Umeå University's logo

umu.sePublications
System disruptions
We are currently experiencing disruptions on the search portals due to high traffic. We are working to resolve the issue, you may temporarily encounter an error message.
Change search
Link to record
Permanent link

Direct link
Morozova-Roche, LudmillaORCID iD iconorcid.org/0000-0001-5886-2023
Alternative names
Publications (10 of 125) Show all publications
Pashaei, S., Shabani, S., Mohammadi, S., Morozova-Roche, L., Salari, N., Rahimi, Z. & Khodarahmi, R. (2025). Differential expression of neurodegeneration-related genes in SH-SY5Y neuroblastoma cells under the influence of Cyclophilin A: could the enzyme be a likely trigger and therapeutic target for Alzheimer's disease?. Neurochemical Research, 50(1), Article ID 47.
Open this publication in new window or tab >>Differential expression of neurodegeneration-related genes in SH-SY5Y neuroblastoma cells under the influence of Cyclophilin A: could the enzyme be a likely trigger and therapeutic target for Alzheimer's disease?
Show others...
2025 (English)In: Neurochemical Research, ISSN 0364-3190, E-ISSN 1573-6903, Vol. 50, no 1, article id 47Article in journal (Refereed) Published
Abstract [en]

The function and mechanism of Cyclophilin A (CypA) in modulating gene expression associated with Alzheimer’s disease (AD) remain unclear. This multifunctional protein is found to be elevated in the cerebrospinal fluid (CSF) of individuals at risk for AD. The cytotoxic effects of CypA, including both wild-type and the mutant R55A, were assessed using the MTT assay. Prior to this evaluation, the purified recombinant protein was validated through enzymatic activity assays and western blot analysis. Following treatment with CypA and transient transfection using the CypA construct, real-time PCR (qRT-PCR) and western blotting were conducted to analyze the expression of factors involved in various signaling pathways, with an emphasis on inflammation, cell death, and intercellular communication. The findings indicate that CypA has a significant impact on the gene expression of factors associated with inflammation and the progression of AD in SH-SY5Y cells. It can be concluded that CypA is capable of regulating gene expression in SH-SY5Y cells, either in a manner dependent on or independent of its enzymatic activity. Additionally, the influence of this multifunctional protein on gene expression is contingent upon the specific site of action, as well as the dosage and duration of exposure to the cells.

Place, publisher, year, edition, pages
Springer Nature, 2025
Keywords
Alzheimer’s disease, Cyclophilin A, Gene expression, Inflammation, Post translational modifications
National Category
Neurosciences Medical Biotechnology (with a focus on Cell Biology (including Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy)
Identifiers
urn:nbn:se:umu:diva-233334 (URN)10.1007/s11064-024-04253-2 (DOI)001371017900002 ()39636462 (PubMedID)2-s2.0-85211328280 (Scopus ID)
Available from: 2025-01-02 Created: 2025-01-02 Last updated: 2025-01-02Bibliographically approved
Ghosh, S., Tamilselvi, S., Williams, C., Jayaweera, S. W., Iashchishyn, I. A., Šulskis, D., . . . Morozova-Roche, L. (2024). ApoE isoforms inhibit amyloid aggregation of proinflammatory protein S100a9. International Journal of Molecular Sciences, 25(4), Article ID 2114.
Open this publication in new window or tab >>ApoE isoforms inhibit amyloid aggregation of proinflammatory protein S100a9
Show others...
2024 (English)In: International Journal of Molecular Sciences, ISSN 1661-6596, E-ISSN 1422-0067, Vol. 25, no 4, article id 2114Article in journal (Refereed) Published
Abstract [en]

