1
|
Litus EA, Shevelyova MP, Vologzhannikova AA, Deryusheva EI, Machulin AV, Nemashkalova EL, Permyakova ME, Sokolov AS, Alikova VD, Uversky VN, Permyakov SE. Binding of Pro-Inflammatory Proteins S100A8 or S100A9 to Amyloid-β Peptide Suppresses Its Fibrillation. Biomolecules 2025; 15:431. [PMID: 40149967 PMCID: PMC11939996 DOI: 10.3390/biom15030431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/07/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
Human serum albumin (HSA) is a natural depot of amyloid-β peptide (Aβ), a key player in Alzheimer's disease (AD). HSA and pro-inflammatory Ca2+-binding proteins S100A8 and S100A9 are involved in Aβ metabolism and its deposition in the brain, serving as probable triggers and therapeutic targets in AD, but their interplay with regard to Aβ binding/fibrillation is unclear. To this end, here we explore the in vitro binding of Ca2+-bound S100A8 or S100A9 to monomeric Aβ and the influence of the S100 proteins on Aβ fibrillation. The equilibrium dissociation constants of the complexes of dimeric S100A8/S100A9 with Aβ40/42 estimated by biolayer interferometry are 1-5 µM. S100A8 and S100A9 interfere with HSA binding to Aβ. Thioflavin T assay and electron microscopy data show that micromolar S100A8/S100A9 inhibit Aβ40 fibrillation, and the inhibitory effect of S100A8 exceeds that for HSA. The competition for Aβ between HSA and S100A8/S100A9 may contribute to the Aβ-HSA imbalance in the pro-inflammatory conditions in AD.
Collapse
Affiliation(s)
- Ekaterina A. Litus
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya Str., 7, Pushchino, 142290 Moscow, Russia; (M.P.S.); (A.A.V.); (E.I.D.); (E.L.N.); (M.E.P.); (A.S.S.); (V.D.A.); (S.E.P.)
| | - Marina P. Shevelyova
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya Str., 7, Pushchino, 142290 Moscow, Russia; (M.P.S.); (A.A.V.); (E.I.D.); (E.L.N.); (M.E.P.); (A.S.S.); (V.D.A.); (S.E.P.)
| | - Alisa A. Vologzhannikova
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya Str., 7, Pushchino, 142290 Moscow, Russia; (M.P.S.); (A.A.V.); (E.I.D.); (E.L.N.); (M.E.P.); (A.S.S.); (V.D.A.); (S.E.P.)
| | - Evgenia I. Deryusheva
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya Str., 7, Pushchino, 142290 Moscow, Russia; (M.P.S.); (A.A.V.); (E.I.D.); (E.L.N.); (M.E.P.); (A.S.S.); (V.D.A.); (S.E.P.)
| | - Andrey V. Machulin
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, pr. Nauki, 5, Pushchino, 142290 Moscow, Russia;
| | - Ekaterina L. Nemashkalova
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya Str., 7, Pushchino, 142290 Moscow, Russia; (M.P.S.); (A.A.V.); (E.I.D.); (E.L.N.); (M.E.P.); (A.S.S.); (V.D.A.); (S.E.P.)
| | - Maria E. Permyakova
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya Str., 7, Pushchino, 142290 Moscow, Russia; (M.P.S.); (A.A.V.); (E.I.D.); (E.L.N.); (M.E.P.); (A.S.S.); (V.D.A.); (S.E.P.)
| | - Andrey S. Sokolov
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya Str., 7, Pushchino, 142290 Moscow, Russia; (M.P.S.); (A.A.V.); (E.I.D.); (E.L.N.); (M.E.P.); (A.S.S.); (V.D.A.); (S.E.P.)
| | - Valeria D. Alikova
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya Str., 7, Pushchino, 142290 Moscow, Russia; (M.P.S.); (A.A.V.); (E.I.D.); (E.L.N.); (M.E.P.); (A.S.S.); (V.D.A.); (S.E.P.)
| | - Vladimir N. Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Sergei E. Permyakov
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya Str., 7, Pushchino, 142290 Moscow, Russia; (M.P.S.); (A.A.V.); (E.I.D.); (E.L.N.); (M.E.P.); (A.S.S.); (V.D.A.); (S.E.P.)
| |
Collapse
|
2
|
Qiao CM, Tan LL, Ma XY, Xia YM, Li T, Li MA, Wu J, Nie X, Cui C, Zhao WJ, Shen YQ. Mechanism of S100A9-mediated astrocyte activation via TLR4/NF-κB in Parkinson's disease. Int Immunopharmacol 2025; 146:113938. [PMID: 39724736 DOI: 10.1016/j.intimp.2024.113938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/16/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Astrocyte-mediated neuroinflammation plays a key role in Parkinson's disease (PD) progression. The proinflammatory protein S100A9 is linked to various neurodegenerative diseases, but its involvement in astrocyte activation in PD remains unclear. Here, we investigate the role of S100A9 in astrocyte-mediated neuroinflammation in PD. C57BL/6J mice were intraperitoneally injected with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP; 15 mg/kg four times daily) and subsequently treated with Paquinimod, a S100A9 inhibitor (7 mg/kg, once daily for 7 days, totaling 8 doses). We observed an abnormal increase in S100A9 protein expression and a rise in S100A9-positive cells in the striatum of PD mice. Paquinimod treatment significantly improved behavioral deficits (pole test, rotarod test, traction test, and open field tests), prevented the reduction in striatal tyrosine hydroxylase (TH) protein and the loss of dopaminergic neurons (TH+) in the substantia nigra (SN) in PD mice. Interestingly, S100A9 was predominantly expressed in astrocytes (GFAP+S100A9+ cells) rather than in neurons or microglia, and its inhibition significantly reduced astrocyte activation (GFAP+ cells), reversed A1 astrocyte gene upregulation (H2-D1, C3, Serping1), and increased A2 astrocyte gene expression (Emp1, Ptx3, S100a10). Moreover, S100A9 inhibition also reduced the expression of inflammatory markers (IL-6, IL-1β, TNF-α) and suppressed the TLR4/NF-κB signaling pathway. In vitro, TLR4/NF-κB inhibitors mitigated inflammation and A1/A2 polarization of astrocytic MA cells induced by recombinant S100A9 (rS100A9). These findings suggest that S100A9 mediates astrocyte neuroinflammation and A1/A2 polarization via TLR4/NF-κB signaling, highlighting its potential as a therapeutic target for PD.
Collapse
Affiliation(s)
- Chen-Meng Qiao
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Lu-Lu Tan
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xiao-Yu Ma
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yi-Meng Xia
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ting Li
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ming-An Li
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jian Wu
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xin Nie
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Chun Cui
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei-Jiang Zhao
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yan-Qin Shen
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
3
|
Gruel R, Bijnens B, Van Den Daele J, Thys S, Willems R, Wuyts D, Van Dam D, Verstraelen P, Verboven R, Roels J, Vandamme N, Mancuso R, Pita‐Almenar JD, De Vos WH. S100A8-enriched microglia populate the brain of tau-seeded and accelerated aging mice. Aging Cell 2024; 23:e14120. [PMID: 38403918 PMCID: PMC11113266 DOI: 10.1111/acel.14120] [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: 06/19/2023] [Revised: 01/29/2024] [Accepted: 02/01/2024] [Indexed: 02/27/2024] Open
Abstract
Long considered to fluctuate between pro- and anti-inflammatory states, it has now become evident that microglia occupy a variegated phenotypic landscape with relevance to aging and neurodegeneration. However, whether specific microglial subsets converge in or contribute to both processes that eventually affect brain function is less clear. To investigate this, we analyzed microglial heterogeneity in a tauopathy mouse model (K18-seeded P301L) and an accelerated aging model (Senescence-Accelerated Mouse-Prone 8, SAMP8) using cellular indexing of transcriptomes and epitopes by sequencing. We found that widespread tau pathology in K18-seeded P301L mice caused a significant change in the number and morphology of microglia, but only a mild overrepresentation of disease-associated microglia. At the cell population-level, we observed a marked upregulation of the calprotectin-encoding genes S100a8 and S100a9. In 9-month-old SAMP8 mice, we identified a unique microglial subpopulation that showed partial similarity with the disease-associated microglia phenotype and was additionally characterized by a high expression of the same calprotectin gene set. Immunostaining for S100A8 revealed that this population was enriched in the hippocampus, correlating with the cognitive impairment observed in this model. However, incomplete colocalization between their residence and markers of neuronal loss suggests regional specificity. Importantly, S100A8-positive microglia were also retrieved in brain biopsies of human AD and tauopathy patients as well as in a biopsy of an aged individual without reported pathology. Thus, the emergence of S100A8-positive microglia portrays a conspicuous commonality between accelerated aging and tauopathy progression, which may have relevance for ensuing brain dysfunction.
Collapse
Affiliation(s)
- Roxane Gruel
- Laboratory of Cell Biology & HistologyUniversity of AntwerpWilrijkBelgium
| | - Baukje Bijnens
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIBAntwerpBelgium
- Department of Biomedical SciencesUniversity of AntwerpAntwerpBelgium
| | | | - Sofie Thys
- Laboratory of Cell Biology & HistologyUniversity of AntwerpWilrijkBelgium
| | - Roland Willems
- Janssen Research and DevelopmentNeuroscience Therapeutic AreaBeerseBelgium
| | - Dirk Wuyts
- Janssen Research and DevelopmentNeuroscience Therapeutic AreaBeerseBelgium
| | - Debby Van Dam
- Laboratory of Neurochemistry & Behaviour, Experimental Neurobiology Unit, Department of Biomedical SciencesUniversity of AntwerpAntwerpBelgium
- Department of Neurology and Alzheimer CenterUniversity of GroningenGroningenThe Netherlands
| | - Peter Verstraelen
- Laboratory of Cell Biology & HistologyUniversity of AntwerpWilrijkBelgium
| | - Rosanne Verboven
- Laboratory of Cell Biology & HistologyUniversity of AntwerpWilrijkBelgium
| | - Jana Roels
- VIB Single Cell Core, VIBGhent‐LeuvenBelgium
- VIB‐UGent Center for Inflammation ResearchGhentBelgium
| | - Niels Vandamme
- VIB Single Cell Core, VIBGhent‐LeuvenBelgium
- VIB‐UGent Center for Inflammation ResearchGhentBelgium
| | - Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIBAntwerpBelgium
- Department of Biomedical SciencesUniversity of AntwerpAntwerpBelgium
| | | | - Winnok H. De Vos
- Laboratory of Cell Biology & HistologyUniversity of AntwerpWilrijkBelgium
- Antwerp Centre for Advanced MicroscopyUniversity of AntwerpAntwerpBelgium
- μNEURO research excellence consortiumUniversity of AntwerpAntwerpBelgium
| |
Collapse
|
4
|
Smith HM, Moodie JE, Monterrubio-Gómez K, Gadd DA, Hillary RF, Chybowska AD, McCartney DL, Campbell A, Redmond P, Page D, Taylor A, Corley J, Harris SE, Valdés Hernández M, Muñoz Maniega S, Bastin ME, Wardlaw JM, Deary IJ, Boardman JP, Mullin DS, Russ TC, Cox SR, Marioni RE. Epigenetic scores of blood-based proteins as biomarkers of general cognitive function and brain health. Clin Epigenetics 2024; 16:46. [PMID: 38528588 PMCID: PMC10962132 DOI: 10.1186/s13148-024-01661-7] [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: 11/30/2023] [Accepted: 03/16/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Epigenetic Scores (EpiScores) for blood protein levels have been associated with disease outcomes and measures of brain health, highlighting their potential usefulness as clinical biomarkers. They are typically derived via penalised regression, whereby a linear weighted sum of DNA methylation (DNAm) levels at CpG sites are predictive of protein levels. Here, we examine 84 previously published protein EpiScores as possible biomarkers of cross-sectional and longitudinal measures of general cognitive function and brain health, and incident dementia across three independent cohorts. RESULTS Using 84 protein EpiScores as candidate biomarkers, associations with general cognitive function (both cross-sectionally and longitudinally) were tested in three independent cohorts: Generation Scotland (GS), and the Lothian Birth Cohorts of 1921 and 1936 (LBC1921 and LBC1936, respectively). A meta-analysis of general cognitive functioning results in all three cohorts identified 18 EpiScore associations (absolute meta-analytic standardised estimates ranged from 0.03 to 0.14, median of 0.04, PFDR < 0.05). Several associations were also observed between EpiScores and global brain volumetric measures in the LBC1936. An EpiScore for the S100A9 protein (a known Alzheimer disease biomarker) was associated with general cognitive functioning (meta-analytic standardised beta: - 0.06, P = 1.3 × 10-9), and with time-to-dementia in GS (Hazard ratio 1.24, 95% confidence interval 1.08-1.44, P = 0.003), but not in LBC1936 (Hazard ratio 1.11, P = 0.32). CONCLUSIONS EpiScores might make a contribution to the risk profile of poor general cognitive function and global brain health, and risk of dementia, however these scores require replication in further studies.
Collapse
Affiliation(s)
- Hannah M Smith
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Joanna E Moodie
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Karla Monterrubio-Gómez
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Danni A Gadd
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Robert F Hillary
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Aleksandra D Chybowska
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Daniel L McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Paul Redmond
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Danielle Page
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Adele Taylor
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Janie Corley
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Sarah E Harris
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Maria Valdés Hernández
- Centre for Clinical Brain Sciences, and Edinburgh Centre in the UK Dementia Research Institute, Chancellor's Building, University of Edinburgh, Little France, Edinburgh, UK
| | - Susana Muñoz Maniega
- Centre for Clinical Brain Sciences, and Edinburgh Centre in the UK Dementia Research Institute, Chancellor's Building, University of Edinburgh, Little France, Edinburgh, UK
| | - Mark E Bastin
- Centre for Clinical Brain Sciences, and Edinburgh Centre in the UK Dementia Research Institute, Chancellor's Building, University of Edinburgh, Little France, Edinburgh, UK
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, and Edinburgh Centre in the UK Dementia Research Institute, Chancellor's Building, University of Edinburgh, Little France, Edinburgh, UK
| | - Ian J Deary
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - James P Boardman
- Centre for Clinical Brain Sciences, and Edinburgh Centre in the UK Dementia Research Institute, Chancellor's Building, University of Edinburgh, Little France, Edinburgh, UK
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Donncha S Mullin
- Centre for Clinical Brain Sciences, Division of Psychiatry, University of Edinburgh, Edinburgh, UK
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
| | - Tom C Russ
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, and Edinburgh Centre in the UK Dementia Research Institute, Chancellor's Building, University of Edinburgh, Little France, Edinburgh, UK
| | - Simon R Cox
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
5
|
Ghosh S, Tamilselvi S, Williams C, Jayaweera SW, Iashchishyn IA, Šulskis D, Gilthorpe JD, Olofsson A, Smirnovas V, Svedružić ŽM, Morozova-Roche LA. ApoE Isoforms Inhibit Amyloid Aggregation of Proinflammatory Protein S100A9. Int J Mol Sci 2024; 25:2114. [PMID: 38396791 PMCID: PMC10889306 DOI: 10.3390/ijms25042114] [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: 12/22/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
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-β (Aβ) 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.
