structure of single protein molecules with over a million-fold lower mass than conventional bulk vibrational spectroscopy. These results pave the way to probe directly the chemical and structural properties of individual biomolecules, as well as their interactions, in a broad range of chemical and biological systems.
- Published
- 2020
- Full Text
- View/download PDF
32. Proteome-wide observation of the phenomenon of life on the edge of solubility.
- Author
-
Vecchi G, Sormanni P, Mannini B, Vandelli A, Tartaglia GG, Dobson CM, Hartl FU, and Vendruscolo M
- Subjects
- Aging physiology, Animals, Caenorhabditis elegans growth & development, Homeostasis, Mass Spectrometry, Molecular Chaperones metabolism, Protein Aggregates physiology, Protein Folding, Solubility, Caenorhabditis elegans metabolism, Caenorhabditis elegans Proteins chemistry, Caenorhabditis elegans Proteins metabolism, Proteome chemistry, Proteome metabolism
- Abstract
To function effectively proteins must avoid aberrant aggregation, and hence they are expected to be expressed at concentrations safely below their solubility limits. By analyzing proteome-wide mass spectrometry data of Caenorhabditis elegans , however, we show that the levels of about three-quarters of the nearly 4,000 proteins analyzed in adult animals are close to their intrinsic solubility limits, indeed exceeding them by about 10% on average. We next asked how aging and functional self-assembly influence these solubility limits. We found that despite the fact that the total quantity of proteins within the cellular environment remains approximately constant during aging, protein aggregation sharply increases between days 6 and 12 of adulthood, after the worms have reproduced, as individual proteins lose their stoichiometric balances and the cellular machinery that maintains solubility undergoes functional decline. These findings reveal that these proteins are highly prone to undergoing concentration-dependent phase separation, which on aging is rationalized in a decrease of their effective solubilities, in particular for proteins associated with translation, growth, reproduction, and the chaperone system., Competing Interests: The authors declare no competing interest., (Copyright © 2020 the Author(s). Published by PNAS.)
- Published
- 2020
- Full Text
- View/download PDF
33. Differential Interactome and Innate Immune Response Activation of Two Structurally Distinct Misfolded Protein Oligomers.
- Author
-
Mannini B, Vecchi G, Labrador-Garrido A, Fabre B, Fani G, Franco JM, Lilley K, Pozo D, Vendruscolo M, Chiti F, Dobson CM, and Roodveldt C
- Subjects
- Animals, Cell Line, Cricetinae, Humans, Mice, Microglia pathology, Protein Aggregation, Pathological pathology, Protein Binding, Carboxyl and Carbamoyl Transferases immunology, Escherichia coli Proteins immunology, Immunity, Innate immunology, Microglia immunology, Protein Aggregation, Pathological immunology
- Abstract
The formation of misfolded protein oligomers during early stages of amyloid aggregation and the activation of neuroinflammatory responses are two key events associated with neurodegenerative diseases. Although it has been established that misfolded oligomers are involved in the neuroinflammatory process, the links between their structural features and their functional effects on the immune response remain unknown. To explore such links, we took advantage of two structurally distinct soluble oligomers (type A and B) of protein HypF-N and compared the elicited microglial inflammatory responses. By using confocal microscopy, protein pull-down, and high-throughput mass spectrometry, we found that, even though both types bound to a common pool of microglial proteins, type B oligomers-with a lower solvent-exposed hydrophobicity-showed enhanced protein binding, correlating with the observed inflammatory response. Furthermore, the interactome associated with inflammatory-mediated neurodegeneration revealed previously unidentified receptors and signaling molecules likely to be involved in the oligomer-elicited innate immune response.
- Published
- 2019
- Full Text
- View/download PDF
34. Trodusquemine enhances Aβ 42 aggregation but suppresses its toxicity by displacing oligomers from cell membranes.
- Author
-
Limbocker R, Chia S, Ruggeri FS, Perni M, Cascella R, Heller GT, Meisl G, Mannini B, Habchi J, Michaels TCT, Challa PK, Ahn M, Casford ST, Fernando N, Xu CK, Kloss ND, Cohen SIA, Kumita JR, Cecchi C, Zasloff M, Linse S, Knowles TPJ, Chiti F, Vendruscolo M, and Dobson CM
- Subjects
- Amyloid beta-Peptides drug effects, Animals, Caenorhabditis elegans, Cell Line, Tumor, Cholestanes pharmacology, Drug Evaluation, Preclinical, Peptide Fragments drug effects, Spermine pharmacology, Spermine therapeutic use, Alzheimer Disease drug therapy, Amyloid beta-Peptides metabolism, Cholestanes therapeutic use, Peptide Fragments metabolism, Spermine analogs & derivatives
- Abstract
Transient oligomeric species formed during the aggregation process of the 42-residue form of the amyloid-β peptide (Aβ
42 ) are key pathogenic agents in Alzheimer's disease (AD). To investigate the relationship between Aβ42 aggregation and its cytotoxicity and the influence of a potential drug on both phenomena, we have studied the effects of trodusquemine. This aminosterol enhances the rate of aggregation by promoting monomer-dependent secondary nucleation, but significantly reduces the toxicity of the resulting oligomers to neuroblastoma cells by inhibiting their binding to the cellular membranes. When administered to a C. elegans model of AD, we again observe an increase in aggregate formation alongside the suppression of Aβ42 -induced toxicity. In addition to oligomer displacement, the reduced toxicity could also point towards an increased rate of conversion of oligomers to less toxic fibrils. The ability of a small molecule to reduce the toxicity of oligomeric species represents a potential therapeutic strategy against AD.- Published
- 2019
- Full Text
- View/download PDF
35. Stabilization and Characterization of Cytotoxic Aβ 40 Oligomers Isolated from an Aggregation Reaction in the Presence of Zinc Ions.
