1
|
Garfagnini T, Ferrari L, Koopman MB, Dekker FA, Halters S, Van Kappel E, Mayer G, Bressler S, Maurice MM, Rüdiger SGD, Friedler A. A Peptide Strategy for Inhibiting Different Protein Aggregation Pathways. Chemistry 2024; 30:e202400080. [PMID: 38972842 DOI: 10.1002/chem.202400080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/27/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
Protein aggregation correlates with many human diseases. Protein aggregates differ in structure and shape. Strategies to develop effective aggregation inhibitors that reach the clinic failed so far. Here, we developed a family of peptides targeting early aggregation stages for both amorphous and fibrillar aggregates of proteins unrelated in sequence and structure. They act on dynamic precursors before mechanistic differentiation takes place. Using peptide arrays, we first identified peptides inhibiting the amorphous aggregation of a molten globular, aggregation-prone mutant of the Axin tumor suppressor. Optimization revealed that the peptides activity did not depend on their sequences but rather on their molecular determinants: a composition of 20-30 % flexible, 30-40 % aliphatic and 20-30 % aromatic residues, a hydrophobicity/hydrophilicity ratio close to 1, and an even distribution of residues of different nature throughout the sequence. The peptides also suppressed fibrillation of Tau, a disordered protein that forms amyloids in Alzheimer's disease, and slowed down that of Huntingtin Exon1, an amyloidogenic protein in Huntington's disease, both entirely unrelated to Axin. Our compounds thus target early stages of different aggregation mechanisms, inhibiting both amorphous and amyloid aggregation. Such cross-mechanistic, multi-targeting aggregation inhibitors may be lead compounds for developing drug candidates against various protein aggregation diseases.
Collapse
Affiliation(s)
- Tommaso Garfagnini
- Institute of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus at Givat Ram, 9190401, Jerusalem, Israel
| | - Luca Ferrari
- Cellular Protein Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584, Utrecht CH, The Netherlands
- Science for Life, Utrecht University, Padualaan 8, 3584, Utrecht CH, The Netherlands
- Max Perutz Labs, Vienna BioCenter (VBC), University of Vienna, Vienna, Austria
| | - Margreet B Koopman
- Cellular Protein Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584, Utrecht CH, The Netherlands
- Science for Life, Utrecht University, Padualaan 8, 3584, Utrecht CH, The Netherlands
| | - Françoise A Dekker
- Cellular Protein Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584, Utrecht CH, The Netherlands
- Science for Life, Utrecht University, Padualaan 8, 3584, Utrecht CH, The Netherlands
| | - Sem Halters
- Cellular Protein Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584, Utrecht CH, The Netherlands
- Science for Life, Utrecht University, Padualaan 8, 3584, Utrecht CH, The Netherlands
| | - Eline Van Kappel
- Oncode Institute, Department of Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, 3584, Utrecht CH, The Netherlands
| | - Guy Mayer
- Institute of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus at Givat Ram, 9190401, Jerusalem, Israel
| | - Shachar Bressler
- Institute of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus at Givat Ram, 9190401, Jerusalem, Israel
| | - Madelon M Maurice
- Oncode Institute, Department of Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, 3584, Utrecht CH, The Netherlands
| | - Stefan G D Rüdiger
- Cellular Protein Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584, Utrecht CH, The Netherlands
- Science for Life, Utrecht University, Padualaan 8, 3584, Utrecht CH, The Netherlands
| | - Assaf Friedler
- Institute of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus at Givat Ram, 9190401, Jerusalem, Israel
| |
Collapse
|
2
|
Cascella R, Banchelli M, Abolghasem Ghadami S, Ami D, Gagliani MC, Bigi A, Staderini T, Tampellini D, Cortese K, Cecchi C, Natalello A, Adibi H, Matteini P, Chiti F. An in situ and in vitro investigation of cytoplasmic TDP-43 inclusions reveals the absence of a clear amyloid signature. Ann Med 2023; 55:72-88. [PMID: 36495262 PMCID: PMC9746631 DOI: 10.1080/07853890.2022.2148734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 11/12/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction: Several neurodegenerative conditions are associated with a common histopathology within neurons of the central nervous system, consisting of the deposition of cytoplasmic inclusions of TAR DNA-binding protein 43 (TDP-43). Such inclusions have variably been described as morphologically and molecularly ordered aggregates having amyloid properties, as filaments without the cross-β-structure and dye binding specific for amyloid, or as amorphous aggregates with no defined structure and fibrillar morphology.Aims and Methods: Here we have expressed human full-length TDP-43 in neuroblastoma x spinal cord 34 (NSC-34) cells to investigate the morphological, structural, and tinctorial properties of TDP-43 inclusions in situ. We have used last-generation amyloid diagnostic probes able to cross the cell membrane and detect amyloid in the cytoplasm and have adopted Raman and Fourier transform infrared microspectroscopies to study in situ the secondary structure of the TDP-43 protein in the inclusions. We have then used transmission electron microscopy to study the morphology of the TDP-43 inclusions.Results: The results show the absence of amyloid dye binding, the lack of an enrichment of cross-β structure in the inclusions, and of a fibrillar texture in the round inclusions. The aggregates formed in vitro from the purified protein under conditions in which it is initially native also lack all these characteristics, ruling out a clear amyloid-like signature.Conclusions: These findings indicate a low propensity of TDP-43 to form amyloid fibrils and even non-amyloid filaments, under conditions in which the protein is initially native and undergoes its typical nucleus-to-cell mislocalization. It cannot be excluded that filaments emerge on the long time scale from such inclusions, but the high propensity of the protein to form initially other types of inclusions appear to be an essential characteristic of TDP-43 proteinopathies.KEY MESSAGESCytoplasmic inclusions of TDP-43 formed in NSC-34 cells do not stain with amyloid-diagnostic dyes, are not enriched with cross-β structure, and do not show a fibrillar morphology.TDP-43 assemblies formed in vitro from pure TDP-43 do not have any hallmarks of amyloid.
Collapse
Affiliation(s)
- Roberta Cascella
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Martina Banchelli
- Institute of Applied Physics “Nello Carrara”, National Research Council, Sesto Fiorentino, Italy
| | | | - Diletta Ami
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy
- Milan Center of Neuroscience (NeuroMI), Milan, Italy
| | - Maria Cristina Gagliani
- Cellular Electron Microscopy Laboratory, Department of Experimental Medicine, University of Genova, Genoa, Italy
| | - Alessandra Bigi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Tommaso Staderini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Davide Tampellini
- U 1195 INSERM-Université Paris-Saclay, Paris, France
- Institut Professeur Baulieu, Paris, France
| | - Katia Cortese
- Cellular Electron Microscopy Laboratory, Department of Experimental Medicine, University of Genova, Genoa, Italy
| | - Cristina Cecchi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Antonino Natalello
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy
- Milan Center of Neuroscience (NeuroMI), Milan, Italy
| | - Hadi Adibi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Paolo Matteini
- Institute of Applied Physics “Nello Carrara”, National Research Council, Sesto Fiorentino, Italy
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| |
Collapse
|
3
|
Perni M, Mannini B, Xu CK, Kumita JR, Dobson CM, Chiti F, Vendruscolo M. Exogenous misfolded protein oligomers can cross the intestinal barrier and cause a disease phenotype in C. elegans. Sci Rep 2021; 11:14391. [PMID: 34257326 PMCID: PMC8277765 DOI: 10.1038/s41598-021-93527-8] [Citation(s) in RCA: 7] [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: 02/14/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
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.
Collapse
Affiliation(s)
- Michele Perni
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Benedetta Mannini
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Catherine K Xu
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Janet R Kumita
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Christopher M Dobson
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy.
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK.
| |
Collapse
|
4
|
Leal-Lasarte M, Mannini B, Chiti F, Vendruscolo M, Dobson CM, Roodveldt C, Pozo D. Distinct responses of human peripheral blood cells to different misfolded protein oligomers. Immunology 2021; 164:358-371. [PMID: 34043816 PMCID: PMC8442237 DOI: 10.1111/imm.13377] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 01/10/2023] Open
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+CD25HighFoxP3+ 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.
