88 results on '"Tahirovic S"'
Search Results
2. Long-term dynamics of aberrant neuronal activity in awake Alzheimer’s disease transgenic mice
- Author
-
Korzhova, V., Marinković, P., Njavro, J. Rudan, Goltstein, P. M., Sun, F., Tahirovic, S., Herms, J., and Liebscher, S.
- Published
- 2021
- Full Text
- View/download PDF
3. Specific NPC1 loss in microglia of the mouse brain leads to neuroinflammation and results in synaptic loss
- Author
-
Hummel, S., additional, Dinkel, L., additional, Bartos, L. M., additional, Wind-Mark, K., additional, Slemann, L., additional, Kunze, L., additional, Englert, A., additional, Hörmann, L., additional, Gnörich, J., additional, Lindner, S., additional, Bartenstein, P., additional, Albert, N. L., additional, Brendel, M., additional, and Tahirovic, S., additional
- Published
- 2023
- Full Text
- View/download PDF
4. Desynchronization of microglial activity is closely associated with cognitive decline in Alzheimer’s disease
- Author
-
Zatcepin, A., additional, Xiang, X., additional, Parhizkar, S., additional, Gnörich, J., additional, Grosch, M., additional, Wind, K., additional, Shi, Y., additional, Beyer, L., additional, Biechele, G., additional, Eckenweber, F., additional, Wiedemann, T., additional, Lindner, S., additional, Rominger, A., additional, Bartenstein, P., additional, Willem, M., additional, Tahirovic, S., additional, Herms, J., additional, Haass, C., additional, Ziegler, S., additional, and Brendel, M., additional
- Published
- 2022
- Full Text
- View/download PDF
5. Asymmetry of plaque burden in amyloid mouse models
- Author
-
Beyer, L, additional, Sacher, C, additional, Blume, T, additional, Sauerbeck, J, additional, Eckenweber, F, additional, Focke, C, additional, Parhizkar, S, additional, Lindner, S, additional, Gildehaus, FJ, additional, von Ungern-Sternberg, B, additional, Neumann, U, additional, Baumann, K, additional, Tahirovic, S, additional, Kleinberger, G, additional, Willem, M, additional, Haass, C, additional, Bartenstein, P, additional, Rominger, A, additional, Herms, J, additional, and Brendel, M, additional
- Published
- 2020
- Full Text
- View/download PDF
6. Opposite microglial phenotypes upon loss of PGRN or TREM2 result in reduced cerebral glucose metabolism
- Author
-
Deussing, M, additional, Götzl, J, additional, Werner, G, additional, Sebastian, L, additional, Kleinberger, G, additional, Parhizkar, S, additional, Colombo, A, additional, Wagner, M, additional, Winkelmann, J, additional, Diehl-Schmid, J, additional, Levin, J, additional, Fellerer, K, additional, Bultmann, S, additional, Bartenstein, P, additional, Rominger, A, additional, Tahirovic, S, additional, Butovsky, O, additional, Capell, A, additional, Haass, C, additional, and Brendel, M, additional
- Published
- 2019
- Full Text
- View/download PDF
7. Bestimmung der Glukoseaufnahme in Mikrogliasubtypen bei Neurodegeneration
- Author
-
Wagner, S., Bartos, L. M., Zenatti, V., Prtvar, D., Fixemer, S., Klaus, C., Herms, J., Paeger, L., Prestel, M., Tahirovic, S., and Brendel, M.
- Published
- 2024
- Full Text
- View/download PDF
8. Multizentrischer Ansatz bei der β-Amyloid-µPET-Bildgebung: Machbarkeit und Grenzen in einem Kopf-an-Kopf-Vergleich mit drei Scannern
- Author
-
Gnörich, J., Koehler, M., Wind, K., Klaus, C., Zatcepin, A., Monasor, L., Beyer, L., Eckenweber, F., Scheifele, M., Gildehaus, F., Ungern-Sternberg, B., Barthel, H., Haass, C., Bartenstein, P., Herms, J., Tahirovic, S., Ziegler, S., and Brendel, M.
- Published
- 2024
- Full Text
- View/download PDF
9. Arginine methylation next to the PY-NLS modulates Transportin binding and nuclear import of FUS
- Author
-
Dorothee Dormann, Madl T, Cf, Valori, Bentmann E, Tahirovic S, Abou-Ajram C, Kremmer E, Ansorge O, Ir, Mackenzie, Neumann M, Haass C, University of Zurich, and Dormann, Dorothee
- Subjects
Have You Seen...? ,Protein-Arginine N-Methyltransferases ,Nuclear Localization Signals ,2400 General Immunology and Microbiology ,metabolism [Protein-Arginine N-Methyltransferases] ,metabolism [Repressor Proteins] ,metabolism [RNA-Binding Protein FUS] ,metabolism [Karyopherins] ,2800 General Neuroscience ,genetics [Amyotrophic Lateral Sclerosis] ,PRMT1 protein, human ,Protein Binding ,Signal Transduction ,Proline ,metabolism [Arginine] ,Molecular Sequence Data ,10208 Institute of Neuropathology ,Active Transport, Cell Nucleus ,610 Medicine & health ,Karyopherins ,Arginine ,Methylation ,Article ,1300 General Biochemistry, Genetics and Molecular Biology ,ddc:570 ,mental disorders ,genetics [Karyopherins] ,1312 Molecular Biology ,amyotrophic lateral sclerosis (ALS) ,arginine methylation ,frontotemporal lobar degeneration (FTLD) ,fused in sarcoma (FUS) ,Transportin (TRN) ,Humans ,Amino Acid Sequence ,Gene Silencing ,genetics [Protein-Arginine N-Methyltransferases] ,metabolism [Cell Nucleus] ,Cell Nucleus ,metabolism [Amyotrophic Lateral Sclerosis] ,Amyotrophic Lateral Sclerosis ,metabolism [Nuclear Localization Signals] ,nervous system diseases ,metabolism [Frontotemporal Lobar Degeneration] ,Repressor Proteins ,genetics [Repressor Proteins] ,metabolism [Tyrosine] ,metabolism [Proline] ,570 Life sciences ,biology ,Tyrosine ,RNA-Binding Protein FUS ,sense organs ,Frontotemporal Lobar Degeneration ,genetics [RNA-Binding Protein FUS] ,HeLa Cells - Abstract
Fused in sarcoma (FUS) is a nuclear protein that carries a proline tyrosine nuclear localization signal (PY NLS) and is imported into the nucleus via Transportin (TRN). Defects in nuclear import of FUS have been implicated in neurodegeneration since mutations in the PY NLS of FUS cause amyotrophic lateral sclerosis (ALS). Moreover FUS is deposited in the cytosol in a subset of frontotemporal lobar degeneration (FTLD) patients. Here we show that arginine methylation modulates nuclear import of FUS via a novel TRN binding epitope. Chemical or genetic inhibition of arginine methylation restores TRN mediated nuclear import of ALS associated FUS mutants. The unmethylated arginine glycine glycine domain preceding the PY NLS interacts with TRN and arginine methylation in this domain reduces TRN binding. Inclusions in ALS FUS patients contain methylated FUS while inclusions in FTLD FUS patients are not methylated. Together with recent findings that FUS co aggregates with two related proteins of the FET family and TRN in FTLD FUS but not in ALS FUS our study provides evidence that these two diseases may be initiated by distinct pathomechanisms and implicates alterations in arginine methylation in pathogenesis. © 2012 European Molecular Biology Organization.
- Published
- 2012
- Full Text
- View/download PDF
10. The FTLD risk factor TMEM106B and MAP6 control dendritic trafficking of lysosomes
- Author
-
Sub Cell Biology, Celbiologie, Schwenk, B.M., Lang, C.M., Hogl, S., Tahirovic, S., Orozco, D., Rentzsch, K., Lichtenthaler, S.F., Hoogenraad, Casper, Capell, A., Haass, C., Edbauer, D., Sub Cell Biology, Celbiologie, Schwenk, B.M., Lang, C.M., Hogl, S., Tahirovic, S., Orozco, D., Rentzsch, K., Lichtenthaler, S.F., Hoogenraad, Casper, Capell, A., Haass, C., and Edbauer, D.
- Published
- 2014
11. Dual Cleavage of Neuregulin 1 Type III by BACE1 and ADAM17 Liberates Its EGF-Like Domain and Allows Paracrine Signaling
- Author
-
Fleck, D., primary, van Bebber, F., additional, Colombo, A., additional, Galante, C., additional, Schwenk, B. M., additional, Rabe, L., additional, Hampel, H., additional, Novak, B., additional, Kremmer, E., additional, Tahirovic, S., additional, Edbauer, D., additional, Lichtenthaler, S. F., additional, Schmid, B., additional, Willem, M., additional, and Haass, C., additional
- Published
- 2013
- Full Text
- View/download PDF
12. Rac1 Regulates Neuronal Polarization through the WAVE Complex
- Author
-
Tahirovic, S., primary, Hellal, F., additional, Neukirchen, D., additional, Hindges, R., additional, Garvalov, B. K., additional, Flynn, K. C., additional, Stradal, T. E., additional, Chrostek-Grashoff, A., additional, Brakebusch, C., additional, and Bradke, F., additional
- Published
- 2010
- Full Text
- View/download PDF
13. Neuronal Polarity
- Author
-
Tahirovic, S., primary and Bradke, F., additional
- Published
- 2009
- Full Text
- View/download PDF
14. Asymmetry of plaque burden in amyloid mouse models.
- Author
-
Beyer, L, Sacher, C, Blume, T, Sauerbeck, J, Eckenweber, F, Focke, C, Parhizkar, S, Lindner, S, Gildehaus, FJ, von Ungern-Sternberg, B, Neumann, U, Baumann, K, Tahirovic, S, Kleinberger, G, Willem, M, Haass, C, Bartenstein, P, Rominger, A, Herms, J, and Brendel, M
- Published
- 2020
- Full Text
- View/download PDF
15. Neuronal polarity
- Author
-
Tahirovic, S. and Frank Bradke
- Subjects
Central Nervous System ,Neurons ,rho GTP-Binding Proteins ,Cell Polarity ,Microtubules ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Actins ,Axons ,nervous system ,Animals ,Humans ,Cell Lineage ,Drosophila ,Caenorhabditis elegans ,Cytoskeleton ,Perspectives ,Developmental Biology - Abstract
The assembly of functional neuronal networks in the developing animal relies on the polarization of neurons, i.e., the formation of a single axon and multiple dendrites. Breaking the symmetry of neurons depends on cytoskeletal rearrangements. In particular, axon specification requires local dynamic instability of actin and stabilization of microtubules. The polarized cytoskeleton also provides the basis for selective trafficking and retention of cellular components in the future somatodendritic or axonal compartments. Hence, these mechanisms are not only essential to achieve neuronal polarization, but also to maintain it. Different extracellular and intracellular signals converge on the regulation of the cytoskeleton. Most notably, Rho GTPases, PI3K, Ena/VASP, cofilin and SAD kinases are major intracellular regulators of neuronal polarity. Analyzing polarity signals under physiological conditions will provide a better understanding of how neurons can be induced to repolarize under pathological conditions, i.e., to regenerate their axons after central nervous system (CNS) injury.