Collapse
Affiliation(s)
- Shamasree Ghosh
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-90187 Umeå, Sweden; (S.G.); (S.T.); (I.A.I.)
| | - Shanmugam Tamilselvi
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-90187 Umeå, Sweden; (S.G.); (S.T.); (I.A.I.)
| | - Chloe Williams
- Department of Medical and Translational Biology, Umeå University, SE-90187 Umeå, Sweden; (C.W.); (J.D.G.)
| | - Sanduni W. Jayaweera
- Department of Clinical Microbiology, Umeå University, SE-90187 Umeå, Sweden; (S.W.J.); (A.O.)
| | - Igor A. Iashchishyn
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-90187 Umeå, Sweden; (S.G.); (S.T.); (I.A.I.)
| | - Darius Šulskis
- Institute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania; (D.Š.); (V.S.)
| | - Jonathan D. Gilthorpe
- Department of Medical and Translational Biology, Umeå University, SE-90187 Umeå, Sweden; (C.W.); (J.D.G.)
| | - Anders Olofsson
- Department of Clinical Microbiology, Umeå University, SE-90187 Umeå, Sweden; (S.W.J.); (A.O.)
| | - Vytautas Smirnovas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania; (D.Š.); (V.S.)
| | | | - Ludmilla A. Morozova-Roche
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-90187 Umeå, Sweden; (S.G.); (S.T.); (I.A.I.)
| |
Collapse
|
6
|
Bai Q, Sun D, Zeng Y, Zhu J, Zhang C, Zhang X, Chen L, Zhou X, Ye L, Tang Y, Liu Y, Morozova-Roche LA. Effect of Proinflammatory S100A9 Protein on Migration and Proliferation of Microglial Cells. J Mol Neurosci 2023; 73:983-995. [PMID: 37947991 DOI: 10.1007/s12031-023-02168-1] [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: 05/19/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
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.
Collapse
Affiliation(s)
- Qiao Bai
- Chongqing Medical University, 1 Medical College Road, Yu Zhong District, Chongqing, China
| | - Dan Sun
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
| | - Yang Zeng
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
| | - Jie Zhu
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
| | - Ce Zhang
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
| | - Xiaoyin Zhang
- Chongqing Medical University, 1 Medical College Road, Yu Zhong District, Chongqing, China
| | - Li Chen
- Chongqing Medical University, 1 Medical College Road, Yu Zhong District, Chongqing, China
| | - Xin Zhou
- Chongqing Medical University, 1 Medical College Road, Yu Zhong District, Chongqing, China
| | - Liu Ye
- Chongqing Medical University, 1 Medical College Road, Yu Zhong District, Chongqing, China
| | - Yong Tang
- Chongqing Medical University, 1 Medical College Road, Yu Zhong District, Chongqing, China
| | - Yonggang Liu
- Chongqing Medical University, 1 Medical College Road, Yu Zhong District, Chongqing, China.
| | | |
Collapse
|
7
|
Zhang X, Sun D, Zhou X, Zhang C, Yin Q, Chen L, Tang Y, Liu Y, Morozova-Roche LA. Proinflammatory S100A9 stimulates TLR4/NF-κB signaling pathways causing enhanced phagocytic capacity of microglial cells. Immunol Lett 2023; 255:54-61. [PMID: 36870421 DOI: 10.1016/j.imlet.2023.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 02/26/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023]
Abstract
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.
Collapse
Affiliation(s)
- Xiaoyin Zhang
- Laboratory of stem cell and Tissue Engineering, Chongqing Medical University, 400016, Chongqing, China
| | - Dan Sun
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon Technology, Northwest University, 710127, Xi'an, China
| | - Xin Zhou
- Laboratory of stem cell and Tissue Engineering, Chongqing Medical University, 400016, Chongqing, China
| | - Ce Zhang
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon Technology, Northwest University, 710127, Xi'an, China
| | - Qing Yin
- Laboratory of stem cell and Tissue Engineering, Chongqing Medical University, 400016, Chongqing, China
| | - Li Chen
- Laboratory of stem cell and Tissue Engineering, Chongqing Medical University, 400016, Chongqing, China
| | - Yong Tang
- Laboratory of stem cell and Tissue Engineering, Chongqing Medical University, 400016, Chongqing, China
| | - Yonggang Liu
- Laboratory of stem cell and Tissue Engineering, Chongqing Medical University, 400016, Chongqing, China.
| | | |
Collapse
|
8
|
Cho Y, Bae HG, Okun E, Arumugam TV, Jo DG. Physiology and pharmacology of amyloid precursor protein. Pharmacol Ther 2022; 235:108122. [PMID: 35114285 DOI: 10.1016/j.pharmthera.2022.108122] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/17/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023]
Abstract
Amyloid precursor protein (APP) is an evolutionarily conserved transmembrane protein and a well-characterized precursor protein of amyloid-beta (Aβ) peptides, which accumulate in the brains of individuals with Alzheimer's disease (AD)-related pathologies. Aβ has been extensively investigated since the amyloid hypothesis in AD was proposed. Besides Aβ, previous studies on APP and its proteolytic cleavage products have suggested their diverse pathological and physiological functions. However, their roles still have not been thoroughly understood. In this review, we extensively discuss the evolutionarily-conserved biology of APP, including its structure and processing pathway, as well as recent findings on the physiological roles of APP and its fragments in the central nervous system and peripheral nervous system. We have also elaborated upon the current status of APP-targeted therapeutic approaches for AD treatment by discussing inhibitors of several proteases participating in APP processing, including α-, β-, and γ-secretases. Finally, we have highlighted the future perspectives pertaining to further research and the potential clinical role of APP.
Collapse
Affiliation(s)
- Yoonsuk Cho
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea
| | - Han-Gyu Bae
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea
| | - Eitan Okun
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; The Pauld Feder Laboratory on Alzheimer's Disease Research, Israel
| | - Thiruma V Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea; School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia.
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea; Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, South Korea; Biomedical Institute for Convergence, Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
9
|
Jauch-Speer SL, Herrera-Rivero M, Ludwig N, Véras De Carvalho BC, Martens L, Wolf J, Imam Chasan A, Witten A, Markus B, Schieffer B, Vogl T, Rossaint J, Stoll M, Roth J, Fehler O. C/EBPδ-induced epigenetic changes control the dynamic gene transcription of S100a8 and S100a9. eLife 2022; 11:75594. [PMID: 35543413 PMCID: PMC9122501 DOI: 10.7554/elife.75594] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 05/06/2022] [Indexed: 11/25/2022] Open
Abstract
The proinflammatory alarmins S100A8 and S100A9 are among the most abundant proteins in neutrophils and monocytes but are completely silenced after differentiation to macrophages. The molecular mechanisms of the extraordinarily dynamic transcriptional regulation of S100a8 and S100a9 genes, however, are only barely understood. Using an unbiased genome-wide CRISPR/Cas9 knockout (KO)-based screening approach in immortalized murine monocytes, we identified the transcription factor C/EBPδ as a central regulator of S100a8 and S100a9 expression. We showed that S100A8/A9 expression and thereby neutrophil recruitment and cytokine release were decreased in C/EBPδ KO mice in a mouse model of acute lung inflammation. S100a8 and S100a9 expression was further controlled by the C/EBPδ antagonists ATF3 and FBXW7. We confirmed the clinical relevance of this regulatory network in subpopulations of human monocytes in a clinical cohort of cardiovascular patients. Moreover, we identified specific C/EBPδ-binding sites within S100a8 and S100a9 promoter regions, and demonstrated that C/EBPδ-dependent JMJD3-mediated demethylation of H3K27me3 is indispensable for their expression. Overall, our work uncovered C/EBPδ as a novel regulator of S100a8 and S100a9 expression. Therefore, C/EBPδ represents a promising target for modulation of inflammatory conditions that are characterized by S100a8 and S100a9 overexpression.
Collapse
Affiliation(s)
| | | | - Nadine Ludwig
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | | | - Leonie Martens
- Institute of Immunology, University of Münster, Münster, Germany
| | - Jonas Wolf
- Institute of Immunology, University of Münster, Münster, Germany
| | | | - Anika Witten
- Department of Genetic Epidemiology, University of Münster, Münster, Germany
| | - Birgit Markus
- Clinic for Cardiology, Angiology and Internal Intensive Medicine, University Hospital Marburg, Marburg, Germany
| | - Bernhard Schieffer
- Clinic for Cardiology, Angiology and Internal Intensive Medicine, University Hospital Marburg, Marburg, Germany
| | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| | - Jan Rossaint
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Monika Stoll
- Department of Genetic Epidemiology, University of Münster, Münster, Germany
| | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany
| | - Olesja Fehler
- Institute of Immunology, University of Münster, Münster, Germany
| |
Collapse
|
10
|
MAEKAWA T, SUGIMOTO M, KUME S, OHTA T. Pathophysiological features in the brains of female Spontaneously Diabetic Torii (SDT) fatty rats. J Vet Med Sci 2022; 84:330-337. [PMID: 35082197 PMCID: PMC8983279 DOI: 10.1292/jvms.21-0654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/17/2022] [Indexed: 11/30/2022] Open
Abstract
Diabetes mellitus (DM) and obesity are associated with neurodegenerative diseases such as Alzheimer's disease and psychiatric disorders such as major depression. In this study, we investigated pathophysiological changes in the brains of female Spontaneously Diabetic Torii (SDT) fatty rats with diabetes and obesity. Brains of Sprague-Dawley (SD), SDT and SDT fatty rats were collected at 58 weeks of age. The parietal cortical thickness was measured and the number of pyramidal cells in the hippocampal cornu ammonis 1 and 3 (CA1 and CA3) and the number of granule cells in the dentate gyrus (DG) regions were counted. The area of glial fibrillary acidic protein (GFAP) positivity in CA1, CA3 and DG regions were measured. The parietal cortical thickness and the number of cells in CA3 and DG regions of SDT and SDT fatty rats did not show obvious changes. On the other hand, in the CA1 region, the number of cells in SDT rats and SDT fatty rats was significantly lower than that in SD rats, and that in SDT fatty rats was significantly lower than that in SDT rats. The GFAP-positive area in SDT fatty rats was significantly reduced compared to that in SD rats only in the DG region. Preliminarily result showed that the expression of S100a9, an inflammation-related gene, was increased in the brains of SDT fatty rats. These results suggest that female SDT fatty rat may exhibit central nervous system diseases due to obesity and DM.
Collapse
Affiliation(s)
- Tatsuya MAEKAWA
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
- Laboratory of Animal Physiology and Functional Anatomy, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Miki SUGIMOTO
- Laboratory of Animal Physiology and Functional Anatomy, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Shinichi KUME
- Laboratory of Animal Physiology and Functional Anatomy, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Takeshi OHTA
- Laboratory of Animal Physiology and Functional Anatomy, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| |
Collapse
|
11
|
González LF, Bevilacqua LE, Naves R. Nanotechnology-Based Drug Delivery Strategies to Repair the Mitochondrial Function in Neuroinflammatory and Neurodegenerative Diseases. Pharmaceutics 2021; 13:2055. [PMID: 34959337 PMCID: PMC8707316 DOI: 10.3390/pharmaceutics13122055] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 12/14/2022] Open
Abstract
Mitochondria are vital organelles in eukaryotic cells that control diverse physiological processes related to energy production, calcium homeostasis, the generation of reactive oxygen species, and cell death. Several studies have demonstrated that structural and functional mitochondrial disturbances are involved in the development of different neuroinflammatory (NI) and neurodegenerative (ND) diseases (NI&NDDs) such as multiple sclerosis, Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Remarkably, counteracting mitochondrial impairment by genetic or pharmacologic treatment ameliorates neurodegeneration and clinical disability in animal models of these diseases. Therefore, the development of nanosystems enabling the sustained and selective delivery of mitochondria-targeted drugs is a novel and effective strategy to tackle NI&NDDs. In this review, we outline the impact of mitochondrial dysfunction associated with unbalanced mitochondrial dynamics, altered mitophagy, oxidative stress, energy deficit, and proteinopathies in NI&NDDs. In addition, we review different strategies for selective mitochondria-specific ligand targeting and discuss novel nanomaterials, nanozymes, and drug-loaded nanosystems developed to repair mitochondrial function and their therapeutic benefits protecting against oxidative stress, restoring cell energy production, preventing cell death, inhibiting protein aggregates, and improving motor and cognitive disability in cellular and animal models of different NI&NDDs.
Collapse
Affiliation(s)
| | | | - Rodrigo Naves
- Immunology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Av. Independencia 1027, Santiago 8380453, Chile; (L.F.G.); (L.E.B.)
| |
Collapse
|
12
|
Chaudhary H, Iashchishyn IA, Romanova NV, Rambaran MA, Musteikyte G, Smirnovas V, Holmboe M, Ohlin CA, Svedružić ŽM, Morozova-Roche LA. Polyoxometalates as Effective Nano-inhibitors of Amyloid Aggregation of Pro-inflammatory S100A9 Protein Involved in Neurodegenerative Diseases. ACS APPLIED MATERIALS & INTERFACES 2021; 13:26721-26734. [PMID: 34080430 PMCID: PMC8289188 DOI: 10.1021/acsami.1c04163] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Pro-inflammatory and amyloidogenic S100A9 protein is central to the amyloid-neuroinflammatory cascade in neurodegenerative diseases. Polyoxometalates (POMs) constitute a diverse group of nanomaterials, which showed potency in amyloid inhibition. Here, we have demonstrated that two selected nanosized niobium POMs, Nb10 and TiNb9, can act as potent inhibitors of S100A9 amyloid assembly. Kinetics analysis based on ThT fluorescence experiments showed that addition of either Nb10 or TiNb9 reduces the S100A9 amyloid formation rate and amyloid quantity. Atomic force microscopy imaging demonstrated the complete absence of long S100A9 amyloid fibrils at increasing concentrations of either POM and the presence of only round-shaped and slightly elongated aggregates. Molecular dynamics simulation revealed that both Nb10 and TiNb9 bind to native S100A9 homo-dimer by forming ionic interactions with the positively charged Lys residue-rich patches on the protein surface. The acrylamide quenching of intrinsic fluorescence showed that POM binding does not perturb the Trp 88 environment. The far and near UV circular dichroism revealed no large-scale perturbation of S100A9 secondary and tertiary structures upon POM binding. These indicate that POM binding involves only local conformational changes in the binding sites. By using intrinsic and 8-anilino-1-naphthalene sulfonate fluorescence titration experiments, we found that POMs bind to S100A9 with a Kd of ca. 2.5 μM. We suggest that the region, including Lys 50 to Lys 54 and characterized by high amyloid propensity, could be the key sequences involved in S1009 amyloid self-assembly. The inhibition and complete hindering of S100A9 amyloid pathways may be used in the therapeutic applications targeting the amyloid-neuroinflammatory cascade in neurodegenerative diseases.
