Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2022 Jul 15;19(1):187.
doi: 10.1186/s12974-022-02549-0.

Nilotinib modulates LPS-induced cognitive impairment and neuroinflammatory responses by regulating P38/STAT3 signaling

Affiliations

Nilotinib modulates LPS-induced cognitive impairment and neuroinflammatory responses by regulating P38/STAT3 signaling

Jieun Kim et al. J Neuroinflammation. .

Abstract

Background: In chronic myelogenous leukemia, reciprocal translocation between chromosome 9 and chromosome 22 generates a chimeric protein, Bcr-Abl, that leads to hyperactivity of tyrosine kinase-linked signaling transduction. The therapeutic agent nilotinib inhibits Bcr-Abl/DDR1 and can cross the blood-brain barrier, but its potential impact on neuroinflammatory responses and cognitive function has not been studied in detail.

Methods: The effects of nilotinib in vitro and in vivo were assessed by a combination of RT-PCR, real-time PCR, western blotting, ELISA, immunostaining, and/or subcellular fractionation. In the in vitro experiments, the effects of 200 ng/mL LPS or PBS on BV2 microglial cells, primary microglia or primary astrocytes pre- or post-treated with 5 µM nilotinib or vehicle were evaluated. The in vivo experiments involved wild-type mice administered a 7-day course of daily injections with 20 mg/kg nilotinib (i.p.) or vehicle before injection with 10 mg/kg LPS (i.p.) or PBS.

Results: In BV2 microglial cells, pre- and post-treatment with nilotinib altered LPS-induced proinflammatory/anti-inflammatory cytokine mRNA levels by suppressing AKT/P38/SOD2 signaling. Nilotinib treatment also significantly downregulated LPS-stimulated proinflammatory cytokine levels in primary microglia and primary astrocytes by altering P38/STAT3 signaling. Experiments in wild-type mice showed that nilotinib administration affected LPS-mediated microglial/astroglial activation in a brain region-specific manner in vivo. In addition, nilotinib significantly reduced proinflammatory cytokine IL-1β, IL-6 and COX-2 levels and P38/STAT3 signaling in the brain in LPS-treated wild-type mice. Importantly, nilotinib treatment rescued LPS-mediated spatial working memory impairment and cortical dendritic spine number in wild-type mice.

Conclusions: Our results indicate that nilotinib can modulate neuroinflammatory responses and cognitive function in LPS-stimulated wild-type mice.

Keywords: Cognitive function; LPS; Microglia; Nilotinib; SOD2; STAT3; p38.

PubMed Disclaimer

Conflict of interest statement

The authors have no competing interests to declare.