16. TDP-43 loss of function inhibits endosomal trafficking and alters trophic signaling in neurons
- Author
-
Bm, Schwenk, Hartmann H, Serdaroglu A, Mh, Schludi, Hornburg D, Felix Meissner, Orozco D, Colombo A, Tahirovic S, Michaelsen M, Schreiber F, Haupt S, Peitz M, Brüstle O, Küpper C, Klopstock T, Otto M, Ac, Ludolph, Arzberger T, and Ph, Kuhn
17. Opposite microglial phenotypes upon loss of PGRN or TREM2 result in reduced cerebral glucose metabolism
- Author
-
Deussing, M, Götzl, J, Werner, G, Sebastian, L, Kleinberger, G, Parhizkar, S, Colombo, A, Wagner, M, Winkelmann, J, Diehl-Schmid, J, Levin, J, Fellerer, K, Bultmann, S, Bartenstein, P, Rominger, A, Tahirovic, S, Butovsky, O, Capell, A, Haass, C, and Brendel, M
- Published
- 2019
- Full Text
- View/download PDF
18. Casein Kinase 2 dependent phosphorylation of eIF4B regulates BACE1 expression in Alzheimer’s disease
- Author
-
Barbara Bettegazzi, Lisa Michelle Restelli, Fabio Grohovaz, Serena Bellani, Alessio Colombo, Daniele Zacchetti, Takashi Saito, Laura Sebastian Monasor, Stephan Frank, Takaomi C. Saido, Sabina Tahirovic, Nikolaus Deigendesch, Sven Lammich, Franca Codazzi, Bettegazzi, B., Sebastian Monasor, L., Bellani, S., Codazzi, F., Restelli, L. M., Colombo, A. V., Deigendesch, N., Frank, S., Saito, T., Saido, T. C., Lammich, S., Tahirovic, S., Grohovaz, F., and Zacchetti, D.
- Subjects
pharmacology [Protein Kinase Inhibitors] ,Cancer Research ,medicine.medical_treatment ,Action Potentials ,Pathogenesis ,drug effects [Protein Biosynthesis] ,pathology [Alzheimer Disease] ,metabolism [Casein Kinase II] ,Aspartic Acid Endopeptidases ,Premovement neuronal activity ,Eukaryotic Initiation Factors ,Phosphorylation ,EIF4B ,Casein Kinase II ,Neurons ,metabolism [Presenilin-1] ,Alzheimer's disease ,metabolism [Aspartic Acid Endopeptidases] ,Up-Regulation ,drug effects [Up-Regulation] ,Cell biology ,metabolism [Neurons] ,Neuronal physiology ,Casein kinase 2 ,metabolism [Alzheimer Disease] ,Amyloid ,Immunology ,metabolism [Amyloid beta-Peptides] ,Kinases ,Biology ,Article ,Cellular and Molecular Neuroscience ,drug effects [Phosphorylation] ,Alzheimer Disease ,ddc:570 ,antagonists & inhibitors [Casein Kinase II] ,mental disorders ,Presenilin-1 ,medicine ,Animals ,Humans ,drug effects [Neurons] ,Gene Silencing ,Protein Kinase Inhibitors ,Amyloid beta-Peptides ,Protease ,QH573-671 ,Mechanism (biology) ,Cell Biology ,metabolism [Amyloid Precursor Protein Secretases] ,Mice, Inbred C57BL ,Disease Models, Animal ,HEK293 Cells ,Protein Biosynthesis ,Amyloid Precursor Protein Secretases ,metabolism [Eukaryotic Initiation Factors] ,Cytology - Abstract
Alzheimer’s disease (AD) is the most common age-related neurodegenerative disorder. Increased Aβ production plays a fundamental role in the pathogenesis of the disease and BACE1, the protease that triggers the amyloidogenic processing of APP, is a key protein and a pharmacological target in AD. Changes in neuronal activity have been linked to BACE1 expression and Aβ generation, but the underlying mechanisms are still unclear. We provide clear evidence for the role of Casein Kinase 2 in the control of activity-driven BACE1 expression in cultured primary neurons, organotypic brain slices, and murine AD models. More specifically, we demonstrate that neuronal activity promotes Casein Kinase 2 dependent phosphorylation of the translation initiation factor eIF4B and this, in turn, controls BACE1 expression and APP processing. Finally, we show that eIF4B expression and phosphorylation are increased in the brain of APPPS1 and APP-KI mice, as well as in AD patients. Overall, we provide a definition of a mechanism linking brain activity with amyloid production and deposition, opening new perspectives from the therapeutic standpoint.
- Published
- 2021
- Full Text
- View/download PDF
19. Loss of CLN3 in microglia leads to impaired lipid metabolism and myelin turnover.
- Author
-
Yasa S, Butz ES, Colombo A, Chandrachud U, Montore L, Tschirner S, Prestel M, Sheridan SD, Müller SA, Groh J, Lichtenthaler SF, Tahirovic S, and Cotman SL
- Subjects
- Animals, Mice, Neuronal Ceroid-Lipofuscinoses metabolism, Neuronal Ceroid-Lipofuscinoses genetics, Neuronal Ceroid-Lipofuscinoses pathology, Mice, Knockout, Lysosomes metabolism, Mice, Inbred C57BL, Autophagy, Microglia metabolism, Microglia pathology, Membrane Glycoproteins metabolism, Membrane Glycoproteins genetics, Lipid Metabolism, Molecular Chaperones metabolism, Molecular Chaperones genetics, Myelin Sheath metabolism
- Abstract
Loss-of-function mutations in CLN3 cause juvenile Batten disease, featuring neurodegeneration and early-stage neuroinflammation. How loss of CLN3 function leads to early neuroinflammation is not yet understood. Here, we have comprehensively studied microglia from Cln3
∆ex7/8 mice, a genetically accurate disease model. Loss of CLN3 function in microglia leads to lysosomal storage material accumulation and abnormal morphology of subcellular organelles. Moreover, pathological proteomic signatures are indicative of defects in lysosomal function and abnormal lipid metabolism. Consistent with these findings, CLN3-deficient microglia are unable to efficiently turnover myelin and metabolize the associated lipids, showing defects in lipid droplet formation and cholesterol accumulation. Accordingly, we also observe impaired myelin integrity in aged Cln3∆ex7/8 mouse brain. Autophagy inducers and cholesterol-lowering drugs correct the observed microglial phenotypes. Taken together, these data implicate a cell-autonomous defect in CLN3-deficient microglia that impacts their ability to support neuronal cell health, suggesting microglial targeted therapies should be considered for CLN3 disease., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
20. Astroglial glucose uptake determines brain FDG-PET alterations and metabolic connectivity during healthy aging in mice.
- Author
-
Bartos LM, Kunte ST, Wagner S, Beumers P, Schaefer R, Zatcepin A, Li Y, Griessl M, Hoermann L, Wind-Mark K, Bartenstein P, Tahirovic S, Ziegler S, Brendel M, and Gnörich J
- Subjects
- Animals, Mice, Male, Female, Aging metabolism, Radiopharmaceuticals pharmacokinetics, Neurons metabolism, Healthy Aging metabolism, Microglia metabolism, Fluorodeoxyglucose F18 pharmacokinetics, Astrocytes metabolism, Positron-Emission Tomography methods, Glucose metabolism, Brain metabolism, Brain diagnostic imaging, Mice, Inbred C57BL
- Abstract
Purpose: 2-Fluorodeoxyglucose-PET (FDG-PET) is a powerful tool to study glucose metabolism in mammalian brains, but cellular sources of glucose uptake and metabolic connectivity during aging are not yet understood., Methods: Healthy wild-type mice of both sexes (2-21 months of age) received FDG-PET and cell sorting after in vivo tracer injection (scRadiotracing). FDG uptake per cell was quantified in isolated microglia, astrocytes and neurons. Cerebral FDG uptake and metabolic connectivity were determined by PET. A subset of mice received measurement of blood glucose levels to study associations with cellular FDG uptake during aging., Results: Cerebral FDG-PET signals in healthy mice increased linearly with age. Cellular FDG uptake of neurons increased between 2 and 12 months of age, followed by a strong decrease towards late ages. Contrarily, FDG uptake in microglia and astrocytes exhibited a U-shaped function with respect to age, comprising the predominant cellular source of higher cerebral FDG uptake in the later stages. Metabolic connectivity was closely associated with the ratio of glucose uptake in astroglial cells relative to neurons. Cellular FDG uptake was not associated with blood glucose levels and increasing FDG brain uptake as a function of age was still observed after adjusting for blood glucose levels., Conclusion: Trajectories of astroglial glucose uptake drive brain FDG-PET alterations and metabolic connectivity during aging., Competing Interests: Declaration of competing interest MB is a member of the Neuroimaging Committee of the EANM. MB received speaker honoraria from Roche, GE healthcare and Life Molecular Imaging, has advised Life Molecular Imaging and is currently in the advisory board of MIAC. All other authors do not report any conflict of interest., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