- Author
-
Mannini B, Habchi J, Chia S, Ruggeri FS, Perni M, Knowles TPJ, Dobson CM, and Vendruscolo M
- Subjects
- Alzheimer Disease metabolism, Amyloid beta-Peptides drug effects, Amyloid beta-Peptides metabolism, Amyloid beta-Peptides pharmacology, Animals, Caenorhabditis elegans drug effects, Cell Line, Tumor, Cell Survival drug effects, Humans, In Vitro Techniques, Movement drug effects, Neuroblastoma, Neurons drug effects, Neurons metabolism, Peptide Fragments drug effects, Peptide Fragments metabolism, Peptide Fragments pharmacology, Polymers metabolism, Polymers pharmacology, Protein Conformation, beta-Strand, Zinc pharmacology, Amyloid beta-Peptides chemistry, Peptide Fragments chemistry, Polymers chemistry, Protein Aggregates, Protein Aggregation, Pathological metabolism, Zinc metabolism
- Abstract
Small oligomers formed during the aggregation of certain peptides and proteins are highly cytotoxic in numerous neurodegenerative disorders. Because of their transient nature and conformational heterogeneity, however, the structural and biological features of these oligomers are still poorly understood. Here, we describe a method of generating stable oligomers formed by the Alzheimer's Aβ
40 peptide by carrying out an aggregation reaction in the presence of zinc ions. The resulting oligomers are amenable to detailed biophysical and biological characterization, which reveals a homogeneous population with small size, high cross-β sheet structure content, and extended hydrophobic surface patches. We also show that these oligomers decrease the viability of neuroblastoma cells and impair the motility of C. elegans. The availability of these oligomers offers novel opportunities for studying the mechanisms of Aβ40 toxicity in vitro and in cellular and animal models of Alzheimer's disease.- Published
- 2018
- Full Text
- View/download PDF
36. Multistep Inhibition of α-Synuclein Aggregation and Toxicity in Vitro and in Vivo by Trodusquemine.
- Author
-
Perni M, Flagmeier P, Limbocker R, Cascella R, Aprile FA, Galvagnion C, Heller GT, Meisl G, Chen SW, Kumita JR, Challa PK, Kirkegaard JB, Cohen SIA, Mannini B, Barbut D, Nollen EAA, Cecchi C, Cremades N, Knowles TPJ, Chiti F, Zasloff M, Vendruscolo M, and Dobson CM
- Subjects
- Animals, Caenorhabditis elegans physiology, Cell Line, Cholestanes therapeutic use, Disease Models, Animal, Humans, Neurons drug effects, Neurons metabolism, Parkinson Disease metabolism, Protein Aggregation, Pathological metabolism, Spermine pharmacology, Spermine therapeutic use, Cholestanes pharmacology, Parkinson Disease drug therapy, Protein Aggregates drug effects, Protein Aggregation, Pathological prevention & control, Spermine analogs & derivatives, alpha-Synuclein metabolism
- Abstract
The aggregation of α-synuclein, an intrinsically disordered protein that is highly abundant in neurons, is closely associated with the onset and progression of Parkinson's disease. We have shown previously that the aminosterol squalamine can inhibit the lipid induced initiation process in the aggregation of α-synuclein, and we report here that the related compound trodusquemine is capable of inhibiting not only this process but also the fibril-dependent secondary pathways in the aggregation reaction. We further demonstrate that trodusquemine can effectively suppress the toxicity of α-synuclein oligomers in neuronal cells, and that its administration, even after the initial growth phase, leads to a dramatic reduction in the number of α-synuclein inclusions in a Caenorhabditis elegans model of Parkinson's disease, eliminates the related muscle paralysis, and increases lifespan. On the basis of these findings, we show that trodusquemine is able to inhibit multiple events in the aggregation process of α-synuclein and hence to provide important information about the link between such events and neurodegeneration, as it is initiated and progresses. Particularly in the light of the previously reported ability of trodusquemine to cross the blood-brain barrier and to promote tissue regeneration, the present results suggest that this compound has the potential to be an important therapeutic candidate for Parkinson's disease and related disorders.