Increasing evidence suggests that the calcium-binding and proinflammatory protein S100A9 is an important player in neuroinflammation-mediated Alzheimer's disease (AD). The amyloid co-aggregation of S100A9 with amyloid-beta (A beta) is an important hallmark of this pathology. Apolipoprotein E (ApoE) is also known to be one of the important genetic risk factors of AD. ApoE primarily exists in three isoforms, ApoE2 (Cys112/Cys158), ApoE3 (Cys112/Arg158), and ApoE4 (Arg112/Arg158). Even though the difference lies in just two amino acid residues, ApoE isoforms produce differential effects on the neuroinflammation and activation of the microglial state in AD. Here, we aim to understand the effect of the ApoE isoforms on the amyloid aggregation of S100A9. We found that both ApoE3 and ApoE4 suppress the aggregation of S100A9 in a concentration-dependent manner, even at sub-stoichiometric ratios compared to S100A9. These interactions lead to a reduction in the quantity and length of S100A9 fibrils. The inhibitory effect is more pronounced if ApoE isoforms are added in the lipid-free state versus lipidated ApoE. We found that, upon prolonged incubation, S100A9 and ApoE form low molecular weight complexes with stochiometric ratios of 1:1 and 2:1, which remain stable under SDS-gel conditions. These complexes self-assemble also under the native conditions; however, their interactions are transient, as revealed by glutaraldehyde cross-linking experiments and molecular dynamics (MD) simulation. MD simulation demonstrated that the lipid-binding C-terminal domain of ApoE and the second EF-hand calcium-binding motif of S100A9 are involved in these interactions. We found that amyloids of S100A9 are cytotoxic to neuroblastoma cells, and the presence of either ApoE isoforms does not change the level of their cytotoxicity. A significant inhibitory effect produced by both ApoE isoforms on S100A9 amyloid aggregation can modulate the amyloid-neuroinflammatory cascade in AD.

Place, publisher, year, edition, pages
MDPI, 2024
Keywords
amyloid, apolipoprotein E, proinflammatory, neurodegeneration, neuroinflammation, Alzheimer's disease, cytotoxicity, fibrils, inhibition
National Category
Neurosciences
Identifiers
urn:nbn:se:umu:diva-228711 (URN)10.3390/ijms25042114 (DOI)001170070200001 ()38396791 (PubMedID)2-s2.0-85187311676 (Scopus ID)
Available from: 2024-08-21 Created: 2024-08-21 Last updated: 2024-08-21Bibliographically approved
Leri, M., Sun, D., Svedružic, Ž. M., Šulskis, D., Smirnovas, V., Stefani, M., . . . Bucciantini, M. (2024). Pro-inflammatory protein S100A9 targeted by a natural molecule to prevent neurodegeneration onset. International Journal of Biological Macromolecules, 276(Part 2), Article ID 133838.
Open this publication in new window or tab >>Pro-inflammatory protein S100A9 targeted by a natural molecule to prevent neurodegeneration onset
Show others...
2024 (English)In: International Journal of Biological Macromolecules, ISSN 0141-8130, E-ISSN 1879-0003, Vol. 276, no Part 2, article id 133838Article in journal (Refereed) Published
Abstract [en]

Accumulation of the pro-inflammatory protein S100A9 has been implicated in neuroinflammatory cascades in neurodegenerative diseases (NDs) such as Alzheimer's disease (AD) and Parkinson's disease (PD). S100A9 co-aggregates with other proteins such as α-synuclein in PD and Aβ in AD, contributing to amyloid plaque formation and neurotoxicity. The amyloidogenic nature of this protein and its role in chronic neuroinflammation suggest that it may play a key role in the pathophysiology of these diseases. Research into molecules targeting S100A9 could be a potential therapeutic strategy to prevent its amyloidogenic self-assembly and to attenuate the neuroinflammatory response in affected brain tissue. This work suggests that bioactive natural molecules, such as those found in the Mediterranean diet, may have the potential to alleviate neuroinflammation associated with the accumulation of proteins such as S100A9 in neurodegenerative diseases. A major component of extra virgin olive oil (EVOO), hydroxytyrosol (HT), with its ability to interact with and modulate S100A9 amyloid self-assembly and expression, offers a compelling approach for the development of novel and effective interventions for the prevention and treatment of ND. The findings highlight the importance of exploring natural compounds, such as HT, as potential therapeutic options for these complex and challenging neurological conditions.