21. Remote Neuroinflammation in Newly Diagnosed Glioblastoma Correlates with Unfavorable Clinical Outcome.
- Author
-
Bartos LM, Quach S, Zenatti V, Kirchleitner SV, Blobner J, Wind-Mark K, Kolabas ZI, Ulukaya S, Holzgreve A, Ruf VC, Kunze LH, Kunte ST, Hoermann L, Härtel M, Park HE, Groß M, Franzmeier N, Zatcepin A, Zounek A, Kaiser L, Riemenschneider MJ, Perneczky R, Rauchmann BS, Stöcklein S, Ziegler S, Herms J, Ertürk A, Tonn JC, Thon N, von Baumgarten L, Prestel M, Tahirovic S, Albert NL, and Brendel M
- Subjects
- Humans, Animals, Mice, Male, Female, Middle Aged, Adult, Positron-Emission Tomography methods, Aged, Prognosis, Tumor Microenvironment immunology, Disease Models, Animal, Glioblastoma pathology, Glioblastoma genetics, Glioblastoma metabolism, Glioblastoma diagnosis, Glioblastoma mortality, Receptors, GABA metabolism, Receptors, GABA genetics, Brain Neoplasms pathology, Brain Neoplasms genetics, Brain Neoplasms metabolism, Brain Neoplasms diagnosis, Neuroinflammatory Diseases pathology, Neuroinflammatory Diseases etiology, Neuroinflammatory Diseases diagnosis
- Abstract
Purpose: Current therapy strategies still provide only limited success in the treatment of glioblastoma, the most frequent primary brain tumor in adults. In addition to the characterization of the tumor microenvironment, global changes in the brain of patients with glioblastoma have been described. However, the impact and molecular signature of neuroinflammation distant of the primary tumor site have not yet been thoroughly elucidated., Experimental Design: We performed translocator protein (TSPO)-PET in patients with newly diagnosed glioblastoma (n = 41), astrocytoma WHO grade 2 (n = 7), and healthy controls (n = 20) and compared TSPO-PET signals of the non-lesion (i.e., contralateral) hemisphere. Back-translation into syngeneic SB28 glioblastoma mice was used to characterize Pet alterations on a cellular level. Ultimately, multiplex gene expression analyses served to profile immune cells in remote brain., Results: Our study revealed elevated TSPO-PET signals in contralateral hemispheres of patients with newly diagnosed glioblastoma compared to healthy controls. Contralateral TSPO was associated with persisting epileptic seizures and shorter overall survival independent of the tumor phenotype. Back-translation into syngeneic glioblastoma mice pinpointed myeloid cells as the predominant source of contralateral TSPO-PET signal increases and identified a complex immune signature characterized by myeloid cell activation and immunosuppression in distant brain regions., Conclusions: Neuroinflammation within the contralateral hemisphere can be detected with TSPO-PET imaging and associates with poor outcome in patients with newly diagnosed glioblastoma. The molecular signature of remote neuroinflammation promotes the evaluation of immunomodulatory strategies in patients with detrimental whole brain inflammation as reflected by high TSPO expression., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
22. Regional desynchronization of microglial activity is associated with cognitive decline in Alzheimer's disease.
- Author
-
Zatcepin A, Gnörich J, Rauchmann BS, Bartos LM, Wagner S, Franzmeier N, Malpetti M, Xiang X, Shi Y, Parhizkar S, Grosch M, Wind-Mark K, Kunte ST, Beyer L, Meyer C, Brösamle D, Wendeln AC, Osei-Sarpong C, Heindl S, Liesz A, Stoecklein S, Biechele G, Finze A, Eckenweber F, Lindner S, Rominger A, Bartenstein P, Willem M, Tahirovic S, Herms J, Buerger K, Simons M, Haass C, Rupprecht R, Riemenschneider MJ, Albert NL, Beyer M, Neher JJ, Paeger L, Levin J, Höglinger GU, Perneczky R, Ziegler SI, and Brendel M
- Subjects
- Animals, Mice, Humans, Disease Models, Animal, Positron-Emission Tomography, Receptors, GABA metabolism, Male, Mice, Transgenic, Connectome methods, Female, Microglia metabolism, Alzheimer Disease metabolism, Alzheimer Disease pathology, Cognitive Dysfunction metabolism, Brain metabolism, Brain pathology
- Abstract
Background: Microglial activation is one hallmark of Alzheimer disease (AD) neuropathology but the impact of the regional interplay of microglia cells in the brain is poorly understood. We hypothesized that microglial activation is regionally synchronized in the healthy brain but experiences regional desynchronization with ongoing neurodegenerative disease. We addressed the existence of a microglia connectome and investigated microglial desynchronization as an AD biomarker., Methods: To validate the concept, we performed microglia depletion in mice to test whether interregional correlation coefficients (ICCs) of 18 kDa translocator protein (TSPO)-PET change when microglia are cleared. Next, we evaluated the influence of dysfunctional microglia and AD pathophysiology on TSPO-PET ICCs in the mouse brain, followed by translation to a human AD-continuum dataset. We correlated a personalized microglia desynchronization index with cognitive performance. Finally, we performed single-cell radiotracing (scRadiotracing) in mice to ensure the microglial source of the measured desynchronization., Results: Microglia-depleted mice showed a strong ICC reduction in all brain compartments, indicating microglia-specific desynchronization. AD mouse models demonstrated significant reductions of microglial synchronicity, associated with increasing variability of cellular radiotracer uptake in pathologically altered brain regions. Humans within the AD-continuum indicated a stage-depended reduction of microglia synchronicity associated with cognitive decline. scRadiotracing in mice showed that the increased TSPO signal was attributed to microglia., Conclusion: Using TSPO-PET imaging of mice with depleted microglia and scRadiotracing in an amyloid model, we provide first evidence that a microglia connectome can be assessed in the mouse brain. Microglia synchronicity is closely associated with cognitive decline in AD and could serve as an independent personalized biomarker for disease progression., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
23. Author Correction: Loss of TREM2 function increases amyloid seeding but reduces plaque-associated ApoE.
- Author
-
Parhizkar S, Arzberger T, Brendel M, Kleinberger G, Deussing M, Focke C, Nuscher B, Xiong M, Ghasemigharagoz A, Katzmarski N, Krasemann S, Lichtenthaler SF, Müller SA, Colombo A, Monasor LS, Tahirovic S, Herms J, Willem M, Pettkus N, Butovsky O, Bartenstein P, Edbauer D, Rominger A, Ertürk A, Grathwohl SA, Neher JJ, Holtzman DM, Meyer-Luehmann M, and Haass C
- Published
- 2024
- Full Text
- View/download PDF
24. Towards multicenter β-amyloid PET imaging in mouse models: A triple scanner head-to-head comparison.
- Author
-
Gnörich J, Koehler M, Wind-Mark K, Klaus C, Zatcepin A, Palumbo G, Lalia M, Monasor LS, Beyer L, Eckenweber F, Scheifele M, Gildehaus FJ, von Ungern-Sternberg B, Barthel H, Sabri O, Bartenstein P, Herms J, Tahirovic S, Franzmeier N, Ziegler S, and Brendel M
- Subjects
- Animals, Mice, Brain diagnostic imaging, Brain metabolism, Aniline Compounds, Male, Stilbenes, Positron-Emission Tomography methods, Mice, Transgenic, Disease Models, Animal, Amyloid beta-Peptides metabolism, Alzheimer Disease diagnostic imaging, Alzheimer Disease metabolism
- Abstract
Aim: β-amyloid (Aβ) small animal PET facilitates quantification of fibrillar amyloidosis in Alzheimer's disease (AD) mouse models. Thus, the methodology is receiving growing interest as a monitoring tool in preclinical drug trials. In this regard, harmonization of data from different scanners at multiple sites would allow the establishment large collaborative cohorts and may facilitate efficacy comparison of different treatments. Therefore, we objected to determine the level of agreement of Aβ-PET quantification by a head-to-head comparison of three different state-of-the-art small animal PET scanners, which could help pave the way for future multicenter studies., Methods: Within a timeframe of 5 ± 2 weeks, transgenic APPPS1 (n = 9) and wild-type (WT) (n = 8) mice (age range: 13-16 months) were examined three times by Aβ-PET ([
18 F]florbetaben) using a Siemens Inveon DPET, a MedisonanoScan PET/MR, and a MedisonanoScan PET/CT with harmonized reconstruction protocols. Cortex-to-white-matter 30-60 min p.i. standardized uptake value ratios (SUVRCTX/WM ) were calculated to compare binding differences, effect sizes (Cohen's d) and z-score values of APPPS1 relative to WT mice. Correlation coefficients (Pearson's r) were calculated for the agreement of individual SUVR between different scanners. Voxel-wise analysis was used to determine the agreement of spatial pathology patterns. For validation of PET imaging against the histological gold standard, individual SUVR values were subject to a correlation analysis with area occupancy of methoxy‑X04 staining., Results: All three small animal PET scanners yielded comparable group differences between APPPS1 and WT mice (∆PET =20.4 % ± 2.9 %, ∆PET/MR =18.4 % ± 4.5 %, ∆PET/CT =18.1 % ± 3.3 %). Voxel-wise analysis confirmed a high degree of congruency of the spatial pattern (Dice coefficient (DC)PETvs.PET/MR =83.0 %, DCPETvs.PET/CT =69.3 %, DCPET/MRvs.PET/CT =81.9 %). Differences in the group level variance of the three scanners resulted in divergent z-scores (zPET =11.5 ± 1.6; zPET/MR =5.3 ± 1.3; zPET/CT =3.4 ± 0.6) and effect sizes (dPET =8.5, dPET/MR =4.5, dPET/CT =4.1). However, correlations at the individual mouse level were still strong between scanners (rPETvs.PET/MR =0.96, rPETvs.PET/CT =0.91, rPET/MRvs.PET/CT =0.87; all p ≤ 0.0001). Methoxy-X04 staining exhibited a significant correlation across all three PET machines combined (r = 0.76, p < 0.0001) but also at individual level (PET: r = 0.81, p = 0.026; PET/MR: r = 0.89, p = 0.0074; PET/CT: r = 0.93, p = 0.0028)., Conclusions: Our comparison of standardized small animal Aβ-PET acquired by three different scanners substantiates the possibility of moving towards a multicentric approach in preclinical AD research. The alignment of image acquisition and analysis methods achieved good overall comparability between data sets. Nevertheless, differences in variance of sensitivity and specificity of different scanners may limit data interpretation at the individual mouse level and deserves methodological optimization., Competing Interests: Declaration of competing interest L.B. is an Novartis Radiopharmaceuticals employee, unrelated to this work. M.B. received speaker honoraria from GE healthcare, Roche and Life Molecular Imaging and is an advisor of Life Molecular Imaging. All other authors do not report a conflict of interest., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
25. T cell-mediated microglial activation triggers myelin pathology in a mouse model of amyloidosis.
- Author
-
Kedia S, Ji H, Feng R, Androvic P, Spieth L, Liu L, Franz J, Zdiarstek H, Anderson KP, Kaboglu C, Liu Q, Mattugini N, Cherif F, Prtvar D, Cantuti-Castelvetri L, Liesz A, Schifferer M, Stadelmann C, Tahirovic S, Gokce O, and Simons M
- Subjects
- Animals, Mice, CD8-Positive T-Lymphocytes immunology, Mice, Transgenic, Oligodendroglia pathology, Oligodendroglia metabolism, Mice, Inbred C57BL, Microglia pathology, Microglia metabolism, Microglia immunology, Myelin Sheath pathology, Myelin Sheath metabolism, Disease Models, Animal, Amyloidosis pathology, Alzheimer Disease pathology, Alzheimer Disease metabolism, Alzheimer Disease immunology
- Abstract
Age-related myelin damage induces inflammatory responses, yet its involvement in Alzheimer's disease remains uncertain, despite age being a major risk factor. Using a mouse model of Alzheimer's disease, we found that amyloidosis itself triggers age-related oligodendrocyte and myelin damage. Mechanistically, CD8
+ T cells promote the progressive accumulation of abnormally interferon-activated microglia that display myelin-damaging activity. Thus, our data suggest that immune responses against myelinating oligodendrocytes may contribute to neurodegenerative diseases with amyloidosis., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
26. Endo-lysosomal dysfunction and neuronal-glial crosstalk in Niemann-Pick type C disease.
- Author
-
Malara M, Prestel M, and Tahirovic S
- Subjects
- Humans, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Neurons, Cholesterol metabolism, Lysosomes metabolism, Lysosomes pathology, Niemann-Pick Disease, Type C genetics, Niemann-Pick Disease, Type C metabolism, Niemann-Pick Disease, Type C pathology
- Abstract
Niemann-Pick type C (NPC) disease is a rare progressive lysosomal lipid storage disorder that manifests with a heterogeneous spectrum of clinical syndromes, including visceral, neurological and psychiatric symptoms. This monogenetic autosomal recessive disease is largely caused by mutations in the NPC1 gene, which controls intracellular lipid homeostasis. Vesicle-mediated endo-lysosomal lipid trafficking and non-vesicular lipid exchange via inter-organelle membrane contact sites are both regulated by the NPC1 protein. Loss of NPC1 function therefore triggers intracellular accumulation of diverse lipid species, including cholesterol, glycosphingolipids, sphingomyelin and sphingosine. The NPC1-mediated dysfunction of lipid transport has severe consequences for all brain cells, leading to neurodegeneration. Besides the cell-autonomous contribution of neuronal NPC1, aberrant NPC1 signalling in other brain cells is critical for the pathology. We discuss here the importance of endo-lysosomal dysfunction and a tight crosstalk between neurons, oligodendrocytes, astrocytes and microglia in NPC pathology. We strongly believe that a cell-specific rescue may not be sufficient to counteract the severity of the NPC pathology, but targeting common mechanisms, such as endo-lysosomal and lipid trafficking dysfunction, may ameliorate NPC pathology. This article is part of a discussion meeting issue 'Understanding the endo-lysosomal network in neurodegeneration'.