- Published
- 2018
- Full Text
- View/download PDF
37. Toxic HypF-N Oligomers Selectively Bind the Plasma Membrane to Impair Cell Adhesion Capability.
- Author
-
Oropesa-Nuñez R, Keshavan S, Dante S, Diaspro A, Mannini B, Capitini C, Cecchi C, Stefani M, Chiti F, and Canale C
- Subjects
- Animals, Bacterial Proteins toxicity, CHO Cells, Cell Membrane drug effects, Cell Proliferation drug effects, Cricetulus, Extracellular Matrix drug effects, Extracellular Matrix metabolism, Protein Binding, Protein Structure, Quaternary, Substrate Specificity, Bacterial Proteins chemistry, Bacterial Proteins metabolism, Cell Adhesion drug effects, Cell Membrane metabolism, Protein Multimerization
- Abstract
The deposition of fibrillar protein aggregates in human organs is the hallmark of several pathological states, including highly debilitating neurodegenerative disorders and systemic amyloidoses. It is widely accepted that small oligomers arising as intermediates in the aggregation process, released by fibrils, or growing in secondary nucleation steps are the cytotoxic entities in protein-misfolding diseases, notably neurodegenerative conditions. Increasing evidence indicates that cytotoxicity is triggered by the interaction between nanosized protein aggregates and cell membranes, even though little information on the molecular details of such interaction is presently available. In this work, we propose what is, to our knowledge, a new approach, based on the use of single-cell force spectroscopy applied to multifunctional substrates, to study the interaction between protein oligomers, cell membranes, and/or the extracellular matrix. We compared the interaction of single Chinese hamster ovary cells with two types of oligomers (toxic and nontoxic) grown from the N-terminal domain of the Escherichia coli protein HypF. We were able to quantify the affinity between both oligomer type and the cell membrane by measuring the mechanical work needed to detach the cells from the aggregates, and we could discriminate the contributions of the membrane lipid and protein fractions to such affinity. The fundamental role of the ganglioside GM1 in the membrane-oligomers interaction was also highlighted. Finally, we observed that the binding of toxic oligomers to the cell membrane significantly affects the functionality of adhesion molecules such as Arg-Gly-Asp binding integrins, and that this effect requires the presence of the negatively charged sialic acid moiety of GM1., (Copyright © 2018. Published by Elsevier Inc.)
- Published
- 2018
- Full Text
- View/download PDF
38. Delivery of Native Proteins into C. elegans Using a Transduction Protocol Based on Lipid Vesicles.
- Author
-
Perni M, Aprile FA, Casford S, Mannini B, Sormanni P, Dobson CM, and Vendruscolo M
- Subjects
- Adsorption, Animals, Animals, Genetically Modified, Caenorhabditis elegans metabolism, Caenorhabditis elegans Proteins metabolism, Disease Models, Animal, Genetic Therapy methods, Humans, Intestinal Absorption, Lipids chemistry, Liposomes administration & dosage, Liposomes chemistry, Liposomes pharmacokinetics, Parkinson Disease metabolism, Parkinson Disease therapy, Reproducibility of Results, alpha-Synuclein chemistry, alpha-Synuclein metabolism, Caenorhabditis elegans genetics, Caenorhabditis elegans Proteins genetics, Parkinson Disease genetics, alpha-Synuclein genetics
- Abstract
The nematode worm Caenorhabditis elegans (C. elegans) is a versatile and widely used animal model for in vivo studies of a broad range of human diseases, in particular for understanding their genetic origins and for screening drug candidates. Nevertheless, the challenges associated with the administration of native proteins to C. elegans have limited the range of applications of this animal model in protein-based drug discovery programs. Here, we describe a readily usable protocol for the transduction of native proteins in C. elegans, which is based on the encapsulation of the proteins of interest within cationic lipid vesicles, prior to their administration to worms. This procedure limits the degradation of the proteins in the guts of the animals, and promotes their adsorption into body tissues. To illustrate the efficacy of this approach we apply it to deliver an antibody designed to inhibit α-synuclein aggregation, and show that it can lead to the rescue of the disease phenotype in a C. elegans model of Parkinson's disease. As this transduction protocol is fast and inexpensive, we anticipate that it will be readily applicable to protein-based drug discovery studies that utilize C. elegans as a model organism.