Collapse
Affiliation(s)
- Himanshu Chaudhary
- Department
of Medical Biochemistry and Biophysics, Umeå University, Umeå 90187, Sweden
| | - Igor A. Iashchishyn
- Department
of Medical Biochemistry and Biophysics, Umeå University, Umeå 90187, Sweden
| | - Nina V. Romanova
- Department
of Medical Biochemistry and Biophysics, Umeå University, Umeå 90187, Sweden
| | | | - Greta Musteikyte
- Institute
of Biotechnology, Life Sciences Center, Vilnius University, Vilnius LT-10257, Lithuania
| | - Vytautas Smirnovas
- Institute
of Biotechnology, Life Sciences Center, Vilnius University, Vilnius LT-10257, Lithuania
| | - Michael Holmboe
- Department
of Chemistry, Umeå University, 90187 Umeå, Sweden
| | - C. André Ohlin
- Department
of Chemistry, Umeå University, 90187 Umeå, Sweden
| | | | - Ludmilla A. Morozova-Roche
- Department
of Medical Biochemistry and Biophysics, Umeå University, Umeå 90187, Sweden
- . Tel.: +46736205283. Fax: +46907865283
| |
Collapse
|
13
|
Contini C, Olianas A, Serrao S, Deriu C, Iavarone F, Boroumand M, Bizzarro A, Lauria A, Faa G, Castagnola M, Messana I, Manconi B, Masullo C, Cabras T. Top-Down Proteomics of Human Saliva Highlights Anti-inflammatory, Antioxidant, and Antimicrobial Defense Responses in Alzheimer Disease. Front Neurosci 2021; 15:668852. [PMID: 34121996 PMCID: PMC8189262 DOI: 10.3389/fnins.2021.668852] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Alzheimer disease (AD) is the most prevalent neurodegenerative disease in the elderly, characterized by accumulation in the brain of misfolded proteins, inflammation, and oxidative damage leading to neuronal cell death. By considering the viewpoint that AD onset and worsening may be influenced by environmental factors causing infection, oxidative stress, and inflammatory reaction, we investigated the changes of the salivary proteome in a population of patients with respect to that in healthy controls (HCs). Indeed, the possible use of saliva as a diagnostic tool has been explored in several oral and systemic diseases. Moreover, the oral cavity continuously established adaptative and protective processes toward exogenous stimuli. In the present study, qualitative/quantitative variations of 56 salivary proteoforms, including post-translationally modified derivatives, have been analyzed by RP-HPLC-ESI-IT-MS and MS/MS analyses, and immunological methods were applied to validate MS results. The salivary protein profile of AD patients was characterized by significantly higher levels of some multifaceted proteins and peptides that were either specific to the oral cavity or also expressed in other body districts: (i) peptides involved in the homeostasis of the oral cavity; (ii) proteins acting as ROS/RNS scavengers and with a neuroprotective role, such as S100A8, S100A9, and their glutathionylated and nitrosylated proteoforms; cystatin B and glutathionylated and dimeric derivatives; (iii) proteins with antimicrobial activity, such as α-defensins, cystatins A and B, histatin 1, statherin, and thymosin β4, this last with a neuroprotective role at the level of microglia. These results suggested that, in response to injured conditions, Alzheimer patients established defensive mechanisms detectable at the oral level. Data are available via ProteomeXchange with identifier PXD021538.
Collapse
Affiliation(s)
- Cristina Contini
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, Cagliari, Italy
| | - Alessandra Olianas
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, Cagliari, Italy
| | - Simone Serrao
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, Cagliari, Italy
| | - Carla Deriu
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, Cagliari, Italy
| | - Federica Iavarone
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| | - Mozhgan Boroumand
- Laboratorio di Proteomica, Centro Europeo di Ricerca sul Cervello, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Alessandra Bizzarro
- UOC Continuità Assistenziale, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| | - Alessandra Lauria
- UOC Continuità Assistenziale, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| | - Gavino Faa
- Dipartimento di Scienze Mediche e Sanità Pubblica, University of Cagliari, Cagliari, Italy
| | - Massimo Castagnola
- Laboratorio di Proteomica, Centro Europeo di Ricerca sul Cervello, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Irene Messana
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Barbara Manconi
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, Cagliari, Italy
| | - Carlo Masullo
- Dipartimento di Neuroscienze, Sez. Neurologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Tiziana Cabras
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, Cagliari, Italy
| |
Collapse
|
14
|
Ribon M, Leone C, Chiot A, Berriat F, Rampanana M, Cottin J, Bohl D, Millecamps S, Lobsiger CS, Heneka MT, Boillée S. Deletion of the inflammatory S100-A9/MRP14 protein does not influence survival in hSOD1 G93A ALS mice. Neurobiol Aging 2021; 101:181-186. [PMID: 33626479 DOI: 10.1016/j.neurobiolaging.2021.01.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/22/2020] [Accepted: 01/15/2021] [Indexed: 11/15/2022]
Abstract
Neuroinflammation is a hallmark of Amyotrophic Lateral Sclerosis (ALS) in hSOD1G93A mouse models where microglial cells contribute to the progressive motor neuron degenerative process. S100-A8 and S100-A9 (also known as MRP8 and MRP14, respectively) are cytoplasmic proteins expressed by inflammatory myeloid cells, including microglia and macrophages. Mainly acting as a heterodimer, S100-A8/A9, when secreted, can activate Toll-like Receptor 4 on immune cells, leading to deleterious proinflammatory cytokine production. Deletion of S100a9 in Alzheimer's disease mouse models showed a positive outcome, reducing pathology. We now assessed its role in ALS. Unexpectedly, our results show that deleting S100a9 in hSOD1G93A ALS mice had no impact on mouse survival, but rather accelerated symptoms with no impact on microglial activation and motor neuron survival, suggesting that blocking S100-A9 would not be a valuable strategy for ALS.
Collapse
Affiliation(s)
- Matthieu Ribon
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Céline Leone
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Aude Chiot
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Félix Berriat
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Martine Rampanana
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Julie Cottin
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Delphine Bohl
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Stéphanie Millecamps
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Christian S Lobsiger
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Michael T Heneka
- Department of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Séverine Boillée
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France.
| |
Collapse
|
15
|
Bjorkli C, Sandvig A, Sandvig I. Bridging the Gap Between Fluid Biomarkers for Alzheimer's Disease, Model Systems, and Patients. Front Aging Neurosci 2020; 12:272. [PMID: 32982716 PMCID: PMC7492751 DOI: 10.3389/fnagi.2020.00272] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is a debilitating neurodegenerative disease characterized by the accumulation of two proteins in fibrillar form: amyloid-β (Aβ) and tau. Despite decades of intensive research, we cannot yet pinpoint the exact cause of the disease or unequivocally determine the exact mechanism(s) underlying its progression. This confounds early diagnosis and treatment of the disease. Cerebrospinal fluid (CSF) biomarkers, which can reveal ongoing biochemical changes in the brain, can help monitor developing AD pathology prior to clinical diagnosis. Here we review preclinical and clinical investigations of commonly used biomarkers in animals and patients with AD, which can bridge translation from model systems into the clinic. The core AD biomarkers have been found to translate well across species, whereas biomarkers of neuroinflammation translate to a lesser extent. Nevertheless, there is no absolute equivalence between biomarkers in human AD patients and those examined in preclinical models in terms of revealing key pathological hallmarks of the disease. In this review, we provide an overview of current but also novel AD biomarkers and how they relate to key constituents of the pathological cascade, highlighting confounding factors and pitfalls in interpretation, and also provide recommendations for standardized procedures during sample collection to enhance the translational validity of preclinical AD models.
Collapse
Affiliation(s)
- Christiana Bjorkli
- Sandvig Group, Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Axel Sandvig
- Sandvig Group, Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.,Institute of Neuromedicine and Movement Science, Department of Neurology, St. Olavs Hospital, Trondheim, Norway.,Department of Pharmacology and Clinical Neurosciences, Division of Neuro, Head, and Neck, University Hospital of Umeå, Umeå, Sweden
| | - Ioanna Sandvig
- Sandvig Group, Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
16
|
Martin NA, Hyrlov KH, Elkjaer ML, Thygesen EK, Wlodarczyk A, Elbaek KJ, Aboo C, Okarmus J, Benedikz E, Reynolds R, Hegedus Z, Stensballe A, Svenningsen ÅF, Owens T, Illes Z. Absence of miRNA-146a Differentially Alters Microglia Function and Proteome. Front Immunol 2020; 11:1110. [PMID: 32582192 PMCID: PMC7292149 DOI: 10.3389/fimmu.2020.01110] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/07/2020] [Indexed: 12/18/2022] Open
Abstract
Background: MiR-146a is an important regulator of innate inflammatory responses and is also implicated in cell death and survival. Methods: By sorting CNS resident cells, microglia were the main cellular source of miR-146a. Therefore, we investigated microglia function and phenotype in miR-146a knock-out (KO) mice, analyzed the proteome of KO and wild-type (WT) microglia by LC-MS/MS, and examined miR-146a expression in different brain lesions of patients with multiple sclerosis (MS). Results: When stimulated with LPS or myelin in vitro, microglia from KO mice expressed higher levels of IL-1β, TNF, IL-6, IL-10, CCL3, and CCL2 compared to WT. Stimulation increased migration and phagocytosis of WT but not KO microglia. CD11c+ microglia were induced by cuprizone (CPZ) in the WT mice but less in the KO. The proteome of ex vivo microglia was not different in miR-146a KO compared to WT mice, but CPZ treatment induced differential and reduced protein responses in the KO: GOT1, COX5b, CRYL1, and cystatin-C were specifically changed in KO microglia. We explored discriminative features of microglia proteomes: sparse Partial Least Squares-Discriminant Analysis showed the best discrimination when control and CPZ-treated conditions were compared. Cluster of ten proteins separated WT and miR-146a KO microglia after CPZ: among them were sensomes allowing to perceive the environment, Atp1a3 that belongs to the signature of CD11c+ microglia, and proteins related to inflammatory responses (S100A9, Ppm1g). Finally, we examined the expression of miR-146a and its validated target genes in different brain lesions of MS patients. MiR-146 was upregulated in all lesion types, and the highest expression was in active lesions. Nineteen of 88 validated target genes were significantly changed in active lesions, while none were changed in NAWM. Conclusion: Our data indicated that microglia is the major source of miR-146a in the CNS. The absence of miR-146a differentially affected microglia function and proteome, and miR-146a may play an important role in gene regulation of active MS lesions.
Collapse
Affiliation(s)
- Nellie A Martin
- Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Kirsten H Hyrlov
- Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Maria L Elkjaer
- Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Eva K Thygesen
- Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Agnieszka Wlodarczyk
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Institute of Clinical Research, BRIDGE, University of Southern Denmark, Odense, Denmark
| | - Kirstine J Elbaek
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Christopher Aboo
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark.,Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, China
| | - Justyna Okarmus
- Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Eirikur Benedikz
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Richard Reynolds
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Zoltan Hegedus
- Laboratory of Bioinformatics, Biological Research Centre, Szeged, Hungary.,Department of Biochemistry and Medical Chemistry, University of Pecs, Pecs, Hungary
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Åsa Fex Svenningsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Trevor Owens
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Institute of Clinical Research, BRIDGE, University of Southern Denmark, Odense, Denmark
| | - Zsolt Illes
- Department of Neurology, Odense University Hospital, Odense, Denmark.,Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Institute of Clinical Research, BRIDGE, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
17
|
Prophylactic effects of biogenic selenium nanoparticles on acute toxoplasmosis: An in vivo study. Ann Med Surg (Lond) 2020; 54:85-88. [PMID: 32405413 PMCID: PMC7212177 DOI: 10.1016/j.amsu.2020.04.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 03/12/2020] [Accepted: 04/01/2020] [Indexed: 11/23/2022] Open
Abstract
Background In this investigation, the in vivo efficacy and safety of biogenic selenium nanoparticles (SeNPs) are assessed against acute toxoplasmosis caused by Toxoplasma gondii (Sarcocystidae) in the mice. Methods Male NMRI mice were orally treated with normal saline (control group) and SeNPs at the doses of 5 and 10 mg/kg once a day for 14 days. On the 15th day, the mice were infected with 104 tachyzoites of T. gondii RH strain by the intraperitoneal route. The mortality rate and parasite load were determined in the infected mice. The mRNA levels of IFN-γ, IL10, IL12, and inducible nitric oxide synthase were also examined in the infected mice by quantitative real-time PCR. Results The rate of mortality in the infected mice receiving SeNPs at the doses of 5 and 10 mg/kg compared with the mice in the control group was 100% on the 9 and 10 days after the administration. The mean number of tachyzoites in the infected mice receiving SeNPs was significantly lower than that in the control group. No significant difference (p > 0.05) was found in the biochemical parameters between the mice treated with SeNPs and the mice in the control group. The results revealed that mRNA levels significantly improved in the infected mice treated with SeNPs compared with those in the control group. Conclusion Findings of the present investigation showed the considerable efficacy of SeNPs with no important toxicity for curing acute toxoplasmosis in the mice model. However, further studies are needed to clarify the accurate anti-Toxoplasma mechanisms of SeNPs. We evaluated the efficacy of Biogenic Selenium Nanoparticles (SeNPs) against acute toxoplasmosis. Mice were orally treated with SeNPs at the doses of 5 and 10 mg/kg once a day for 14 days. The average number of tachyzoites and mortality rate was significantly lower in treated mice. No significant difference (p > 0.05) observed in the biochemical parameters between the mice treated. The mRNA levels of meaningfully improved in infected mice treated with SeNPs.