Collapse
Affiliation(s)
- Magdalena Leal-Lasarte
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Seville, Spain
| | - Benedetta Mannini
- Section of Biochemistry, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy.,Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Fabrizio Chiti
- Section of Biochemistry, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Michele Vendruscolo
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Christopher M Dobson
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Cintia Roodveldt
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Seville, Spain.,Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - David Pozo
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Seville, Spain.,Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| |
Collapse
|
5
|
Cariati I, Bonanni R, Marini M, Rinaldi AM, Zarrilli B, Tancredi V, Frank C, D’Arcangelo G, Diociaiuti M. Role of Electrostatic Interactions in Calcitonin Prefibrillar Oligomer-Induced Amyloid Neurotoxicity and Protective Effect of Neuraminidase. Int J Mol Sci 2021; 22:ijms22083947. [PMID: 33920464 PMCID: PMC8070249 DOI: 10.3390/ijms22083947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/01/2021] [Accepted: 04/09/2021] [Indexed: 01/06/2023] Open
Abstract
Salmon calcitonin is a good model for studying amyloid behavior and neurotoxicity. Its slow aggregation rate allows the purification of low molecular weight prefibrillar oligomers, which are the most toxic species. It has been proposed that these species may cause amyloid pore formation in neuronal membranes through contact with negatively charged sialic acid residues of the ganglioside GM1. In particular, it has been proposed that an electrostatic interaction may be responsible for the initial contact between prefibrillar oligomers and GM1 contained in lipid rafts. Based on this evidence, the aim of our work was to investigate whether the neurotoxic action induced by calcitonin prefibrillar oligomers could be counteracted by treatment with neuraminidase, an enzyme that removes sialic acid residues from gangliosides. Therefore, we studied cell viability in HT22 cell lines and evaluated the effects on synaptic transmission and long-term potentiation by in vitro extracellular recordings in mouse hippocampal slices. Our results showed that treatment with neuraminidase alters the surface charges of lipid rafts, preventing interaction between the calcitonin prefibrillar oligomers and GM1, and suggesting that the enzyme, depending on the concentration used, may have a partial or total protective action in terms of cell survival and modulation of synaptic transmission.
Collapse
Affiliation(s)
- Ida Cariati
- Medical-Surgical Biotechnologies and Translational Medicine (Phd), Department of Clinical Sciences and Translational Medicine, “Tor Vergata” University of Rome, Via Montpellier 1, 00133 Rome, Italy
- Correspondence:
| | - Roberto Bonanni
- Department of Systems Medicine, “Tor Vergata” University of Rome, Via Montpellier 1, 00133 Rome, Italy; (R.B.); (M.M.); (A.M.R.); (B.Z.); (V.T.); (G.D.)
| | - Mario Marini
- Department of Systems Medicine, “Tor Vergata” University of Rome, Via Montpellier 1, 00133 Rome, Italy; (R.B.); (M.M.); (A.M.R.); (B.Z.); (V.T.); (G.D.)
| | - Anna Maria Rinaldi
- Department of Systems Medicine, “Tor Vergata” University of Rome, Via Montpellier 1, 00133 Rome, Italy; (R.B.); (M.M.); (A.M.R.); (B.Z.); (V.T.); (G.D.)
| | - Beatrice Zarrilli
- Department of Systems Medicine, “Tor Vergata” University of Rome, Via Montpellier 1, 00133 Rome, Italy; (R.B.); (M.M.); (A.M.R.); (B.Z.); (V.T.); (G.D.)
| | - Virginia Tancredi
- Department of Systems Medicine, “Tor Vergata” University of Rome, Via Montpellier 1, 00133 Rome, Italy; (R.B.); (M.M.); (A.M.R.); (B.Z.); (V.T.); (G.D.)
- Centre of Space Bio-Medicine, “Tor Vergata” University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Claudio Frank
- UniCamillus-Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy;
| | - Giovanna D’Arcangelo
- Department of Systems Medicine, “Tor Vergata” University of Rome, Via Montpellier 1, 00133 Rome, Italy; (R.B.); (M.M.); (A.M.R.); (B.Z.); (V.T.); (G.D.)
- Centre of Space Bio-Medicine, “Tor Vergata” University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Marco Diociaiuti
- Centro Nazionale Malattie Rare, Istituto Superiore di Sanità, 00161 Rome, Italy;
| |
Collapse
|
6
|
Farrugia MY, Caruana M, Ghio S, Camilleri A, Farrugia C, Cauchi RJ, Cappelli S, Chiti F, Vassallo N. Toxic oligomers of the amyloidogenic HypF-N protein form pores in mitochondrial membranes. Sci Rep 2020; 10:17733. [PMID: 33082392 PMCID: PMC7575562 DOI: 10.1038/s41598-020-74841-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 10/06/2020] [Indexed: 12/30/2022] Open
Abstract
Studies on the amyloidogenic N-terminal domain of the E. coli HypF protein (HypF-N) have contributed significantly to a detailed understanding of the pathogenic mechanisms in neurodegenerative diseases characterised by the formation of misfolded oligomers, by proteins such as amyloid-β, α-synuclein and tau. Given that both cell membranes and mitochondria are increasingly recognised as key targets of oligomer toxicity, we investigated the damaging effects of aggregates of HypF-N on mitochondrial membranes. Essentially, we found that HypF-N oligomers characterised by high surface hydrophobicity (type A) were able to trigger a robust permeabilisation of mito-mimetic liposomes possessing cardiolipin-rich membranes and dysfunction of isolated mitochondria, as demonstrated by a combination of mitochondrial shrinking, lowering of mitochondrial membrane potential and cytochrome c release. Furthermore, using single-channel electrophysiology recordings we obtained evidence that the type A aggregates induced currents reflecting formation of ion-conducting pores in mito-mimetic planar phospholipid bilayers, with multi-level conductances ranging in the hundreds of pS at negative membrane voltages. Conversely, HypF-N oligomers with low surface hydrophobicity (type B) could not permeabilise or porate mitochondrial membranes. These results suggest an inherent toxicity of membrane-active aggregates of amyloid-forming proteins to mitochondria, and that targeting of oligomer-mitochondrial membrane interactions might therefore afford protection against such damage.
Collapse
Affiliation(s)
- Maria Ylenia Farrugia
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Mario Caruana
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Stephanie Ghio
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Angelique Camilleri
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | | | - Ruben J Cauchi
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Sara Cappelli
- Section of Biochemistry, Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy
| | - Fabrizio Chiti
- Section of Biochemistry, Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy
| | - Neville Vassallo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta.
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta.
| |
Collapse
|
7
|
Skorupa P, Lindenstrauß U, Burschel S, Blumenscheit C, Friedrich T, Pinske C. The N-terminal domains of the paralogous HycE and NuoCD govern assembly of the respective formate hydrogenlyase and NADH dehydrogenase complexes. FEBS Open Bio 2020; 10:371-385. [PMID: 31925988 PMCID: PMC7050243 DOI: 10.1002/2211-5463.12787] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/17/2019] [Accepted: 01/08/2020] [Indexed: 11/23/2022] Open
Abstract
Formate hydrogenlyase (FHL) is the main hydrogen-producing enzyme complex in enterobacteria. It converts formate to CO2 and H2 via a formate dehydrogenase and a [NiFe]-hydrogenase. FHL and complex I are evolutionarily related and share a common core architecture. However, complex I catalyses the fundamentally different electron transfer from NADH to quinone and pumps protons. The catalytic FHL subunit, HycE, resembles NuoCD of Escherichia coli complex I; a fusion of NuoC and NuoD present in other organisms. The C-terminal domain of HycE harbours the [NiFe]-active site and is similar to other hydrogenases, while this domain in NuoCD is involved in quinone binding. The N-terminal domains of these proteins do not bind cofactors and are not involved in electron transfer. As these N-terminal domains are separate proteins in some organisms, we removed them in E. coli and observed that both FHL and complex I activities were essentially absent. This was due to either a disturbed assembly or to complex instability. Replacing the N-terminal domain of HycE with a 180 amino acid E. coli NuoC protein fusion did not restore activity, indicating that the domains have complex-specific functions. A FHL complex in which the N- and C-terminal domains of HycE were physically separated still retained most of its FHL activity, while the separation of NuoCD abolished complex I activity completely. Only the FHL complex tolerates physical separation of the HycE domains. Together, the findings strongly suggest that the N-terminal domains of these proteins are key determinants in complex assembly.
