2024

Focal clusters of peri-synaptic matrix contribute to activity-dependent plasticity and memory in mice. Chelini G., Mirzapourdelavar H., Durning P., Baidoe-Ansah D., Sethi M.K., O’Donovan S.M., Klengel T., Balasco L., Berciu C., Boyer-Boiteau A., McCullumsmith R., Ressler K.J., Zaia J., Bozzi Y., Dityatev A., Berretta S. (2024) Cell Reports; 43; 114112; doi: https://doi.org/10.1016/j.celrep.2024.114112

Risk factors for intracerebral hemorrhage in small-vessel disease and non-small-vessel disease etiologies—an observational proof-of-concept study. Arndt Ph., Chahem Ch., Luchtmann M., Kuschel J.-N., Behme D., Pfister M., Neumann J., Görtler M., Dörner M., Pawlitzki M., Jansen R., Meuth S.G., Vielhaber St., Henneicke S., Schreiber St. (2024) Frontiers in Neurology; 15; 1322442; doi: http://doi.org/10.3389/fneur.2024.1322442

Microvascular damage, neuroinflammation and extracellular matrix remodeling in Col18a1 knockout mice as a model for early cerebral small vessel disease. Khoshneviszadeh M., Henneicke S., Piracy D., Senthilnathan A., Morton L., Arndt P., Kaushik R., Norman O., Jukkola J., Dunay I.R., Seidenbecher C., Heikkinen A., Schreiber St., Dityatev A. (2024) Matrix Biology; 128; 39-64; doi: https://doi.org/10.1016/j.matbio.2024.02.007

2023

Noradrenergic neuromodulation in ageing and disease. Krohn F., Lancini E., Ludwig M., Leiman M., Guruprasath G., Haag L., Panczyszyn J., Düzel E., Haemmerer D., Betts M. (2023) Neuroscience & Biobehavioral Reviews; Volume 152, 105311, doi: https://doi.org/10.1016/j.neubiorev.2023.105311

Brain microvascular disease and functional network connectivity – a call for a stage-based approach. Schreiber S., Arndt P., Meuth S.G., Dityatev A., Mattern H. (2023) Brain Communications;, fcad135, doi: https://doi.org/10.1093/braincomms/fcad135

Microvascular damage, neuroinflammation and extracellular matrix remodeling in Col18a1 knockout mice as a model for early cerebral small vessel disease. Khoshneviszadeh M., Jandke S., Morton L., Kaushik R., Arndt P., Norman O., Jukkola J., Dunay I.R., Seidenbecher C., Heikkinen A., Schreiber S., Dityatev A. (2023) PREPRINT (Version 2) Research Square, 19 April 2023; doi: https://doi.org/10.21203/rs.3.rs-1980968/v2

Depletion of neurocan in the prefrontal cortex impairs temporal order recognition, cognitive flexibility and perisomatic GABAergic innervation. Baidoe-Ansah D.*Mirzapourdelavar H.*, Carceller H., Perez M., Strackeljan L., Garcia B., Seidenbecher C.I.Kaushik R., Nacher J., Dityatev A. (2023) bioRxiv. 04.18.537277; doi: https://doi.org/10.1101/2023.04.18.537277 *shared first-authorship

Sex differences in anxiety and threat avoidance in GAD65 knock-out mice. Ulrich M., Pollali E., Çalışkan G.Stork O.Albrecht A. (2023)
bioRxiv 2023.03.16.532980; doi: https://doi.org/10.1101/2023.03.16.532980

Neural Extracellular Matrix Remodeling Signatures in Genetic and Acquired Mouse Models of Epilepsy. Blondiaux A., Jia S., Annamneedi A.Çalışkan G.Schulze J., Montenegro-Venegas C., Wykes R.C., Fejtova M., Walker M.C., Stork O.Gundelfinger E.D., Dityatev A., Seidenbecher C.I. (2023) BioRxiv. 2023.04.19.537468; doi: https://doi.org/10.1101/2023.04.19.537468