Collapse
|
18
|
Sreejit G, Flynn MC, Patil M, Krishnamurthy P, Murphy AJ, Nagareddy PR. S100 family proteins in inflammation and beyond. Adv Clin Chem 2020; 98:173-231. [PMID: 32564786 DOI: 10.1016/bs.acc.2020.02.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The S100 family proteins possess a variety of intracellular and extracellular functions. They interact with multiple receptors and signal transducers to regulate pathways that govern inflammation, cell differentiation, proliferation, energy metabolism, apoptosis, calcium homeostasis, cell cytoskeleton and microbial resistance. S100 proteins are also emerging as novel diagnostic markers for identifying and monitoring various diseases. Strategies aimed at targeting S100-mediated signaling pathways hold a great potential in developing novel therapeutics for multiple diseases. In this chapter, we aim to summarize the current knowledge about the role of S100 family proteins in health and disease with a major focus on their role in inflammatory conditions.
Collapse
Affiliation(s)
| | - Michelle C Flynn
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mallikarjun Patil
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andrew J Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Department of Immunology, Monash University, Melbourne, VIC, Australia
| | | |
Collapse
|
19
|
Qiu X, Ping S, Kyle M, Longo J, Chin L, Zhao LR. S100 Calcium-Binding Protein A9 Knockout Contributes to Neuroprotection and Functional Improvement after Traumatic Brain Injury. J Neurotrauma 2019; 37:950-965. [PMID: 31621496 DOI: 10.1089/neu.2018.6170] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
S100 calcium-binding protein A9 (S100a9), a proinflammatory protein, has been shown to be involved in the development of neuroinflammatory disorders and neurodegenerative diseases. Upregulation of S100a9 in the brain during acute brain injury has been proposed to be associated with acute neuroinflammation. However, it remains unclear whether eliminating S100a9 expression will show beneficial outcomes after traumatic brain injury (TBI). Using S100a9 knockout mice, this study has demonstrated that S100a9 deletion ameliorates post-TBI anxiety, improves TBI-impaired motor and cognitive function, reduces lesion size, prevents perilesional neuron loss and neurodegeneration, diminishes neuroinflammation and TBI-induced neurogenesis, and enhances perilesional expression of neuroplasticity protein. These findings suggest that S100a9 plays a detrimental role in TBI. Genetic deletion of S100a9 enhances neuroprotection and improves functional outcome after TBI. This study sheds light on the pathological involvement of S100a9 in TBI, which would provide a new therapeutic target to minimize TBI-induced brain damage.
Collapse
Affiliation(s)
- Xuecheng Qiu
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, New York
| | - Suning Ping
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, New York
| | - Michele Kyle
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, New York
| | - John Longo
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, New York
| | - Lawrence Chin
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, New York
| | - Li-Ru Zhao
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, New York.,VA Health Care Upstate New York, Syracuse VA Medical Center, Syracuse, New York
| |
Collapse
|
20
|
Cristóvão JS, Gomes CM. S100 Proteins in Alzheimer's Disease. Front Neurosci 2019; 13:463. [PMID: 31156365 PMCID: PMC6532343 DOI: 10.3389/fnins.2019.00463] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/24/2019] [Indexed: 01/05/2023] Open
Abstract
S100 proteins are calcium-binding proteins that regulate several processes associated with Alzheimer's disease (AD) but whose contribution and direct involvement in disease pathophysiology remains to be fully established. Due to neuroinflammation in AD patients, the levels of several S100 proteins are increased in the brain and some S100s play roles related to the processing of the amyloid precursor protein, regulation of amyloid beta peptide (Aβ) levels and Tau phosphorylation. S100 proteins are found associated with protein inclusions, either within plaques or as isolated S100-positive puncta, which suggests an active role in the formation of amyloid aggregates. Indeed, interactions between S100 proteins and aggregating Aβ indicate regulatory roles over the aggregation process, which may either delay or aggravate aggregation, depending on disease stage and relative S100 and Aβ levels. Additionally, S100s are also known to influence AD-related signaling pathways and levels of other cytokines. Recent evidence also suggests that metal-ligation by S100 proteins influences trace metal homeostasis in the brain, particularly of zinc, which is also a major deregulated process in AD. Altogether, this evidence strongly suggests a role of S100 proteins as key players in several AD-linked physiopathological processes, which we discuss in this review.
Collapse
Affiliation(s)
- Joana S. Cristóvão
- Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
- Departamento de Química e Bioquímica, Universidade de Lisboa, Lisbon, Portugal
| | - Cláudio M. Gomes
- Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
- Departamento de Química e Bioquímica, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
21
|
Wang C, Iashchishyn IA, Kara J, Foderà V, Vetri V, Sancataldo G, Marklund N, Morozova-Roche LA. Proinflammatory and amyloidogenic S100A9 induced by traumatic brain injury in mouse model. Neurosci Lett 2019; 699:199-205. [PMID: 30753908 DOI: 10.1016/j.neulet.2019.02.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 02/07/2019] [Accepted: 02/08/2019] [Indexed: 12/27/2022]
Abstract
Traumatic brain injury (TBI) represents a significant risk factor for development of neurodegenerative diseases such as Alzheimer's and Parkinson's. The S100A9-driven amyloid-neuroinflammatory cascade occurring during primary and secondary TBI events can serve as a mechanistic link between TBI and Alzheimer's as demonstrated recently in the human brain tissues. Here by using immunohistochemistry in the controlled cortical impact TBI mouse model we have found pro-inflammatory S100A9 in the brain tissues of all mice on the first and third post-TBI days, while 70% of mice did not show any S100A9 presence on seventh post-TBI day similar to controls. This indicates that defensive mechanisms effectively cleared S100A9 in these mouse brain tissues during post-TBI recovery. By using sequential immunohistochemistry we have shown that S100A9 was produced by both neuronal and microglial cells. However, Aβ peptide deposits characteristic for Alzheimer's disease were not detected in any post-TBI animals. On the first and third post-TBI days S100A9 was found to aggregate intracellularly into amyloid oligomers, similar to what was previously observed in human TBI tissues. Complementary, by using Rayleigh scatting, intrinsic fluorescence and atomic force microscopy we demonstrated that in vitro S100A9 self-assembles into amyloid oligomers within minutes. Its amyloid aggregation is highly dependent on changes of environmental conditions such as variation of calcium levels, pH, temperature and reduction/oxidation, which might be relevant to perturbation of cellular and tissues homeostasis under TBI. Present results demonstrate that S100A9 induction mechanisms in TBI are similar in mice and humans, emphasizing that S100A9 is an important marker of brain injury and therefore can be a potential therapeutic target.
Collapse
Affiliation(s)
- Chao Wang
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187 Umeå, Sweden
| | - Igor A Iashchishyn
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187 Umeå, Sweden
| | - John Kara
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187 Umeå, Sweden
| | - Vito Foderà
- Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Valeria Vetri
- Dipartimento di Fisica e Chimica e Aten Center Universitá di Palermo, 90128 Palermo, Italy
| | - Giuseppe Sancataldo
- Dipartimento di Fisica e Chimica e Aten Center Universitá di Palermo, 90128 Palermo, Italy
| | - Niklas Marklund
- Department of Neurosurgery, Uppsala University Hospital, 751 85 Uppsala, Sweden
| | | |
Collapse
|
22
|
The human olfactory cleft mucus proteome and its age-related changes. Sci Rep 2018; 8:17170. [PMID: 30464187 PMCID: PMC6249231 DOI: 10.1038/s41598-018-35102-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 10/22/2018] [Indexed: 12/14/2022] Open
Abstract
Age-related decreases in olfactory sensitivity are often accompanied by a decrease in the quality of life. However, the molecular mechanisms underlying these changes are not well described. Inhaled substances including odorants are detected by sensory neurons in the olfactory cleft covered with a layer of mucus. This olfactory mucus is the first molecular machinery responsible for tissue protection and for detection of environmental odorants. Yet, little is known about the molecular identities of the actors because of the lack of information on the mucus proteome and its age-related changes. Here, we sampled human mucus from different nasal locations and from young and elderly subjects. The composition of the mucus was extensively analyzed by shotgun proteomic analysis for a vast array of proteins. We also explored correlations between the levels of each mucus proteins with the olfactory sensitivity of subjects. This analysis revealed previously unrecognized proteins with potentially important functions in olfaction. Taken together, this report describes the most comprehensive catalogue of the nasal mucus proteins to date, their positional and age-related differences, and candidate proteins associated with olfaction. This catalogue will provide fundamental information useful for future studies, such as identification of olfactory auxiliary proteins, causes of age-related declines in olfaction, and biomarkers for neurodegenerative disorders.
Collapse
|
23
|
Karthick C, Nithiyanandan S, Essa MM, Guillemin GJ, Jayachandran SK, Anusuyadevi M. Time-dependent effect of oligomeric amyloid-β (1-42)-induced hippocampal neurodegeneration in rat model of Alzheimer's disease. Neurol Res 2018; 41:139-150. [PMID: 30453864 DOI: 10.1080/01616412.2018.1544745] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Alzheimer's disease (AD) is characterized with an abnormal deposition of insoluble amyloid-beta (Aβ) peptide plaques, tangles formation and synaptic dysfunction. These result in impaired functioning of neuronal circuits and alter the behavioral response owing to activation of neurotransmitter receptors. Recently, it has been implicated that Aβ influences N-methyl d-aspartate (NMDA) receptor activation in AD; however, the molecular mechanism underlying remains unclear. Thus, emerged specific aim to study the time-course effect of oligomeric Aβ(1-42) (oAβ1-42) on the mRNA expression of genes encoding NMDA and acetylcholine receptors in the rat model of AD. METHODS Aggregated forms of synthetic Aβ peptides were injected bilaterally into the intrahippocampal region of rat brain using stereotaxic surgery. Behavioral analysis was performed using eight-arm Radial Arm Maze task at the end of experimental period. Euthanized rat brain hippocampal tissue was used to study the mRNA expression of glutamatergic and cholinergic receptor using semiquantitative reverse transcription-polymerase chain reaction. RESULTS oAβ1-42 decreased the gene expression level of α7-nicotinic acetylcholine receptor and increased the mRNA expression of NMDA receptor 2A, and -2B subunits. In particular, oAβ1-42 aggregates increased the retention time and altered the behavioral response in rats after 15 days of injection. Further, amyloid-β1-42 are highly expressed in 15 days after postinjection in hippocampus of adult rats. CONCLUSION Acute exposure of oAβ1-42 modulated differential gene expression of glutamatergic and cholinergic receptors in hippocampus of adult rats and is duration dependent reflecting changes in hippocampal circuitry system underlying learning and memory impairments. ABBREVIATIONS AD: Alzheimer's disease, Aβ: amyloid-β; oAβ1-42: oligomeric amyloid-β 1-42 full length peptide; CAM: calmodulin; CNS: central nervous system; CR: Congo red; DG: dentate gyrus; EC: entorhinal cortex; HFIP: 1,1,1,3,3,3-hexafluoro-2-propanol; IBO: ibotenic acid; NMDA: N-methyl d-aspartate; NMDAR: N-methyl d-aspartate receptor; NR2A: N-methyl d-aspartate receptor 2A; NR2B: N-methyl d-aspartate receptor 2B; ACh: acetylcholine; α7-nAChR: α7-nicotinic acetylcholine receptor; PBS: phosphate buffered saline; RAM: Radial Arm Maze; ThT: thioflavin T.
Collapse
Affiliation(s)
- Chennakesavan Karthick
- a Molecular Gerontology Laboratory, Department of Biochemistry , School of Life Sciences, Bharathidasan University , Tiruchirappalli , India
| | - Saravanan Nithiyanandan
- a Molecular Gerontology Laboratory, Department of Biochemistry , School of Life Sciences, Bharathidasan University , Tiruchirappalli , India
| | - Musthafa Mohamed Essa
- b Department of Food Science and Nutrition , College of Agriculture and Marine Sciences, Sultan Qaboos University , Muscat , Oman
| | - Gilles J Guillemin
- c Neuroinflammation group, Faculty of Medicine and Health Sciences, Deb Bailey MND Research Laboratory , Macquaire University , Sydney , Australia
| | - Swaminathan K Jayachandran
- d Molecular Cardiology and Drug Discovery Laboratory, Department of Bioinformatics , School of Life Sciences, Bharathidasan University , Tiruchirappalli , India
| | - Muthuswamy Anusuyadevi
- a Molecular Gerontology Laboratory, Department of Biochemistry , School of Life Sciences, Bharathidasan University , Tiruchirappalli , India
| |
Collapse
|
24
|
Maekawa T, Ohta T, Kume S. Pathophysiological abnormalities in the brains of Spontaneously Diabetic Torii-Lepr fa (SDT fatty) rats, a novel type 2 diabetic model. J Vet Med Sci 2018; 80:1385-1391. [PMID: 30012919 PMCID: PMC6160888 DOI: 10.1292/jvms.18-0296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
In recent years, a relationship between diabetes and neurodegenerative diseases, such as Parkinson's disease, Alzheimer disease or depression, has been proposed. In this study, pathophysiological changes in the brain, especially in the hippocampus, of male SDT fatty rats with obesity and hyperglycemia were investigated. Brains of SD rats and SDT fatty rats were collected at 32 and 58 weeks of age, and parietal cortical thickness and number of pyramidal cells in the hippocampal cornu ammonis 1 and 3 (CA1 and CA3) regions were measured. At 58 weeks of age, the parietal cortical thickness and number of pyramidal cells in the hippocampal CA1 and CA3 regions were lower in SDT fatty rats than in age-matched SD rats. Measurements of mRNA in rat brains at 58 weeks of age showed that the expression of genes related to inflammatory responses (S100a9, TNFα, NF-κB) was elevated in SDT fatty rats. From the aforementioned results, changes suggestive of brain atrophy and impairment in cognitive function were observed in male SDT fatty rat brains.