Place, publisher, year, edition, pages
Elsevier, 2024
Keywords
Amyloid aggregation, Natural polyphenols, Neuro-inflammation, S100A9 protein
National Category
Medical Biotechnology (with a focus on Cell Biology (including Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy) Neurosciences
Identifiers
urn:nbn:se:umu:diva-228101 (URN)10.1016/j.ijbiomac.2024.133838 (DOI)001278870400001 ()39002917 (PubMedID)2-s2.0-85199166676 (Scopus ID)
Funder
Swedish Society of Medicine, 2022-00638
Available from: 2024-08-06 Created: 2024-08-06 Last updated: 2024-08-06Bibliographically approved
Naaman, E., Qarawani, A., Ben-Zvi Elimelech, R., Harel, M., Sigal-Dror, S., Safuri, S., . . . Zayit-Soudry, S. (2024). The surprising nonlinear effects of S100A9 proteins in the retina. ACS Chemical Neuroscience, 15(4), 735-744
Open this publication in new window or tab >>The surprising nonlinear effects of S100A9 proteins in the retina
Show others...
2024 (English)In: ACS Chemical Neuroscience, E-ISSN 1948-7193, Vol. 15, no 4, p. 735-744Article in journal (Refereed) Published
Abstract [en]

Age-related macular degeneration (AMD) is a complex disease in which inflammation is implicated as a key factor but the precise molecular mechanisms are poorly understood. AMD lesions contain an excess of the pro-inflammatory S100A9 protein, but its retinal significance was yet unexplored. S100A9 was shown to be intrinsically amyloidogenic in vitro and in vivo. Here, we hypothesized that the retinal effects of S100A9 are related to its supramolecular conformation. ARPE-19 cultures were treated with native dimeric and fibrillar S100A9 preparations, and cell viability was determined. Wild-type rats were treated intravitreally with the S100A9 solutions in the right eye and with the vehicle in the left. Retinal function was assessed longitudinally by electroretinography (ERG), comparing the amplitudes and configurations for each intervention. Native S100A9 had no impact on cellular viability in vitro or on the retinal function in vivo. Despite dispersed intracellular uptake, fibrillar S100A9 did not decrease ARPE-19 cell viability. In contrast, S100A9 fibrils impaired retinal function in vivo following intravitreal injection in rats. Intriguingly, low-dose fibrillar S100A9 induced contrasting in vivo effects, significantly increasing the ERG responses, particularly over 14 days postinjection. The retinal effects of S100A9 were further characterized by glial and microglial cell activation. We provide the first indication for the retinal effects of S100A9, showing that its fibrils inflicted retinal dysfunction and glial activation in vivo, while low dose of the same assemblies resulted in an unpredicted enhancement of the ERG amplitudes. These nonlinear responses highlight the consequences of self-assembly of S100A9 and provide insight into its pathophysiological and possibly physiological roles in the retina.

Place, publisher, year, edition, pages
American Chemical Society (ACS), 2024
Keywords
age-related macular degeneration, neurodegeneration, retina, retinal amyloids, retinal inflammation, S100A9
National Category
Ophthalmology Neurosciences
Identifiers
urn:nbn:se:umu:diva-221649 (URN)10.1021/acschemneuro.3c00650 (DOI)38324770 (PubMedID)2-s2.0-85185332117 (Scopus ID)
Available from: 2024-03-05 Created: 2024-03-05 Last updated: 2024-03-05Bibliographically approved
Sohrabi, T., Mirzaei-Behbahani, B., Zadali, R., Pirhaghi, M., Morozova-Roche, L. & Meratan, A. A. (2023). Common mechanisms underlying α-synuclein-induced mitochondrial dysfunction in Parkinson's disease. Journal of Molecular Biology, 435(12), Article ID 167992.
Open this publication in new window or tab >>Common mechanisms underlying α-synuclein-induced mitochondrial dysfunction in Parkinson's disease
Show others...
2023 (English)In: Journal of Molecular Biology, ISSN 0022-2836, E-ISSN 1089-8638, Vol. 435, no 12, article id 167992Article, review/survey (Refereed) Published
Abstract [en]