Figures

Fig. 1
Fig. 1
The multi-tyrosine kinase inhibitor nilotinib suppresses LPS-mediated proinflammatory responses and increases anti-inflammatory responses in BV2 microglial cells. a Structure of nilotinib. b Impact of nilotinib or vehicle on cell viability as assessed by the MTT assay (n = 14/group). c, d Immunocytochemistry of c-Abl expression in LPS-treated BV2 microglial cells pre-treated with nilotinib as shown (C: n = 54; L: n = 141; Nil + L: n = 73). e, f Immunocytochemistry of c-Abl expression in LPS-treated BV2 microglial cells post-treated with nilotinib as shown (C: n = 160; L: n = 130; L + Nil: n = 95). g Real-time PCR analysis of proinflammatory cytokine expression in LPS-treated BV2 microglial cells pre-treated with nilotinib as shown (n = 8/group). h Real-time PCR analysis of proinflammatory cytokine expression in LPS-treated BV2 microglial cells post-treated with nilotinib as shown (n = 6/group). i Real-time PCR analysis of anti-inflammatory cytokine expression in LPS-treated BV2 microglial cells post-treated with nilotinib as shown (n = 5–8/group). C: control, L: LPS, Nil + L: nilotinib + LPS, L + Nil: LPS + nilotinib, *p < 0.05, **p < 0.01, ***p < 0.001, scale bar = 20 μm
Fig. 2
Fig. 2
Nilotinib suppresses LPS-stimulated AKT/P38/STAT3 signaling in BV2 microglial cells. a, b RT-PCR analysis of proinflammatory cytokine IL-1β expression in BV2 microglial cells treated sequentially with LPS, TLR4 inhibitor (TAK-242) and nilotinib as shown (n = 7/group). c Real-time PCR analysis of proinflammatory cytokine COX-2 mRNA expression in BV2 microglial cells treated sequentially with LPS, TLR4 inhibitor (TAK-242) and nilotinib as shown (n = 8/group). d, e Western blotting analysis of AKT/P38 signaling in LPS-treated BV2 microglial cells post-treated with nilotinib as shown (n = 5/group). fi Immunocytochemistry analysis of AKT/P38 signaling in LPS-treated BV2 microglial cells post-treated with nilotinib (p-AKT, C: n = 181; L: n = 162; L + Nil: n = 111, p-P38, C: n = 197; L: n = 246; L + Nil: n = 157). j Western blot analysis of nuclear p-STAT3 expression in LPS-treated BV2 microglial post-treated with nilotinib as shown (n = 4/group). k Immunocytochemistry analysis of p-STAT3 levels in LPS-treated BV2 microglial cells post-treated with nilotinib as shown in j (C: n = 110; L: n = 161; L + Nil: n = 94). C: control, L: LPS, Nil + L: nilotinib + LPS, *p < 0.05, **p < 0.01, ***p < 0.001, scale bar = 20 μm
Fig. 3
Fig. 3
Nilotinib decreases LPS-mediated microglial neuroinflammatory responses in a Sod2-dependent manner. a Real-time PCR analysis of Sod2 gene expression in LPS-treated BV2 microglial cells post-treated with nilotinib as shown (n = 6/group). be Real-time PCR analysis of relative Sod2, IL-6, IL-1β, and COX-2 mRNA levels in BV2 microglial cells transfected with Sod2 siRNA (30 nM) or control (scramble) siRNA for 24 h and subsequently treated with LPS and nilotinib as shown (n = 8/group). C: control, L: LPS, Nil + L: nilotinib + LPS, *p < 0.05, ***p < 0.001
Fig. 4
Fig. 4
Nilotinib alters LPS-evoked changes in pro- and anti-inflammatory cytokine levels via P38/STAT signaling and suppresses Sod2 mRNA levels in primary microglia. a Assessment of primary microglial purity based on the CD11b/DAPI ratio (C: n = 371; L: n = 287; L + Nil: n = 216). b Immunocytochemistry analysis of c-Abl levels in LPS-treated primary microglia post-treated with nilotinib as shown (n = C: n = 561; L: n = 577; Nil + L: n = 298). c, d Real-time PCR analysis of pro- and anti-inflammatory cytokine levels in LPS-treated primary microglia post-treated with nilotinib as shown (n = 8/group). eg Western blotting analysis and immunohistochemistry staining of p-P38 and p-STAT3 levels in LPS-treated primary microglia post-treated with nilotinib as shown (p-P38 for western blot: n = 8/group; p-P38 for ICC: C: n = 526; L: n = 464; Nil + L: n = 352; p-STAT3 for ICC: C: n = 422; L: n = 358; Nil + L: n = 213). h Real-time PCR analysis of Sod2 mRNA in LPS-treated primary microglia post-treated with nilotinib (n = 8/group). *p < 0.05, **p < 0.01, ***p < 0.001. Scale bar = 20 μM
Fig. 5
Fig. 5
Nilotinib downregulates LPS-induced proinflammatory responses in primary astrocytes. a Assessment of primary astrocyte purity based on the GFAP/DAPI ratio (C: n = 253; L: n = 231; L + Nil: n = 236). b Immunocytochemistry analysis of c-Abl expression in LPS-treated primary astrocytes pre-treated with nilotinib as shown (C: n = 59; L: n = 43; Nil + L: n = 42). c Real-time PCR analysis of proinflammatory cytokine levels in LPS-treated primary astrocytes pre-treated with nilotinib as shown (n = 8/group). d Immunocytochemistry analysis of c-Abl expression in LPS-treated primary astrocytes post-treated with nilotinib as shown (C: n = 45; L: n = 39; L + Nil: n = 18). e Real-time PCR analysis of proinflammatory cytokine levels in LPS-treated primary astrocytes post-treated with nilotinib as shown (n = 8/group). C: control, L: LPS, Nil + L: nilotinib + LPS, L + Nil: LPS + nilotinib, **p < 0.01, ***p < 0.001, scale bar = 20 μm