Collapse
Affiliation(s)
- Tatsuya Maekawa
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan.,Laboratory of Animal Physiology and Functional Anatomy, Graduate School of Agriculture, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Takeshi Ohta
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Shinichi Kume
- Laboratory of Animal Physiology and Functional Anatomy, Graduate School of Agriculture, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan
| |
Collapse
|
25
|
Wang C, Iashchishyn IA, Pansieri J, Nyström S, Klementieva O, Kara J, Horvath I, Moskalenko R, Rofougaran R, Gouras G, Kovacs GG, Shankar SK, Morozova-Roche LA. S100A9-Driven Amyloid-Neuroinflammatory Cascade in Traumatic Brain Injury as a Precursor State for Alzheimer's Disease. Sci Rep 2018; 8:12836. [PMID: 30150640 PMCID: PMC6110751 DOI: 10.1038/s41598-018-31141-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 08/13/2018] [Indexed: 12/22/2022] Open
Abstract
Pro-inflammatory and amyloidogenic S100A9 protein is an important contributor to Alzheimer's disease (AD) pathology. Traumatic brain injury (TBI) is viewed as a precursor state for AD. Here we have shown that S100A9-driven amyloid-neuroinflammatory cascade was initiated in TBI and may serve as a mechanistic link between TBI and AD. By analyzing the TBI and AD human brain tissues, we demonstrated that in post-TBI tissues S100A9, produced by neurons and microglia, becomes drastically abundant compared to Aβ and contributes to both precursor-plaque formation and intracellular amyloid oligomerization. Conditions implicated in TBI, such as elevated S100A9 concentration, acidification and fever, provide strong positive feedback for S100A9 nucleation-dependent amyloid formation and delay in its proteinase clearance. Consequently, both intracellular and extracellular S100A9 oligomerization correlated with TBI secondary neuronal loss. Common morphology of TBI and AD plaques indicated their similar initiation around multiple aggregation centers. Importantly, in AD and TBI we found S100A9 plaques without Aβ. S100A9 and Aβ plaque pathology was significantly advanced in AD cases with TBI history at earlier age, signifying TBI as a risk factor. These new findings highlight the detrimental consequences of prolonged post-TBI neuroinflammation, which can sustain S100A9-driven amyloid-neurodegenerative cascade as a specific mechanism leading to AD development.
Collapse
Affiliation(s)
- Chao Wang
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden.
| | - Igor A Iashchishyn
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden.,Department of General Chemistry, Sumy State University, Sumy, 40000, Ukraine
| | - Jonathan Pansieri
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden
| | - Sofie Nyström
- IFM-Department of Chemistry, Linköping University, 58183, Linköping, Sweden
| | - Oxana Klementieva
- Department of Experimental Medical Sciences, Lund University, 22184, Lund, Sweden
| | - John Kara
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden
| | - Istvan Horvath
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden
| | - Roman Moskalenko
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden.,Department of Pathology, Sumy State University, Sumy, 40000, Ukraine
| | - Reza Rofougaran
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden
| | - Gunnar Gouras
- Department of Experimental Medical Sciences, Lund University, 22184, Lund, Sweden
| | - Gabor G Kovacs
- Institute of Neurology, Medical University of Vienna, 1097, Vienna, Austria
| | - S K Shankar
- Human Brain Tissue Repository, Department of Neuropathology, National Institute of Mental Health and Neurosciences, 560029, Bangalore, India
| | | |
Collapse
|
26
|
Iashchishyn IA, Gruden MA, Moskalenko RA, Davydova TV, Wang C, Sewell RDE, Morozova-Roche LA. Intranasally Administered S100A9 Amyloids Induced Cellular Stress, Amyloid Seeding, and Behavioral Impairment in Aged Mice. ACS Chem Neurosci 2018; 9:1338-1348. [PMID: 29618200 DOI: 10.1021/acschemneuro.7b00512] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Amyloid formation and neuroinflammation are major features of Alzheimer's disease pathology. Proinflammatory mediator S100A9 was shown to act as a link between the amyloid and neuroinflammatory cascades in Alzheimer's disease, leading together with Aβ to plaque formation, neuronal loss and memory impairment. In order to examine if S100A9 alone in its native and amyloid states can induce neuronal stress and memory impairment, we have administered S100A9 species intranasally to aged mice. Single and sequential immunohistochemistry and passive avoidance behavioral test were conducted to evaluate the consequences. Administered S100A9 species induced widespread cellular stress responses in cerebral structures, including frontal lobe, hippocampus and cerebellum. These were manifested by increased levels of S100A9, Bax, and to a lesser extent activated caspase-3 immunopositive cells. Upon administration of S100A9 fibrils, the amyloid oligomerization was observed in the brain tissues, which can further exacerbate cellular stress. The cellular stress responses correlated with significantly increased training and decreased retention latencies measured in the passive avoidance test for the S100A9 treated animal groups. Remarkably, the effect size in the behavioral tests was moderate already in the group treated with native S100A9, while the effect sizes were large in the groups administered S100A9 amyloid oligomers or fibrils. The findings demonstrate the brain susceptibility to neurotoxic damage of S100A9 species leading to behavioral and memory impairments. Intranasal administration of S100A9 species proved to be an effective method to study amyloid induced brain dysfunctions, and S100A9 itself may be postulated as a target to allay early stage neurodegenerative and neuroinflammatory processes.
Collapse
Affiliation(s)
- Igor A. Iashchishyn
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå SE-90187, Sweden
- Department of General Chemistry, Sumy State University, Sumy 40007, Ukraine
| | - Marina A. Gruden
- Department of Functional Neurochemistry, P. K. Anokhin Research Institute of Normal Physiology, Moscow 125315, Russia
| | - Roman A. Moskalenko
- Department of Pathology, Sumy State University, Sumy 40007, Ukraine
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå SE-90187, Sweden
| | - Tatiana V. Davydova
- Department of Neuroimmunopathology, Research Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
| | - Chao Wang
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå SE-90187, Sweden
| | - Robert D. E. Sewell
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3NB, United Kingdom
| | | |
Collapse
|
27
|
Horvath I, Iashchishyn IA, Moskalenko RA, Wang C, Wärmländer SKTS, Wallin C, Gräslund A, Kovacs GG, Morozova-Roche LA. Co-aggregation of pro-inflammatory S100A9 with α-synuclein in Parkinson's disease: ex vivo and in vitro studies. J Neuroinflammation 2018; 15:172. [PMID: 29866153 PMCID: PMC5987543 DOI: 10.1186/s12974-018-1210-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/20/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Chronic neuroinflammation is a hallmark of Parkinson's disease (PD) pathophysiology, associated with increased levels of pro-inflammatory factors in PD brain tissues. The pro-inflammatory mediator and highly amyloidogenic protein S100A9 is involved in the amyloid-neuroinflammatory cascade in Alzheimer's disease. This is the first report on the co-aggregation of α-synuclein (α-syn) and S100A9 both in vitro and ex vivo in PD brain. METHODS Single and sequential immunohistochemistry, immunofluorescence, scanning electron and atomic force (AFM) microscopies were used to analyze the ex vivo PD brain tissues for S100A9 and α-syn location and aggregation. In vitro studies revealing S100A9 and α-syn interaction and co-aggregation were conducted by NMR, circular dichroism, Thioflavin-T fluorescence, AFM, and surface plasmon resonance methods. RESULTS Co-localized and co-aggregated S100A9 and α-syn were found in 20% Lewy bodies and 77% neuronal cells in the substantia nigra; both proteins were also observed in Lewy bodies in PD frontal lobe (Braak stages 4-6). Lewy bodies were characterized by ca. 10-23 μm outer diameter, with S100A9 and α-syn being co-localized in the same lamellar structures. S100A9 was also detected in neurons and blood vessels of the aged patients without PD, but in much lesser extent. In vitro S100A9 and α-syn were shown to interact with each other via the α-syn C-terminus with an apparent dissociation constant of ca. 5 μM. Their co-aggregation occurred significantly faster and led to formation of larger amyloid aggregates than the self-assembly of individual proteins. S100A9 amyloid oligomers were more toxic than those of α-syn, while co-aggregation of both proteins mitigated the cytotoxicity of S100A9 oligomers. CONCLUSIONS We suggest that sustained neuroinflammation promoting the spread of amyloidogenic S100A9 in the brain tissues may trigger the amyloid cascade involving α-syn and S100A9 and leading to PD, similar to the effect of S100A9 and Aβ co-aggregation in Alzheimer's disease. The finding of S100A9 involvement in PD may open a new avenue for therapeutic interventions targeting S100A9 and preventing its amyloid self-assembly in affected brain tissues.
Collapse
Affiliation(s)
- Istvan Horvath
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden
| | - Igor A Iashchishyn
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden
- Department of General Chemistry, Sumy State University, Sumy, 40007, Ukraine
| | - Roman A Moskalenko
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden
- Department of Pathology, Sumy State University, Sumy, 40007, Ukraine
| | - Chao Wang
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden
| | | | - Cecilia Wallin
- Department of Biochemistry and Biophysics, Stockholm University, 10691, Stockholm, Sweden
| | - Astrid Gräslund
- Department of Biochemistry and Biophysics, Stockholm University, 10691, Stockholm, Sweden
| | - Gabor G Kovacs
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
28
|
Cao T, Zhou X, Zheng X, Cui Y, Tsien JZ, Li C, Wang H. Histone Deacetylase Inhibitor Alleviates the Neurodegenerative Phenotypes and Histone Dysregulation in Presenilins-Deficient Mice. Front Aging Neurosci 2018; 10:137. [PMID: 29867447 PMCID: PMC5962686 DOI: 10.3389/fnagi.2018.00137] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 04/24/2018] [Indexed: 11/28/2022] Open
Abstract
Histone acetylation has been shown to play a crucial role in memory formation, and histone deacetylase (HDAC) inhibitor sodium butyrate (NaB) has been demonstrated to improve memory performance and rescue the neurodegeneration of several Alzheimer’s Disease (AD) mouse models. The forebrain presenilin-1 and presenilin-2 conditional double knockout (cDKO) mice showed memory impairment, forebrain degeneration, tau hyperphosphorylation and inflammation that closely mimics AD-like phenotypes. In this article, we have investigated the effects of systemic administration of NaB on neurodegenerative phenotypes in cDKO mice. We found that chronic NaB treatment significantly restored contextual memory but did not alter cued memory in cDKO mice while such an effect was not permanent after treatment withdrawal. We further revealed that NaB treatment did not rescue reduced synaptic numbers and cortical shrinkage in cDKO mice, but significantly increased the neurogenesis in subgranular zone of dentate gyrus (DG). We also observed that tau hyperphosphorylation and inflammation related protein glial fibrillary acidic protein (GFAP) level were decreased in cDKO mice by NaB. Furthermore, GO and pathway analysis for the RNA-Seq data demonstrated that NaB treatment induced enrichment of transcripts associated with inflammation/immune processes and cytokine-cytokine receptor interactions. RT-PCR confirmed that NaB treatment inhibited the expression of inflammation related genes such as S100a9 and Ccl4 found upregulated in the brain of cDKO mice. Surprisingly, the level of brain histone acetylation in cDKO mice was dramatically increased and was decreased by the administration of NaB, which may reflect dysregulation of histone acetylation underlying memory impairment in cDKO mice. These results shed some lights on the possible molecular mechanisms of HDAC inhibitor in alleviating the neurodegenerative phenotypes of cDKO mice and provide a promising target for treating AD.
Collapse
Affiliation(s)
- Ting Cao
- Shanghai Key Laboratory of Brain Functional Genomics, Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Psychology and Cognitive Science, East China Normal University Shanghai, China
| | - Xiaojuan Zhou
- Shanghai Key Laboratory of Brain Functional Genomics, Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Psychology and Cognitive Science, East China Normal University Shanghai, China
| | - Xianjie Zheng
- Shanghai Key Laboratory of Brain Functional Genomics, Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Psychology and Cognitive Science, East China Normal University Shanghai, China
| | - Yue Cui
- Shanghai Key Laboratory of Brain Functional Genomics, Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Psychology and Cognitive Science, East China Normal University Shanghai, China
| | - Joe Z Tsien
- Brain and Behavior Discovery Institute and Department of Neurology, Medical College of Georgia at Augusta University Augusta, GA, United States
| | - Chunxia Li
- Shanghai Key Laboratory of Brain Functional Genomics, Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Psychology and Cognitive Science, East China Normal University Shanghai, China
| | - Huimin Wang
- Shanghai Key Laboratory of Brain Functional Genomics, Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Psychology and Cognitive Science, East China Normal University Shanghai, China.,NYU-ECNU Institute of Brain and Cognitive Science, New York University Shanghai Shanghai, China.,Shanghai Changning-ECNU Mental Health Center Shanghai, China
| |
Collapse
|
29
|
Jhang KA, Park JS, Kim HS, Chong YH. Sulforaphane rescues amyloid-β peptide-mediated decrease in MerTK expression through its anti-inflammatory effect in human THP-1 macrophages. J Neuroinflammation 2018. [PMID: 29530050 PMCID: PMC5848511 DOI: 10.1186/s12974-018-1112-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background Mer tyrosine kinase (MerTK) activity necessary for amyloid-stimulated phagocytosis strongly implicates that MerTK dysregulation might contribute to chronic inflammation implicated in Alzheimer’s disease (AD) pathology. However, the precise mechanism involved in the regulation of MerTK expression by amyloid-β (Aβ) in proinflammatory environment has not yet been ascertained. Methods The objective of this study was to determine the underlying mechanism involved in Aβ-mediated decrease in MerTK expression through Aβ-mediated regulation of MerTK expression and its modulation by sulforaphane in human THP-1 macrophages challenged with Aβ1-42. We used protein preparation, Ca2+ influx fluorescence imaging, nuclear fractionation, Western blotting techniques, and small interfering RNA (siRNA) knockdown to perform our study. Results Aβ1-42 elicited a marked decrease in MerTK expression along with increased intracellular Ca2+ level and induction of proinflammatory cytokines such as IL-1β and TNF-α. Ionomycin A and thapsigargin also increased intracellular Ca2+ levels and production of IL-1β and TNF-α, mimicking the effect of Aβ1-42. In contrast, the Aβ1-42-evoked responses were attenuated by depletion of Ca2+ with ethylene glycol tetraacetic acid. Furthermore, recombinant IL-1β or TNF-α elicited a decrease in MerTK expression. However, immunodepletion of IL-1β or TNF-α with neutralizing antibodies significantly inhibited Aβ1-42-mediated downregulation of MerTK expression. Notably, sulforaphane treatment potently inhibited Aβ1-42-induced intracellular Ca2+ level and rescued the decrease in MerTK expression by blocking nuclear factor-κB (NF-κB) nuclear translocation, thereby decreasing IL-1β and TNF-α production upon Aβ1-42 stimulation. Such adverse effects of sulforaphane were replicated by BAY 11-7082, a NF-κB inhibitor. Moreover, sulforaphane’s anti-inflammatory effects on Aβ1-42-induced production of IL-1β and TNF-α were significantly diminished by siRNA-mediated knockdown of MerTK, confirming a critical role of MerTK in suppressing Aβ1-42-induced innate immune response. Conclusion These findings implicate that targeting of MerTK with phytochemical sulforaphane as a mechanism for preventing Aβ1-42-induced neuroinflammation has potential to be applied in AD therapeutics.