- Published
- 2024
- Full Text
- View/download PDF
27. The COP9 signalosome reduces neuroinflammation and attenuates ischemic neuronal stress in organotypic brain slice culture model.
- Author
-
Tian Y, Milic J, Monasor LS, Chakraborty R, Wang S, Yuan Y, Asare Y, Behrends C, Tahirovic S, and Bernhagen J
- Subjects
- Humans, Animals, Mice, COP9 Signalosome Complex, Cullin Proteins, Endothelial Cells, Brain, Inflammation drug therapy, Cytokines, Neuroinflammatory Diseases, NF-kappa B
- Abstract
The constitutive photomorphogenesis 9 (COP9) signalosome (CSN) is a deNEDDylase controlling ubiquitination activity of cullin-RING-E3 ligases (CRLs) and thus the levels of key cellular proteins. While the CSN and its catalytic subunit CSN5 have been extensively studied in cancer, its role in inflammatory and neurological diseases is less understood. Following verification that CSN5 is expressed in mouse and human brain, here we studied the role of the CSN in neuroinflammation and ischemic neuronal damage employing models of relevant brain-resident cell types, an ex vivo organotypic brain slice culture model, and the CRL NEDDylation state-modifying drugs MLN4924 and CSN5i-3, which mimic and inhibit, respectively, CSN5 deNEDDylase activity. Untargeted mass spectrometry-based proteomics revealed that MLN4924 and CSN5i-3 substantially alter the microglial proteome, including inflammation-related proteins. Applying these drugs and mimicking microglial and endothelial inflammation as well as ischemic neuronal stress by TNF and oxygen-glucose-deprivation/reoxygenation (OGD/RO) treatment, respectively, we could link CSN5/CSN-mediated cullin deNEDDylation to reduction of microglial inflammation, attenuated cerebral endothelial inflammation, improved barrier integrity, as well as protection from ischemic stress-induced neuronal cell death. Specifically, MLN4924 reduced phagocytic activity, motility, and inflammatory cytokine expression of microglial cells, and this was linked to inhibition of inflammation-induced NF-κB and Akt signaling. Inversely, Csn5 knockdown and CSN5i-3 increased NF-κB signaling. Moreover, MLN4924 abrogated TNF-induced NF-κB signaling in cerebral microvascular endothelial cells (hCMECs) and rescued hCMEC monolayers from OGD/RO-triggered barrier leakage, while CSN5i-3 exacerbated permeability. In an ex vivo organotypic brain slice model of ischemia/reperfusion stress, MLN4924 protected from neuronal death, while CSN5i-3 impaired neuronal survival. Neuronal damage was attributable to microglial activation and inflammatory cytokines, as indicated by microglial shape tracking and TNF-blocking experiments. Our results indicate a protective role of the CSN in neuroinflammation via brain-resident cell types involved in ischemic brain disease and implicate CSN activity-mimicking deNEDDylating drugs as potential therapeutics., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
28. Long-Term Pioglitazone Treatment Has No Significant Impact on Microglial Activation and Tau Pathology in P301S Mice.
- Author
-
Kunze LH, Ruch F, Biechele G, Eckenweber F, Wind-Mark K, Dinkel L, Feyen P, Bartenstein P, Ziegler S, Paeger L, Tahirovic S, Herms J, and Brendel M
- Subjects
- Mice, Animals, Pioglitazone pharmacology, Microglia metabolism, Amyloid beta-Peptides metabolism, PPAR gamma metabolism, Mice, Transgenic, tau Proteins metabolism, Disease Models, Animal, Tauopathies metabolism, Alzheimer Disease metabolism
- Abstract
Neuroinflammation is one disease hallmark on the road to neurodegeneration in primary tauopathies. Thus, immunomodulation might be a suitable treatment strategy to delay or even prevent the occurrence of symptoms and thus relieve the burden for patients and caregivers. In recent years, the peroxisome proliferator-activated receptor γ (PPARγ) has received increasing attention as it is immediately involved in the regulation of the immune system and can be targeted by the anti-diabetic drug pioglitazone. Previous studies have shown significant immunomodulation in amyloid-β (Aβ) mouse models by pioglitazone. In this study, we performed long-term treatment over six months in P301S mice as a tauopathy model with either pioglitazone or placebo. We performed serial 18 kDa translocator protein positron-emission-tomography (TSPO-PET) imaging and terminal immunohistochemistry to assess microglial activation during treatment. Tau pathology was quantified via immunohistochemistry at the end of the study. Long-term pioglitazone treatment had no significant effect on TSPO-PET, immunohistochemistry read-outs of microglial activation, or tau pathology levels in P301S mice. Thus, we conclude that pioglitazone modifies the time course of Aβ-dependent microglial activation, but does not significantly modulate microglial activation in response to tau pathology.
- Published
- 2023
- Full Text
- View/download PDF
29. Depletion and activation of microglia impact metabolic connectivity of the mouse brain.
- Author
-
Gnörich J, Reifschneider A, Wind K, Zatcepin A, Kunte ST, Beumers P, Bartos LM, Wiedemann T, Grosch M, Xiang X, Fard MK, Ruch F, Werner G, Koehler M, Slemann L, Hummel S, Briel N, Blume T, Shi Y, Biechele G, Beyer L, Eckenweber F, Scheifele M, Bartenstein P, Albert NL, Herms J, Tahirovic S, Haass C, Capell A, Ziegler S, and Brendel M
- Subjects
- Animals, Mice, Progranulins metabolism, Brain metabolism, Positron-Emission Tomography, Membrane Glycoproteins metabolism, Receptors, Immunologic metabolism, Microglia metabolism, Fluorodeoxyglucose F18 metabolism
- Abstract
Aim: We aimed to investigate the impact of microglial activity and microglial FDG uptake on metabolic connectivity, since microglial activation states determine FDG-PET alterations. Metabolic connectivity refers to a concept of interacting metabolic brain regions and receives growing interest in approaching complex cerebral metabolic networks in neurodegenerative diseases. However, underlying sources of metabolic connectivity remain to be elucidated., Materials and Methods: We analyzed metabolic networks measured by interregional correlation coefficients (ICCs) of FDG-PET scans in WT mice and in mice with mutations in progranulin (Grn) or triggering receptor expressed on myeloid cells 2 (Trem2) knockouts (
-/- ) as well as in double mutant Grn-/- /Trem2-/- mice. We selected those rodent models as they represent opposite microglial signatures with disease associated microglia in Grn-/- mice and microglia locked in a homeostatic state in Trem2-/- mice; however, both resulting in lower glucose uptake of the brain. The direct influence of microglia on metabolic networks was further determined by microglia depletion using a CSF1R inhibitor in WT mice at two different ages. Within maps of global mean scaled regional FDG uptake, 24 pre-established volumes of interest were applied and assigned to either cortical or subcortical networks. ICCs of all region pairs were calculated and z-transformed prior to group comparisons. FDG uptake of neurons, microglia, and astrocytes was determined in Grn-/- and WT mice via assessment of single cell tracer uptake (scRadiotracing)., Results: Microglia depletion by CSF1R inhibition resulted in a strong decrease of metabolic connectivity defined by decrease of mean cortical ICCs in WT mice at both ages studied (6-7 m; p = 0.0148, 9-10 m; p = 0.0191), when compared to vehicle-treated age-matched WT mice. Grn-/- , Trem2-/- and Grn-/- /Trem2-/- mice all displayed reduced FDG-PET signals when compared to WT mice. However, when analyzing metabolic networks, a distinct increase of ICCs was observed in Grn-/- mice when compared to WT mice in cortical (p < 0.0001) and hippocampal (p < 0.0001) networks. In contrast, Trem2-/- mice did not show significant alterations in metabolic connectivity when compared to WT. Furthermore, the increased metabolic connectivity in Grn-/- mice was completely suppressed in Grn-/- /Trem2-/- mice. Grn-/- mice exhibited a severe loss of neuronal FDG uptake (- 61%, p < 0.0001) which shifted allocation of cellular brain FDG uptake to microglia (42% in Grn-/- vs. 22% in WT)., Conclusions: Presence, absence, and activation of microglia have a strong impact on metabolic connectivity of the mouse brain. Enhanced metabolic connectivity is associated with increased microglial FDG allocation., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
30. Beneficial Effect of ACI-24 Vaccination on Aβ Plaque Pathology and Microglial Phenotypes in an Amyloidosis Mouse Model.