Fig. 6
Fig. 6
Nilotinib suppresses LPS-stimulated P38/STAT3 signaling in primary astrocytes. a, c, e Western blotting analysis of p-P38/p-AKT signaling and p-STAT3/NF-kB transcription factors in LPS-treated primary astrocytes post-treated with nilotinib as shown (n = 5/group). b, d, f, g Immunocytochemistry analysis of p-P38, p-AKT, p-STAT3, and p-NF-kB levels in LPS-treated primary astrocytes post-treated with nilotinib as shown (p-P38, C: n = 17; L: n = 26; L + Nil: n = 15, p-AKT, C: n = 25; L: n = 30; L + Nil: n = 18, p-STAT3, C: n = 126; L: n = 128; L + Nil: n = 173, p-NF-kB, C: n = 203; L: n = 229; L + Nil: n = 213). C: control, L: LPS, L + Nil: LPS + nilotinib, *p < 0.05, **p < 0.01, ***p < 0.001, scale bar = 20 μm
Fig. 7
Fig. 7
Nilotinib reduces LPS-evoked c-Abl levels in the brain in wild-type mice. a Immunofluorescence staining of c-Abl expression in brain slices from wild-type mice injected with vehicle (5% DMSO, 10% PEG and 20% Tween 80 in deionized water) or nilotinib (20 mg/kg, i.p.) daily for 7 days followed by LPS (10 mg/kg, i.p.) or PBS on day 7. b Quantification of the data in a (n = 12–23 brain slices from 3–4 mice/group). c Western blotting analysis of c-Abl protein levels in the cortex and hippocampus (n = 4/group). *p < 0.05, **p < 0.01, ***p < 0.001. Scale bar = 100 μM
Fig. 8
Fig. 8
Nilotinib decreases LPS-induced microglial activation/morphology and astrocyte morphology in the brain in wild-type mice. a, c Immunofluorescence staining of microglial or astrocyte expression in brain slices from wild-type mice injected with vehicle (5% DMSO, 10% PEG and 20% Tween 80 in deionized water) or nilotinib (20 mg/kg, i.p.) daily for 7 days followed by LPS (10 mg/kg, i.p.) or PBS on day 7. b, d Quantification of data in a and c (Iba-1, n = 15–22 brain slices from 3–4 mice/group; GFAP, n = 15–22 brain slices from 3 to 4 mice/group). *p < 0.05, **p < 0.01, ***p < 0.001. Scale bar = 100 μM
Fig. 9
Fig. 9
Nilotinib reduces LPS-mediated IL-1β, IL-6, and COX-2 mRNA and protein levels in wild-type mice. ac Real-time PCR of IL-1β, IL-6, and COX-2 mRNA levels in the cortex and hippocampus (n = 8/group). df Immunofluorescence staining of IL-1β, IL-6, and COX-2 in brain slices from wild-type mice injected with vehicle (5% DMSO, 10% PEG and 20% Tween 80 in deionized water) or nilotinib (20 mg/kg, i.p.) daily for 7 days followed by LPS (10 mg/kg, i.p.) or PBS on day 7. The graphs present the quantification of the raw data from the IF staining in df (IL-1β, n = 14–20 brain slices from 3 to 4 mice/group: IL-6, and COX-2, n = 20–22 brain slices from 3 to 4 mice/group). gi ELISA of IL-1β, IL-6, and COX-2 protein levels in the cortex and hippocampus (n = 8/group). *p < 0.05, **p < 0.01, ***p < 0.001. Scale bar = 100 μM
Fig. 10
Fig. 10
Nilotinib reduces LPS-induced p-P38 and p-STAT3 protein levels in wild-type mice. a, c Immunofluorescence staining of p-P38 and p-STAT3 in brain slices from wild-type mice injected with vehicle (5% DMSO, 10% PEG and 20% Tween 80 in deionized water) or nilotinib (20 mg/kg, i.p.) daily for 7 days followed by LPS (10 mg/kg, i.p.) or PBS on day 7. b, d Quantification of data in a and c (p-P38, n = 14–24 brain slices from 3 to 4 mice/group, p-STAT3, n = 21–24 brain slices from 3 to 4 mice/group). e, f Western blotting analysis of p-P38 and p-STAT3 in the cortex and hippocampus in wild-type mice (n = 4/group). *p < 0.05, **p < 0.01, ***p < 0.001. Scale bar = 100 μM
Fig. 11
Fig. 11
Nilotinib reverses LPS-induced spatial memory impairment and cortical dendritic spine number in wild-type mice. a, b Y-maze and novel object recognition (NOR) tests were performed on days 7 and 8, respectively. Spontaneous alternations and number of total entries are represented in a, and object preference is shown in b (n = 8–11 mice/group). c, d After the behavior experiments, Golgi staining was performed, and dendritic spine number was measured in the cortical and hippocampal AO and BS regions (n = 25–26 neurons from 4 to 5 mice/group). *p < 0.05, **p < 0.01, ***p < 0.001. Scale bar = 10 μM
Fig. 12
Fig. 12
Diagram of the effects of nilotinib on LPS-stimulated neuroinflammatory responses and cognitive function. In microglial cells, nilotinib alters LPS-induced proinflammatory cytokine levels via AKT/P38/STAT3 activation and/or in a Sod2-dependent manner. In primary astrocytes, nilotinib downregulates the LPS-stimulated increase in proinflammatory cytokine levels by inhibiting the P38/STAT3 axis. In wild-type mice, nilotinib reduces LPS-induced gliosis, proinflammatory cytokine levels, and downstream P38/STAT3 signaling. Importantly, nilotinib rescues the short-term memory impairment and decrease in dendritic spinogenesis induced by LPS in wild-type mice. The ability of nilotinib to alter LPS-induced gliosis in vitro/in vivo and to modulate neuroinflammation-linked behavior suggests that nilotinib may have potential utility as a therapeutic drug for neuroinflammation/cognitive-associated disease

