In vivo nanoparticle imaging of innate immune cells can serve as a marker of disease severity in a model of multiple sclerosis

Standard

In vivo nanoparticle imaging of innate immune cells can serve as a marker of disease severity in a model of multiple sclerosis. / Kirschbaum, Klara; Sonner, Jana K; Zeller, Matthias W; Deumelandt, Katrin; Bode, Julia; Sharma, Rakesh; Krüwel, Thomas; Fischer, Manuel; Hoffmann, Angelika; Costa da Silva, Milene; Muckenthaler, Martina U; Wick, Wolfgang; Tews, Björn; Chen, John W; Heiland, Sabine; Bendszus, Martin; Platten, Michael; Breckwoldt, Michael O.

In: P NATL ACAD SCI USA, Vol. 113, No. 46, 15.11.2016, p. 13227-13232.

Research output: SCORING: Contribution to journalSCORING: Journal articleResearchpeer-review

Harvard

Kirschbaum, K, Sonner, JK, Zeller, MW, Deumelandt, K, Bode, J, Sharma, R, Krüwel, T, Fischer, M, Hoffmann, A, Costa da Silva, M, Muckenthaler, MU, Wick, W, Tews, B, Chen, JW, Heiland, S, Bendszus, M, Platten, M & Breckwoldt, MO 2016, 'In vivo nanoparticle imaging of innate immune cells can serve as a marker of disease severity in a model of multiple sclerosis', P NATL ACAD SCI USA, vol. 113, no. 46, pp. 13227-13232. https://doi.org/10.1073/pnas.1609397113

APA

Kirschbaum, K., Sonner, J. K., Zeller, M. W., Deumelandt, K., Bode, J., Sharma, R., Krüwel, T., Fischer, M., Hoffmann, A., Costa da Silva, M., Muckenthaler, M. U., Wick, W., Tews, B., Chen, J. W., Heiland, S., Bendszus, M., Platten, M., & Breckwoldt, M. O. (2016). In vivo nanoparticle imaging of innate immune cells can serve as a marker of disease severity in a model of multiple sclerosis. P NATL ACAD SCI USA, 113(46), 13227-13232. https://doi.org/10.1073/pnas.1609397113

Vancouver

Bibtex

@article{e2cfd28b49c34387a6a5287b3be9c459,
title = "In vivo nanoparticle imaging of innate immune cells can serve as a marker of disease severity in a model of multiple sclerosis",
abstract = "Innate immune cells play a key role in the pathogenesis of multiple sclerosis and experimental autoimmune encephalomyelitis (EAE). Current clinical imaging is restricted to visualizing secondary effects of inflammation, such as gliosis and blood-brain barrier disruption. Advanced molecular imaging, such as iron oxide nanoparticle imaging, can allow direct imaging of cellular and molecular activity, but the exact cell types that phagocytose nanoparticles in vivo and how phagocytic activity relates to disease severity is not well understood. In this study we used MRI to map inflammatory infiltrates using high-field MRI and fluorescently labeled cross-linked iron oxide nanoparticles for cell tracking. We confirmed nanoparticle uptake and MR detectability ex vivo. Using in vivo MRI, we identified extensive nanoparticle signal in the cerebellar white matter and circumscribed cortical gray matter lesions that developed during the disease course (4.6-fold increase of nanoparticle accumulation in EAE compared with healthy controls, P < 0.001). Nanoparticles showed good cellular specificity for innate immune cells in vivo, labeling activated microglia, infiltrating macrophages, and neutrophils, whereas there was only sparse uptake by adaptive immune cells. Importantly, nanoparticle signal correlated better with clinical disease than conventional gadolinium (Gd) imaging (r, 0.83 for nanoparticles vs. 0.71 for Gd-imaging, P < 0.001). We validated our approach using the Food and Drug Administration-approved iron oxide nanoparticle ferumoxytol. Our results show that noninvasive molecular imaging of innate immune responses can serve as an imaging biomarker of disease activity in autoimmune-mediated neuroinflammation with potential clinical applications in a wide range of inflammatory diseases.",
keywords = "Animals, Brain/diagnostic imaging, Encephalomyelitis, Autoimmune, Experimental/diagnostic imaging, Female, Immunity, Innate, Macrophages/immunology, Magnetic Resonance Imaging, Magnetite Nanoparticles/administration & dosage, Mice, Microglia/immunology, Multiple Sclerosis/diagnostic imaging, Phagocytosis, Severity of Illness Index",
author = "Klara Kirschbaum and Sonner, {Jana K} and Zeller, {Matthias W} and Katrin Deumelandt and Julia Bode and Rakesh Sharma and Thomas Kr{\"u}wel and Manuel Fischer and Angelika Hoffmann and {Costa da Silva}, Milene and Muckenthaler, {Martina U} and Wolfgang Wick and Bj{\"o}rn Tews and Chen, {John W} and Sabine Heiland and Martin Bendszus and Michael Platten and Breckwoldt, {Michael O}",
year = "2016",
month = nov,
day = "15",
doi = "10.1073/pnas.1609397113",
language = "English",
volume = "113",
pages = "13227--13232",
journal = "P NATL ACAD SCI USA",
issn = "0027-8424",
publisher = "National Academy of Sciences",
number = "46",