- Author
-
Rudan Njavro J, Vukicevic M, Fiorini E, Dinkel L, Müller SA, Berghofer A, Bordier C, Kozlov S, Halle A, Buschmann K, Capell A, Giudici C, Willem M, Feederle R, Lichtenthaler SF, Babolin C, Montanari P, Pfeifer A, Kosco-Vilbois M, and Tahirovic S
- Subjects
- Mice, Animals, Microglia metabolism, Amyloid beta-Protein Precursor metabolism, Mice, Transgenic, Amyloid beta-Peptides metabolism, Plaque, Amyloid metabolism, Phenotype, Vaccination, Alzheimer Disease genetics, Alzheimer Disease therapy, Alzheimer Disease metabolism, Amyloidosis metabolism
- Abstract
Amyloid-β (Aβ) deposition is an initiating factor in Alzheimer's disease (AD). Microglia are the brain immune cells that surround and phagocytose Aβ plaques, but their phagocytic capacity declines in AD. This is in agreement with studies that associate AD risk loci with genes regulating the phagocytic function of immune cells. Immunotherapies are currently pursued as strategies against AD and there are increased efforts to understand the role of the immune system in ameliorating AD pathology. Here, we evaluated the effect of the Aβ targeting ACI-24 vaccine in reducing AD pathology in an amyloidosis mouse model. ACI-24 vaccination elicited a robust and sustained antibody response in APPPS1 mice with an accompanying reduction of Aβ plaque load, Aβ plaque-associated ApoE and dystrophic neurites as compared to non-vaccinated controls. Furthermore, an increased number of NLRP3-positive plaque-associated microglia was observed following ACI-24 vaccination. In contrast to this local microglial activation at Aβ plaques, we observed a more ramified morphology of Aβ plaque-distant microglia compared to non-vaccinated controls. Accordingly, bulk transcriptomic analysis revealed a trend towards the reduced expression of several disease-associated microglia (DAM) signatures that is in line with the reduced Aβ plaque load triggered by ACI-24 vaccination. Our study demonstrates that administration of the Aβ targeting vaccine ACI-24 reduces AD pathology, suggesting its use as a safe and cost-effective AD therapeutic intervention.
- Published
- 2022
- Full Text
- View/download PDF
31. Single-Cell Radiotracer Allocation via Immunomagnetic Sorting to Disentangle PET Signals at Cellular Resolution.
- Author
-
Bartos LM, Kunte ST, Beumers P, Xiang X, Wind K, Ziegler S, Bartenstein P, Choi H, Lee DS, Haass C, von Baumgarten L, Tahirovic S, Albert NL, Lindner S, and Brendel M
- Subjects
- Animals, Glucose, Hexokinase, Mice, Radiochemistry, Fluorodeoxyglucose F18, Positron-Emission Tomography methods
- Abstract
With great interest, our independent groups of scientists located in Korea and Germany recognized the use of a very similar methodologic approach to quantify the uptake of radioactive glucose (
18 F-FDG) at the cellular level. The focus of our investigations was to disentangle microglial18 F-FDG uptake. To do so, CD11b immunomagnetic cell sorting was applied to isolate microglia cells after in vivo18 F-FDG injection, to allow simple quantification via a γ-counter. Importantly, this technique reveals a snapshot of cellular glucose uptake in living mice at the time of injection since18 F-FDG is trapped by hexokinase phosphorylation without a further opportunity to be metabolized. Both studies indicated high18 F-FDG uptake of single CD11b-positive microglia cells and a significant increase in microglial18 F-FDG uptake when this cell type is activated in the presence of amyloid pathology. Furthermore, another study noticed that immunomagnetic cell sorting after tracer injection facilitated determination of high18 F-FDG uptake in myeloid cells in a range of tumor models. Here, we aim to discuss the rationale for single-cell radiotracer allocation via immunomagnetic cell sorting (scRadiotracing) by providing examples of promising applications of this innovative technology in neuroscience, oncology, and radiochemistry., (© 2022 by the Society of Nuclear Medicine and Molecular Imaging.)- Published
- 2022
- Full Text
- View/download PDF
32. Response to Comment on "Microglial activation states drive glucose uptake and FDG-PET alterations in neurodegenerative diseases".
- Author
-
Xiang X, Tahirovic S, Ziegler S, Haass C, and Brendel M
- Subjects
- Glucose, Humans, Microglia, Positron-Emission Tomography, Fluorodeoxyglucose F18, Neurodegenerative Diseases diagnostic imaging
- Abstract
Microglial FDG uptake alterations are the source of FDG-PET changes in models of neurodegenerative diseases.
- Published
- 2022
- Full Text
- View/download PDF
33. Glitter in the Darkness? Nonfibrillar β-Amyloid Plaque Components Significantly Impact the β-Amyloid PET Signal in Mouse Models of Alzheimer Disease.
- Author
-
Biechele G, Monasor LS, Wind K, Blume T, Parhizkar S, Arzberger T, Sacher C, Beyer L, Eckenweber F, Gildehaus FJ, von Ungern-Sternberg B, Willem M, Bartenstein P, Cumming P, Rominger A, Herms J, Lichtenthaler SF, Haass C, Tahirovic S, and Brendel M
- Abstract
β-amyloid (Aβ) PET is an important tool for quantification of amyloidosis in the brain of suspected Alzheimer disease (AD) patients and transgenic AD mouse models. Despite the excellent correlation of Aβ PET with gold standard immunohistochemical assessments, the relative contributions of fibrillar and nonfibrillar Aβ components to the in vivo Aβ PET signal remain unclear. Thus, we obtained 2 murine cerebral amyloidosis models that present with distinct Aβ plaque compositions and performed regression analysis between immunohistochemistry and Aβ PET to determine the biochemical contributions to Aβ PET signal in vivo. Methods: We investigated groups of App
NL-G-F and APPPS1 mice at 3, 6, and 12 mo of age by longitudinal18 F-florbetaben Aβ PET and with immunohistochemical analysis of the fibrillar and total Aβ burdens. We then applied group-level intermodality regression models using age- and genotype-matched sets of fibrillar and nonfibrillar Aβ data (predictors) and Aβ PET results (outcome) for both Aβ mouse models. An independent group of double-hit APPPS1 mice with dysfunctional microglia due to knockout of triggering receptor expression on myeloid cells 2 (Trem2-/- ) served for validation and evaluation of translational impact. Results: Neither fibrillar nor nonfibrillar Aβ content alone sufficed to explain the Aβ PET findings in either AD model. However, a regression model compiling fibrillar and nonfibrillar Aβ together with the estimate of individual heterogeneity and age at scanning could explain a 93% of variance of the Aβ PET signal ( P < 0.001). Fibrillar Aβ burden had a 16-fold higher contribution to the Aβ PET signal than nonfibrillar Aβ. However, given the relatively greater abundance of nonfibrillar Aβ, we estimate that nonfibrillar Aβ produced 79% ± 25% of the net in vivo Aβ PET signal in AppNL-G-F mice and 25% ± 12% in APPPS1 mice. Corresponding results in separate groups of APPPS1/Trem2-/- and APPPS1/Trem2+/+ mice validated the calculated regression factors and revealed that the altered fibrillarity due to Trem2 knockout impacts the Aβ PET signal. Conclusion: Taken together, the in vivo Aβ PET signal derives from the composite of fibrillar and nonfibrillar Aβ plaque components. Although fibrillar Aβ has inherently higher PET tracer binding, the greater abundance of nonfibrillar Aβ plaque in AD-model mice contributes importantly to the PET signal., (© 2022 by the Society of Nuclear Medicine and Molecular Imaging.)- Published
- 2022
- Full Text
- View/download PDF
34. Microglia contribute to the propagation of Aβ into unaffected brain tissue.
- Author
-
d'Errico P, Ziegler-Waldkirch S, Aires V, Hoffmann P, Mezö C, Erny D, Monasor LS, Liebscher S, Ravi VM, Joseph K, Schnell O, Kierdorf K, Staszewski O, Tahirovic S, Prinz M, and Meyer-Luehmann M
- Subjects
- Brain metabolism, Humans, Neurons metabolism, Plaque, Amyloid pathology, Amyloid beta-Peptides metabolism, Microglia metabolism
- Abstract
Microglia appear activated in the vicinity of amyloid beta (Aβ) plaques, but whether microglia contribute to Aβ propagation into unaffected brain regions remains unknown. Using transplantation of wild-type (WT) neurons, we show that Aβ enters WT grafts, and that this is accompanied by microglia infiltration. Manipulation of microglia function reduced Aβ deposition within grafts. Furthermore, in vivo imaging identified microglia as carriers of Aβ pathology in previously unaffected tissue. Our data thus argue for a hitherto unexplored mechanism of Aβ propagation., (© 2021. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
35. Impaired Retromer Function in Niemann-Pick Type C Disease Is Dependent on Intracellular Cholesterol Accumulation.
- Author
-
Dominko K, Rastija A, Sobocanec S, Vidatic L, Meglaj S, Lovincic Babic A, Hutter-Paier B, Colombo AV, Lichtenthaler SF, Tahirovic S, and Hecimovic S
- Subjects
- Animals, CHO Cells, Cells, Cultured, Cricetulus, Disease Models, Animal, Female, Humans, Male, Membrane Transport Proteins metabolism, Mice, Neurons cytology, Niemann-Pick Disease, Type C metabolism, Primary Cell Culture, Receptors, LDL metabolism, Vesicular Transport Proteins metabolism, Cholesterol metabolism, Loss of Function Mutation, Neurons metabolism, Niemann-Pick C1 Protein genetics, Niemann-Pick Disease, Type C genetics
- Abstract
Niemann-Pick type C disease (NPC) is a rare inherited neurodegenerative disorder characterized by an accumulation of intracellular cholesterol within late endosomes and lysosomes due to NPC1 or NPC2 dysfunction. In this work, we tested the hypothesis that retromer impairment may be involved in the pathogenesis of NPC and may contribute to increased amyloidogenic processing of APP and enhanced BACE1-mediated proteolysis observed in NPC disease. Using NPC1 -null cells, primary mouse NPC1-deficient neurons and NPC1-deficient mice (BALB/cNctr- Npc1m1N ), we show that retromer function is impaired in NPC. This is manifested by altered transport of the retromer core components Vps26, Vps35 and/or retromer receptor sorLA and by retromer accumulation in neuronal processes, such as within axonal swellings. Changes in retromer distribution in NPC1 mouse brains were observed already at the presymptomatic stage (at 4-weeks of age), indicating that the retromer defect occurs early in the course of NPC disease and may contribute to downstream pathological processes. Furthermore, we show that cholesterol depletion in NPC1 -null cells and in NPC1 mouse brains reverts retromer dysfunction, suggesting that retromer impairment in NPC is mechanistically dependent on cholesterol accumulation. Thus, we characterized retromer dysfunction in NPC and propose that the rescue of retromer impairment may represent a novel therapeutic approach against NPC.