Collapse
Affiliation(s)
- Philipp Skorupa
- Institute of Biology/MicrobiologyMartin‐Luther University Halle‐WittenbergGermany
| | - Ute Lindenstrauß
- Institute of Biology/MicrobiologyMartin‐Luther University Halle‐WittenbergGermany
| | - Sabrina Burschel
- Institute of Biochemistry/Molecular BioenergeticsAlbert‐Ludwigs‐University FreiburgGermany
| | | | - Thorsten Friedrich
- Institute of Biochemistry/Molecular BioenergeticsAlbert‐Ludwigs‐University FreiburgGermany
| | - Constanze Pinske
- Institute of Biology/MicrobiologyMartin‐Luther University Halle‐WittenbergGermany
| |
Collapse
|
8
|
Paredes-Rosan CA, Valencia DE, Barazorda-Ccahuana HL, Aguilar-Pineda JA, Gómez B. Amyloid beta oligomers: how pH influences over trimer and pentamer structures? J Mol Model 2019; 26:1. [PMID: 31834477 DOI: 10.1007/s00894-019-4247-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 10/30/2019] [Indexed: 02/07/2023]
Abstract
The aggregation of proteins in the brain is one of the main features of neurodegenerative diseases. In Alzheimer's disease, the abnormal aggregation of Aβ-42 is due to intrinsic and extrinsic factors. The latter is due to variations in the environment, such as temperature, salt concentration, and pH. We evaluated the effect of protonation/deprotonation of residues that are part of trimeric and pentameric oligomers at pH 5, pH 6, and pH 7. Molecular dynamics simulation at 200 ns in the canonical ensemble was implemented. The results have revealed that histidine, glutamic acid, and aspartic acid residues showed a protonation/deprotonation effect in oligomers. The root mean square deviation analysis was used to analyze the structural stability at different pHs. We found an increase in hydrophobicity in the side chains of the trimer, while in the pentamer, the structural instability of a compact structure at pH 5 caused the hydrophobic core to open, revealing the hydrophobic region to the environment. At this point, we believe that conformational changes mediated by pH are essential in the aggregation of Aβ-42 oligomers.
Collapse
Affiliation(s)
- Carla A Paredes-Rosan
- Centro de Investigación en Ingeniería Molecular - CIIM, Vicerrectorado de Investigación, Universidad Católica de Santa María, Samuel Velarde 320, Arequipa, Peru
| | - Diego E Valencia
- Centro de Investigación en Ingeniería Molecular - CIIM, Vicerrectorado de Investigación, Universidad Católica de Santa María, Samuel Velarde 320, Arequipa, Peru
| | - Haruna L Barazorda-Ccahuana
- Centro de Investigación en Ingeniería Molecular - CIIM, Vicerrectorado de Investigación, Universidad Católica de Santa María, Samuel Velarde 320, Arequipa, Peru
| | - Jorge A Aguilar-Pineda
- Centro de Investigación en Ingeniería Molecular - CIIM, Vicerrectorado de Investigación, Universidad Católica de Santa María, Samuel Velarde 320, Arequipa, Peru
| | - Badhin Gómez
- Centro de Investigación en Ingeniería Molecular - CIIM, Vicerrectorado de Investigación, Universidad Católica de Santa María, Samuel Velarde 320, Arequipa, Peru. .,Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Departamento de Farmacia, Bioquímica y Biotecnología, Universidad Católica de Santa María, Samuel Velarde 320, Arequipa, Peru.
| |
Collapse
|
9
|
Cinnamaldehyde and Phenyl Ethyl Alcohol promote the entrapment of intermediate species of HEWL, as revealed by structural, kinetics and thermal stability studies. Sci Rep 2019; 9:18615. [PMID: 31819148 PMCID: PMC6901479 DOI: 10.1038/s41598-019-55082-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/19/2019] [Indexed: 12/25/2022] Open
Abstract
Numerous efforts have been directed towards investigating the different stages leading to the fibrillation process in neurodegenerative diseases and finding the factors modulating it. In this study, using a wide range of molecular techniques as well as fibrillation kinetics coupled with differential scanning fluorimetry (DSF) and crystal structure determination of HEWL treated with cinnamaldehyde (Cin) and Phenyl ethyl alcohol (PEA) in their aroma form during fibrillation, we were able to identify the binding positions of Cin and PEA in HEWL. Additionally, crystal structures were used to suggest residues Thr43, Asn44, Arg45 and Arg68 as a plausible ‘hotspot’ promoting entrapment of intermediate species in the process of fibril formation in HEWL. We were also able to use DSF to show that Cin can significantly decrease the thermal stability of HEWL, promoting the formation of partially unfolded intermediate species. In conclusion, our data led us to emphasize that compounds in their ‘aroma form’ can influence the structure and stability of protein molecules and suggest reconsideration of HEWL as a model protein for fibrillation studies related to neurodegenerative diseases based on the initial structure of the proteins, whether globular (HEWL) or intrinsically disordered.
Collapse
|
10
|
Mannini B, Vecchi G, Labrador-Garrido A, Fabre B, Fani G, Franco JM, Lilley K, Pozo D, Vendruscolo M, Chiti F, Dobson CM, Roodveldt C. Differential Interactome and Innate Immune Response Activation of Two Structurally Distinct Misfolded Protein Oligomers. ACS Chem Neurosci 2019; 10:3464-3478. [PMID: 31313906 DOI: 10.1021/acschemneuro.9b00088] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
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.
Collapse
Affiliation(s)
- Benedetta Mannini
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, U.K
| | - Giulia Vecchi
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, U.K
| | - Adahir Labrador-Garrido
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER) - Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, 41092 Seville, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, University of Seville, 41092 Seville, Spain
| | - Bertrand Fabre
- Cambridge Centre for Proteomics, Systems Biology Centre, Department of Biochemistry, University of Cambridge, CB2 1GA Cambridge, U.K
| | - Giulia Fani
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, U.K
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, 50134 Florence, Italy
| | - Jaime M. Franco
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER) - Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, 41092 Seville, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, University of Seville, 41092 Seville, Spain
| | - Kathryn Lilley
- Cambridge Centre for Proteomics, Systems Biology Centre, Department of Biochemistry, University of Cambridge, CB2 1GA Cambridge, U.K
| | - David Pozo
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER) - Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, 41092 Seville, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, University of Seville, 41092 Seville, Spain
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, U.K
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, 50134 Florence, Italy
| | - Christopher M. Dobson
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, U.K
| | - Cintia Roodveldt
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER) - Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, 41092 Seville, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, University of Seville, 41092 Seville, Spain
| |
Collapse
|
11
|
Oropesa-Nuñez R, Seghezza S, Dante S, Diaspro A, Cascella R, Cecchi C, Stefani M, Chiti F, Canale C. Interaction of toxic and non-toxic HypF-N oligomers with lipid bilayers investigated at high resolution with atomic force microscopy. Oncotarget 2018; 7:44991-45004. [PMID: 27391440 PMCID: PMC5216700 DOI: 10.18632/oncotarget.10449] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 06/26/2016] [Indexed: 11/25/2022] Open
Abstract
Protein misfolded oligomers are considered the most toxic species amongst those formed in the process of amyloid formation and the molecular basis of their toxicity, although not completely understood, is thought to originate from the interaction with the cellular membrane. Here, we sought to highlight the molecular determinants of oligomer-membrane interaction by atomic force microscopy. We monitored the interaction between multiphase supported lipid bilayers and two types of HypF-N oligomers displaying different structural features and cytotoxicities. By our approach we imaged with unprecedented resolution the ordered and disordered lipid phases of the bilayer and different oligomer structures interacting with either phase. We identified the oligomers and lipids responsible for toxicity and, more generally, we established the importance of the membrane lipid component in mediating oligomer toxicity. Our findings support the importance of GM1 ganglioside in mediating the oligomer-bilayer interaction and support a mechanism of oligomer cytotoxicity involving bilayer destabilization by globular oligomers within GM1-rich ordered raft regions rather than by annular oligomers in the surrounding disordered membrane domains.