Brain Vascular Health in ALS Is Mediated through Motor Cortex Microvascular Integrity. Schreiber S., Bernal J., Arndt P., Schreiber F., Müller P., Morton L., Braun-Dullaeus R.C., Valdés-Hernández M.D.C., Duarte R., Wardlaw J.M., Meuth S.G., Mietzner G., Vielhaber S., Dunay I.R., Dityatev A., Jandke S., Mattern H. (2023) Cells. Mar 21; doi:https://www.mdpi.com/2073-4409/12/6/957.

Real-time mechanisms of exacerbated synaptic remodeling by microglia in acute models of systemic inflammation and tauopathy. Cangalaya C., Wegmann S., Sun W., Diez L., Gottfried A., Richter K., Stoyanov S., Pakan J., Fischer K.D., Dityatev A. (2023) Brain Behav Immun. Mar 9;110:245-259. https://doi.org/10.1016/j.bbi.2023.02.023

Extracellular Matrix Changes in Subcellular Brain Fractions and Cerebrospinal Fluid of Alzheimer’s Disease Patients. Höhn L., Hußler W., Richter A., Smalla K.-H., Birkl-Toeglhofer A.-M., Birkl C., Vielhaber S., Leber S.L., Gundelfinger E.D., Haybaeck J., Schreiber S., Seidenbecher C.I. (2023) Int. J. Mol. Sci. 24, 5532. https://doi.org/10.3390/ijms24065532

Mice deficient in synaptic protease neurotrypsin show impaired spaced long-term potentiation and blunted learning-induced modulation of dendritic spines. Ferrer-Ferrer M., Jia S., Kaushik R., Schneeberg J., Figiel I., Aleshin S., Mironov A., Safari M., Frischknecht R., Wlodarczyk J., Senkov A., Dityatev A. (2023) Cell. Mol. Life Sci.80, 82. https://doi.org/10.1007/s00018-023-04720-z

Initial and ongoing tobacco smoking elicits vascular damage and distinct inflammatory response linked to neurodegeneration. Garza AP*, Morton L*, Pállinger É, Buzás EI, Schreiber S., Schott B.H., Dunay I.R. (2023) Jan 29. Brain, Behavior, & Immunity – Health, Vol. 28, 2023, 100597. https://doi.org/10.1016/j.bbih.2023.100597

Heparan sulfates regulate axonal excitability and context generalization through Ca2+/calmodulin-dependent protein kinase II. Song I., Kuznetsova T., Baidoe-Ansah D., Mirzapourdelavar H., Senkov O., Hayani H., Mironov A., Kaushik R., Druzin M., Johansson S., Dityatev A. (2023) bioRxiv 2023.01.26.524131; https://doi.org/10.1101/2023.01.26.524131

Vascular and neural transcriptomics reveal stage-dependent pathways to inflammation and cognitive dysfunction in a rat model of hypertension. Ulbrich P., Morton L., Briese M., Lämmlin N., Mattern H., Hasanuzzaman M.d., Westhues M.,  Khoshneviszadeh M., Appenzeller S.,  Guendel D., Toussaint M., Brust P., Kniess T., Oelschlegel A., Goldschmidt J., Meuth S., Heinze H.J., Vielhaber G.D., Vielhaber S., Becker A., Dityatev A., Jandke S., Sendtner M., Dunay I., Schreiber S. (2023) bioRxiv 2023.01.20.524921; DOI: 10.1101/2023.01.20.524921

Priorities for research on neuromodulatory subcortical systems in Alzheimer’s disease: Position paper from the NSS PIA of ISTAART. Ehrenberg A.J.,  Kelberman M.A.,  Liu K.Y., …, Betts M.J., Lancini E., et al. Alzheimer’s Dement.  2023; 1- 15. https://doi.org/10.1002/alz.12937

