ArticlePDF Available

The stability of the metabolic turnover of arachidonic acid in human unruptured intracranial aneurysmal walls is sustained

Authors:

Abstract and Figures

Objective Intracranial aneurysm (IA) is considered a chronic inflammatory condition that affects intracranial arteries. Cyclooxygenase 2 (COX2) and prostaglandin E2 (PGE2) are considered potential targets of specific medical treatment for IAs. Previous studies have reported the elevated COX2 expression in the IA wall. However, not much has been studied about the upstream regulation of COX2 and PGE2, and the metabolism of arachidonic acid (AA) in human IAs. In this study, we aimed to elucidate the distribution of fatty acids in human IA walls for the first time. Methods Samples from 6 ruptured and 5 unruptured human IAs were surgically resected after the aneurysmal clipping and analyzed using desorption electrospray ionization imaging mass spectrometry. Results AA and AA-containing phospholipids were not detected in the unruptured IA walls. On the contrast, significantly larger amounts of AA and AA-containing phospholipids were detected in the ruptured IA walls compared to unruptured IA walls. Conclusions This study showed for the first time that AA was not detected in unruptured human IA walls. Our findings suggest that the stability of the turnover of AA in human unruptured IA walls is sustained. In contrast, this study showed that larger amounts of AA and AA-containing phospholipids were detected in the ruptured IA walls. More cases and further analysis are necessary to interpret our present results.
Content may be subject to copyright.
Clinical Neurology and Neurosurgery 208 (2021) 106881
Available online 8 August 2021
0303-8467/© 2021 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
The stability of the metabolic turnover of arachidonic acid in human
unruptured intracranial aneurysmal walls is sustained
Ririko Takeda
a
,
b
,
*
,
1
, Ariful Islam
c
,
1
, Tomohito Sato
c
,
d
, Hiroki Kurita
b
, Tomoaki Kahyo
c
,
d
,
Tetsumei Urano
e
, Mitsutoshi Setou
c
,
d
,
f
a
Department of Neurosurgery, Teikyo University Hospital, Mizonokuchi, Kawasaki, Japan
b
Department of Cerebrovascular Surgery, International Medical Center, Saitama Medical University, Hidaka, Japan
c
Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
d
International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
e
Department of Physiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
f
Department of Systems Molecular Anatomy, Institute for Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu, Japan
ARTICLE INFO
Keywords:
Intracranial aneurysm
Imaging mass spectroscopy
Arachidonic acid
Phospholipid
ABSTRACT
Objective: Intracranial aneurysm (IA) is considered a chronic inammatory condition that affects intracranial
arteries. Cyclooxygenase 2 (COX2) and prostaglandin E2 (PGE2) are considered potential targets of specic
medical treatment for IAs. Previous studies have reported the elevated COX2 expression in the IA wall. However,
not much has been studied about the upstream regulation of COX2 and PGE2, and the metabolism of arachidonic
acid (AA) in human IAs. In this study, we aimed to elucidate the distribution of fatty acids in human IA walls for
the rst time.
Methods: Samples from 6 ruptured and 5 unruptured human IAs were surgically resected after the aneurysmal
clipping and analyzed using desorption electrospray ionization imaging mass spectrometry.
Results: AA and AA-containing phospholipids were not detected in the unruptured IA walls. On the contrast,
signicantly larger amounts of AA and AA-containing phospholipids were detected in the ruptured IA walls
compared to unruptured IA walls.
Conclusions: This study showed for the rst time that AA was not detected in unruptured human IA walls. Our
ndings suggest that the stability of the turnover of AA in human unruptured IA walls is sustained. In contrast,
this study showed that larger amounts of AA and AA-containing phospholipids were detected in the ruptured IA
walls. More cases and further analysis are necessary to interpret our present results.
1. Introduction
Intracranial aneurysm (IA) is currently considered a chronic in-
ammatory disease that affects the intracranial arteries [1]. As an un-
derlying mechanism, the prostaglandin E2 (PGE2)-EP2-NF-κB signaling
cascade in macrophages was considered by Aoki et al. as a factor regu-
lating such chronic inammation involved in the progression and
rupture of IAs [1]. PGE2 is a lipid mediator and an arachidonic acid (AA)