Collapse
Affiliation(s)
- Reinier Oropesa-Nuñez
- Department of Nanophysics, Istituto Italiano di Tecnologia, Genova, Italy.,DIBRIS Department, University of Genova, Genova, Italy
| | - Silvia Seghezza
- Department of Nanophysics, Istituto Italiano di Tecnologia, Genova, Italy
| | - Silvia Dante
- Department of Nanophysics, Istituto Italiano di Tecnologia, Genova, Italy
| | - Alberto Diaspro
- Department of Nanophysics, Istituto Italiano di Tecnologia, Genova, Italy.,Department of Physics, University of Genova, Genova, Italy
| | - Roberta Cascella
- Section of Biochemistry, Department of Biomedical Experimental and Clinical Sciences, University of Florence, Firenze, Italy
| | - Cristina Cecchi
- Section of Biochemistry, Department of Biomedical Experimental and Clinical Sciences, University of Florence, Firenze, Italy
| | - Massimo Stefani
- Section of Biochemistry, Department of Biomedical Experimental and Clinical Sciences, University of Florence, Firenze, Italy
| | - Fabrizio Chiti
- Section of Biochemistry, Department of Biomedical Experimental and Clinical Sciences, University of Florence, Firenze, Italy
| | - Claudio Canale
- Department of Nanophysics, Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
12
|
Abdolvahabi A, Shi Y, Rasouli S, Croom CM, Chuprin A, Shaw BF. How Do Gyrating Beads Accelerate Amyloid Fibrillization? Biophys J 2017; 112:250-264. [PMID: 28122213 PMCID: PMC5266089 DOI: 10.1016/j.bpj.2016.12.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 11/22/2016] [Accepted: 12/05/2016] [Indexed: 01/21/2023] Open
Abstract
The chemical and physical mechanisms by which gyrating beads accelerate amyloid fibrillization in microtiter plate assays are unclear. Identifying these mechanisms will help optimize high-throughput screening assays for molecules and mutations that modulate aggregation and might explain why different research groups report different rates of aggregation for identical proteins. This article investigates how the rate of superoxide dismutase-1 (SOD1) fibrillization is affected by 12 different beads with a wide range of hydrophobicity, mass, stiffness, and topology but identical diameter. All assays were performed on D90A apo-SOD1, which is a stable and wild-type-like variant of SOD1. The most significant and uniform correlation between any material property of each bead and that bead's effect on SOD1 fibrillization rate was with regard to bead mass. A linear correlation existed between bead mass and rate of fibril elongation (R2 = 0.7): heavier beads produced faster rates and shorter fibrils. Nucleation rates (lag time) also correlated with bead mass, but only for non-polymeric beads (i.e., glass, ceramic, metallic). The effect of bead mass on fibrillization correlated (R2 = 0.96) with variations in buoyant forces and contact forces (between bead and microplate well), and was not an artifact of residual momentum during intermittent gyration. Hydrophobic effects were observed, but only for polymeric beads: lag times correlated negatively with contact angle of water and degree of protein adhesion (surface adhesion and hydrophobic effects were negligible for non-polymeric beads). These results demonstrate that contact forces (alone) explain kinetic variation among non-polymeric beads, whereas surface hydrophobicity and contact forces explain kinetic variation among polymeric beads. This study also establishes conditions for high-throughput amyloid assays of SOD1 that enable the control over fibril morphologies and produce eightfold faster lag times and fourfold less stochasticity than in previous studies.
Collapse
Affiliation(s)
| | - Yunhua Shi
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas
| | - Sanaz Rasouli
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas; Institute of Biomedical Studies, Baylor University, Waco, Texas
| | - Corbin M Croom
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas
| | - Aleksandra Chuprin
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas
| | - Bryan F Shaw
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas.
| |
Collapse
|
13
|
Kisilevsky R, Raimondi S, Bellotti V. Historical and Current Concepts of Fibrillogenesis and In vivo Amyloidogenesis: Implications of Amyloid Tissue Targeting. Front Mol Biosci 2016; 3:17. [PMID: 27243018 PMCID: PMC4860540 DOI: 10.3389/fmolb.2016.00017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 04/21/2016] [Indexed: 12/22/2022] Open
Abstract
Historical and current concepts of in vitro fibrillogenesis are considered in the light of disorders in which amyloid is deposited at anatomic sites remote from the site of synthesis of the corresponding precursor protein. These clinical conditions set constraints on the interpretation of information derived from in vitro fibrillogenesis studies. They suggest that in addition to kinetic and thermodynamic factors identified in vitro, fibrillogenesis in vivo is determined by site specific factors most of which have yet to be identified.
Collapse
Affiliation(s)
- Robert Kisilevsky
- Department of Pathology and Molecular Medicine, Queen's University Kingston, ON, Canada
| | - Sara Raimondi
- Unit of Biochemistry, Department of Molecular Medicine, University of Pavia Pavia, Italy
| | - Vittorio Bellotti
- Unit of Biochemistry, Department of Molecular Medicine, University of PaviaPavia, Italy; Wolfson Drug Discovery Unit, Division of Medicine, Centre for Amyloidosis and Acute Phase Proteins, University College LondonLondon, UK
| |
Collapse
|
14
|
Iannuzzi C, Irace G, Sirangelo I. The effect of glycosaminoglycans (GAGs) on amyloid aggregation and toxicity. Molecules 2015; 20:2510-28. [PMID: 25648594 PMCID: PMC6272481 DOI: 10.3390/molecules20022510] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 01/29/2015] [Indexed: 01/15/2023] Open
Abstract
Amyloidosis is a protein folding disorder in which normally soluble proteins are deposited extracellularly as insoluble fibrils, impairing tissue structure and function. Charged polyelectrolytes such as glycosaminoglycans (GAGs) are frequently found associated with the proteinaceous deposits in tissues of patients affected by amyloid diseases. Experimental evidence indicate that they can play an active role in favoring amyloid fibril formation and stabilization. Binding of GAGs to amyloid fibrils occurs mainly through electrostatic interactions involving the negative polyelectrolyte charges and positively charged side chains residues of aggregating protein. Similarly to catalyst for reactions, GAGs favor aggregation, nucleation and amyloid fibril formation functioning as a structural templates for the self-assembly of highly cytotoxic oligomeric precursors, rich in β-sheets, into harmless amyloid fibrils. Moreover, the GAGs amyloid promoting activity can be facilitated through specific interactions via consensus binding sites between amyloid polypeptide and GAGs molecules. We review the effect of GAGs on amyloid deposition as well as proteins not strictly related to diseases. In addition, we consider the potential of the GAGs therapy in amyloidosis.
Collapse
Affiliation(s)
- Clara Iannuzzi
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università di Napoli, Via L. De Crecchio 7, Napoli 80138, Italy.
| | - Gaetano Irace
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università di Napoli, Via L. De Crecchio 7, Napoli 80138, Italy.
| | - Ivana Sirangelo
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università di Napoli, Via L. De Crecchio 7, Napoli 80138, Italy.
| |
Collapse
|
15
|
Wagoner VA, Cheon M, Chang I, Hall CK. Impact of sequence on the molecular assembly of short amyloid peptides. Proteins 2014; 82:1469-83. [PMID: 24449257 DOI: 10.1002/prot.24515] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 12/17/2013] [Accepted: 12/27/2013] [Indexed: 11/08/2022]
Abstract
The goal of this work is to understand how the sequence of a protein affects the likelihood that it will form an amyloid fibril and the kinetics along the fibrillization pathway. The focus is on very short fragments of amyloid proteins since these play a role in the fibrillization of the parent protein and can form fibrils themselves. Discontinuous molecular dynamics simulations using the PRIME20 force field were performed of the aggregation of 48-peptide systems containing SNQNNF (PrP (170-175)), SSTSAA (RNaseA(15-20)), MVGGVV (Aβ(35-40)), GGVVIA (Aβ(37-42)), and MVGGVVIA (Aβ(35-42)). In our simulations SNQQNF, SSTTSAA, and MVGGVV form large numbers of fibrillar structures spontaneously (as in experiment). GGVVIA forms β-sheets that do not stack into fibrils (unlike experiment). The combination sequence MVGGVVIA forms less fibrils than MVGGVV, hindered by the presence of the hydrophobic residues at the C-terminal. Analysis of the simulation kinetics and energetics reveals why MVGGVV forms fibrils and GGVVIA does not, and why adding I and A to MVGGVVIA reduces fibrillization and enhances amorphous aggregation into oligomeric structures. The latter helps explain why Aβ(1-42) assembles into more complex oligomers than Aβ(1-40), a consequence of which is that it is more strongly associated with Alzheimer's disease.
Collapse
Affiliation(s)
- Victoria A Wagoner
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, 27695-7905
| | | | | | | |
Collapse
|
16
|
Deshmukh RS, Chaudhary RK, Roy I. Effect of pesticides on the aggregation of mutant huntingtin protein. Mol Neurobiol 2012; 45:405-14. [PMID: 22415443 DOI: 10.1007/s12035-012-8252-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 02/28/2012] [Indexed: 01/08/2023]
Abstract
The classical reports on neurodegeneration concentrate on studying disruption of signalling cascades. Although it is now well recognized that misfolding and aggregation of specific proteins are associated with a majority of these diseases, their role in aggravating the symptoms is not so well understood. Huntington's disease (HD) is a neurodegenerative disorder that results from damage to complex II of mitochondria. In this work, we have studied the effect of mitochondrial complex I inhibitors, viz. 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and rotenone, and complex II inhibitor, viz. 3-nitropropionic acid, on the aggregation of mutant huntingtin (mthtt) protein, whose misfolding and aggregation results in cellular abnormalities characteristic of HD. All three inhibitors were found to accelerate the aggregation of mthtt in vitro, although the amounts of aggregates formed were different in all cases. Thus, apart from their effect on mitochondrial viability, these neurotoxins are capable of interfering with the protein aggregation process and thus, hastening the onset of the disease.