Parkinson's disease (PD) is the most common neurological movement disorder characterized by the selective and irreversible loss of dopaminergic neurons in substantia nigra pars compacta resulting in dopamine deficiency in the striatum. While most cases are sporadic or environmental, about 10% of patients have a positive family history with a genetic cause. The misfolding and aggregation of α-synuclein (α-syn) as a casual factor in the pathogenesis of PD has been supported by a great deal of literature. Extensive studies of mechanisms underpinning degeneration of the dopaminergic neurons induced by α-syn dysfunction suggest a complex process that involves multiple pathways, including mitochondrial dysfunction and increased oxidative stress, impaired calcium homeostasis through membrane permeabilization, synaptic dysfunction, impairment of quality control systems, disruption of microtubule dynamics and axonal transport, endoplasmic reticulum/Golgi dysfunction, nucleus malfunction, and microglia activation leading to neuroinflammation. Among them mitochondrial dysfunction has been considered as the most primary target of α-syn-induced toxicity, leading to neuronal cell death in both sporadic and familial forms of PD. Despite reviewing many aspects of PD pathogenesis related to mitochondrial dysfunction, a systemic study on how α-syn malfunction/aggregation damages mitochondrial functionality and leads to neurodegeneration is missing in the literature. In this review, we give a detailed molecular overview of the proposed mechanisms by which α-syn, directly or indirectly, contributes to mitochondrial dysfunction. This may provide valuable insights for development of new therapeutic approaches in relation to PD. Antioxidant-based therapy as a potential strategy to protect mitochondria against oxidative damage, its challenges, and recent developments in the field are discussed.

Place, publisher, year, edition, pages
Elsevier, 2023
Keywords
mitochondrial dysfunction, neurodegeneration, oxidative stress, Parkinson's disease, α-synuclein
National Category
Neurosciences
Identifiers
urn:nbn:se:umu:diva-205371 (URN)10.1016/j.jmb.2023.167992 (DOI)001013488800001 ()36736886 (PubMedID)2-s2.0-85148351527 (Scopus ID)
Funder
Umeå University
Available from: 2023-03-27 Created: 2023-03-27 Last updated: 2024-08-23Bibliographically approved
Bai, Q., Sun, D., Zeng, Y., Zhu, J., Zhang, C., Zhang, X., . . . Morozova-Roche, L. (2023). Effect of proinflammatory S100A9 protein on migration and proliferation of microglial cells. Journal of Molecular Neuroscience, 73(11-12), 983-995
Open this publication in new window or tab >>Effect of proinflammatory S100A9 protein on migration and proliferation of microglial cells
Show others...
2023 (English)In: Journal of Molecular Neuroscience, ISSN 0895-8696, E-ISSN 1559-1166, Vol. 73, no 11-12, p. 983-995Article in journal (Refereed) Published
Abstract [en]

Alzheimer’s disease (AD) is a multifactorial disease affecting aging population worldwide. Neuroinflammation became a focus of research as one of the major pathologic processes relating to the disease onset and progression. Proinflammatory S100A9 is the central culprit in the amyloid-neuroinflammatory cascade implicated in AD and other neurodegenerative diseases. We studied the effect of S100A9 on microglial BV-2 cell proliferation and migration. The responses of BV-2 cells to S100A9 stimulation were monitored in real-time using live cell microscopy, transcriptome sequencing, immunofluorescence staining, western blot analysis, and ELISA. We observed that a low dose of S100A9 promotes migration and proliferation of BV-2 cells. However, acute inflammatory condition (i.e., high S100A9 doses) causes diminished cell viability; it is uncovered that S100A9 activates TLR-4 and TLR-7 signaling pathways, leading to TNF-α and IL-6 expression, which affect BV-2 cell migration and proliferation in a concentration-dependent manner. Interestingly, the effects of S100A9 are not only inhibited by TNF-α and IL-6 antibodies. The addition of amyloid-β (Aβ) 1–40 peptide resumes the capacities of BV-2 cells to the level of low S100A9 concentrations. Based on these results, we conclude that in contrast to the beneficial effects of low S100A9 dose, high S100A9 concentration leads to impaired mobility and proliferation of immune cells, reflecting neurotoxicity at acute inflammatory conditions. However, the formation of Aβ plaques may be a natural mechanism that rescues cells from the proinflammatory and cytotoxic effects of S100A9, especially considering that inflammation is one of the primary causes of AD.