Jacob-induced transcriptional inactivation of CREB promotes Aβ-induced synapse loss in Alzheimer’s disease. Grochowska K.M., Gomes G.M., Raman R., Kaushik R., Sosulina L., Kaneko H., Oelschlegel A.M., Yuanxiang P., Reyes-Resina I., Bayraktar G., Samer S., Spilker C., Woo M.S., Morawski M., Goldschmidt J., Friese M.A., Rossner S., Navarro G., Remy S., Reissner C., Karpova A.Kreutz M.R. EMBO J. 2023 Jan 3:e112453. doi: https://doi.org/10.15252/embj.20221124531

2022

Arterial hypertension and β-amyloid accumulation have spatially overlapping effects on posterior white matter hyperintensity volume: A cross-sectional study. Bernal J.,  Schreiber S., Menze I., Ostendorf A., Pfister M., Geisendoerfer J., Nemali A., Maass A., YakupovR., Peters O., Preis L., Schneider L., Herrera A.L., Priller J., Spruth E.J.,  Altenstein S.,  Schneider A., Fliessbach K., Wiltfang J., Schott B.H., Rostamzadeh A.,  Glanz W.,  Buerger K., Janowitz D., Ewers M., Perneczky R., RauchmannB.S., Teipel S., Kilimann I., Laske C., Munk M.H., Spottke A., Roy N., Dobisch L., Dechent P., Scheffler K., Hetzer S., Wolfsgruber, S., Kleineidam L., Schmid M., Berger M., Jessen F., Wirth M., Duezel E., Ziegler G. (2022)medRxiv 2022.07.19.22277546; DOI: https://doi.org/10.1101/2022.07.19.22277546

IFN-γ-mediated neuronal defense mechanism targets Toxoplasma. Figueiredo C.A., Dunay I.R. (2022) Trends in Parasitology. ISSN 1471-4922. https://doi.org/10.1016/j.pt.2022.10.001

The neuropeptide PACAP alleviates T. gondii infection-induced neuroinflammation and neuronal impairment. Figueiredo C.A., Düsedau H.P., Steffen J., Ehrentraut S., Dunay M.P., Toth G., Reglödi D., Heimesaat M.M., Dunay I.R. (2022) J Neuroinflammation 19, 274.https://doi.org/10.21203/rs.3.rs-1498493/v2

Error-driven associative learning: a multivariate cross-classification fMRI study on interactions between performance monitoring and memory formation. Weuthen A., Kirschner H., Ullsperger M. (2022) PsyArXiv. DOI: https://doi.org/10.31234/osf.io/3f689

Microvascular damage, neuroinflammation and extracellular matrix remodeling in Col18a1 knockout mice as a model for early cerebral small vessel disease. Khoshneviszadeh M., Jandke S., Kaushik R., Ulbrich P., Norman O., Jukkola J., Heikkinen A., Schreiber S., Dityatev A. (2022) PREPRINT (Version 1) Research Square. https://doi.org/10.21203/rs.3.rs-1980968/v1

Stage-dependent responses of vascular and parenchymal cells in the hypertensive rat brain. Ulbrich P., Morton L., Briese M., Lämmlin N., Mattern H., Hasanuzzaman M.d., Westhues M.,  Khoshneviszadeh M., Garz C., Becker A., Dityatev A., Jandke S., Yilmazer-Hanke D., Sendtner M., Dunay I.R.Schreiber S. (2022) Journal of Hypertension 40(Suppl 1):e146. https://doi.org/10.1097/01.hjh.0000836812.14897.03

Fear engrams and NPYergic circuit in the dorsal dentate gyrus determine remote fear memory generalization. Raza S.A., Klinger K.del Ángel M.Demiray Y.E.Çalışkan G.Kreutz M.R.Stork O. (2022) bioRxiv 2022.04.26.489543;  https://doi.org/10.1101/2022.04.26.489543

Persisting Microbiota and Neuronal Imbalance Following T. gondii Infection Reliant on the Infection Route. French T., Steffen J., Glas A., Osbelt L., Strowig T., Schott B.H., Schüler T., Dunay I.R. (2022) Front Immunol. Jul 11;13:920658. https://doi.org/10.3389/fimmu.2022.920658

The needs of a synapse-How local organelles serve synaptic proteostasis. Grochowska K.M., Andres-Alonso M., Karpova A.Kreutz M.R. EMBO J. 2022 Apr 4;41(7):e110057. DOI: https://doi.org/10.15252/embj.2021110057.