Collapse
Affiliation(s)
- Ruhi S Deshmukh
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, 160 062, India
| | | | | |
Collapse
|
17
|
Shahnawaz M, Soto C. Microcin amyloid fibrils A are reservoir of toxic oligomeric species. J Biol Chem 2012; 287:11665-76. [PMID: 22337880 DOI: 10.1074/jbc.m111.282533] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Microcin E492 (Mcc), a low molecular weight bacteriocin produced by Klebsiella pneumoniae RYC492, has been shown to exist in two forms: soluble forms that are believed to be toxic to the bacterial cell by forming pores and non-toxic fibrillar forms that share similar biochemical and biophysical properties with amyloids associated with several human diseases. Here we report that fibrils polymerized in vitro from soluble forms sequester toxic species that can be released upon changing environmental conditions such as pH, ionic strength, and upon dilution. Our results indicate that basic pH (≥8.5), low NaCl concentrations (≤50 mm), and dilution (>10-fold) destabilize Mcc fibrils into more soluble species that are found to be toxic to the target cells. Additionally, we also found a similar conversion of non-toxic fibrils into highly toxic oligomers using Mcc aggregates produced in vivo. Moreover, the soluble protein released from fibrils is able to rapidly polymerize into amyloid fibrils under fibril-forming conditions and to efficiently seed aggregation of monomeric Mcc. Our findings indicate that fibrillar forms of Mcc constitute a reservoir of toxic oligomeric species that is released into the medium upon changing the environmental conditions. These findings may have substantial implications to understand the dynamic process of interconversion between toxic and non-toxic aggregated species implicated in protein misfolding diseases.
Collapse
Affiliation(s)
- Mohammad Shahnawaz
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, University of Texas Houston Medical School, Houston, Texas 77030, USA
| | | |
Collapse
|
18
|
Zampagni M, Cascella R, Casamenti F, Grossi C, Evangelisti E, Wright D, Becatti M, Liguri G, Mannini B, Campioni S, Chiti F, Cecchi C. A comparison of the biochemical modifications caused by toxic and non-toxic protein oligomers in cells. J Cell Mol Med 2012; 15:2106-16. [PMID: 21155974 PMCID: PMC4394221 DOI: 10.1111/j.1582-4934.2010.01239.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
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 Ca2+ 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.
Collapse
|
19
|
Influence of the electric field on supramolecular structure and properties of amyloid-specific reagent Congo red. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2011; 40:1187-96. [PMID: 21947508 PMCID: PMC3181408 DOI: 10.1007/s00249-011-0750-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 09/08/2011] [Indexed: 11/13/2022]
Abstract
Among specific amyloid ligands, Congo red and its analogues are often considered potential therapeutic compounds. However, the results of the studies so far have not been univocal because the properties of this dye, derived mostly from its supramolecular nature, are still poorly understood. The supramolecular structure of Congo red, formed by π–π stacking of dye molecules, is susceptible to the influence of the electric field, which may significantly facilitate electron delocalization. Consequently, the electric field may generate altered physico-chemical properties of the dye. Enhanced electron delocalization, induced by the electric field, alters the total charge of Congo red, making the dye more acidic (negatively charged). This is a consequence of withdrawing electrons from polar substituents of aromatic rings—sulfonic and amino groups—thus increasing their tendency to dissociate protons. The electric field-induced charge alteration observed in electrophoresis depends on dye concentration. This concentration-dependent charge alteration effect disappears when the supramolecular structure disintegrates in DMSO. Dipoles formed from supramolecular fibrillar species in the electric field become ordered in the solution, introducing the modified arrangement to liquid crystalline phase. Experimental results and theoretical studies provide evidence confirming predictions that the supramolecular character of Congo red is the main reason for its specific properties and reactivity.
Collapse
|
20
|
Wang YK, Huang SC, Wu YF, Chen YC, Lin YL, Nayak M, Lin YR, Chen WH, Chiu YR, Li TTH, Yeh BS, Wu TK. Site-directed mutations of thermostable direct hemolysin from Grimontia hollisae alter its arrhenius effect and biophysical properties. Int J Biol Sci 2011; 7:333-46. [PMID: 21494434 PMCID: PMC3076507 DOI: 10.7150/ijbs.7.333] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2011] [Accepted: 03/23/2011] [Indexed: 11/23/2022] Open
Abstract
Recombinant thermostable direct hemolysin from Grimontia hollisae (Gh-rTDH) exhibits paradoxical Arrhenius effect, where the hemolytic activity is inactivated by heating at 60 °C but is reactivated by additional heating above 80 °C. This study investigated individual or collective mutational effect of Tyr53, Thr59, and Ser63 positions of Gh-rTDH on hemolytic activity, Arrhenius effect, and biophysical properties. In contrast to the Gh-rTDH wild-type (Gh-rTDH(WT)) protein, a 2-fold decrease of hemolytic activity and alteration of Arrhenius effect could be detected from the Gh-rTDH(Y53H/T59I) and Gh-rTDH(T59I/S63T) double-mutants and the Gh-rTDH(Y53H/T59I/S63T) triple-mutant. Differential scanning calorimetry results showed that the Arrhenius effect-loss and -retaining mutants consistently exhibited higher and lower endothermic transition temperatures, respectively, than that of the Gh-rTDH(WT). Circular dichroism measurements of Gh-rTDH(WT) and Gh-rTDH(mut) showed a conspicuous change from a β-sheet to α-helix structure around the endothermic transition temperature. Consistent with the observation is the conformational change of the proteins from native globular form into fibrillar form, as determined by Congo red experiments and transmission electron microscopy.
Collapse
Affiliation(s)
- Yu-Kuo Wang
- 1. Department of Biological Science and Technology, National Chiao Tung University, 30068, Hsin-Chu, Taiwan, Republic of China
| | - Sheng-Cih Huang
- 1. Department of Biological Science and Technology, National Chiao Tung University, 30068, Hsin-Chu, Taiwan, Republic of China
| | - Yi-Fang Wu
- 1. Department of Biological Science and Technology, National Chiao Tung University, 30068, Hsin-Chu, Taiwan, Republic of China
| | - Yu-Ching Chen
- 1. Department of Biological Science and Technology, National Chiao Tung University, 30068, Hsin-Chu, Taiwan, Republic of China
| | - Yen-Ling Lin
- 1. Department of Biological Science and Technology, National Chiao Tung University, 30068, Hsin-Chu, Taiwan, Republic of China
| | - Manoswini Nayak
- 1. Department of Biological Science and Technology, National Chiao Tung University, 30068, Hsin-Chu, Taiwan, Republic of China
| | - Yan Ren Lin
- 1. Department of Biological Science and Technology, National Chiao Tung University, 30068, Hsin-Chu, Taiwan, Republic of China
| | - Wen-Hung Chen
- 1. Department of Biological Science and Technology, National Chiao Tung University, 30068, Hsin-Chu, Taiwan, Republic of China
| | - Yi-Rong Chiu
- 1. Department of Biological Science and Technology, National Chiao Tung University, 30068, Hsin-Chu, Taiwan, Republic of China
| | - Thomas Tien-Hsiung Li
- 2. Institute of Biochemistry, National Chung Hsing University, 40227, Taichung, Taiwan, Republic of China
| | - Bo-Sou Yeh
- 3. Hsin Chu General Hospital, Department of Health, Executive Yuan, Taiwan, Republic of China
| | - Tung-Kung Wu
- 1. Department of Biological Science and Technology, National Chiao Tung University, 30068, Hsin-Chu, Taiwan, Republic of China
| |
Collapse
|
21
|
Ramshini H, Parrini C, Relini A, Zampagni M, Mannini B, Pesce A, Saboury AA, Nemat-Gorgani M, Chiti F. Large proteins have a great tendency to aggregate but a low propensity to form amyloid fibrils. PLoS One 2011; 6:e16075. [PMID: 21249193 PMCID: PMC3020945 DOI: 10.1371/journal.pone.0016075] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Accepted: 12/06/2010] [Indexed: 11/19/2022] Open
Abstract
The assembly of soluble proteins into ordered fibrillar aggregates with cross-β structure is an essential event of many human diseases. The polypeptides undergoing aggregation are generally small in size. To explore if the small size is a primary determinant for the formation of amyloids under pathological conditions we have created two databases of proteins, forming amyloid-related and non-amyloid deposits in human diseases, respectively. The size distributions of the two protein populations are well separated, with the systems forming non-amyloid deposits appearing significantly larger. We have then investigated the propensity of the 486-residue hexokinase-B from Saccharomyces cerevisiae (YHKB) to form amyloid-like fibrils in vitro. This size is intermediate between the size distributions of amyloid and non-amyloid forming proteins. Aggregation was induced under conditions known to be most effective for amyloid formation by normally globular proteins: (i) low pH with salts, (ii) pH 5.5 with trifluoroethanol. In both situations YHKB aggregated very rapidly into species with significant β-sheet structure, as detected using circular dichroism and X-ray diffraction, but a weak Thioflavin T and Congo red binding. Moreover, atomic force microscopy indicated a morphology distinct from typical amyloid fibrils. Both types of aggregates were cytotoxic to human neuroblastoma cells, as indicated by the MTT assay. This analysis indicates that large proteins have a high tendency to form toxic aggregates, but low propensity to form regular amyloid in vivo and that such a behavior is intrinsically determined by the size of the protein, as suggested by the in vitro analysis of our sample protein.