Collapse
Affiliation(s)
- Kyoung A Jhang
- Department of Microbiology, Division of Molecular Biology and Neuroscience, School of Medicine, Ewha Medical Research Institute, Ewha Womans University, Seoul, 158-710, Republic of Korea
| | - Jin-Sun Park
- Department of Molecular Medicine, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, 158-710, Republic of Korea
| | - Hee-Sun Kim
- Department of Molecular Medicine, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, 158-710, Republic of Korea.
| | - Young Hae Chong
- Department of Microbiology, Division of Molecular Biology and Neuroscience, School of Medicine, Ewha Medical Research Institute, Ewha Womans University, Seoul, 158-710, Republic of Korea.
| |
Collapse
|
30
|
Swindell WR, Xing X, Fritz Y, Diaconu D, Simon DI, Ward NL, Gudjonsson JE. Deficiency of myeloid-related proteins 8 and 14 (Mrp8/Mrp14) does not block inflammaging but prevents steatosis. Oncotarget 2018; 7:35535-35551. [PMID: 27224926 PMCID: PMC5094943 DOI: 10.18632/oncotarget.9550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 05/13/2016] [Indexed: 12/24/2022] Open
Abstract
The Mrp8 and Mrp14 proteins (calprotectin) accumulate within tissues during aging and may contribute to chronic inflammation. To address this possibility, we evaluated female calprotectin-deficient Mrp14-KO and wild-type (WT) mice at 5 and 24 months of age. However, there was no evidence that age-related inflammation is blunted in KO mice. Inflammation markers were in fact elevated in livers from old KO mice, and microarray analysis revealed more consistent elevation of genes specifically expressed by B-cells and T-cells. Adipose-specific genes, however, were less consistently elevated in aged KO mice, suggesting an anti-steatosis effect of Mrp8/14 deficiency. Consistent with this, genes decreased by the anti-steatosis agent SRT1720 were decreased in old KO compared to old WT mice. Expression of lipid metabolism genes was altered in KO mice at 5 months of age, along with genes associated with development, biosynthesis and immunity. These early-age effects of Mrp8/14 deficiency, in the absence of any external stressor, were unexpected. Taken together, our findings demonstrate a pro-steatosis rather than pro-inflammatory role of calprotectin within the aging liver. This appears to reflect a developmental-metabolic phenotype of Mrp14-KO mice that is manifest at a young age in the absence of pro-inflammatory stimuli.
Collapse
Affiliation(s)
- William R Swindell
- Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA.,Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Xianying Xing
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Yi Fritz
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA
| | - Doina Diaconu
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA
| | - Daniel I Simon
- Harrington Heart and Vascular Institute, University Hospitals Case Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Nicole L Ward
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA.,The Murdough Family Center for Psoriasis, Case Western Reserve University, Cleveland, OH, USA
| | | |
Collapse
|
31
|
Lee HJ, Savelieff MG, Kang J, Brophy MB, Nakashige TG, Lee SJC, Nolan EM, Lim MH. Calprotectin influences the aggregation of metal-free and metal-bound amyloid-β by direct interaction. Metallomics 2018; 10:1116-1127. [DOI: 10.1039/c8mt00091c] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
CP-Ser [S100A8(C42S)/S100A9(C3S) oligomer] interacts with metal-free and metal-bound Aβ40 peptides and modulates their aggregation in the absence and presence of metal ions.
Collapse
Affiliation(s)
- Hyuck Jin Lee
- Department of Chemistry
- Korea Advanced Institute of Science and Technology (KAIST)
- Daejeon 34141
- Republic of Korea
| | | | - Juhye Kang
- Department of Chemistry
- Korea Advanced Institute of Science and Technology (KAIST)
- Daejeon 34141
- Republic of Korea
- Department of Chemistry
| | | | | | - Shin Jung C. Lee
- Department of Chemistry
- Ulsan National Institute of Science and Technology (UNIST)
- Ulsan 44919
- Republic of Korea
| | - Elizabeth M. Nolan
- Department of Chemistry
- Massachusetts Institute of Technology (MIT)
- Cambridge
- USA
| | - Mi Hee Lim
- Department of Chemistry
- Korea Advanced Institute of Science and Technology (KAIST)
- Daejeon 34141
- Republic of Korea
| |
Collapse
|
32
|
Iashchishyn IA, Sulskis D, Nguyen Ngoc M, Smirnovas V, Morozova-Roche LA. Finke-Watzky Two-Step Nucleation-Autocatalysis Model of S100A9 Amyloid Formation: Protein Misfolding as "Nucleation" Event. ACS Chem Neurosci 2017; 8:2152-2158. [PMID: 28759719 DOI: 10.1021/acschemneuro.7b00251] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Quantitative kinetic analysis is critical for understanding amyloid mechanisms. Here we demonstrate the application of generic Finke-Watzky (F-W) two-step nucleation-autocatalytic growth model to the concentration-dependent amyloid kinetics of proinflammatory α-helical S100A9 protein at pH 7.4 and at 37 and 42 °C. The model is based on two pseudoelementary reaction steps applied without further analytical constraints, and its treatment of S100A9 amyloid self-assembly demonstrates that initial misfolding and β-sheet formation, defined as "nucleation" step, spontaneously takes place within individual S100A9 molecules at higher rate than the subsequent fibrillar growth. The latter, described as an autocatalytic process, will proceed if misfolded amyloid-prone S100A9 is populated on a macroscopic time scale. Short lengths of S100A9 fibrils are consistent with the F-W model. The analysis of fibrillar length distribution by the Beker-Döring model demonstrates independently that such distribution is solely determined by slow fibril growth and there is no fragmentation or secondary pathways decreasing fibrillar length.
Collapse
Affiliation(s)
- Igor A. Iashchishyn
- Department
of Medical Biochemistry and Biophysics, Umeå University, SE-901 87 Umeå, Sweden
- Department
of General Chemistry, Sumy State University, 40007 Sumy, Ukraine
| | - Darius Sulskis
- Department
of Medical Biochemistry and Biophysics, Umeå University, SE-901 87 Umeå, Sweden
- Department
of Biothermodynamics and Drug Design, Institute of Biotechnology, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Mai Nguyen Ngoc
- Department
of Biothermodynamics and Drug Design, Institute of Biotechnology, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Vytautas Smirnovas
- Department
of Biothermodynamics and Drug Design, Institute of Biotechnology, Vilnius University, LT-10257 Vilnius, Lithuania
| | | |
Collapse
|
33
|
Small things matter: Implications of APP intracellular domain AICD nuclear signaling in the progression and pathogenesis of Alzheimer’s disease. Prog Neurobiol 2017; 156:189-213. [DOI: 10.1016/j.pneurobio.2017.05.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/25/2017] [Accepted: 05/30/2017] [Indexed: 01/08/2023]
|
34
|
Lodeiro M, Puerta E, Ismail MAM, Rodriguez-Rodriguez P, Rönnbäck A, Codita A, Parrado-Fernandez C, Maioli S, Gil-Bea F, Merino-Serrais P, Cedazo-Minguez A. Aggregation of the Inflammatory S100A8 Precedes Aβ Plaque Formation in Transgenic APP Mice: Positive Feedback for S100A8 and Aβ Productions. J Gerontol A Biol Sci Med Sci 2017; 72:319-328. [PMID: 27131040 DOI: 10.1093/gerona/glw073] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 04/07/2016] [Indexed: 11/12/2022] Open
Abstract
Inflammation plays an important role in Alzheimer's disease (AD) and other neurodegenerative disorders. Although chronic inflammation in later stages of AD is well described, little is known about the inflammatory processes in preclinical or early stages of the disease prior to plaque deposition. In this study, we report that the inflammatory mediator S100A8 is increased with aging in the mouse brain. It is observed as extracellular aggregates, which do not correspond to corpora amylacea. S100A8 aggregation is enhanced in the hippocampi of two different mouse models for amyloid-β (Aβ) overproduction (Tg2576 and TgAPParctic mice). S100A8 aggregates are seen prior the formation of Aβ plaques and do not colocalize. In vitro treatment of glial cells from primary cultures with Aβ42 resulted in an increased production of S100A8. In parallel, treatment of a neuronal cell line with recombinant S100A8 protein resulted in enhanced Aβ42 and decreased Aβ40 production. Our results suggest that important inflammatory processes are occurring prior to Aβ deposition and the existence of a positive feedback between S100A8 and Aβ productions. The possible relevance of aging- or AD-dependent formation of S100A8 aggregates in the hippocampus thus affecting learning and memory processes is discussed.
Collapse
Affiliation(s)
- Maria Lodeiro
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Elena Puerta
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Muhammad-Al-Mustafa Ismail
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Patricia Rodriguez-Rodriguez
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Annica Rönnbäck
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Alina Codita
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Cristina Parrado-Fernandez
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Silvia Maioli
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Francisco Gil-Bea
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Division of Neurosciences, Department of Cellular and Molecular Neuropharmacology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Paula Merino-Serrais
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Angel Cedazo-Minguez
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
35
|
Mahmoudvand H, Ziaali N, Ghazvini H, Shojaee S, Keshavarz H, Esmaeilpour K, Sheibani V. Toxoplasma gondii Infection Promotes Neuroinflammation Through Cytokine Networks and Induced Hyperalgesia in BALB/c Mice. Inflammation 2016; 39:405-412. [PMID: 26490968 DOI: 10.1007/s10753-015-0262-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
We hypothesized that in Toxoplasma gondii infection, communication among immune cells promotes neuroinflammation through cytokine networks and induces pain sensitivity under conditions of neuropathic pain. The animal model of Toxoplasma infection was established by the intraperitoneal inoculation of 20-25 tissue cysts from Tehran strain of T. gondii to BALB/c mice. Amitriptyline (20 mg/kg, i.p., 1/day) administrated to animals for 7 days before behavioral tests. Pain behavioral tests including tail flick, hot plate, and formalin test were evaluated in all the groups. The mRNA levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6 were examined by real-time PCR. Results revealed that T. gondii induce hyperalgesia in the infected mice, whereas amitriptyline showed a promising effect against the hyperalgesia induced by Toxoplasma infection. The mRNA levels of the aforementioned cytokines significantly (P < 0.05) increased in the infected mice compared to the uninfected ones. Obtained findings suggested that T. gondii infection could promote neuroinflammation through cytokine networks and induced hyperalgesia in BALB/c mice, whereas amitriptyline as an analgesic drug reverses them.
Collapse
Affiliation(s)
- Hossein Mahmoudvand
- Research Center for Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran
| | - Naser Ziaali
- Research Center for Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamed Ghazvini
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Saeideh Shojaee
- Department of Medical Parasitology & Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Keshavarz
- Department of Medical Parasitology & Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Khadijeh Esmaeilpour
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Sheibani
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
36
|
Mahmoudvand H, Ziaali N, Aghaei I, Sheibani V, Shojaee S, Keshavarz H, Shabani M. The possible association between Toxoplasma gondii infection and risk of anxiety and cognitive disorders in BALB/c mice. Pathog Glob Health 2016; 109:369-76. [PMID: 26924347 DOI: 10.1080/20477724.2015.1117742] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
There are conflicting reports concerning the association of Toxoplasma gondii infection with increased risk of mental disorders. This investigation will provide a good understanding about defining the possible association between T. gondii exposure and risk of anxiety and cognitive alterations. Besides, a secondary objective of this study was to determine the effect of pioglitazone administration on the possible alterations induced by T. gondii exposure. Male BALB/c mice were used for this study. The animal model of Toxoplasma infection was established by the intraperitoneal inoculation of 20-25 tissue cysts from Tehran strain of T. gondii. Pioglitazone (20 mg/kg, i.p.1/day) was administered to the animals for 2 weeks before behavioural tests. Behavioural tests including open-field, elevated plus-maze and passive avoidance learning were evaluated in the groups. Since cytokines were implicated as a contributing factor for mood disorders, the mRNA levels of TNF-α, IL-1β, IL-6 as well as inducible nitric oxide synthase (iNOs) were examined by real-time PCR. Findings demonstrated that T. gondii caused anxiety-like symptoms and impaired cognitive functions of the infected BALB/c mice, whereas pioglitazone, a peroxisome proliferator-activated receptor agonist, showed a promising effect against the cognitive impairments induced by Toxoplasma infection. The results also revealed that the mRNA levels of the aforementioned cytokines were significantly (p < 0.05) increased in the infected mice compared to the uninfected BALB/c ones. Pioglitazone can be offered as a potential neuroprotective agent in the treatment of patients with T. gondii infection that manifests anxiety and cognitive impairments; however, further studies are needed to clarify the exact mechanisms.
Collapse
Affiliation(s)
- Hossein Mahmoudvand
- 1 Department of Medical Parasitology and Mycology, Lorestan University of Medical Sciences , Khorramabad, Iran
| | | | | | | | | | | | | |
Collapse
|
37
|
Mahmoudvand H, Sheibani V, Shojaee S, Mirbadie SR, Keshavarz H, Esmaeelpour K, Keyhani AR, Ziaali N. Toxoplasma gondii Infection Potentiates Cognitive Impairments of Alzheimer's Disease in the BALB/c Mice. J Parasitol 2016; 102:629-635. [PMID: 27513205 DOI: 10.1645/16-28] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
This study tests the hypothesis that in chronic Toxoplasma gondii infection communication among immune cells promotes neuroinflammation through cytokine networks and potentiate cognitive impairments in BALB/c mice with Alzheimer's disease (AD). The animal model of Toxoplasma infection was established by the intraperitoneal inoculation of 20-25 tissue cysts from the Tehran strain of T. gondii . We injected amyloid-beta 1-42 peptide (Aβ1-42, 1 and 2 μl) into the hippocampus of BALB/c mice to establish an animal model of AD. The behavioral experiments such as spatial learning and memory were performed using the Morris water maze test. The mRNA levels of TNF-α, IL-1β, IFN-γ, and inducible nitric oxide synthase (iNOS) were examined by real-time PCR. We found that T. gondii infection caused AD-like symptoms and impaired learning and memory functions of the infected BALB/c mice. We also found that in Toxoplasma infection + Aβ1-42 (1 μl) group, T. gondii infection could potentiate AD in infected mice receiving subdoses of Aβ1-42 (1 μl) and caused considerable impairment in learning and memory functions similar to AD group. Comparison of the results demonstrated that mRNA levels of IL-1β, TNF-α, IFN-γ, and iNOS significantly (P < 0.001) increased in T. gondii + Aβ1-42 (1 μl) in comparison with the other tested groups. The obtained results showed that chronic T. gondii infection communication among immune cells promotes neuroinflammation through cytokine networks and induces pathological progression of AD in the mice brain, whereas the presence of neuroanatomical Toxoplasma tissue cysts in the brain could also affect the behavioral functions in T. gondii -infected mice.