Choosing memory retrieval strategies: a critical role for inhibition in the dentate gyrus. Albrecht A., Müller I., Weiglein A., Pollali E., Çalışkan G.Stork O. (2022) Neurobiol Stress. Jul 31;20:100474.https://doi.org/10.1016/j.ynstr.2022.100474

Feedback-related EEG dynamics separately reflect decision parameters, biases, and future choices. Kirschner H., Fischer A.G., Ullsperger M. (2022) NeuroImage, Volume 259, 119437, ISSN 1053-8119, https://doi.org/10.1016/j.neuroimage.2022.119437

Monitoring regional astrocyte diversity by cell-type specific proteomic labeling in vivo. Prabhakar P., Pielot R., Landgraf P., Wissing J., Bayrhammer A., van Ham M., Gundelfinger E.D., Jänsch L., Dieterich D.C.Müller A. (2022) Glia,  1– 22. https://doi.org/10.1002/glia.24304

Neddylation-dependent protein degradation is a nexus between synaptic insulin resistance, neuroinflammation and Alzheimer’s disease. Confettura A.D., Cuboni E., Ammar M.R., Jia S., Gomes G.M., Yuanxiang P., Raman R., Li T., Grochowska K.M., Ahrends R., Karpova A., Dityatev A., Kreutz M.R. (2022) Transl Neurodegener. Jan 6;11(1):2. https://doi.org/10.1186/s40035-021-00277-8

Aging-Associated Changes in Cognition, Expression and Epigenetic Regulation of Chondroitin 6-Sulfotransferase Chst3. Baidoe-Ansah D., Sakib S., Jia S., Mirzapourdelavar H., Strackeljan L., Fischer A., Aleshin S., Kaushik R., Dityatev A. (2022) Cells. Jun 27;11(13):2033. https://doi.org/10.3390/cells11132033

Brevican and Neurocan Cleavage Products in the Cerebrospinal Fluid – Differential Occurrence in ALS, Epilepsy and Small Vessel Disease. Hußler W., Höhn L., Stolz C., Vielhaber S., Garz C., Schmitt F.C., Gundelfinger E.D., Schreiber S. and Seidenbecher C.I. (2022) Front. Cell. Neurosci. 16:838432. https://doi.org/10.3389/fncel.2022.838432

2021

Influenza A Virus (H1N1) Infection Induces Microglial Activation and Temporal Dysbalance in Glutamatergic Synaptic Transmission. Düsedau H.P., Steffen J., Figueiredo C.A., Boehme J.D., Schultz K., Erck C., Korte M., Faber-Zuschratter H., Smalla K.H., Dieterich D., Kröger A., Bruder D., Dunay IR. (2021) mBio. Oct 26;12(5):e0177621. https://doi.org/10.1128/mBio.01776-21

Extracellular matrix remodeling through endocytosis and resurfacing of Tenascin-R. Dankovich T.M., Kaushik R., Olsthoorn L.H.M., Petersen G.C., Giro P.E., Kluever V., Agüi-Gonzalez P., Grewe K., Bao G., Beuermann S., Hadi H.A., Doeren J., Klöppner S., Cooper B.H., Dityatev A., Rizzoli S.O. (2021) Nat Commun. Dec 8;12(1):7129. https://doi.org/10.1038/s41467-021-27462-7