Collapse
Affiliation(s)
- Hassan Ramshini
- Dipartimento di Scienze Biochimiche, Università di Firenze, Florence, Italy
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | | | | | | | - Benedetta Mannini
- Dipartimento di Scienze Biochimiche, Università di Firenze, Florence, Italy
| | | | - Ali Akbar Saboury
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | | | - Fabrizio Chiti
- Dipartimento di Scienze Biochimiche, Università di Firenze, Florence, Italy
- * E-mail:
| |
Collapse
|
22
|
Martin DJ, Ramirez-Alvarado M. Comparison of amyloid fibril formation by two closely related immunoglobulin light chain variable domains. Amyloid 2010; 17:129-36. [PMID: 21077798 PMCID: PMC3018850 DOI: 10.3109/13506129.2010.530081] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Light chain amyloidosis (AL amyloidosis) is a haematological disorder in which a clonal population of B cells expands and secretes enormous amounts of the immunoglobulin light chain protein. These light chains misfold and aggregate into amyloid fibrils, leading to organ dysfunction and death. We have studied the in vitro fibril formation kinetics of two patient-derived immunoglobulin light chain variable domain proteins, designated AL-09 and AL-103, in response to changes in solution conditions. Both proteins are members of the κI O18:O8 germline and therefore are highly similar in sequence, but they presented with different clinical phenotypes. We find that AL-09 forms fibrils more readily and more rapidly than AL-103 in vitro, mirroring the clinical phenotypes of the patients and suggesting a possible connection between the fibril kinetics of the disease protein and the disease progression.
Collapse
Affiliation(s)
- Douglas J Martin
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
23
|
Wang L, Schubert D, Sawaya MR, Eisenberg D, Riek R. Multidimensional structure-activity relationship of a protein in its aggregated states. Angew Chem Int Ed Engl 2010; 49:3904-8. [PMID: 20397175 PMCID: PMC3004770 DOI: 10.1002/anie.201000068] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Lei Wang
- Laboratorium für Physikalische Chemie, ETH Zürich, 8093 Zürich, Switzerland
| | | | | | | | | |
Collapse
|
24
|
Arad-Haase G, Chuartzman SG, Dagan S, Nevo R, Kouza M, Mai BK, Nguyen HT, Li MS, Reich Z. Mechanical unfolding of acylphosphatase studied by single-molecule force spectroscopy and MD simulations. Biophys J 2010; 99:238-47. [PMID: 20655852 PMCID: PMC2895382 DOI: 10.1016/j.bpj.2010.04.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Revised: 03/28/2010] [Accepted: 04/01/2010] [Indexed: 11/30/2022] Open
Abstract
Single-molecule manipulation methods provide a powerful means to study protein transitions. Here we combined single-molecule force spectroscopy and steered molecular-dynamics simulations to study the mechanical properties and unfolding behavior of the small enzyme acylphosphatase (AcP). We find that mechanical unfolding of AcP occurs at relatively low forces in an all-or-none fashion and is decelerated in the presence of a ligand, as observed in solution measurements. The prominent energy barrier for the transition is separated from the native state by a distance that is unusually long for alpha/beta proteins. Unfolding is initiated at the C-terminal strand (beta(T)) that lies at one edge of the beta-sheet of AcP, followed by unraveling of the strand located at the other. The central strand of the sheet and the two helices in the protein unfold last. Ligand binding counteracts unfolding by stabilizing contacts between an arginine residue (Arg-23) and the catalytic loop, as well as with beta(T) of AcP, which renders the force-bearing units of the protein resistant to force. This stabilizing effect may also account for the decelerated unfolding of ligand-bound AcP in the absence of force.
Collapse
Affiliation(s)
- Gali Arad-Haase
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Silvia G. Chuartzman
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Shlomi Dagan
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Reinat Nevo
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Maksim Kouza
- Department of Physics, Michigan Technological University, Houghton, Michigan
| | - Binh Khanh Mai
- Saigon Institute for Computational Science and Technology, Ho Chi Minh City, Vietnam
| | - Hung Tien Nguyen
- Saigon Institute for Computational Science and Technology, Ho Chi Minh City, Vietnam
| | - Mai Suan Li
- Institute of Physics, Polish Academy of Sciences, Warsaw, Poland
| | - Ziv Reich
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
25
|
Radu LC, Yang J, Kopeček J. Self-assembling diblock copolymers of poly[N-(2-hydroxypropyl)methacrylamide] and a beta-sheet peptide. Macromol Biosci 2009; 9:36-44. [PMID: 18855948 PMCID: PMC4599367 DOI: 10.1002/mabi.200800193] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The self-assembly of hybrid diblock copolymers composed of poly(HPMA) and beta-sheet peptide P11 (CH(3)CO-QQRFQWQFEQQ-NH(2)) blocks was investigated. Copolymers were synthesized via thiol-maleimide coupling reaction, by conjugation of semitelechelic poly(HPMA)-SH with maleimide-modified beta-sheet peptide. As expected, CD and CR binding studies showed that the peptide block imposed its beta-sheet structural arrangement on the structure of diblock copolymers. TEM and AFM proved that peptide and these copolymers had the ability to self-assemble into fibrils.
Collapse
Affiliation(s)
- Larisa Cristina Radu
- Department of Bioengineering, University of Utah, Salt Lake City, Utah 84112 USA
| | - Jiyuan Yang
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112 USA
| | - Jindřich Kopeček
- Department of Bioengineering, University of Utah, Salt Lake City, Utah 84112 USA. Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112 USA
| |
Collapse
|
26
|
Shin TM, Isas JM, Hsieh CL, Kayed R, Glabe CG, Langen R, Chen J. Formation of soluble amyloid oligomers and amyloid fibrils by the multifunctional protein vitronectin. Mol Neurodegener 2008; 3:16. [PMID: 18939994 PMCID: PMC2577670 DOI: 10.1186/1750-1326-3-16] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Accepted: 10/21/2008] [Indexed: 11/10/2022] Open
Abstract
Background The multifunctional protein vitronectin is present within the deposits associated with Alzheimer disease (AD), age-related macular degeneration (AMD), atherosclerosis, systemic amyloidoses, and glomerulonephritis. The extent to which vitronectin contributes to amyloid formation within these plaques, which contain misfolded, amyloidogenic proteins, and the role of vitronectin in the pathophysiology of the aforementioned diseases is currently unknown. The investigation of vitronectin aggregation is significant since the formation of oligomeric and fibrillar structures are common features of amyloid proteins. Results We observed vitronectin immunoreactivity in senile plaques of AD brain, which exhibited overlap with the amyloid fibril-specific OC antibody, suggesting that vitronectin is deposited at sites of amyloid formation. Of particular interest is the growing body of evidence indicating that soluble nonfibrillar oligomers may be responsible for the development and progression of amyloid diseases. In this study we demonstrate that both plasma-purified and recombinant human vitronectin readily form spherical oligomers and typical amyloid fibrils. Vitronectin oligomers are toxic to cultured neuroblastoma and retinal pigment epithelium (RPE) cells, possibly via a membrane-dependent mechanism, as they cause leakage of synthetic vesicles. Oligomer toxicity was attenuated in RPE cells by the anti-oligomer A11 antibody. Vitronectin fibrils contain a C-terminal protease-resistant fragment, which may approximate the core region of residues essential to amyloid formation. Conclusion These data reveal the propensity of vitronectin to behave as an amyloid protein and put forth the possibilities that accumulation of misfolded vitronectin may contribute to aggregate formation seen in age-related amyloid diseases.