}

RIS

TY - JOUR

T1 - In vivo nanoparticle imaging of innate immune cells can serve as a marker of disease severity in a model of multiple sclerosis

AU - Kirschbaum, Klara

AU - Sonner, Jana K

AU - Zeller, Matthias W

AU - Deumelandt, Katrin

AU - Bode, Julia

AU - Sharma, Rakesh

AU - Krüwel, Thomas

AU - Fischer, Manuel

AU - Hoffmann, Angelika

AU - Costa da Silva, Milene

AU - Muckenthaler, Martina U

AU - Wick, Wolfgang

AU - Tews, Björn

AU - Chen, John W

AU - Heiland, Sabine

AU - Bendszus, Martin

AU - Platten, Michael

AU - Breckwoldt, Michael O

PY - 2016/11/15

Y1 - 2016/11/15

N2 - Innate immune cells play a key role in the pathogenesis of multiple sclerosis and experimental autoimmune encephalomyelitis (EAE). Current clinical imaging is restricted to visualizing secondary effects of inflammation, such as gliosis and blood-brain barrier disruption. Advanced molecular imaging, such as iron oxide nanoparticle imaging, can allow direct imaging of cellular and molecular activity, but the exact cell types that phagocytose nanoparticles in vivo and how phagocytic activity relates to disease severity is not well understood. In this study we used MRI to map inflammatory infiltrates using high-field MRI and fluorescently labeled cross-linked iron oxide nanoparticles for cell tracking. We confirmed nanoparticle uptake and MR detectability ex vivo. Using in vivo MRI, we identified extensive nanoparticle signal in the cerebellar white matter and circumscribed cortical gray matter lesions that developed during the disease course (4.6-fold increase of nanoparticle accumulation in EAE compared with healthy controls, P < 0.001). Nanoparticles showed good cellular specificity for innate immune cells in vivo, labeling activated microglia, infiltrating macrophages, and neutrophils, whereas there was only sparse uptake by adaptive immune cells. Importantly, nanoparticle signal correlated better with clinical disease than conventional gadolinium (Gd) imaging (r, 0.83 for nanoparticles vs. 0.71 for Gd-imaging, P < 0.001). We validated our approach using the Food and Drug Administration-approved iron oxide nanoparticle ferumoxytol. Our results show that noninvasive molecular imaging of innate immune responses can serve as an imaging biomarker of disease activity in autoimmune-mediated neuroinflammation with potential clinical applications in a wide range of inflammatory diseases.

AB - Innate immune cells play a key role in the pathogenesis of multiple sclerosis and experimental autoimmune encephalomyelitis (EAE). Current clinical imaging is restricted to visualizing secondary effects of inflammation, such as gliosis and blood-brain barrier disruption. Advanced molecular imaging, such as iron oxide nanoparticle imaging, can allow direct imaging of cellular and molecular activity, but the exact cell types that phagocytose nanoparticles in vivo and how phagocytic activity relates to disease severity is not well understood. In this study we used MRI to map inflammatory infiltrates using high-field MRI and fluorescently labeled cross-linked iron oxide nanoparticles for cell tracking. We confirmed nanoparticle uptake and MR detectability ex vivo. Using in vivo MRI, we identified extensive nanoparticle signal in the cerebellar white matter and circumscribed cortical gray matter lesions that developed during the disease course (4.6-fold increase of nanoparticle accumulation in EAE compared with healthy controls, P < 0.001). Nanoparticles showed good cellular specificity for innate immune cells in vivo, labeling activated microglia, infiltrating macrophages, and neutrophils, whereas there was only sparse uptake by adaptive immune cells. Importantly, nanoparticle signal correlated better with clinical disease than conventional gadolinium (Gd) imaging (r, 0.83 for nanoparticles vs. 0.71 for Gd-imaging, P < 0.001). We validated our approach using the Food and Drug Administration-approved iron oxide nanoparticle ferumoxytol. Our results show that noninvasive molecular imaging of innate immune responses can serve as an imaging biomarker of disease activity in autoimmune-mediated neuroinflammation with potential clinical applications in a wide range of inflammatory diseases.

KW - Animals

KW - Brain/diagnostic imaging

KW - Encephalomyelitis, Autoimmune, Experimental/diagnostic imaging

KW - Female

KW - Immunity, Innate

KW - Macrophages/immunology

KW - Magnetic Resonance Imaging

KW - Magnetite Nanoparticles/administration & dosage

KW - Mice

KW - Microglia/immunology

KW - Multiple Sclerosis/diagnostic imaging

KW - Phagocytosis

KW - Severity of Illness Index

U2 - 10.1073/pnas.1609397113

DO - 10.1073/pnas.1609397113

M3 - SCORING: Journal article

C2 - 27799546

VL - 113

SP - 13227

EP - 13232

JO - P NATL ACAD SCI USA

JF - P NATL ACAD SCI USA

SN - 0027-8424

IS - 46

ER -