- Published
- 2017
- Full Text
- View/download PDF
39. Chaperones as Suppressors of Protein Misfolded Oligomer Toxicity.
- Author
-
Mannini B and Chiti F
- Abstract
Chaperones have long been recognized to play well defined functions such as to: (i) assist protein folding and promote formation and maintenance of multisubunit complexes; (ii) mediate protein degradation; (iii) inhibit protein aggregation; and (iv) promote disassembly of undesired aberrant protein aggregates. In addition to these well-established functions, it is increasingly clear that chaperones can also interact with aberrant protein aggregates, such as pre-fibrillar oligomers and fibrils, and inhibit their toxicity commonly associated with neurodegenerative diseases without promoting their disassembly. In particular, the evidence collected so far in different labs, exploiting different experimental approaches and using different chaperones and client aggregated proteins, indicates the existence of two distinct mechanisms of action mediated by the chaperones to neutralize the toxicity of aberrant proteins oligomers: (i) direct binding of the chaperones to the hydrophobic patches exposed on the oligomer/fibril surface, with resulting shielding or masking of the moieties responsible for the aberrant interactions with cellular targets; (ii) chaperone-mediated conversion of aberrant protein aggregates into large and more innocuous species, resulting in a decrease of their surface-to-volume ratio and diffusibility and in deposits more easily manageable by clearance mechanisms, such as autophagy. In this review article we will describe the in vitro and in vivo evidence supporting both mechanisms and how this results in a suppression of the detrimental effects caused by protein misfolded aggregates.
- Published
- 2017
- Full Text
- View/download PDF
40. Systematic development of small molecules to inhibit specific microscopic steps of Aβ42 aggregation in Alzheimer's disease.
- Author
-
Habchi J, Chia S, Limbocker R, Mannini B, Ahn M, Perni M, Hansson O, Arosio P, Kumita JR, Challa PK, Cohen SI, Linse S, Dobson CM, Knowles TP, and Vendruscolo M
- Subjects
- Alzheimer Disease, Amyloid beta-Peptides metabolism, Animals, Caenorhabditis elegans, Cerebrospinal Fluid chemistry, Humans, Peptide Fragments metabolism, Small Molecule Libraries, Amyloid beta-Peptides chemistry, Drug Discovery, Peptide Fragments chemistry
- Abstract
The aggregation of the 42-residue form of the amyloid-β peptide (Aβ42) is a pivotal event in Alzheimer's disease (AD). The use of chemical kinetics has recently enabled highly accurate quantifications of the effects of small molecules on specific microscopic steps in Aβ42 aggregation. Here, we exploit this approach to develop a rational drug discovery strategy against Aβ42 aggregation that uses as a read-out the changes in the nucleation and elongation rate constants caused by candidate small molecules. We thus identify a pool of compounds that target specific microscopic steps in Aβ42 aggregation. We then test further these small molecules in human cerebrospinal fluid and in a Caenorhabditis elegans model of AD. Our results show that this strategy represents a powerful approach to identify systematically small molecule lead compounds, thus offering an appealing opportunity to reduce the attrition problem in drug discovery., Competing Interests: Part of the work described in this paper has been the subject of a patent application filed by Cambridge Enterprise, a wholly owned subsidiary of the University of Cambridge (now licensed to Wren Therapeutics Ltd., where M.V. is Chief Scientific Officer; S.I.A.C., C.M.D., and M.V. are members of the Board of Directors; and S.I.A.C., S.L., C.M.D., and T.P.J.K. are consultants).
- Published
- 2017
- Full Text
- View/download PDF
41. Bis(indolyl)phenylmethane derivatives are effective small molecules for inhibition of amyloid fibril formation by hen lysozyme.
- Author
-
Ramshini H, Mannini B, Khodayari K, Ebrahim-Habibi A, Moghaddasi AS, Tayebee R, and Chiti F
- Subjects
- Dose-Response Relationship, Drug, Humans, Indoles toxicity, Kinetics, MCF-7 Cells, Amyloid chemistry, Indoles chemistry, Indoles pharmacology, Muramidase chemistry, Protein Aggregates drug effects
- Abstract
Amyloid or similar protein aggregates are the hallmarks of many disorders, including Alzheimer's, Parkinson's, Huntington's diseases and amyloidoses. The inhibition of the formation of these aberrant species by small molecules is a promising strategy for disease treatment. However, at present, all such diseases lack an appropriate therapeutic approach based on small molecules. In this work we have evaluated five bis(indolyl)phenylmethane derivatives to reduce amyloid fibril formation by hen egg white lysozyme (HEWL) and its associated cytotoxicity. HEWL is a widely used model system to study the fundamentals of amyloid fibril formation and is heterologous to human lysozyme, which forms amyloid fibrils in a familial form of systemic amyloidosis. HEWL aggregation was tested in the presence and absence of the five compounds, under conditions in which the protein is partially unfolded. To this purpose, various techniques were used, including Congo red and Thioflavin T binding assays, atomic force microscopy, Fourier-Transform Infrared spectroscopy and cell-based cytotoxicity assays, such as the MTT reduction test and the trypan blue test. It was found that all compounds inhibited the formation of amyloid fibrils and their associated toxicity, diverging the aggregation process towards the formation of large, morphologically amorphous, unstructured, nontoxic aggregates, thus resembling class I molecules defined previously. In addition, the five compounds also appeared to disaggregate pre-formed fibrils of HEWL, which categorizes them into class IA. The half maximal inhibitory concentration (IC50) was found to be ca 12.3 ± 1.0 μM for the forefather compound., (Copyright © 2016 Elsevier Masson SAS. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
42. Effect of molecular chaperones on aberrant protein oligomers in vitro: super-versus sub-stoichiometric chaperone concentrations.