Collapse
Affiliation(s)
- Thuzar M Shin
- Zilhka Neurogenetic Institute, University of Southern California, Los Angeles, California 90033, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Pellistri F, Bucciantini M, Relini A, Nosi D, Gliozzi A, Robello M, Stefani M. Nonspecific interaction of prefibrillar amyloid aggregates with glutamatergic receptors results in Ca2+ increase in primary neuronal cells. J Biol Chem 2008; 283:29950-60. [PMID: 18676369 DOI: 10.1074/jbc.m803992200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
It is widely reported that the Ca(2+) increase following nonspecific cell membrane permeabilization is among the earliest biochemical modifications in cells exposed to toxic amyloid aggregates. However, more recently receptors with Ca(2+) channel activity such as alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA), N-methyl D-aspartate (NMDA), ryanodine, and inositol 1,4,5-trisphosphate receptors have been proposed as mediators of the Ca(2+) increase in neuronal cells challenged with beta-amyloid peptides. We previously showed that prefibrillar aggregates of proteins not associated with amyloid diseases are toxic to exposed cells similarly to comparable aggregates of disease-associated proteins. In particular, prefibrillar aggregates of the prokaryotic HypF-N were shown to be toxic to different cultured cell lines by eliciting Ca(2+) and reactive oxygen species increases. This study was aimed at assessing whether NMDA and AMPA receptor activations could be considered a generic feature of cell interaction with amyloid aggregates rather than a specific effect of some aggregated protein. Therefore, we investigated whether NMDA and AMPA receptors were involved in the Ca(2+) increase following exposure of rat cerebellar granule cells to HypF-N prefibrillar aggregates. We found that the intracellular Ca(2+) increase was associated with the early activation of NMDA and AMPA receptors, although some nonspecific membrane permeabilization was also observed at longer times of exposure. This result matched a significant co-localization of the aggregates with both receptors on the plasma membrane. Our data support the possibility that glutamatergic channels are generic sites of interaction with the cell membrane of prefibrillar aggregates of different peptides and proteins as well as the key structures responsible for the resulting early membrane permeabilization to Ca(2+).
Collapse
Affiliation(s)
- Francesca Pellistri
- Department of Physics, University of Genoa, Via Dodecaneso, 33, I-16146 Genoa, Italy
| | | | | | | | | | | | | |
Collapse
|
28
|
Rahimi F, Shanmugam A, Bitan G. Structure-function relationships of pre-fibrillar protein assemblies in Alzheimer's disease and related disorders. Curr Alzheimer Res 2008; 5:319-41. [PMID: 18537546 DOI: 10.2174/156720508784533358] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Several neurodegenerative diseases, including Alzheimer's, Parkinson's, Huntington's and prion diseases, are characterized pathognomonically by the presence of intra- and/or extracellular lesions containing proteinaceous aggregates, and by extensive neuronal loss in selective brain regions. Related non-neuropathic systemic diseases, e.g., light-chain and senile systemic amyloidoses, and other organ-specific diseases, such as dialysis-related amyloidosis and type-2 diabetes mellitus, also are characterized by deposition of aberrantly folded, insoluble proteins. It is debated whether the hallmark pathologic lesions are causative. Substantial evidence suggests that these aggregates are the end state of aberrant protein folding whereas the actual culprits likely are transient, pre-fibrillar assemblies preceding the aggregates. In the context of neurodegenerative amyloidoses, the proteinaceous aggregates may eventuate as potentially neuroprotective sinks for the neurotoxic, oligomeric protein assemblies. The pre-fibrillar, oligomeric assemblies are believed to initiate the pathogenic mechanisms that lead to synaptic dysfunction, neuronal loss, and disease-specific regional brain atrophy. The amyloid beta-protein (Abeta), which is believed to cause Alzheimer's disease (AD), is considered an archetypal amyloidogenic protein. Intense studies have led to nominal, functional, and structural descriptions of oligomeric Abeta assemblies. However, the dynamic and metastable nature of Abeta oligomers renders their study difficult. Different results generated using different methodologies under different experimental settings further complicate this complex area of research and identification of the exact pathogenic assemblies in vivo seems daunting. Here we review structural, functional, and biological experiments used to produce and study pre-fibrillar Abeta assemblies, and highlight similar studies of proteins involved in related diseases. We discuss challenges that contemporary researchers are facing and future research prospects in this demanding yet highly important field.
Collapse
Affiliation(s)
- F Rahimi
- Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095-7334, USA
| | | | | |
Collapse
|
29
|
Differentiation increases the resistance of neuronal cells to amyloid toxicity. Neurochem Res 2008; 33:2516-31. [PMID: 18307032 DOI: 10.1007/s11064-008-9627-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2007] [Accepted: 02/06/2008] [Indexed: 10/22/2022]
Abstract
A substantial lack of information is recognized on the features underlying the variable susceptibility to amyloid aggregate toxicity of cells with different phenotypes. Recently, we showed that different cell types are variously affected by early aggregates of a prokaryotic hydrogenase domain (HypF-N). In the present study we investigated whether differentiation affects cell susceptibility to amyloid injury using a human neurotypic SH-SY5Y cell differentiation model. We found that retinoic acid-differentiated cells were significantly more resistant against Abeta1-40, Abeta1-42 and HypF-N prefibrillar aggregate toxicity respect to undifferentiated cells treated similarly. Earlier and sharper increases in cytosolic Ca(2+) and ROS with marked lipid peroxidation and mitochondrial dysfunction were also detected in exposed undifferentiated cells resulting in apoptosis activation. The reduced vulnerability of differentiated cells matched a more efficient Ca(2+)-ATPase equipment and a higher total antioxidant capacity. Finally, increasing the content of membrane cholesterol resulted in a remarkable reduction of vulnerability and ability to bind the aggregates in either undifferentiated and differentiated cells.
Collapse
|
30
|
Canale C, Torrassa S, Rispoli P, Relini A, Rolandi R, Bucciantini M, Stefani M, Gliozzi A. Natively folded HypF-N and its early amyloid aggregates interact with phospholipid monolayers and destabilize supported phospholipid bilayers. Biophys J 2006; 91:4575-88. [PMID: 16997875 PMCID: PMC1779933 DOI: 10.1529/biophysj.106.089482] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Recent data depict membranes as the main sites where proteins/peptides are recruited and concentrated, misfold, and nucleate amyloids; at the same time, membranes are considered key triggers of amyloid toxicity. The N-terminal domain of the prokaryotic hydrogenase maturation factor HypF (HypF-N) in 30% trifluoroethanol undergoes a complex path of fibrillation starting with initial 2-3-nm oligomers and culminating with the appearance of mature fibrils. Oligomers are highly cytotoxic and permeabilize lipid membranes, both biological and synthetic. In this article, we report an in-depth study aimed at providing information on the surface activity of HypF-N and its interaction with synthetic membranes of different lipid composition, either in the native conformation or as amyloid oligomers or fibrils. Like other amyloidogenic peptides, the natively folded HypF-N forms stable films at the air/water interface and inserts into synthetic phospholipid bilayers with efficiencies depending on the type of phospholipid. In addition, HypF-N prefibrillar aggregates interact with, insert into, and disassemble supported phospholipid bilayers similarly to other amyloidogenic peptides. These results support the idea that, at least in most cases, early amyloid aggregates of different peptides and proteins produce similar effects on the integrity of membrane assembly and hence on cell viability.
Collapse
Affiliation(s)
- Claudio Canale
- Department of Physics, University of Genoa, Genoa, Italy
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Baglioni S, Casamenti F, Bucciantini M, Luheshi LM, Taddei N, Chiti F, Dobson CM, Stefani M. Prefibrillar amyloid aggregates could be generic toxins in higher organisms. J Neurosci 2006; 26:8160-7. [PMID: 16885229 PMCID: PMC6673784 DOI: 10.1523/jneurosci.4809-05.2006] [Citation(s) in RCA: 190] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
More than 40 human diseases are associated with fibrillar deposits of specific peptides or proteins in tissue. Amyloid fibrils, or their precursors, can be highly toxic to cells, suggesting their key role in disease pathogenesis. Proteins not associated with any disease are able to form oligomers and amyloid assemblies in vitro displaying structures and cytotoxicity comparable with those of aggregates of disease-related polypeptides. In isolated cells, such toxicity has been shown to result from increased membrane permeability with disruption of ion homeostasis and oxidative stress. Here we microinjected into the nucleus basalis magnocellularis of rat brains aggregates of an Src homology 3 domain and the N-terminal domain of the prokaryotic HypF, neither of which is associated with amyloid disease. Prefibrillar aggregates of both proteins, but not their mature fibrils or soluble monomers, impaired cholinergic neuron viability in a dose-dependent manner similar to that seen in cell cultures. Contrary to the situation with cultured cells, however, under our experimental conditions, cell stress in tissue is not followed by a comparable level of cell death, a result that is very likely to reflect the presence of protective mechanisms reducing aggregate toxicity. These findings support the hypothesis that neurodegenerative disorders result primarily from a generic cell dysfunction caused by early misfolded species in the aggregation process.