Collapse
Affiliation(s)
- Hossein Mahmoudvand
- Department of Medical Parasitology and Mycology, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Vahid Sheibani
- Department of Medical Parasitology and Mycology, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Saeideh Shojaee
- Department of Medical Parasitology and Mycology, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Seyed Reza Mirbadie
- Department of Medical Parasitology and Mycology, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hossein Keshavarz
- Department of Medical Parasitology and Mycology, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Khadijeh Esmaeelpour
- Department of Medical Parasitology and Mycology, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Ali Reza Keyhani
- Department of Medical Parasitology and Mycology, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Naser Ziaali
- Department of Medical Parasitology and Mycology, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
38
|
Chang KA, Shin KY, Nam E, Lee YB, Moon C, Suh YH, Lee SH. Plasma soluble neuregulin-1 as a diagnostic biomarker for Alzheimer's disease. Neurochem Int 2016; 97:1-7. [PMID: 27133777 DOI: 10.1016/j.neuint.2016.04.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 04/12/2016] [Accepted: 04/26/2016] [Indexed: 01/14/2023]
Abstract
To identify some apparent biomarker candidates for the diagnosis of Alzheimer's disease (AD) pathology, we investigated whether there would be a significant difference between the levels of the plasma proteins of AD patients and healthy people. A total of 115 subjects were enrolled, 60 individuals with AD and 55 healthy controls. There was a statistical difference in the mini-mental status exam (MMSE) scores and the clinical dementia rating (CDR) scores between the two groups. We used the immunoblotting assay to analyze several plasma proteins in the subjects. Amyloid-β (Aβ), S100a9, and soluble neuregulin-1 (sNRG-1), including α-synuclein (α-Syn) as a detection control were detected in the plasma samples. Unlike Aβ, S100a9 and α-Syn, the level of sNRG-1 of the AD patients was significantly higher than that of the healthy control subjects. The AD patients were divided into mild and moderate groups according to their MMSE and CDR scores. We found a significant correlation between the level of sNRG-1 and MMSE scores. The sNRG-1 level was significantly higher in mild AD patients as well as in moderate AD patients compared with that of the control subjects. These new findings indicate that increased plasma sNRG-1 levels might be a novel and reliable biological marker for the early diagnosis of AD.
Collapse
Affiliation(s)
- Keun-A Chang
- Department of Pharmacology, College of Medicine, Neuroscience Research Institute, Gachon University, Incheon, 406-799, South Korea
| | - Ki Young Shin
- Department of Microbiology, College of Natural Science, Dankook University, Cheonan-si, Chungnam, 330-714, South Korea
| | - Eunjoo Nam
- Department of Pharmacology, College of Medicine, Neuroscience Research Institute, Gachon University, Incheon, 406-799, South Korea
| | - Yeong-Bae Lee
- Department of Neurology, Gil Medical Center, Gachon University, Incheon, 405-760, South Korea
| | - Cheil Moon
- Department of Brain Science, Graduate School, Daegu Gyeungbuk Institute of Science and Technology, Daegu, 711-873, South Korea
| | - Yoo-Hun Suh
- Department of Pharmacology, College of Medicine, Neuroscience Research Institute, Gachon University, Incheon, 406-799, South Korea.
| | - Sang Hyung Lee
- Department of Neurosurgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, 110-799, South Korea.
| |
Collapse
|
39
|
Gonzalez-Pena D, Nixon SE, O’Connor JC, Southey BR, Lawson MA, McCusker RH, Borras T, Machuca D, Hernandez AG, Dantzer R, Kelley KW, Rodriguez-Zas SL. Microglia Transcriptome Changes in a Model of Depressive Behavior after Immune Challenge. PLoS One 2016; 11:e0150858. [PMID: 26959683 PMCID: PMC4784788 DOI: 10.1371/journal.pone.0150858] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 02/20/2016] [Indexed: 12/20/2022] Open
Abstract
Depression symptoms following immune response to a challenge have been reported after the recovery from sickness. A RNA-Seq study of the dysregulation of the microglia transcriptome in a model of inflammation-associated depressive behavior was undertaken. The transcriptome of microglia from mice at day 7 after Bacille Calmette Guérin (BCG) challenge was compared to that from unchallenged Control mice and to the transcriptome from peripheral macrophages from the same mice. Among the 562 and 3,851 genes differentially expressed between BCG-challenged and Control mice in microglia and macrophages respectively, 353 genes overlapped between these cells types. Among the most differentially expressed genes in the microglia, serum amyloid A3 (Saa3) and cell adhesion molecule 3 (Cadm3) were over-expressed and coiled-coil domain containing 162 (Ccdc162) and titin-cap (Tcap) were under-expressed in BCG-challenged relative to Control. Many of the differentially expressed genes between BCG-challenged and Control mice were associated with neurological disorders encompassing depression symptoms. Across cell types, S100 calcium binding protein A9 (S100A9), interleukin 1 beta (Il1b) and kynurenine 3-monooxygenase (Kmo) were differentially expressed between challenged and control mice. Immune response, chemotaxis, and chemokine activity were among the functional categories enriched by the differentially expressed genes. Functional categories enriched among the 9,117 genes differentially expressed between cell types included leukocyte regulation and activation, chemokine and cytokine activities, MAP kinase activity, and apoptosis. More than 200 genes exhibited alternative splicing events between cell types including WNK lysine deficient protein kinase 1 (Wnk1) and microtubule-actin crosslinking factor 1(Macf1). Network visualization revealed the capability of microglia to exhibit transcriptome dysregulation in response to immune challenge still after resolution of sickness symptoms, albeit lower than that observed in macrophages. The persistent transcriptome dysregulation in the microglia shared patterns with neurological disorders indicating that the associated persistent depressive symptoms share a common transcriptome basis.
Collapse
Affiliation(s)
- Dianelys Gonzalez-Pena
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States of America
| | - Scott E. Nixon
- Illinois Informatics Institute, University of Illinois Urbana-Champaign, Urbana, IL, United States of America
| | - Jason C. O’Connor
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Bruce R. Southey
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States of America
| | - Marcus A. Lawson
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States of America
| | - Robert H. McCusker
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States of America
| | - Tania Borras
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States of America
| | - Debbie Machuca
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States of America
| | - Alvaro G. Hernandez
- High-Throughput Sequencing and Genotyping Unit, Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Champaign, IL, United States of America
| | - Robert Dantzer
- Department of Symptom Research, University of Texas M. D. Anderson Cancer Center, Houston, TX, United States of America
| | - Keith W. Kelley
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States of America
- Integrative Immunology and Behavior Program and Department of Pathology, College of Medicine, University of Illinois at Urbana-Champaign, Champaign, IL, United States of America
| | - Sandra L. Rodriguez-Zas
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States of America
- Department of Statistics and Carle Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
- * E-mail:
| |
Collapse
|
40
|
The misfolded pro-inflammatory protein S100A9 disrupts memory via neurochemical remodelling instigating an Alzheimer's disease-like cognitive deficit. Behav Brain Res 2016; 306:106-16. [PMID: 26965570 DOI: 10.1016/j.bbr.2016.03.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 02/15/2016] [Accepted: 03/05/2016] [Indexed: 12/14/2022]
Abstract
Memory deficits may develop from a variety of neuropathologies including Alzheimer's disease dementia. During neurodegenerative conditions there are contributory factors such as neuroinflammation and amyloidogenesis involved in memory impairment. In the present study, dual properties of S100A9 protein as a pro-inflammatory and amyloidogenic agent were explored in the passive avoidance memory task along with neurochemical assays in the prefrontal cortex and hippocampus of aged mice. S100A9 oligomers and fibrils were generated in vitro and verified by AFM, Thioflavin T and A11 antibody binding. Native S100A9 as well as S100A9 oligomers and fibrils or their combination were administered intranasally over 14 days followed by behavioral and neurochemical analysis. Both oligomers and fibrils evoked amnestic activity which correlated with disrupted prefrontal cortical and hippocampal dopaminergic neurochemistry. The oligomer-fibril combination produced similar but weaker neurochemistry to the fibrils administered alone but without passive avoidance amnesia. Native S100A9 did not modify memory task performance even though it generated a general and consistent decrease in monoamine levels (DA, 5-HT and NA) and increased metabolic marker ratios of DA and 5-HT turnover (DOPAC/DA, HVA/DA and 5-HIAA) in the prefrontal cortex. These results provide insight into a novel pathogenetic mechanism underlying amnesia in a fear-aggravated memory task based on amyloidogenesis of a pro-inflammatory factor leading to disrupted brain neurochemistry in the aged brain. The data further suggests that amyloid species of S100A9 create deleterious effects principally on the dopaminergic system and this novel finding might be potentially exploited during dementia management through a neuroprotective strategy.
Collapse
|
41
|
Adav SS, Gallart-Palau X, Tan KH, Lim SK, Tam JP, Sze SK. Dementia-linked amyloidosis is associated with brain protein deamidation as revealed by proteomic profiling of human brain tissues. Mol Brain 2016; 9:20. [PMID: 26892330 PMCID: PMC4759965 DOI: 10.1186/s13041-016-0200-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/11/2016] [Indexed: 01/25/2023] Open
Abstract
Background Aggregation of malformed proteins is a key feature of many neurodegenerative diseases, but the mechanisms that drive proteinopathy in the brain are poorly understood. We aimed to characterize aggregated proteins in human brain tissues affected by dementia. Results To characterize amyloidal plaque purified from post-mortem brain tissue of dementia patient, we applied ultracentrifugation-electrostatic repulsion hydrophilic interaction chromatography (UC-ERLIC) coupled mass spectrometry-based proteomics technologies. Proteomics profiling of both soluble and aggregated amyloidal plaque demonstrated significant enrichment and deamidation of S100A9, ferritin, hemoglobin subunits, creatine kinase and collagen protein among the aggregated brain proteins. Amyloidal plaques were enriched in the deamidated variant of protein S100A9, and structural analysis indicated that both the low- and high-affinity calcium binding motifs of S100A9 were deamidated exclusively in the aggregated fraction, suggesting altered charge state and function of this protein in brain tissues affected by dementia. The multiple deamidated residues of S100A9 predicts introduction of negative charge that alter Ca++ binding, suggesting increased capacity to form pathological aggregates in the brain. Conclusion UC-coupled proteomics revealed that brain amyloidal plaques are enriched in deamidated proteins, and suggested that altered charge state and calcium-binding capacity of S100A9 may enhance protein aggregation and promote neurodegeneration in the human brain. Electronic supplementary material The online version of this article (doi:10.1186/s13041-016-0200-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sunil S Adav
- School of Biological Sciences, Division of Structural Biology and Biochemistry, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore. .,Department of Maternal Fetal Medicine, KK Women's and Children's Hospital, 100 Bukit Timah Road, 229899, Singapore, Singapore.
| | - Xavier Gallart-Palau
- School of Biological Sciences, Division of Structural Biology and Biochemistry, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore.
| | - Kok Hian Tan
- Department of Maternal Fetal Medicine, KK Women's and Children's Hospital, 100 Bukit Timah Road, 229899, Singapore, Singapore.
| | - Sai Kiang Lim
- Institute of Medical Biology, A*STAR, 8A Biomedical Grove, 138648, Singapore, Singapore.
| | - James P Tam
- School of Biological Sciences, Division of Structural Biology and Biochemistry, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore.
| | - Siu Kwan Sze
- School of Biological Sciences, Division of Structural Biology and Biochemistry, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore.
| |
Collapse
|
42
|
Horvath I, Jia X, Johansson P, Wang C, Moskalenko R, Steinau A, Forsgren L, Wågberg T, Svensson J, Zetterberg H, Morozova-Roche LA. Pro-inflammatory S100A9 Protein as a Robust Biomarker Differentiating Early Stages of Cognitive Impairment in Alzheimer's Disease. ACS Chem Neurosci 2016; 7:34-9. [PMID: 26550994 DOI: 10.1021/acschemneuro.5b00265] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pro-inflammatory protein S100A9 was established as a biomarker of dementia progression and compared with others such as Aβ(1-42) and tau-proteins. CSF samples from 104 stringently diagnosed individuals divided into five subgroups were analyzed, including nondemented controls, stable mild cognitive impairment (SMCI), mild cognitive impairment due to Alzheimer's disease (MCI-AD), Alzheimer's disease (AD), and vascular dementia (VaD) patients. ELISA, dot-blotting, and electrochemical impedance spectroscopy were used as research methods. The S100A9 and Aβ(1-42) levels correlated with each other: their CSF content decreased already at the SMCI stage and declined further under MCI-AD, AD, and VaD conditions. Immunohistochemical analysis also revealed involvement of both Aβ(1-42) and S100A9 in the amyloid-neuroinflammatory cascade already during SMCI. Tau proteins were not yet altered in SMCI; however their contents increased during MCI-AD and AD, diagnosing later dementia stages. Thus, four biomarkers together, reflecting different underlying pathological causes, can accurately differentiate dementia progression and also distinguish AD from VaD.