Place, publisher, year, edition, pages
Springer Nature, 2023
Keywords
Alzheimer’s disease, Amyloid β peptide, Interleukin, Neuroinflammation, S100A9
National Category
Medical Biotechnology (with a focus on Cell Biology (including Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy)
Identifiers
urn:nbn:se:umu:diva-216892 (URN)10.1007/s12031-023-02168-1 (DOI)001101691400001 ()37947991 (PubMedID)2-s2.0-85176226906 (Scopus ID)
Available from: 2023-12-08 Created: 2023-12-08 Last updated: 2024-01-12Bibliographically approved
Zhang, X., Sun, D., Zhou, X., Zhang, C., Yin, Q., Chen, L., . . . Morozova-Roche, L. A. (2023). Proinflammatory S100A9 stimulates TLR4/NF-κB signaling pathways causing enhanced phagocytic capacity of microglial cells. Immunology Letters, 255, 54-61
Open this publication in new window or tab >>Proinflammatory S100A9 stimulates TLR4/NF-κB signaling pathways causing enhanced phagocytic capacity of microglial cells
Show others...
2023 (English)In: Immunology Letters, ISSN 0165-2478, E-ISSN 1879-0542, Vol. 255, p. 54-61Article in journal (Refereed) Published
Abstract [en]

Alzheimer's disease (AD) is the main cause of dementia, affecting the increasingly aging population. Growing evidence indicates that neuro-inflammation plays crucial roles, e.g., the association between AD risk genes with innate immune functions. In this study, we demonstrate that moderate concentrations of pro-inflammatory cytokine S100A9 regulate immune response of BV2 microglial cells, i.e., the phagocytic capacity, reflected by elevated number of 1 μm diameter Dsred-stained latex beads in the cytoplasm. In contrast, at high S100A9 concentrations, both the viability and phagocytic capacity of BV2 cells drop substantially. Furthermore, it is uncovered that S100A9 affects phagocytosis of microglia via NF-κB signaling pathways. Application of related target-specific drugs, i.e., IKK and TLR4 inhibitors, effectively suppresses BV2 cells’ immune responses. These results suggest that pro-inflammatory S100A9 activates microglial phagocytosis, and possibly contributes to the clearance of amyloidogenic species at the early stage of AD.

Place, publisher, year, edition, pages
Elsevier, 2023
Keywords
Alzheimer's disease, Microglia, Neuroinflammation, Phagocytosis, S100A9
National Category
Immunology Immunology in the medical area
Identifiers
urn:nbn:se:umu:diva-206026 (URN)10.1016/j.imlet.2023.02.008 (DOI)000951654500001 ()36870421 (PubMedID)2-s2.0-85150356339 (Scopus ID)
Available from: 2023-03-28 Created: 2023-03-28 Last updated: 2023-09-05Bibliographically approved
Berntsson, E., Vosough, F., Svantesson, T., Pansieri, J., Iashchishyn, I., Ostojic, L., . . . Wärmländer, S. K. (2023). Residue-specific binding of Ni(II) ions influences the structure and aggregation of amyloid beta (Aβ) peptides. Scientific Reports, 13(1), Article ID 3341.
Open this publication in new window or tab >>Residue-specific binding of Ni(II) ions influences the structure and aggregation of amyloid beta (Aβ) peptides
Show others...
2023 (English)In: Scientific Reports, E-ISSN 2045-2322, Vol. 13, no 1, article id 3341Article in journal (Refereed) Published
Abstract [en]