Collapse
|
32
|
Carny O, Gazit E. A model for the role of short self-assembled peptides in the very early stages of the origin of life. FASEB J 2006; 19:1051-5. [PMID: 15985527 DOI: 10.1096/fj.04-3256hyp] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The molecular basis of the origin of life is one of the most fundamental questions in modern biology. While the "RNA world" hypothesis offers a very sensible model for the evolvement of the current biochemical networks, there is a lack of knowledge about the early steps that led to the formation of the first RNA molecules. This issue is essential as it is practically impossible that complex molecules as functional RNA oligonucleotides had evolved spontaneously. It was recently demonstrated that peptide molecules as simple as dipeptides can self-assemble into well-ordered tubular, fibrilar, and closed-cage structures. Other studies have confirmed the ability of dipeptides to act as catalysts and the capability of other peptides, as short as tripeptides, to serve as a template for nucleotide binding and orientation. Unlike complex RNA molecules, the spontaneous formation of functional short peptides in the primordial earth conditions is very likely. We suggest a novel mechanism for the origin of life that is based on the ability of short peptides to form encapsulated structures, catalyst chemical reaction, and serve as highly ordered template for the assembly of nucleotides. This model may explain the early events that led to the formation of the current biochemical machinery that combines the elaborated and coordinated interaction between nucleic acids and proteins to allow the function of living systems.
Collapse
Affiliation(s)
- Ohad Carny
- Department of Molecular Microbiology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | | |
Collapse
|
33
|
Nguyen HD, Hall CK. Spontaneous fibril formation by polyalanines; discontinuous molecular dynamics simulations. J Am Chem Soc 2006; 128:1890-901. [PMID: 16464090 PMCID: PMC3215763 DOI: 10.1021/ja0539140] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Fibrillary protein aggregates rich in beta-sheet structure have been implicated in the pathology of several neurodegenerative diseases. In this work, we investigate the formation of fibrils by performing discontinuous molecular dynamics simulations on systems containing 12 to 96 model Ac-KA(14)K-NH(2) peptides using our newly developed off-lattice, implicit-solvent, intermediate-resolution model, PRIME. We find that, at a low concentration, random-coil peptides assemble into alpha-helices at low temperatures. At intermediate concentrations, random-coil peptides assemble into alpha-helices at low temperatures and large beta-sheet structures at high temperatures. At high concentrations, the system forms beta-sheets over a wide range of temperatures. These assemble into fibrils above a critical temperature which decreases with concentration and exceeds the isolated peptide's folding temperature. At very high temperatures and all concentrations, the system is in a random-coil state. All of these results are in good qualitative agreement with those by Blondelle and co-workers on Ac-KA(14)K-NH(2) peptides. The fibrils observed in our simulations mimic the structural characteristics observed in experiments in terms of the number of sheets formed, the values of the intra- and intersheet separations, and the parallel peptide arrangement within each beta-sheet. Finally, we find that when the strength of the hydrophobic interaction between nonpolar side chains is high compared to the strength of hydrogen bonding, amorphous aggregates, rather than fibrillar aggregates, are formed.
Collapse
Affiliation(s)
- Hung D Nguyen
- Department of Chemical Engineering, North Carolina State University, Raleigh, NC 27695-7905, USA
| | | |
Collapse
|
34
|
Stathopulos PB, Scholz GA, Hwang YM, Rumfeldt JAO, Lepock JR, Meiering EM. Sonication of proteins causes formation of aggregates that resemble amyloid. Protein Sci 2004; 13:3017-27. [PMID: 15459333 PMCID: PMC2286572 DOI: 10.1110/ps.04831804] [Citation(s) in RCA: 306] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Despite the widespread use of sonication in medicine, industry, and research, the effects of sonication on proteins remain poorly characterized. We report that sonication of a range of structurally diverse proteins results in the formation of aggregates that have similarities to amyloid aggregates. The formation of amyloid is associated with, and has been implicated in, causing of a wide range of protein conformational disorders including Alzheimer's disease, Huntington's disease, Parkinson's disease, and prion diseases. The aggregates cause large enhancements in fluorescence of the dye thioflavin T, exhibit green-gold birefringence upon binding the dye Congo red, and cause a red-shift in the absorbance spectrum of Congo red. In addition, circular dichroism reveals that sonication-induced aggregates have high beta-content, and proteins with significant native alpha-helical structure show increased beta-structure in the aggregates. Ultrastructural analysis by electron microscopy reveals a range of morphologies for the sonication-induced aggregates, including fibrils with diameters of 5-20 nm. The addition of preformed aggregates to unsonicated protein solutions results in accelerated and enhanced formation of additional aggregates upon heating. The dye-binding and structural characteristics, as well as the ability of the sonication-induced aggregates to seed the formation of new aggregates are all similar to the properties of amyloid. These results have important implications for the use of sonication in food, biotechnological and medical applications, and for research on protein aggregation and conformational disorders.
Collapse
Affiliation(s)
- Peter B Stathopulos
- Guelph-Waterloo Centre for Graduate Studies in Chemistry and Biochemistry, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | | | | | | | | | | |
Collapse
|
35
|
Stefani M, Dobson CM. Protein aggregation and aggregate toxicity: new insights into protein folding, misfolding diseases and biological evolution. J Mol Med (Berl) 2003; 81:678-99. [PMID: 12942175 DOI: 10.1007/s00109-003-0464-5] [Citation(s) in RCA: 1234] [Impact Index Per Article: 56.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2003] [Accepted: 06/03/2003] [Indexed: 10/26/2022]
Abstract
The deposition of proteins in the form of amyloid fibrils and plaques is the characteristic feature of more than 20 degenerative conditions affecting either the central nervous system or a variety of peripheral tissues. As these conditions include Alzheimer's, Parkinson's and the prion diseases, several forms of fatal systemic amyloidosis, and at least one condition associated with medical intervention (haemodialysis), they are of enormous importance in the context of present-day human health and welfare. Much remains to be learned about the mechanism by which the proteins associated with these diseases aggregate and form amyloid structures, and how the latter affect the functions of the organs with which they are associated. A great deal of information concerning these diseases has emerged, however, during the past 5 years, much of it causing a number of fundamental assumptions about the amyloid diseases to be re-examined. For example, it is now apparent that the ability to form amyloid structures is not an unusual feature of the small number of proteins associated with these diseases but is instead a general property of polypeptide chains. It has also been found recently that aggregates of proteins not associated with amyloid diseases can impair the ability of cells to function to a similar extent as aggregates of proteins linked with specific neurodegenerative conditions. Moreover, the mature amyloid fibrils or plaques appear to be substantially less toxic than the pre-fibrillar aggregates that are their precursors. The toxicity of these early aggregates appears to result from an intrinsic ability to impair fundamental cellular processes by interacting with cellular membranes, causing oxidative stress and increases in free Ca2+ that eventually lead to apoptotic or necrotic cell death. The 'new view' of these diseases also suggests that other degenerative conditions could have similar underlying origins to those of the amyloidoses. In addition, cellular protection mechanisms, such as molecular chaperones and the protein degradation machinery, appear to be crucial in the prevention of disease in normally functioning living organisms. It also suggests some intriguing new factors that could be of great significance in the evolution of biological molecules and the mechanisms that regulate their behaviour.
Collapse
Affiliation(s)
- Massimo Stefani
- Department of Biochemical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy.
| | | |
Collapse
|
36
|
Graether SP, Slupsky CM, Sykes BD. Freezing of a fish antifreeze protein results in amyloid fibril formation. Biophys J 2003; 84:552-7. [PMID: 12524307 PMCID: PMC1302635 DOI: 10.1016/s0006-3495(03)74874-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Amyloid is associated with a number of diseases including Alzheimer's, Huntington's, Parkinson's, and the spongiform encephalopathies. Amyloid fibrils have been formed in vitro from both disease and nondisease related proteins, but the latter requires extremes of pH, heat, or the presence of a chaotropic agent. We show, using fluorescence spectroscopy, electron microscopy, and solid-state NMR spectroscopy, that the alpha-helical type I antifreeze protein from the winter flounder forms amyloid fibrils at pH 4 and 7 upon freezing and thawing. Our results demonstrate that the freezing of some proteins may accelerate the formation of amyloid fibrils.
Collapse
Affiliation(s)
- Steffen P Graether
- Canadian Institute of Health Research Group in Protein Structure and Function, Department of Biochemistry and Protein Engineering Network of Centres of Excellence, University of Alberta, Edmonton, T6G 2H7 Canada
| | | | | |
Collapse
|