- Published
- 2021
- Full Text
- View/download PDF
36. Microglial activation states drive glucose uptake and FDG-PET alterations in neurodegenerative diseases.
- Author
-
Xiang X, Wind K, Wiedemann T, Blume T, Shi Y, Briel N, Beyer L, Biechele G, Eckenweber F, Zatcepin A, Lammich S, Ribicic S, Tahirovic S, Willem M, Deussing M, Palleis C, Rauchmann BS, Gildehaus FJ, Lindner S, Spitz C, Franzmeier N, Baumann K, Rominger A, Bartenstein P, Ziegler S, Drzezga A, Respondek G, Buerger K, Perneczky R, Levin J, Höglinger GU, Herms J, Haass C, and Brendel M
- Subjects
- Humans, Glucose, Microglia, Positron-Emission Tomography, Animals, Mice, Fluorodeoxyglucose F18, Neurodegenerative Diseases diagnostic imaging
- Abstract
2-Deoxy-2-[
18 F]fluoro-d-glucose positron emission tomography (FDG-PET) is widely used to study cerebral glucose metabolism. Here, we investigated whether the FDG-PET signal is directly influenced by microglial glucose uptake in mouse models and patients with neurodegenerative diseases. Using a recently developed approach for cell sorting after FDG injection, we found that, at cellular resolution, microglia displayed higher glucose uptake than neurons and astrocytes. Alterations in microglial glucose uptake were responsible for both the FDG-PET signal decrease in Trem2 -deficient mice and the FDG-PET signal increase in mouse models for amyloidosis. Thus, opposite microglial activation states determine the differential FDG uptake. Consistently, 12 patients with Alzheimer’s disease and 21 patients with four-repeat tauopathies also exhibited a positive association between glucose uptake and microglial activity as determined by18 F-GE-180 18-kDa translocator protein PET (TSPO-PET) in preserved brain regions, indicating that the cerebral glucose uptake in humans is also strongly influenced by microglial activity. Our findings suggest that microglia activation states are responsible for FDG-PET signal alterations in patients with neurodegenerative diseases and mouse models for amyloidosis. Microglial activation states should therefore be considered when performing FDG-PET.- Published
- 2021
- Full Text
- View/download PDF
37. Casein Kinase 2 dependent phosphorylation of eIF4B regulates BACE1 expression in Alzheimer's disease.
- Author
-
Bettegazzi B, Sebastian Monasor L, Bellani S, Codazzi F, Restelli LM, Colombo AV, Deigendesch N, Frank S, Saito T, Saido TC, Lammich S, Tahirovic S, Grohovaz F, and Zacchetti D
- Subjects
- Action Potentials, Alzheimer Disease pathology, Amyloid beta-Peptides metabolism, Animals, Casein Kinase II antagonists & inhibitors, Disease Models, Animal, Gene Silencing, HEK293 Cells, Humans, Mice, Inbred C57BL, Neurons drug effects, Neurons metabolism, Phosphorylation drug effects, Presenilin-1 metabolism, Protein Biosynthesis drug effects, Protein Kinase Inhibitors pharmacology, Up-Regulation drug effects, Mice, Alzheimer Disease metabolism, Amyloid Precursor Protein Secretases metabolism, Aspartic Acid Endopeptidases metabolism, Casein Kinase II metabolism, Eukaryotic Initiation Factors metabolism
- Abstract
Alzheimer's disease (AD) is the most common age-related neurodegenerative disorder. Increased Aβ production plays a fundamental role in the pathogenesis of the disease and BACE1, the protease that triggers the amyloidogenic processing of APP, is a key protein and a pharmacological target in AD. Changes in neuronal activity have been linked to BACE1 expression and Aβ generation, but the underlying mechanisms are still unclear. We provide clear evidence for the role of Casein Kinase 2 in the control of activity-driven BACE1 expression in cultured primary neurons, organotypic brain slices, and murine AD models. More specifically, we demonstrate that neuronal activity promotes Casein Kinase 2 dependent phosphorylation of the translation initiation factor eIF4B and this, in turn, controls BACE1 expression and APP processing. Finally, we show that eIF4B expression and phosphorylation are increased in the brain of APPPS1 and APP-KI mice, as well as in AD patients. Overall, we provide a definition of a mechanism linking brain activity with amyloid production and deposition, opening new perspectives from the therapeutic standpoint., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
38. Do heterozygous mutations of Niemann-Pick type C predispose to late-onset neurodegeneration: a review of the literature.
- Author
-
Schneider SA, Tahirovic S, Hardy J, Strupp M, and Bremova-Ertl T
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Animals, Child, Glucosylceramidase genetics, Heterozygote, Humans, Middle Aged, Mutation, Young Adult, Gaucher Disease, Neurodegenerative Diseases genetics, Niemann-Pick Disease, Type C genetics
- Abstract
Background/methods: Monogenic diseases are important models for the study of neurodegenerative diseases, such as Parkinson's disease (PD) and dementia. Notably, for some disorders, homozygosity is associated with a complex metabolic disease, while heterozygosity predisposes to late-onset neurodegeneration. For instance, biallelic glucocerebrosidase gene mutations cause Gaucher's disease, while heterozygous mutations are a common genetic risk factor for late-onset PD. Little is known about similar risks of related diseases, such as Niemann-Pick type C (NPC). Given that both conditions map into related, i.e., lysosomal, pathways, we hypothesize a similar risk of single-NPC gene mutations. Indeed, there is increasing evidence based on clinical observations in humans and animal studies. Here we review the current knowledge of NPC heterozygosity., Results: Family history studies suggest a high proportion of late-onset neurodegenerative diseases in NPC families. We identified 19 cases with heterozygous NPC mutations in the literature who presented with a neurodegenerative disease, including levodopa-responsive PD, atypical parkinsonism (PSP, CBD), dystonia or dementia with a mean age at onset of about 57 years (range 8-87). Consistent splenomegaly and mildly abnormal filipin staining results have also been reported in heterozygous gene mutation carriers. Imaging and pathological data support this notion., Discussion/conclusion: This finding has wider implications in so far as NPC-related forms of Parkinsonian syndromes, dementia, motor neuron disease and other neurodegenerative disorders may benefit from NPC-mechanistic therapies, in particular related to lysosomal dysfunction. Further research is warranted to generate systematic data of heterozygous mutation carriers, including longitudinal data.
- Published
- 2021
- Full Text
- View/download PDF
39. Microglial activation in the right amygdala-entorhinal-hippocampal complex is associated with preserved spatial learning in App NL-G-F mice.
- Author
-
Biechele G, Wind K, Blume T, Sacher C, Beyer L, Eckenweber F, Franzmeier N, Ewers M, Zott B, Lindner S, Gildehaus FJ, von Ungern-Sternberg B, Tahirovic S, Willem M, Bartenstein P, Cumming P, Rominger A, Herms J, and Brendel M
- Subjects
- Alzheimer Disease genetics, Alzheimer Disease metabolism, Amyloid beta-Protein Precursor genetics, Animals, Female, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Positron-Emission Tomography methods, Receptors, GABA biosynthesis, Receptors, GABA genetics, Amygdala metabolism, Amyloid beta-Protein Precursor biosynthesis, Entorhinal Cortex metabolism, Hippocampus metabolism, Microglia metabolism, Spatial Learning physiology
- Abstract
Background: In Alzheimer`s disease (AD), regional heterogeneity of β-amyloid burden and microglial activation of individual patients is a well-known phenomenon. Recently, we described a high incidence of inter-individual regional heterogeneity in terms of asymmetry of plaque burden and microglial activation in β-amyloid mouse models of AD as assessed by positron-emission-tomography (PET). We now investigate the regional associations between amyloid plaque burden, microglial activation, and impaired spatial learning performance in transgenic mice in vivo., Methods: In 30 App
NL-G-F mice (15 female, 15 male) we acquired cross-sectional 18 kDa translocator protein (TSPO-PET,18 F-GE-180) and β-amyloid-PET (18 F-florbetaben) scans at ten months of age. Control data were obtained from age- and sex-matched C57BI/6 wild-type mice. We assessed spatial learning (i.e. Morris water maze) within two weeks of PET scanning and correlated the principal component of spatial learning performance scores with voxel-wise β-amyloid and TSPO tracer uptake maps in AppNL-G-F mice, controlled for age and sex. In order to assess the effects of hemispheric asymmetry, we also analyzed correlations of spatial learning performance with tracer uptake in bilateral regions of interest for frontal cortex, entorhinal/piriform cortex, amygdala, and hippocampus, using a regression model. We tested the correlation between regional asymmetry of PET biomarkers with individual spatial learning performance., Results: Voxel-wise analyses in AppNL-G-F mice revealed that higher TSPO-PET signal in the amygdala, entorhinal and piriform cortices, the hippocampus and the hypothalamus correlated with spatial learning performance. Region-based analysis showed significant correlations between TSPO expression in the right entorhinal/piriform cortex and the right amygdala and spatial learning performance, whereas there were no such correlations in the left hemisphere. Right lateralized TSPO expression in the amygdala predicted better performance in the Morris water maze (β = -0.470, p = 0.013), irrespective of the global microglial activation and amyloid level. Region-based results for amyloid-PET showed no significant associations with spatial learning., Conclusion: Elevated microglial activation in the right amygdala-entorhinal-hippocampal complex of AppNL-G-F mice is associated with better spatial learning. Our findings support a protective role of microglia on cognitive function when they highly express TSPO in specific brain regions involved in spatial memory., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
40. Microbiota-derived short chain fatty acids modulate microglia and promote Aβ plaque deposition.
- Author
-
Colombo AV, Sadler RK, Llovera G, Singh V, Roth S, Heindl S, Sebastian Monasor L, Verhoeven A, Peters F, Parhizkar S, Kamp F, Gomez de Aguero M, MacPherson AJ, Winkler E, Herms J, Benakis C, Dichgans M, Steiner H, Giera M, Haass C, Tahirovic S, and Liesz A
- Subjects
- Alzheimer Disease metabolism, Animals, Female, Male, Mice, Specific Pathogen-Free Organisms, Fatty Acids, Volatile metabolism, Gastrointestinal Microbiome, Microglia metabolism, Plaque, Amyloid metabolism
- Abstract
Previous studies have identified a crucial role of the gut microbiome in modifying Alzheimer's disease (AD) progression. However, the mechanisms of microbiome-brain interaction in AD were so far unknown. Here, we identify microbiota-derived short chain fatty acids (SCFA) as microbial metabolites which promote Aβ deposition. Germ-free (GF) AD mice exhibit a substantially reduced Aβ plaque load and markedly reduced SCFA plasma concentrations; conversely, SCFA supplementation to GF AD mice increased the Aβ plaque load to levels of conventionally colonized (specific pathogen-free [SPF]) animals and SCFA supplementation to SPF mice even further exacerbated plaque load. This was accompanied by the pronounced alterations in microglial transcriptomic profile, including upregulation of ApoE. Despite increased microglial recruitment to Aβ plaques upon SCFA supplementation, microglia contained less intracellular Aβ. Taken together, our results demonstrate that microbiota-derived SCFA are critical mediators along the gut-brain axis which promote Aβ deposition likely via modulation of the microglial phenotype., Competing Interests: AC, RS, GL, VS, SR, SH, LS, AV, FP, SP, FK, MG, EW, JH, CB, MD, HS, MG, CH, ST, AL No competing interests declared, AM Reviewing editor, eLife, (© 2021, Colombo et al.)