Antibiotic-induced gut dysbiosis leads to activation of microglia and impairment of cholinergic gamma oscillations in the hippocampus. Çalışkan G., French T., Enrile Lacalle S., Del Angel M., Steffen J., Heimesaat MM., Rita Dunay I., Stork O. (2021) Brain, Behavior, and Immunity 99:203-217. https://doi.org/10.1016/j.bbi.2021.10.007

Brain extracellular matrix: An upcoming target in neurological and psychiatric disorders. Dityatev A., Seidenbecher C. and Morawski, M. (2021) Eur J Neurosci 2021 53: 3807-3810. https://doi.org/10.1111/ejn.15336

CSF and PET biomarkers for noradrenergic dysfunction in neurodegenerative diseases: a systematic review and meta-analysis. Lancini E.Haag L., Bartl F., Rühling M., Ashton N.J., Zetterberg H., Düzel E.Hämmerer D.Betts M.J. (2021) Alzheimer’s Dement., 17: e055942. https://doi.org/10.1002/alz.055942

Microglia Depletion-Induced Remodeling of Extracellular Matrix and Excitatory Synapses in the Hippocampus of Adult Mice. Strackeljan L., Baczynska E., Cangalaya C., Baidoe-Ansah D., Wlodarczyk, J.; Kaushik R., Dityatev A.  (2021) Cells 10, 1862. https://doi.org/10.3390/cells10081862

Transgenic modelling of Ndr2 gene amplification reveals disturbance of hippocampus circuitry and function. Madencioglu D.A., Çalışkan G., Yuanxiang P., Rehberg K., Demiray Y.E., Kul E., Engler A., Hayani H., Bergado-Acosta J.R., Kummer A., Müller I., Song I., Dityatev A., Kähne T., Kreutz M.R., Stork O. (2021) iScience Volume 24, Issue 8, 102868, ISSN 2589-0042. https://doi.org/10.1016/j.isci.2021.102868

The Presynaptic Scaffold Protein Bassoon in Forebrain Excitatory Neurons Mediates Hippocampal Circuit Maturation: Potential Involvement of TrkB Signalling. Annamneedi A., del Angel C.M., Gundelfinger E.D., Stork O., Caliskan G.. (2021) International Journal of Molecular Sciences 22(15):7944. https://doi.org/10.3390/ijms22157944

Autism-associated SHANK3 missense point mutations impact conformational fluctuations and protein turnover at synapses. Bucher M., Niebling S., Han Y., Molodenskiy D., Hassani Nia F., Kreienkamp H.J., Svergun D., Kim E., Kostyukova A.S., Kreutz M.R., Mikhaylova M. (2021) Elife. 10:e66165. https://doi.org/10.7554/eLife.66165 

Met carriers of the BDNF Val66Met polymorphism show reduced Glx/NAA in the pregenual ACC in two independent cohorts. Martens, L., Herrmann, L., Colic, L., Li, M., Richter, A., Behnisch, G., Stork, O., Seidenbecher, C., Schott, B.H., Walter, M. (2021) Sci Rep. Mar 24;11(1):6742. https://doi.org/10.1038/s41598-021-86220-3

To err is (not only) human: Mechanisms of post-error attentional regulation illuminated in mice. Ullsperger, M., Stork, O. (2021) Neuron 109(7):1074-1076. https://doi.org/10.1016/j.neuron.2021.03.014

Short antisense oligonucleotides alleviate the pleiotropic toxicity of RNA harboring expanded CGG repeats. Derbis M, Kul E, Niewiadomska, D., Sekrecki, M., Piasecka, A., Taylor, K., Hukema, R.K., Stork, O., Sobczak, K. (2021) Nat Commun. Feb 24;12(1):1265.PMID: 33627639 https://doi.org/10.1038/s41467-021-21021-w

The nuclear lamina is a hub for the nuclear function of Jacob. Samer, S., Raman, R., Laube, G., Kreutz, M.R., Karpova, A. (2021) Mol Brain. 14(1):9. https://doi.org/10.1186/s13041-020-00722-1