- Author
-
Cappelli S, Penco A, Mannini B, Cascella R, Wilson MR, Ecroyd H, Li X, Buxbaum JN, Dobson CM, Cecchi C, Relini A, and Chiti F
- Subjects
- Animals, Carboxyl and Carbamoyl Transferases metabolism, Cell Line, Tumor, Clusterin genetics, Clusterin metabolism, Escherichia coli Proteins metabolism, Humans, Mice, Prealbumin chemistry, Prealbumin genetics, Prealbumin metabolism, Protein Aggregates, Protein Folding, Recombinant Proteins chemistry, Recombinant Proteins genetics, alpha-Crystallin B Chain genetics, alpha-Crystallin B Chain metabolism, Carboxyl and Carbamoyl Transferases chemistry, Clusterin chemistry, Escherichia coli Proteins chemistry, alpha-Crystallin B Chain chemistry
- Abstract
Living systems protect themselves from aberrant proteins by a network of chaperones. We have tested in vitro the effects of different concentrations, ranging from 0 to 16 μm, of two molecular chaperones, namely αB-crystallin and clusterin, and an engineered monomeric variant of transthyretin (M-TTR), on the morphology and cytotoxicity of preformed toxic oligomers of HypF-N, which represent a useful model of misfolded protein aggregates. Using atomic force microscopy imaging and static light scattering analysis, all were found to bind HypF-N oligomers and increase the size of the aggregates, to an extent that correlates with chaperone concentration. SDS-PAGE profiles have shown that the large aggregates were predominantly composed of the HypF-N protein. ANS fluorescence measurements show that the chaperone-induced clustering of HypF-N oligomers does not change the overall solvent exposure of hydrophobic residues on the surface of the oligomers. αB-crystallin, clusterin and M-TTR can diminish the cytotoxic effects of the HypF-N oligomers at all chaperone concentration, as demonstrated by MTT reduction and Ca2+ influx measurements. The observation that the protective effect is primarily at all concentrations of chaperones, both when the increase in HypF-N aggregate size is minimal and large, emphasizes the efficiency and versatility of these protein molecules.
- Published
- 2016
- Full Text
- View/download PDF
43. SERS Detection of Amyloid Oligomers on Metallorganic-Decorated Plasmonic Beads.
- Author
-
Guerrini L, Arenal R, Mannini B, Chiti F, Pini R, Matteini P, and Alvarez-Puebla RA
- Subjects
- Animals, Benzoates chemistry, Cattle, Humans, Metal Nanoparticles ultrastructure, Microspheres, Solutions, Sulfhydryl Compounds chemistry, Amyloid analysis, Gold chemistry, Metal Nanoparticles chemistry, Polystyrenes chemistry, Protein Multimerization, Spectrum Analysis, Raman methods
- Abstract
Protein misfolded proteins are among the most toxic endogenous species of macromolecules. These chemical entities are responsible for neurodegenerative disorders such as Alzheimer's, Parkinson's, Creutzfeldt-Jakob's and different non-neurophatic amyloidosis. Notably, these oligomers show a combination of marked heterogeneity and low abundance in body fluids, which have prevented a reliable detection by immunological methods so far. Herein we exploit the selectivity of proteins to react with metallic ions and the sensitivity of surface-enhanced Raman spectroscopy (SERS) toward small electronic changes in coordination compounds to design and engineer a reliable optical sensor for protein misfolded oligomers. Our strategy relies on the functionalization of Au nanoparticle-decorated polystyrene beads with an effective metallorganic Raman chemoreceptor, composed by Al(3+) ions coordinated to 4-mercaptobenzoic acid (MBA) with high Raman cross-section, that selectively binds aberrant protein oligomers. The mechanical deformations of the MBA phenyl ring upon complexation with the oligomeric species are registered in its SERS spectrum and can be quantitatively correlated with the concentration of the target biomolecule. The SERS platform used here appears promising for future implementation of diagnostic tools of aberrant species associated with protein deposition diseases, including those with a strong social and economic impact, such as Alzheimer's and Parkinson's diseases.