- Published
- 2021
- Full Text
- View/download PDF
41. Loss of NPC1 enhances phagocytic uptake and impairs lipid trafficking in microglia.
- Author
-
Colombo A, Dinkel L, Müller SA, Sebastian Monasor L, Schifferer M, Cantuti-Castelvetri L, König J, Vidatic L, Bremova-Ertl T, Lieberman AP, Hecimovic S, Simons M, Lichtenthaler SF, Strupp M, Schneider SA, and Tahirovic S
- Subjects
- Animals, Blotting, Western, Cells, Cultured, Female, Humans, Intracellular Signaling Peptides and Proteins genetics, Male, Mass Spectrometry, Mice, Mice, Inbred C57BL, Mice, Knockout, Myelin Sheath metabolism, Niemann-Pick C1 Protein, Niemann-Pick Disease, Type C genetics, Phagocytosis genetics, Phagocytosis physiology, Proteomics methods, Cholesterol metabolism, Intracellular Signaling Peptides and Proteins metabolism, Microglia metabolism, Niemann-Pick Disease, Type C metabolism
- Abstract
Niemann-Pick type C disease is a rare neurodegenerative disorder mainly caused by mutations in NPC1, resulting in abnormal late endosomal/lysosomal lipid storage. Although microgliosis is a prominent pathological feature, direct consequences of NPC1 loss on microglial function remain not fully characterized. We discovered pathological proteomic signatures and phenotypes in NPC1-deficient murine models and demonstrate a cell autonomous function of NPC1 in microglia. Loss of NPC1 triggers enhanced phagocytic uptake and impaired myelin turnover in microglia that precede neuronal death. Npc1
-/- microglia feature a striking accumulation of multivesicular bodies and impaired trafficking of lipids to lysosomes while lysosomal degradation function remains preserved. Molecular and functional defects were also detected in blood-derived macrophages of NPC patients that provide a potential tool for monitoring disease. Our study underscores an essential cell autonomous role for NPC1 in immune cells and implies microglial therapeutic potential.- Published
- 2021
- Full Text
- View/download PDF
42. Emerging Microglia Biology Defines Novel Therapeutic Approaches for Alzheimer's Disease.
- Author
-
Lewcock JW, Schlepckow K, Di Paolo G, Tahirovic S, Monroe KM, and Haass C
- Subjects
- Alzheimer Disease genetics, Animals, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal therapeutic use, Brain drug effects, Genome-Wide Association Study trends, Humans, Membrane Glycoproteins genetics, Membrane Glycoproteins immunology, Membrane Glycoproteins therapeutic use, Microglia drug effects, Receptors, Immunologic genetics, Receptors, Immunologic immunology, Receptors, Immunologic therapeutic use, Alzheimer Disease immunology, Alzheimer Disease therapy, Brain immunology, Genetic Therapy trends, Immunotherapy trends, Microglia immunology
- Abstract
Alzheimer's disease (AD) is currently untreatable, and therapeutic strategies aimed to slow cognitive decline have not yet been successful. Many of these approaches have targeted the amyloid cascade, indicating that novel treatment strategies are required. Recent genome-wide association studies (GWASs) have identified a number of risk factors in genes expressed in microglia, underscoring their therapeutic potential in neurodegeneration. In this review, we discuss how the recently defined functions of these AD risk genes can be targeted therapeutically to modulate microglial cell state and slow the progression of AD. Antibody-mediated stimulation of the triggering receptor of myeloid cells 2 (TREM2) is on the forefront of these candidate therapeutic approaches based on a combination of compelling human genetics and emerging preclinical data. This and other approaches to modify microglial function are a topic of intensive study and provide an opportunity for innovative AD treatments, which may be applied alone or potentially in combination with classical anti-amyloid therapies., Competing Interests: Declaration of Interests C.H. collaborates with Denali Therapeutics, participated on one advisory board meeting of Biogen, and received a speaker honorarium from Novartis and Roche. C.H. is chief advisor of ISAR Bioscience. J.W.L., K.M.M., and G.D.P. are employees and shareholders of Denali Therapeutics. J.W.L., K.M.M., K.S., and C.H. have patents pending describing TREM2 antibodies., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
43. Asymmetry of Fibrillar Plaque Burden in Amyloid Mouse Models.
- Author
-
Sacher C, Blume T, Beyer L, Biechele G, Sauerbeck J, Eckenweber F, Deussing M, Focke C, Parhizkar S, Lindner S, Gildehaus FJ, von Ungern-Sternberg B, Baumann K, Tahirovic S, Kleinberger G, Willem M, Haass C, Bartenstein P, Cumming P, Rominger A, Herms J, and Brendel M
- Subjects
- Animals, Disease Models, Animal, Image Processing, Computer-Assisted, Mice, Mice, Inbred C57BL, Plaque, Amyloid diagnostic imaging, Positron-Emission Tomography, Amyloid beta-Peptides chemistry, Plaque, Amyloid metabolism, Protein Aggregates
- Abstract
Asymmetries of amyloid-β (Aβ) burden are well known in Alzheimer disease (AD) but did not receive attention in Aβ mouse models of Alzheimer disease. Therefore, we investigated Aβ asymmetries in Aβ mouse models examined by Aβ small-animal PET and tested if such asymmetries have an association with microglial activation. Methods: We analyzed 523 cross-sectional Aβ PET scans of 5 different Aβ mouse models (APP/PS1, PS2APP, APP-SL70, App
NL-G-F , and APPswe) together with 136 18-kDa translocator protein (TSPO) PET scans for microglial activation. The asymmetry index (AI) was calculated between tracer uptake in both hemispheres. AIs of Aβ PET were analyzed in correlation with TSPO PET AIs. Extrapolated required sample sizes were compared between analyses of single and combined hemispheres. Results: Relevant asymmetries of Aβ deposition were identified in at least 30% of all investigated mice. There was a significant correlation between AIs of Aβ PET and TSPO PET in 4 investigated Aβ mouse models (APP/PS1: R = 0.593, P = 0.001; PS2APP: R = 0.485, P = 0.019; APP-SL70: R = 0.410, P = 0.037; AppNL-G-F : R = 0.385, P = 0.002). Asymmetry was associated with higher variance of tracer uptake in single hemispheres, leading to higher required sample sizes. Conclusion: Asymmetry of fibrillar plaque neuropathology occurs frequently in Aβ mouse models and acts as a potential confounder in experimental designs. Concomitant asymmetry of microglial activation indicates a neuroinflammatory component to hemispheric predominance of fibrillary amyloidosis., (© 2020 by the Society of Nuclear Medicine and Molecular Imaging.)- Published
- 2020
- Full Text
- View/download PDF
44. An optimized quantitative proteomics method establishes the cell type-resolved mouse brain secretome.
- Author
-
Tüshaus J, Müller SA, Kataka ES, Zaucha J, Sebastian Monasor L, Su M, Güner G, Jocher G, Tahirovic S, Frishman D, Simons M, and Lichtenthaler SF
- Subjects
- ADAM Proteins cerebrospinal fluid, ADAM Proteins metabolism, Amyloid Precursor Protein Secretases antagonists & inhibitors, Amyloid Precursor Protein Secretases cerebrospinal fluid, Animals, Antigens, CD cerebrospinal fluid, Antigens, CD metabolism, Aspartic Acid Endopeptidases antagonists & inhibitors, Aspartic Acid Endopeptidases cerebrospinal fluid, Brain cytology, Cells, Cultured, Cerebrospinal Fluid Proteins, Chromatography, Liquid, Gene Ontology, Lipopolysaccharides pharmacology, Mice, Mice, Inbred C57BL, Nerve Tissue Proteins cerebrospinal fluid, Nerve Tissue Proteins metabolism, Principal Component Analysis, Proteome metabolism, Tandem Mass Spectrometry, Amyloid Precursor Protein Secretases metabolism, Aspartic Acid Endopeptidases metabolism, Astrocytes metabolism, Brain metabolism, Microglia metabolism, Neurons metabolism, Oligodendroglia metabolism, Proteomics methods, Software
- Abstract
To understand how cells communicate in the nervous system, it is essential to define their secretome, which is challenging for primary cells because of large cell numbers being required. Here, we miniaturized secretome analysis by developing the "high-performance secretome protein enrichment with click sugars" (hiSPECS) method. To demonstrate its broad utility, hiSPECS was used to identify the secretory response of brain slices upon LPS-induced neuroinflammation and to establish the cell type-resolved mouse brain secretome resource using primary astrocytes, microglia, neurons, and oligodendrocytes. This resource allowed mapping the cellular origin of CSF proteins and revealed that an unexpectedly high number of secreted proteins in vitro and in vivo are proteolytically cleaved membrane protein ectodomains. Two examples are neuronally secreted ADAM22 and CD200, which we identified as substrates of the Alzheimer-linked protease BACE1. hiSPECS and the brain secretome resource can be widely exploited to systematically study protein secretion and brain function and to identify cell type-specific biomarkers for CNS diseases., (© 2020 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2020
- Full Text
- View/download PDF
45. Fibrillar Aβ triggers microglial proteome alterations and dysfunction in Alzheimer mouse models.
- Author
-
Sebastian Monasor L, Müller SA, Colombo AV, Tanrioever G, König J, Roth S, Liesz A, Berghofer A, Piechotta A, Prestel M, Saito T, Saido TC, Herms J, Willem M, Haass C, Lichtenthaler SF, and Tahirovic S
- Subjects
- Alzheimer Disease pathology, Amyloid beta-Peptides genetics, Amyloid beta-Protein Precursor genetics, Amyloid beta-Protein Precursor metabolism, Animals, Disease Models, Animal, Female, Gene Knock-In Techniques, Male, Mice, Mice, Transgenic, Microglia pathology, Alzheimer Disease metabolism, Amyloid beta-Peptides metabolism, Microglia metabolism, Proteome metabolism
- Abstract
Microglial dysfunction is a key pathological feature of Alzheimer's disease (AD), but little is known about proteome-wide changes in microglia during the course of AD and their functional consequences. Here, we performed an in-depth and time-resolved proteomic characterization of microglia in two mouse models of amyloid β (Aβ) pathology, the overexpression APPPS1 and the knock-in APP-NL-G-F (APP-KI) model. We identified a large panel of Microglial Aβ Response Proteins (MARPs) that reflect heterogeneity of microglial alterations during early, middle and advanced stages of Aβ deposition and occur earlier in the APPPS1 mice. Strikingly, the kinetic differences in proteomic profiles correlated with the presence of fibrillar Aβ, rather than dystrophic neurites, suggesting that fibrillar Aβ may trigger the AD-associated microglial phenotype and the observed functional decline. The identified microglial proteomic fingerprints of AD provide a valuable resource for functional studies of novel molecular targets and potential biomarkers for monitoring AD progression or therapeutic efficacy., Competing Interests: LS, SM, AC, GT, JK, SR, AL, AB, AP, MP, TS, TS, JH, MW, CH, SL, ST No competing interests declared, (© 2020, Sebastian Monasor et al.)