Collapse
Affiliation(s)
- Istvan Horvath
- Department
of Medical Biochemistry and Biophysics, Umeå University, Umeå 901 87, Sweden
| | - Xueen Jia
- Department
of Medical Biochemistry and Biophysics, Umeå University, Umeå 901 87, Sweden
- Department
of Physics, Umeå University, Umeå 901 87, Sweden
| | - Per Johansson
- Department
of Neuropsychiatry, Skaraborg Central Hospital, Falköping 521 85, Sweden
- Department
of Internal Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 45, Sweden
| | - Chao Wang
- Department
of Medical Biochemistry and Biophysics, Umeå University, Umeå 901 87, Sweden
| | - Roman Moskalenko
- Department
of Medical Biochemistry and Biophysics, Umeå University, Umeå 901 87, Sweden
- Department
of Pathology, Sumy State University, Sumy 40000, Ukraine
| | - Andreas Steinau
- Department
of Medical Biochemistry and Biophysics, Umeå University, Umeå 901 87, Sweden
| | - Lars Forsgren
- Department
of Pharmacology and Clinical Neuroscience, Umeå University, Umeå 901 87, Sweden
| | - Thomas Wågberg
- Department
of Physics, Umeå University, Umeå 901 87, Sweden
| | - Johan Svensson
- Department
of Internal Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 45, Sweden
- Department
of Endocrinology, Skaraborg Central Hospital, Skövde 541 85, Sweden
| | - Henrik Zetterberg
- Clinical
Neurochemistry Laboratory, Institute of Neuroscience and Physiology,
the Sahlgrenska Academy, University of Gothenburg, Mölndal 431 80, Sweden
- University College London, Institute of Neurology, Queen Square, London WC1E 6BT, United Kingdom
| | | |
Collapse
|
43
|
Metals and Neuronal Metal Binding Proteins Implicated in Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:9812178. [PMID: 26881049 PMCID: PMC4736980 DOI: 10.1155/2016/9812178] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/17/2015] [Indexed: 11/18/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent age-related dementia affecting millions of people worldwide. Its main pathological hallmark feature is the formation of insoluble protein deposits of amyloid-β and hyperphosphorylated tau protein into extracellular plaques and intracellular neurofibrillary tangles, respectively. Many of the mechanistic details of this process remain unknown, but a well-established consequence of protein aggregation is synapse dysfunction and neuronal loss in the AD brain. Different pathways including mitochondrial dysfunction, oxidative stress, inflammation, and metal metabolism have been suggested to be implicated in this process. In particular, a body of evidence suggests that neuronal metal ions such as copper, zinc, and iron play important roles in brain function in health and disease states and altered homeostasis and distribution as a common feature across different neurodegenerative diseases and aging. In this focused review, we overview neuronal proteins that are involved in AD and whose metal binding properties may underlie important biochemical and regulatory processes occurring in the brain during the AD pathophysiological process.
Collapse
|
44
|
Kim JA, Ha S, Shin KY, Kim S, Lee KJ, Chong YH, Chang KA, Suh YH. Neural stem cell transplantation at critical period improves learning and memory through restoring synaptic impairment in Alzheimer's disease mouse model. Cell Death Dis 2015; 6:e1789. [PMID: 26086962 PMCID: PMC4669825 DOI: 10.1038/cddis.2015.138] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 04/05/2015] [Accepted: 04/22/2015] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is characterized by neuronal loss in several regions of the brain. Recent studies have suggested that stem cell transplantation could serve as a potential therapeutic strategy to halt or ameliorate the inexorable disease progression. However, the optimal stage of the disease for stem cell transplantation to have a therapeutic effect has yet to be determined. Here, we demonstrated that transplantation of neural stem cells into 12-month-old Tg2576 brains markedly improved both cognitive impairments and neuropathological features by reducing β-amyloid processing and upregulating clearance of β-amyloid, secretion of anti-inflammatory cytokines, endogenous neurogenesis, as well as synapse formation. In contrast, the stem cell transplantation did not recover cognitive dysfunction and β-amyloid neuropathology in Tg2576 mice aged 15 months when the memory loss is manifest. Overall, this study underscores that stem cell therapy at optimal time frame is crucial to obtain maximal therapeutic effects that can restore functional deficits or stop the progression of AD.
Collapse
Affiliation(s)
- J A Kim
- Department of Pharmacology, College of Medicine, Neuroscience Research Institute, MRC, Seoul National University, Seoul, 110-799, Korea
| | - S Ha
- Department of Pharmacology, College of Medicine, Neuroscience Research Institute, Gachon University, Incheon, 405-760, Korea
| | - K Y Shin
- Department of Pharmacology, College of Medicine, Neuroscience Research Institute, MRC, Seoul National University, Seoul, 110-799, Korea
| | - S Kim
- Department of Pharmacology, College of Medicine, Neuroscience Research Institute, Gachon University, Incheon, 405-760, Korea
| | - K J Lee
- Synaptic Circuit Plasticity Laboratory, Department of Structure & Function of Neural Network, Korea Brain Research Institute, 61 Cheomdan-ro, Dong-gu, Daegu 701-300, Korea
| | - Y H Chong
- Division of Molecular Biology and Neuroscience, Department of Microbiology, School of Medicine, Ewha Medical Research Institute, Ewha Womans University, Seoul, 158-710, Korea
| | - K-A Chang
- Department of Pharmacology, College of Medicine, Neuroscience Research Institute, Gachon University, Incheon, 405-760, Korea
| | - Y-H Suh
- 1] Department of Pharmacology, College of Medicine, Neuroscience Research Institute, MRC, Seoul National University, Seoul, 110-799, Korea [2] Synaptic Circuit Plasticity Laboratory, Department of Structure & Function of Neural Network, Korea Brain Research Institute, 61 Cheomdan-ro, Dong-gu, Daegu 701-300, Korea
| |
Collapse
|
45
|
Iyalomhe O, Chen Y, Allard J, Ntekim O, Johnson S, Bond V, Goerlitz D, Li J, Obisesan TO. A standardized randomized 6-month aerobic exercise-training down-regulated pro-inflammatory genes, but up-regulated anti-inflammatory, neuron survival and axon growth-related genes. Exp Gerontol 2015; 69:159-69. [PMID: 25981742 DOI: 10.1016/j.exger.2015.05.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 05/12/2015] [Accepted: 05/13/2015] [Indexed: 12/18/2022]
Abstract
There is considerable support for the view that aerobic exercise may confer cognitive benefits to mild cognitively impaired elderly persons. However, the biological mechanisms mediating these effects are not entirely clear. As a preliminary step towards informing this gap in knowledge, we enrolled older adults confirmed to have mild cognitive impairment (MCI) in a 6-month exercise program. Male and female subjects were randomized into a 6-month program of either aerobic or stretch (control) exercise. Data collected from the first 10 completers, aerobic exercise (n=5) or stretch (control) exercise (n=5), were used to determine intervention-induced changes in the global gene expression profiles of the aerobic and stretch groups. Using microarray, we identified genes with altered expression (relative to baseline values) in response to the 6-month exercise intervention. Genes whose expression were altered by at least two-fold, and met the p-value cutoff of 0.01 were inputted into the Ingenuity Pathway Knowledge Base Library to generate gene-interaction networks. After a 6-month aerobic exercise-training, genes promoting inflammation became down-regulated, whereas genes having anti-inflammatory properties and those modulating immune function or promoting neuron survival and axon growth, became up-regulated (all fold change≥±2.0, p<0.01). These changes were not observed in the stretch group. Importantly, the differences in the expression profiles correlated with significant improvement in maximal oxygen uptake (VO2max) in the aerobic program as opposed to the stretch group. We conclude that three distinct cellular pathways may collectively influence the training effects of aerobic exercise in MCI subjects. We plan to confirm these effects using rt-PCR and correlate such changes with the cognitive phenotype.
Collapse
Affiliation(s)
- Osigbemhe Iyalomhe
- Division of Geriatrics, Department of Medicine, Howard University Hospital, 2041 Georgia Ave NW, Washington, DC 20060, USA
| | - Yuanxiu Chen
- Clinical Translational Science Center, Howard University Hospital, 2041 Georgia Ave NW, Washington, DC 20060, USA
| | - Joanne Allard
- Howard University College of Medicine, 520 W Street NW, Washington, DC 20059, USA
| | - Oyonumo Ntekim
- Department of Health, Human Performance, and Leisure Studies, College of Arts and Science, Howard University College of Medicine, 520 W St NW, Washington, DC 20059, USA
| | - Sheree Johnson
- Howard University College of Medicine, 520 W Street NW, Washington, DC 20059, USA
| | - Vernon Bond
- Division of Geriatrics, Department of Medicine, Howard University Hospital, 2041 Georgia Ave NW, Washington, DC 20060, USA
| | - David Goerlitz
- Department of Molecular Biology and Informatics, Georgetown University Medical Center, 400 Reservoir Rd NW, Washington, DC 20057, USA
| | - James Li
- Department of Molecular Biology and Informatics, Georgetown University Medical Center, 400 Reservoir Rd NW, Washington, DC 20057, USA
| | - Thomas O Obisesan
- Division of Geriatrics, Department of Medicine, Howard University Hospital, 2041 Georgia Ave NW, Washington, DC 20060, USA; Clinical Translational Science Center, Howard University Hospital, 2041 Georgia Ave NW, Washington, DC 20060, USA.
| |
Collapse
|
46
|
Li G, Chen H, Cheng L, Zhao R, Zhao J, Xu Y. Amyloid precursor-like protein 2 C-terminal fragments upregulate S100A9 gene and protein expression in BV2 cells. Neural Regen Res 2014; 9:1923-8. [PMID: 25558244 PMCID: PMC4281433 DOI: 10.4103/1673-5374.145362] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2014] [Indexed: 11/16/2022] Open
Abstract
The murine microglial cell line BV2 has neuroprotective effects, but is toxic to neurons by secreting inflammatory cytokines, and is an important target in the treatment of nerve inflammation and neurodegenerative diseases. In the present study, we observed the effects of transfecting three amyloid precursor-like protein 2 (APLP2) C-terminal fragments (CTFs; C57, C50 and C31) in the pEGFP-N1 vector on S100A9 expression in BV2 cells. Reverse transcription-PCR, western blot assay and immunocytochemistry revealed that S100A9 protein and mRNA expression was greater in BV2 cells after CTF transfection than after mock transfection with an empty vector. Furthermore, transfection of full-length APLP2-751 resulted in low levels of S100A9 protein expression. Our results show that APLP2-CTFs upregulate S100A9 protein and mRNA expression in BV2 cells, and identify a novel pathway involved in neuronal injury and apoptosis, and repair and protection in Alzheimer's disease.
Collapse
Affiliation(s)
- Guangzhe Li
- Department of Psychology, Yanbian Brain Hospital, Yanji, Jilin Province, China
| | - Hui Chen
- Department of Preventive Medicine, Medical College, Yanbian University, Yanji, Jilin Province, China
| | - Lin Cheng
- Department of Preventive Medicine, Medical College, Yanbian University, Yanji, Jilin Province, China
| | - Rongjie Zhao
- Department of Pharmacology, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Junchang Zhao
- Department of Pharmacology, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Yanji Xu
- Department of Preventive Medicine, Medical College, Yanbian University, Yanji, Jilin Province, China
| |
Collapse
|
47
|
Target-selective protein S-nitrosylation by sequence motif recognition. Cell 2014; 159:623-34. [PMID: 25417112 DOI: 10.1016/j.cell.2014.09.032] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 08/07/2014] [Accepted: 09/10/2014] [Indexed: 11/20/2022]
Abstract
S-nitrosylation is a ubiquitous protein modification emerging as a principal mechanism of nitric oxide (NO)-mediated signal transduction and cell function. S-nitrosylases can use NO synthase (NOS)-derived NO to modify selected cysteines in target proteins. Despite proteomic identification of over a thousand S-nitrosylated proteins, few S-nitrosylases have been identified. Moreover, mechanisms underlying site-selective S-nitrosylation and the potential role of specific sequence motifs remain largely unknown. Here, we describe a stimulus-inducible, heterotrimeric S-nitrosylase complex consisting of inducible NOS (iNOS), S100A8, and S100A9. S100A9 exhibits transnitrosylase activity, shuttling NO from iNOS to the target protein, whereas S100A8 and S100A9 coordinately direct site selection. A family of proteins S-nitrosylated by iNOS-S100A8/A9 were revealed by proteomic analysis. A conserved I/L-X-C-X2-D/E motif was necessary and sufficient for iNOS-S100A8/A9-mediated S-nitrosylation. These results reveal an elusive parallel between protein S-nitrosylation and phosphorylation, namely, stimulus-dependent posttranslational modification of selected targets by primary sequence motif recognition.
Collapse
|
48
|
Afanador L, Roltsch EA, Holcomb L, Campbell KS, Keeling DA, Zhang Y, Zimmer DB. The Ca2+ sensor S100A1 modulates neuroinflammation, histopathology and Akt activity in the PSAPP Alzheimer's disease mouse model. Cell Calcium 2014; 56:68-80. [DOI: 10.1016/j.ceca.2014.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 11/25/2022]
|
49
|
Wang J, Yan K, Wu ZQ, Zheng CY, Xu RX, Chen LH, Wen ZM, Zhao HQ, Ma QH. TDP-43 interaction with the intracellular domain of amyloid precursor protein induces p53-associated apoptosis. Neurosci Lett 2014; 569:131-6. [DOI: 10.1016/j.neulet.2014.03.075] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/10/2014] [Accepted: 03/29/2014] [Indexed: 12/13/2022]
|
50
|
The role of pro-inflammatory S100A9 in Alzheimer's disease amyloid-neuroinflammatory cascade. Acta Neuropathol 2014; 127:507-22. [PMID: 24240735 PMCID: PMC4148179 DOI: 10.1007/s00401-013-1208-4] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 10/18/2013] [Accepted: 11/04/2013] [Indexed: 12/12/2022]
Abstract
Pro-inflammatory S100A9 protein is increasingly recognized as an important contributor to inflammation-related neurodegeneration. Here, we provide insights into S100A9 specific mechanisms of action in Alzheimer's disease (AD). Due to its inherent amyloidogenicity S100A9 contributes to amyloid plaque formation together with Aβ. In traumatic brain injury (TBI) S100A9 itself rapidly forms amyloid plaques, which were reactive with oligomer-specific antibodies, but not with Aβ and amyloid fibrillar antibodies. They may serve as precursor-plaques for AD, implicating TBI as an AD risk factor. S100A9 was observed in some hippocampal and cortical neurons in TBI, AD and non-demented aging. In vitro S100A9 forms neurotoxic linear and annular amyloids resembling Aβ protofilaments. S100A9 amyloid cytotoxicity and native S100A9 pro-inflammatory signaling can be mitigated by its co-aggregation with Aβ, which results in a variety of micron-scale amyloid complexes. NMR and molecular docking demonstrated transient interactions between native S100A9 and Aβ. Thus, abundantly present in AD brain pro-inflammatory S100A9, possessing also intrinsic amyloidogenic properties and ability to modulate Aβ aggregation, can serve as a link between the AD amyloid and neuroinflammatory cascades and as a prospective therapeutic target.
Collapse
|