- Published
- 2015
- Full Text
- View/download PDF
44. Toxicity of protein oligomers is rationalized by a function combining size and surface hydrophobicity.
- Author
-
Mannini B, Mulvihill E, Sgromo C, Cascella R, Khodarahmi R, Ramazzotti M, Dobson CM, Cecchi C, and Chiti F
- Subjects
- Carboxyl and Carbamoyl Transferases genetics, Carboxyl and Carbamoyl Transferases toxicity, Circular Dichroism, Escherichia coli Proteins genetics, Escherichia coli Proteins toxicity, Hydrogen-Ion Concentration, Mutation genetics, Carboxyl and Carbamoyl Transferases chemistry, Carboxyl and Carbamoyl Transferases metabolism, Cell Proliferation drug effects, Escherichia coli metabolism, Escherichia coli Proteins chemistry, Escherichia coli Proteins metabolism, Hydrophobic and Hydrophilic Interactions drug effects, Protein Multimerization
- Abstract
The misfolding and aberrant assembly of peptides and proteins into fibrillar aggregates is the hallmark of many pathologies. Fibril formation is accompanied by oligomeric species thought to be the primary pathogenic agents in many of these diseases. With the aim of identifying the structural determinants responsible for the toxicity of misfolded oligomers, we created 12 oligomeric variants from the N-terminal domain of the E. coli HypF protein (HypF-N) by replacing one or more charged amino acid residues with neutral apolar residues and allowing the mutated proteins to aggregate under two sets of conditions. The resulting oligomeric species have different degrees of cytotoxicity when added to the extracellular medium of the cells, as assessed by the extent of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction, apoptosis, and influx of Ca2+ into the cells. The structural properties of the oligomeric variants were characterized by evaluating their surface hydrophobicity with 8-anilinonaphthalene-1-sulfonate (ANS) binding and by measuring their size by means of turbidimetry as well as light scattering. We find that increases in the surface hydrophobicity of the oligomers following mutation can promote the formation of larger assemblies and that the overall toxicity correlates with a combination of both surface hydrophobicity and size, with the most toxic oligomers having high hydrophobicity and small size. These results have allowed the relationships between these three parameters to be studied simultaneously and quantitatively, and have enabled the generation of an equation that is able to rationalize and even predict toxicity of the oligomers resulting from their surface hydrophobicity and size.
- Published
- 2014
- Full Text
- View/download PDF
45. Transthyretin suppresses the toxicity of oligomers formed by misfolded proteins in vitro.
- Author
-
Cascella R, Conti S, Mannini B, Li X, Buxbaum JN, Tiribilli B, Chiti F, and Cecchi C
- Subjects
- Animals, Calcium metabolism, Cells, Cultured, Humans, In Vitro Techniques, Mice, Microscopy, Atomic Force, Models, Molecular, Neuroblastoma metabolism, Neuroblastoma pathology, Neurons metabolism, Neurons pathology, Protein Conformation, Protein Multimerization, Rats, Amyloid beta-Peptides adverse effects, Amyloidogenic Proteins adverse effects, Carboxyl and Carbamoyl Transferases adverse effects, Escherichia coli Proteins adverse effects, Neuroblastoma drug therapy, Neurons drug effects, Prealbumin pharmacology, Protein Folding drug effects
- Abstract
Although human transthyretin (TTR) is associated with systemic amyloidoses, an anti-amyloidogenic effect that prevents Aβ fibril formation in vitro and in animal models has been observed. Here we studied the ability of three different types of TTR, namely human tetramers (hTTR), mouse tetramers (muTTR) and an engineered monomer of the human protein (M-TTR), to suppress the toxicity of oligomers formed by two different amyloidogenic peptides/proteins (HypF-N and Aβ42). muTTR is the most stable homotetramer, hTTR can dissociate into partially unfolded monomers, whereas M-TTR maintains a monomeric state. Preformed toxic HypF-N and Aβ42 oligomers were incubated in the presence of each TTR then added to cell culture media. hTTR, and to a greater extent M-TTR, were found to protect human neuroblastoma cells and rat primary neurons against oligomer-induced toxicity, whereas muTTR had no protective effect. The thioflavin T assay and site-directed labeling experiments using pyrene ruled out disaggregation and structural reorganization within the discrete oligomers following incubation with TTRs, while confocal microscopy, SDS-PAGE, and intrinsic fluorescence measurements indicated tight binding between oligomers and hTTR, particularly M-TTR. Moreover, atomic force microscopy (AFM), light scattering and turbidimetry analyses indicated that larger assemblies of oligomers are formed in the presence of M-TTR and, to a lesser extent, with hTTR. Overall, the data suggest a generic capacity of TTR to efficiently neutralize the toxicity of oligomers formed by misfolded proteins and reveal that such neutralization occurs through a mechanism of TTR-mediated assembly of protein oligomers into larger species, with an efficiency that correlates inversely with TTR tetramer stability., (© 2013.)