- Published
- 2020
- Full Text
- View/download PDF
46. Enhancing protective microglial activities with a dual function TREM2 antibody to the stalk region.
- Author
-
Schlepckow K, Monroe KM, Kleinberger G, Cantuti-Castelvetri L, Parhizkar S, Xia D, Willem M, Werner G, Pettkus N, Brunner B, Sülzen A, Nuscher B, Hampel H, Xiang X, Feederle R, Tahirovic S, Park JI, Prorok R, Mahon C, Liang CC, Shi J, Kim DJ, Sabelström H, Huang F, Di Paolo G, Simons M, Lewcock JW, and Haass C
- Subjects
- Amyloid beta-Peptides, Animals, Cell Line, Tumor, Female, Macrophages, Mice, Rats, Rats, Wistar, Antibodies, Monoclonal pharmacology, Membrane Glycoproteins immunology, Microglia pathology, Multiple Myeloma, Receptors, Immunologic immunology
- Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) is essential for the transition of homeostatic microglia to a disease-associated microglial state. To enhance TREM2 activity, we sought to selectively increase the full-length protein on the cell surface via reducing its proteolytic shedding by A Disintegrin And Metalloproteinase (i.e., α-secretase) 10/17. We screened a panel of monoclonal antibodies against TREM2, with the aim to selectively compete for α-secretase-mediated shedding. Monoclonal antibody 4D9, which has a stalk region epitope close to the cleavage site, demonstrated dual mechanisms of action by stabilizing TREM2 on the cell surface and reducing its shedding, and concomitantly activating phospho-SYK signaling. 4D9 stimulated survival of macrophages and increased microglial uptake of myelin debris and amyloid β-peptide in vitro. In vivo target engagement was demonstrated in cerebrospinal fluid, where nearly all soluble TREM2 was 4D9-bound. Moreover, in a mouse model for Alzheimer's disease-related pathology, 4D9 reduced amyloidogenesis, enhanced microglial TREM2 expression, and reduced a homeostatic marker, suggesting a protective function by driving microglia toward a disease-associated state., (© 2020 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2020
- Full Text
- View/download PDF
47. Opposite microglial activation stages upon loss of PGRN or TREM2 result in reduced cerebral glucose metabolism.
- Author
-
Götzl JK, Brendel M, Werner G, Parhizkar S, Sebastian Monasor L, Kleinberger G, Colombo AV, Deussing M, Wagner M, Winkelmann J, Diehl-Schmid J, Levin J, Fellerer K, Reifschneider A, Bultmann S, Bartenstein P, Rominger A, Tahirovic S, Smith ST, Madore C, Butovsky O, Capell A, and Haass C
- Subjects
- Alzheimer Disease diagnostic imaging, Alzheimer Disease genetics, Alzheimer Disease metabolism, Animals, Frontotemporal Lobar Degeneration diagnostic imaging, Frontotemporal Lobar Degeneration genetics, Frontotemporal Lobar Degeneration metabolism, Mice, Mice, Knockout, Cerebellum diagnostic imaging, Cerebellum metabolism, Glucose metabolism, Membrane Glycoproteins deficiency, Microglia metabolism, Positron-Emission Tomography, Progranulins deficiency, Receptors, Immunologic deficiency
- Abstract
Microglia adopt numerous fates with homeostatic microglia (HM) and a microglial neurodegenerative phenotype (MGnD) representing two opposite ends. A number of variants in genes selectively expressed in microglia are associated with an increased risk for neurodegenerative diseases such as Alzheimer's disease (AD) and frontotemporal lobar degeneration (FTLD). Among these genes are progranulin ( GRN ) and the triggering receptor expressed on myeloid cells 2 ( TREM2 ). Both cause neurodegeneration by mechanisms involving loss of function. We have now isolated microglia from Grn
-/- mice and compared their transcriptomes to those of Trem2-/- mice Surprisingly, while loss of Trem2 enhances the expression of genes associated with a homeostatic state, microglia derived from Grn-/- mice showed a reciprocal activation of the MGnD molecular signature and suppression of gene characteristic for HM The opposite mRNA expression profiles are associated with divergent functional phenotypes. Although loss of TREM2 and progranulin resulted in opposite activation states and functional phenotypes of microglia, FDG (fluoro-2-deoxy-d-glucose)-μPET of brain revealed reduced glucose metabolism in both conditions, suggesting that opposite microglial phenotypes result in similar wide spread brain dysfunction., (© 2019 The Authors. Published under the terms of the CC BY 4.0 license.)- Published
- 2019
- Full Text
- View/download PDF
48. In vivo Ca 2+ imaging of astrocytic microdomains reveals a critical role of the amyloid precursor protein for mitochondria.
- Author
-
Montagna E, Crux S, Luckner M, Herber J, Colombo AV, Marinković P, Tahirovic S, Lichtenthaler SF, Wanner G, Müller UC, Sgobio C, and Herms J
- Subjects
- Amyloid beta-Protein Precursor genetics, Animals, Animals, Newborn, Brain metabolism, Cells, Cultured, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Mitochondria ultrastructure, Transduction, Genetic, Transfection, Amyloid beta-Protein Precursor deficiency, Astrocytes ultrastructure, Brain cytology, Calcium metabolism, Membrane Microdomains metabolism, Mitochondria metabolism
- Abstract
The investigation of amyloid precursor protein (APP) has been mainly confined to its neuronal functions, whereas very little is known about its physiological role in astrocytes. Astrocytes exhibit a particular morphology with slender extensions protruding from somata and primary branches. Along these fine extensions, spontaneous calcium transients occur in spatially restricted microdomains. Within these microdomains mitochondria are responsible for local energy supply and Ca
2+ buffering. Using two-photon in vivo Ca2+ imaging, we report a significant decrease in the density of active microdomains, frequency of spontaneous Ca2+ transients and slower Ca2+ kinetics in mice lacking APP. Mechanistically, these changes could be potentially linked to mitochondrial malfunction as our in vivo and in vitro data revealed severe, APP-dependent structural mitochondrial fragmentation in astrocytes. Functionally, such mitochondria exhibited prolonged kinetics and morphology dependent signal size of ATP-induced Ca2+ transients. Our results highlight a prominent role of APP in the modulation of Ca2+ activity in astrocytic microdomains whose precise functioning is crucial for the reinforcement and modulation of synaptic function. This study provides novel insights in APP physiological functions which are important for the understanding of the effects of drugs validated in Alzheimer's disease treatment that affect the function of APP., (© 2019 Wiley Periodicals, Inc.)- Published
- 2019
- Full Text
- View/download PDF
49. Human stem cell-derived monocytes and microglia-like cells reveal impaired amyloid plaque clearance upon heterozygous or homozygous loss of TREM2.
- Author
-
Claes C, Van Den Daele J, Boon R, Schouteden S, Colombo A, Monasor LS, Fiers M, Ordovás L, Nami F, Bohrmann B, Tahirovic S, De Strooper B, and Verfaillie CM
- Subjects
- Amyloid beta-Protein Precursor genetics, Amyloid beta-Protein Precursor metabolism, Animals, Brain, CRISPR-Cas Systems, Cells, Cultured, Escherichia coli, Membrane Glycoproteins genetics, Mice, Transgenic, Phagocytosis, Pluripotent Stem Cells, Presenilin-1 genetics, Presenilin-1 metabolism, Receptors, Immunologic genetics, Membrane Glycoproteins deficiency, Microglia metabolism, Monocytes metabolism, Plaque, Amyloid metabolism, Receptors, Immunologic deficiency
- Abstract
Introduction: Murine microglia expressing the Alzheimer's disease-linked TREM2
R47H mutation display variable decrease in phagocytosis, while impaired phagocytosis is reported following loss of TREM2. However, no data exist on TREM2+/R47H human microglia. Therefore, we created human pluripotent stem cell (hPSC) monocytes and transdifferentiated microglia-like cells (tMGs) to examine the effect of the TREM2+/R47H mutation and loss of TREM2 on phagocytosis., Methods: We generated isogenic TREM2+/R47H , TREM2+/- , and TREM2-/- hPSCs using CRISPR/Cas9. Following differentiation to monocytes and tMGs, we studied the uptake of Escherichia coli fragments and analyzed amyloid plaque clearance from cryosections of APP/PS1+/- mouse brains., Results: We demonstrated that tMGs resemble cultured human microglia. TREM2+/- and TREM2-/- hPSC monocytes and tMGs phagocytosed significantly less E. coli fragments and cleared less amyloid plaques than wild-type hPSC progeny, with no difference for TREM2+/R47H progeny., Discussion: In vitro phagocytosis of hPSC monocytes and tMGs was not affected by the TREM2+/R47H mutation but was significantly impaired in TREM2+/- and TREM2-/- progeny., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
50. Loss of TREM2 function increases amyloid seeding but reduces plaque-associated ApoE.
- Author
-
Parhizkar S, Arzberger T, Brendel M, Kleinberger G, Deussing M, Focke C, Nuscher B, Xiong M, Ghasemigharagoz A, Katzmarski N, Krasemann S, Lichtenthaler SF, Müller SA, Colombo A, Monasor LS, Tahirovic S, Herms J, Willem M, Pettkus N, Butovsky O, Bartenstein P, Edbauer D, Rominger A, Ertürk A, Grathwohl SA, Neher JJ, Holtzman DM, Meyer-Luehmann M, and Haass C
- Subjects
- Alzheimer Disease metabolism, Alzheimer Disease pathology, Amyloid beta-Peptides genetics, Amyloid beta-Peptides metabolism, Amyloid beta-Protein Precursor genetics, Amyloid beta-Protein Precursor metabolism, Animals, Brain metabolism, Disease Models, Animal, Genotype, Humans, Membrane Glycoproteins metabolism, Mice, Mice, Transgenic, Microglia metabolism, Microglia pathology, Phagocytosis physiology, Plaque, Amyloid metabolism, Plaque, Amyloid pathology, Receptors, Immunologic metabolism, Alzheimer Disease genetics, Amyloid metabolism, Apolipoproteins E metabolism, Brain pathology, Membrane Glycoproteins genetics, Plaque, Amyloid genetics, Receptors, Immunologic genetics
- Abstract
Coding variants in the triggering receptor expressed on myeloid cells 2 (TREM2) are associated with late-onset Alzheimer's disease (AD). We demonstrate that amyloid plaque seeding is increased in the absence of functional Trem2. Increased seeding is accompanied by decreased microglial clustering around newly seeded plaques and reduced plaque-associated apolipoprotein E (ApoE). Reduced ApoE deposition in plaques is also observed in brains of AD patients carrying TREM2 coding variants. Proteomic analyses and microglia depletion experiments revealed microglia as one origin of plaque-associated ApoE. Longitudinal amyloid small animal positron emission tomography demonstrates accelerated amyloidogenesis in Trem2 loss-of-function mutants at early stages, which progressed at a lower rate with aging. These findings suggest that in the absence of functional Trem2, early amyloidogenesis is accelerated due to reduced phagocytic clearance of amyloid seeds despite reduced plaque-associated ApoE.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.