References

    1. Pagan FL, Hebron ML, Wilmarth B, Torres-Yaghi Y, Lawler A, Mundel EE, Yusuf N, Starr NJ, Arellano J, Howard HH, et al. Pharmacokinetics and pharmacodynamics of a single dose nilotinib in individuals with Parkinson’s disease. Pharmacol Res Perspect. 2019;7(2):e00470. doi: 10.1002/prp2.470. - DOI - PMC - PubMed
    1. Reinwald M, Schleyer E, Kiewe P, Blau IW, Burmeister T, Pursche S, Neumann M, Notter M, Thiel E, Hofmann WK, et al. Efficacy and pharmacologic data of second-generation tyrosine kinase inhibitor nilotinib in BCR-ABL-positive leukemia patients with central nervous system relapse after allogeneic stem cell transplantation. Biomed Res Int. 2014;2014:637059. doi: 10.1155/2014/637059. - DOI - PMC - PubMed
    1. Manley PW, Cowan-Jacob SW, Mestan J. Advances in the structural biology, design and clinical development of Bcr-Abl kinase inhibitors for the treatment of chronic myeloid leukaemia. Biochim Biophys Acta. 2005;1754(1–2):3–13. doi: 10.1016/j.bbapap.2005.07.040. - DOI - PubMed
    1. Jabbour E, Cortes J, Kantarjian H. Nilotinib for the treatment of chronic myeloid leukemia: an evidence-based review. Core Evid. 2010;4:207–213. - PMC - PubMed
    1. Olivieri A, Manzione L. Dasatinib: a new step in molecular target therapy. Ann Oncol. 2007;18(Suppl 6):vi42–vi46. doi: 10.1093/annonc/mdm223. - DOI - PubMed

MeSH terms