- Published
- 2013
- Full Text
- View/download PDF
46. Amyloid-β oligomer synaptotoxicity is mimicked by oligomers of the model protein HypF-N.
- Author
-
Tatini F, Pugliese AM, Traini C, Niccoli S, Maraula G, Ed Dami T, Mannini B, Scartabelli T, Pedata F, Casamenti F, and Chiti F
- Subjects
- Agglutination, Animals, Cells, Cultured, Hippocampus physiopathology, Humans, Long-Term Potentiation drug effects, Male, Neurons physiology, Peptide Fragments, Post-Synaptic Density drug effects, Protein Multimerization, Rats, Rats, Wistar, Structure-Activity Relationship, Alzheimer Disease genetics, Amyloid beta-Peptides chemistry, Amyloid beta-Peptides toxicity, Carboxyl and Carbamoyl Transferases chemistry, Carboxyl and Carbamoyl Transferases toxicity, Escherichia coli Proteins chemistry, Escherichia coli Proteins toxicity, Hippocampus drug effects, Neurons drug effects, Synapses drug effects
- Abstract
Protein misfolded oligomers are thought to be the primary pathogenic species in many protein deposition diseases. Oligomers by the amyloid-β peptide play a central role in Alzheimer's disease pathogenesis, being implicated in synaptic dysfunction. Here we show that the oligomers formed by a protein that has no link with human disease, namely the N-terminal domain of HypF from Escherichia coli (HypF-N), are also synaptotoxic. HypF-N oligomers were found to (i) colocalize with post-synaptic densities in primary rat hippocampal neurons; (ii) induce impairment of long-term potentiation in rat hippocampal slices; and (iii) impair spatial learning of rats in the Morris Water Maze test. By contrast, the native protein and control nontoxic oligomers had none of such effects. These results raise the importance of using HypF-N oligomers as a valid tool to investigate the pathogenesis of Alzheimer's disease, with advantages over other systems for their stability, reproducibility, and costs. The results also suggest that, in the context of a compromised protein homeostasis resulting from aggregation of the amyloid β peptide, a number of oligomeric species sharing common synaptotoxic activity can arise and cooperate in the pathogenesis of the disease., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
47. Salt anions promote the conversion of HypF-N into amyloid-like oligomers and modulate the structure of the oligomers and the monomeric precursor state.
- Author
-
Campioni S, Mannini B, López-Alonso JP, Shalova IN, Penco A, Mulvihill E, Laurents DV, Relini A, and Chiti F
- Subjects
- Circular Dichroism, Hydrogen-Ion Concentration, Microscopy, Atomic Force, Protein Binding, Protein Conformation, Protein Denaturation, Spectrum Analysis, Anions chemistry, Anions metabolism, Carboxyl and Carbamoyl Transferases chemistry, Carboxyl and Carbamoyl Transferases metabolism, Escherichia coli Proteins chemistry, Escherichia coli Proteins metabolism, Protein Multimerization, Salts chemistry, Salts metabolism
- Abstract
An understanding of the solution factors contributing to the rate of aggregation of a protein into amyloid oligomers, to the modulation of the conformational state populated prior to aggregation and to the structure/morphology of the resulting oligomers is one of the goals of present research in this field. We have studied the influence of six different salts on the conversion of the N-terminal domain of Escherichiacoli HypF (HypF-N) into amyloid-like oligomers under conditions of acidic pH. Our results show that salts having different anions (NaCl, NaClO(4), NaI, Na(2)SO(4)) accelerate oligomerization with an efficacy that follows the electroselectivity series of the anions (SO(4)(2-)≥ ClO(4)(-)>I(-)>Cl(-)). By contrast, salts with different cations (NaCl, LiCl, KCl) have similar effects. We also investigated the effect of salts on the structure of the final and initial states of HypF-N aggregation. The electroselectivity series does not apply to the effect of anions on the structure of the oligomers. By contrast, it applies to their effect on the content of secondary structure and on the exposure of hydrophobic clusters of the monomeric precursor state. The results therefore indicate that the binding of anions to the positively charged residues of HypF-N at low pH is the mechanism by which salts modulate the rate of oligomerization and the structure of the monomeric precursor state but not the structure of the resulting oligomers. Overall, the data contribute to rationalize the effect of salts on amyloid-like oligomer formation and to explain the role of charged biological macromolecules in protein aggregation processes., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
48. Glycosaminoglycans (GAGs) suppress the toxicity of HypF-N prefibrillar aggregates.
- Author
-
Saridaki T, Zampagni M, Mannini B, Evangelisti E, Taddei N, Cecchi C, and Chiti F
- Subjects
- Amyloid toxicity, Animals, CHO Cells, Carboxyl and Carbamoyl Transferases toxicity, Cricetinae, Cricetulus, Escherichia coli chemistry, Escherichia coli Proteins toxicity, Protein Denaturation, Amyloid metabolism, Carboxyl and Carbamoyl Transferases metabolism, Escherichia coli Proteins metabolism, Glycosaminoglycans metabolism, Protein Multimerization
- Abstract
A group of diverse human pathologies is associated with proteins unable to retain their native state and convert into prefibrillar and fibrillar amyloid aggregates that are then deposited in the extracellular space. Glycosaminoglycans (GAGs) have been found to physically associate with these deposits and also to promote their formation in vitro. However, the effect of GAGs on the toxicity of these aggregates has been investigated in only one protein system, the amyloid β peptide associated with Alzheimer's disease. In this study, we investigate whether GAGs affect the toxicity of the N-terminal domain of Escherichia coli HypF (HypF-N) oligomers on Chinese hamster ovarian cells and the mechanism by which such suppression is mediated. The results show that heparin and other GAGs inhibit the toxicity observed by HypF-N oligomers in a dose-dependent manner. GAGs were not found to bind preformed HypF-N oligomers, change their morphological and structural characteristics or disaggregate them. Nevertheless, they were found to bind to the cells' surface and prevent the interaction of the oligomers with the cells. Overall, the results indicate that GAGs have a generic ability to inhibit the toxicity of aberrant protein oligomers and that such toxicity suppression can occur through different mechanisms, such as through binding to the oligomers with consequent loss of interaction of the oligomers to the GAGs present on the cell surface, as proposed previously for amyloid β aggregates, or through mechanisms independent of direct GAG-oligomer binding, as shown here for HypF-N aggregates., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