Alzheimer's disease (AD) is the most common cause of dementia worldwide. AD brains display deposits of insoluble amyloid plaques consisting mainly of aggregated amyloid-β (Aβ) peptides, and Aβ oligomers are likely a toxic species in AD pathology. AD patients display altered metal homeostasis, and AD plaques show elevated concentrations of metals such as Cu, Fe, and Zn. Yet, the metal chemistry in AD pathology remains unclear. Ni(II) ions are known to interact with Aβ peptides, but the nature and effects of such interactions are unknown. Here, we use numerous biophysical methods-mainly spectroscopy and imaging techniques-to characterize Aβ/Ni(II) interactions in vitro, for different Aβ variants: Aβ(1-40), Aβ(1-40)(H6A, H13A, H14A), Aβ(4-40), and Aβ(1-42). We show for the first time that Ni(II) ions display specific binding to the N-terminal segment of full-length Aβ monomers. Equimolar amounts of Ni(II) ions retard Aβ aggregation and direct it towards non-structured aggregates. The His6, His13, and His14 residues are implicated as binding ligands, and the Ni(II)·Aβ binding affinity is in the low µM range. The redox-active Ni(II) ions induce formation of dityrosine cross-links via redox chemistry, thereby creating covalent Aβ dimers. In aqueous buffer Ni(II) ions promote formation of beta sheet structure in Aβ monomers, while in a membrane-mimicking environment (SDS micelles) coil-coil helix interactions appear to be induced. For SDS-stabilized Aβ oligomers, Ni(II) ions direct the oligomers towards larger sizes and more diverse (heterogeneous) populations. All of these structural rearrangements may be relevant for the Aβ aggregation processes that are involved in AD brain pathology.

Place, publisher, year, edition, pages
Nature Publishing Group, 2023
National Category
Structural Biology
Identifiers
urn:nbn:se:umu:diva-205490 (URN)10.1038/s41598-023-29901-5 (DOI)000986236800026 ()36849796 (PubMedID)2-s2.0-85148966000 (Scopus ID)
Funder
Swedish Research CouncilThe Swedish Brain FoundationMagnus Bergvall FoundationThe Kamprad Family FoundationKarolinska InstituteUmeå UniversityOlle Engkvists stiftelseRegion Stockholm
Available from: 2023-03-14 Created: 2023-03-14 Last updated: 2023-09-05Bibliographically approved
Qi, X., Wang, Y., Yu, H., Liu, R., Leppert, A., Zheng, Z., . . . Chen, G. (2023). Spider silk protein forms amyloid-like nanofibrils through a non-nucleation-dependent polymerization mechanism. Small, 18(46), Article ID 2304031.
Open this publication in new window or tab >>Spider silk protein forms amyloid-like nanofibrils through a non-nucleation-dependent polymerization mechanism
Show others...
2023 (English)In: Small, ISSN 1613-6810, E-ISSN 1613-6829, Vol. 18, no 46, article id 2304031Article in journal (Refereed) Published
Abstract [en]

Amyloid fibrils—nanoscale fibrillar aggregates with high levels of order—are pathogenic in some today incurable human diseases; however, there are also many physiologically functioning amyloids in nature. The process of amyloid formation is typically nucleation-elongation-dependent, as exemplified by the pathogenic amyloid-β peptide (Aβ) that is associated with Alzheimer's disease. Spider silk, one of the toughest biomaterials, shares characteristics with amyloid. In this study, it is shown that forming amyloid-like nanofibrils is an inherent property preserved by various spider silk proteins (spidroins). Both spidroins and Aβ capped by spidroin N- and C-terminal domains, can assemble into macroscopic spider silk-like fibers that consist of straight nanofibrils parallel to the fiber axis as observed in native spider silk. While Aβ forms amyloid nanofibrils through a nucleation-dependent pathway and exhibits strong cytotoxicity and seeding effects, spidroins spontaneously and rapidly form amyloid-like nanofibrils via a non-nucleation-dependent polymerization pathway that involves lateral packing of fibrils. Spidroin nanofibrils share amyloid-like properties but lack strong cytotoxicity and the ability to self-seed or cross-seed human amyloidogenic peptides. These results suggest that spidroins' unique primary structures have evolved to allow functional properties of amyloid, and at the same time direct their fibrillization pathways to avoid formation of cytotoxic intermediates.