Age impairs mnemonic discrimination of objects more than scenes: A web-based, large-scale approach across the lifespan. J. Gusten, G. Ziegler, E.Duzel, D. Berron. (2021) Cortex. Apr 137, 138-148; https://doi.org/10.1016/j.cortex.2020.12.017

Multiomics of synaptic junctions reveals altered lipid metabolism and signaling following environmental enrichment. M. Borgmeyer, C. Coman, C. Has, H.F. Schött, T. Li, P. Westhoff, Y.F.H. Cheung, N. Hoffmann, P. Yuanxiang, T. Behnisch, G.M. Gomes, M. Dumenieu, M. Schweizer, M. Chocholoušková, M. Holčapek, M. Mikhaylova, M.R.*Kreutz, R*. Ahrends. (2021) Cell Rep Oct 5; 37(1):109797; https://doi.org10.1016/j.celrep.2021.109797

2020

Allostatic gene regulation of inhibitory synaptic factors in the rat ventral hippocampus in a juvenile/adult stress model of psychopathology. Albrecht A., Segal M., Stork O. (2020) Eur J Neurosci. 55:2142-2153. https://doi.org/10.1111/ejn.15091

International Consensus Based Review and Recommendations for Minimum Reporting Standards in Research on Transcutaneous Vagus Nerve Stimulation Farmer A.D., Strzelczyk A., Finisguerra A., Gourine A.V., …, Haemmerer D., …, Betts M.J., Koenig J (2020). Frontiers. Hum. Neurosci 14: 409 https://doi.org/10.3389%2Ffnhum.2020.568051

Noradrenergic-dependent functions are associated with age-related locus coeruleus signal intensity differences. Liu, K.Y., Kievit, R.A., Tsvetanov, K.A., Betts M.J., Düzel E., Rowe J.B., CAN-Cam, Howard R., Haemmerer D. (2020) Nat Commun. 11, 1712. https://doi.org/10.1038/s41467-020-15410-w

Attenuation of the extracellular matrix restores microglial activity during the early stage of amyloidosis. Stoyanov, S., Sun, W., Düsedau, H.P., Cangalaya, C., Choi, I., Mirzapourdelavar, H., Baidoe‐Ansah, D., Kaushik, R., Neumann, J., Dunay, I.R., Dityatev, A. (2020) Glia 2021;69:182-200. https://doi.org/10.1002/glia.23894

Interplay between perivascular and perineuronal extracellular matrix remodelling in neurological and psychiatric diseases. Ulbrich, P., Khoshneviszadeh, M., Jandke, S., Schreiber, S., Dityatev, A. (2020) European Journal of Neuroscience 2021; 53:3811-3830. https://doi.org/10.1111/ejn.14887

Synaptic control of DNA methylation involves activity-dependent degradation of DNMT3A1 in the nucleus. Bayraktar, G., Yuanxiang, P., Confettura, A.D., Gomes, G.M., Raza, S.A., Stork, O., Tajima, S., Suetake, I., Karpova, A., Yildirim, F., Kreutz, M.R. (2020) Neuropsychopharmacol. 45, 2120–2130. https://doi.org/10.1038/s41386-020-0780-2

2019

Locus coeruleus MRI contrast is reduced in Alzheimer’s disease dementia and correlates with CSF Abeta levels. M.J. Betts, A. Cardenas-Blanco, M. Kanowski, A. Spottke, S.J. Teipel, I. Kilimann, F. Jessen, E.Duzel. (2019) Alzheimers Dement. Dec 281-285. https://doi.org/10.1016/j.dadm.2019.02.001

Epigenetic mechanism of carbohydrate sulfotransferase 3 (CHST3) downregulation in the aging brain. Baidoe-Ansah D., Sakib M.S., Jia S., Fischer A., Kaushik R.Dityatev A. (2019) bioRxiv 741355. https://doi.org/10.1101/741355