49. Molecular mechanisms used by chaperones to reduce the toxicity of aberrant protein oligomers.
- Author
-
Mannini B, Cascella R, Zampagni M, van Waarde-Verhagen M, Meehan S, Roodveldt C, Campioni S, Boninsegna M, Penco A, Relini A, Kampinga HH, Dobson CM, Wilson MR, Cecchi C, and Chiti F
- Subjects
- Cell Line, Tumor, Humans, Molecular Chaperones genetics, Molecular Chaperones metabolism, Protein Folding, Protein Multimerization
- Abstract
Chaperones are the primary regulators of the proteostasis network and are known to facilitate protein folding, inhibit protein aggregation, and promote disaggregation and clearance of misfolded aggregates inside cells. We have tested the effects of five chaperones on the toxicity of misfolded oligomers preformed from three different proteins added extracellularly to cultured cells. All the chaperones were found to decrease oligomer toxicity significantly, even at very low chaperone/protein molar ratios, provided that they were added extracellularly rather than being overexpressed in the cytosol. Infrared spectroscopy and site-directed labeling experiments using pyrene ruled out structural reorganizations within the discrete oligomers. Rather, confocal microscopy, SDS-PAGE, and intrinsic fluorescence measurements indicated tight binding between oligomers and chaperones. Moreover, atomic force microscopy imaging indicated that larger assemblies of oligomers are formed in the presence of the chaperones. This suggests that the chaperones bind to the oligomers and promote their assembly into larger species, with consequent shielding of the reactive surfaces and a decrease in their diffusional mobility. Overall, the data indicate a generic ability of chaperones to neutralize extracellular misfolded oligomers efficiently and reveal that further assembly of protein oligomers into larger species can be an effective strategy to neutralize such extracellular species.
- Published
- 2012
- Full Text
- View/download PDF
50. A comparison of the biochemical modifications caused by toxic and non-toxic protein oligomers in cells.
- Author
-
Zampagni M, Cascella R, Casamenti F, Grossi C, Evangelisti E, Wright D, Becatti M, Liguri G, Mannini B, Campioni S, Chiti F, and Cecchi C
- Subjects
- Amyloidogenic Proteins chemistry, Amyloidogenic Proteins metabolism, Animals, Apoptosis drug effects, Cell Membrane Permeability drug effects, Cells, Cultured, Cholinergic Neurons chemistry, Disease Models, Animal, Humans, Lipid Peroxidation drug effects, Molecular Targeted Therapy, Neurodegenerative Diseases metabolism, Neurodegenerative Diseases pathology, Rats, Rats, Wistar, Reactive Oxygen Species metabolism, Calcium metabolism, Carboxyl and Carbamoyl Transferases chemistry, Carboxyl and Carbamoyl Transferases pharmacology, Cholinergic Neurons metabolism, Escherichia coli Proteins chemistry, Escherichia coli Proteins pharmacology, Peptides chemistry, Peptides pharmacology
- Abstract
Peptides and proteins can convert from their soluble forms into highly ordered fibrillar aggregates, giving rise to pathological conditions ranging from neurodegenerative disorders to systemic amyloidoses. It is increasingly recognized that protein oligomers forming early in the process of fibril aggregation represent the pathogenic species in protein deposition diseases. The N-terminal domain of the HypF protein from Escherichia coli (HypF-N) has previously been shown to form, under distinct conditions, two types of HypF-N oligomers with indistinguishable morphologies but distinct structural features at the molecular level. Only the oligomer type exposing hydrophobic surfaces and possessing sufficient structural plasticity is toxic (type A), whereas the other type is benign to cultured cells (type B). Here we show that only type A oligomers are able to induce a Ca(2+) influx from the cell medium to the cytosol, to penetrate the plasma membrane, to increase intracellular reactive oxygen species production, lipid peroxidation and release of intracellular calcein, resulting in the activation of the apoptotic pathway. Remarkably, these oligomers can also induce a loss of cholinergic neurons when injected into rat brains. By contrast, markers of cellular stress and viability were unaffected in cultured and rat neuronal cells exposed to type B oligomers. The analysis of the time scales of such effects indicates that the difference of toxicity between the two oligomer types involve the early events of the toxicity cascade, shedding new light on the mechanism of action of protein oligomers and on the molecular targets for the therapeutic intervention against protein deposition diseases., (© 2011 The Authors Journal of Cellular and Molecular Medicine © 2011 Foundation for Cellular and Molecular Medicine/Blackwell Publishing Ltd.)
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.