Place, publisher, year, edition, pages
John Wiley & Sons, 2023
Keywords
cytotoxicity, nanofibril, non-nucleation-dependent polymerization, seeding, spidroin
National Category
Biochemistry Molecular Biology
Identifiers
urn:nbn:se:umu:diva-212398 (URN)10.1002/smll.202304031 (DOI)001029348000001 ()37455347 (PubMedID)2-s2.0-85165008500 (Scopus ID)
Funder
The Karolinska Institutet's Research FoundationAlzheimerfondenGun och Bertil Stohnes StiftelseHedlund foundationStiftelsen Gamla TjänarinnorKarolinska InstituteOlle Engkvists stiftelseMagnus Bergvall FoundationStiftelsen Sigurd och Elsa Goljes minne
Note

Graphical Abstract: Qi, X., Wang, Y., Yu, H., Liu, R., Leppert, A., Zheng, Z., Zhong, X., Jin, Z., Wang, H., Li, X., Wang, X., Landreh, M., A. Morozova-Roche, L., Johansson, J., Xiong, S., Iashchishyn, I. and Chen, G. (2023), Spider Silk Protein Forms Amyloid-Like Nanofibrils through a Non-Nucleation-Dependent Polymerization Mechanism (Small 46/2023). Small, 19: 2370388. DOI: 10.1002/smll.202370388

Available from: 2023-07-31 Created: 2023-07-31 Last updated: 2025-02-20Bibliographically approved
Sanders, E., Csondor, R., Šulskis, D., Baronaitė, I., Smirnovas, V., Maheswaran, L., . . . Williamson, P. T. F. (2023). The stabilization of S100A9 structure by calcium inhibits the formation of amyloid fibrils. International Journal of Molecular Sciences, 24(17), Article ID 13200.
Open this publication in new window or tab >>The stabilization of S100A9 structure by calcium inhibits the formation of amyloid fibrils
Show others...
2023 (English)In: International Journal of Molecular Sciences, ISSN 1661-6596, E-ISSN 1422-0067, Vol. 24, no 17, article id 13200Article in journal (Refereed) Published
Abstract [en]

The calcium-binding protein S100A9 is recognized as an important component of the brain neuroinflammatory response to the onset and development of neurodegenerative disease. S100A9 is intrinsically amyloidogenic and in vivo co-aggregates with amyloid-β peptide and α-synuclein in Alzheimer’s and Parkinson’s diseases, respectively. It is widely accepted that calcium dyshomeostasis plays an important role in the onset and development of these diseases, and studies have shown that elevated levels of calcium limit the potential for S100A9 to adopt a fibrillar structure. The exact mechanism by which calcium exerts its influence on the aggregation process remains unclear. Here we demonstrate that despite S100A9 exhibiting α-helical secondary structure in the absence of calcium, the protein exhibits significant plasticity with interconversion between different conformational states occurring on the micro- to milli-second timescale. This plasticity allows the population of conformational states that favour the onset of fibril formation. Magic-angle spinning solid-state NMR studies of the resulting S100A9 fibrils reveal that the S100A9 adopts a single structurally well-defined rigid fibrillar core surrounded by a shell of approximately 15–20 mobile residues, a structure that persists even when fibrils are produced in the presence of calcium ions. These studies highlight how the dysregulation of metal ion concentrations can influence the conformational equilibria of this important neuroinflammatory protein to influence the rate and nature of the amyloid deposits formed.

Place, publisher, year, edition, pages
MDPI, 2023
Keywords
Alzheimer’s disease, amyloid, neurodegenerative disease, Parkinson’s disease, protein stability, S100A9
National Category
Biophysics Neurology
Identifiers
urn:nbn:se:umu:diva-214519 (URN)10.3390/ijms241713200 (DOI)001061117100001 ()37686007 (PubMedID)2-s2.0-85170341086 (Scopus ID)
Available from: 2023-09-27 Created: 2023-09-27 Last updated: 2025-02-20Bibliographically approved
Projects
Understanding the Origins of Neurodegeneration: Cross-Talk Between the Amyloid and Neuroinflammatory Cascades Studied by Advanced Biophysical Methods. [2019-01232_VR]; Umeå University
Organisations
Identifiers
ORCID iD: ORCID iD iconorcid.org/0000-0001-5886-2023

Search in DiVA

Show all publications