metabolite generated from sequential enzymatic reactions, including
cyclooxygenase (COX), and a potential therapeutic target for IAs [1].
Increased levels of free fatty acids (FFAs), including AA, in several brain
regions after subarachnoid hemorrhage were reported in a previous
study using a rat model [2]. The association of AA-containing phos-
pholipids in the development and rupture of IA walls in a rat model were
also reported in a previous study [3]. However, to date, no report has
described the distribution of FFAs in the walls of IA in humans.
Therefore, we investigated the extent of FFA distribution in the walls
of IAs in human subjects by using a recently developed imaging mass
spectrometry (IMS) technique, desorption electrospray ionization IMS
(DESI-IMS).
* Correspondence to: Department of Neurosurgery, Teikyo University Hospital, 5-1-1 futako, Takatsu-ku, Mizonokuchi, Kawasaki, Kanagawa 213-8507, Japan.
E-mail address: rtakeda@med.teikyo-u.ac.jp (R. Takeda).
1
These authors have equal contribution.
Contents lists available at ScienceDirect
Clinical Neurology and Neurosurgery
journal homepage: www.elsevier.com/locate/clineuro
https://doi.org/10.1016/j.clineuro.2021.106881
Received 8 January 2021; Received in revised form 3 August 2021; Accepted 4 August 2021
Clinical Neurology and Neurosurgery 208 (2021) 106881
2
2. Materials and methods
2.1. Specimen collection and preparation
We included in this study a series of 11 patients with 5 unruptured
and 6 ruptured saccular IAs who underwent surgery in the Department
of Cerebrovascular Surgery of Saitama Medical University International
Medical Center in Hidaka, Japan (supplementary Table 1). The IA
samples were obtained by resecting a small part of the aneurysmal wall
intraoperatively after the clipping of the aneurysmal neck. The speci-
mens were immediately frozen on dry ice and stored at 80 C until
sectioning. All the specimens were sectioned at a 10 µm thickness using a
cryostat for DESI-IMS analysis.
2.2. DESI-IMS data acquisition from the IAs and standard lipids
Sectioned slides were kept at room temperature just before DESI-MS
acquisition. Mass spectra were acquired in the negative ion mode. All
the experiments were performed with the DESI source attached to a
quadrupole time-of-ight (Q-TOF) mass spectrometer (Xevo G2-XS Q-
TOF; Waters, Milford, MA, USA). The mass spectra were calibrated
externally prior to measurement using a sodium formate solution (500
µM) in 90:10 2-propanol-to-water ratio (v/v). The parameters used for
the optimization of DESI and acquisition of data from IA tissues are
given in supplementary Table 2. Tandem mass spectrometry (MS/MS)
using same instrument was performed to conrm the candidate mole-
cule corresponded to m/z 303.23 applying a collision energy of 10 eV,
source temperature 120º C, capillary voltage 4.0 kV, and 98% methanol
(98:2; methanol: water) as spray solvent at a ow rate of 2 µL/min. Two
standard FFAs (arachidonic acid and oleic acid) and those fatty acids
(FAs) containing standard lipids were also measured using the same MS
instrument and parameters (supplementary Table 2) to conrm whether
FAs were detected in IA walls as FFAs or fragment of lipids. For that
purpose, 1 µL solution of standard lipids (10 µg/mL in ethanol) was
applied on glass slide and acquired DESI-MS data.
2.3. Fold changes in FAs and lipid contents between ruptured and
unruptured IA walls
Fold changes (rupture IA vs unruptured IA walls) of detected FAs and
lipids were also analyzed using the average intensity of each FA and
lipid. In mass spectrometric measurement, noise is a common fact which
can hide MS peaks with small intensities. Therefore, intensities of noise
nearby the MS peaks of candidate lipids which were not detected in one
group of IA samples were used as their intensities and calculated their
fold changes [4].
2.4. Data analyses
The MassLynx 4.1 (Waters) software was used for data acquisition
and processing; The HDImaging (Waters) software was used for the
image analysis. The MS Excel and SPSS version 16 software were used
for the statistical analysis. All the values are expressed as mean ±
standard error of mean (SEM). Differences with P values <0.05 (two-
tailed t-test) were considered signicant.
2.5. Chemical and reagents
Liquid chromatography/MS-grade methanol, 2-propanol, and ultra-
pure water were purchased from Wako Pure Chemical Industries (Osaka,
Japan); leucine enkephalin was purchased from Waters (Germany); and
sodium formate, AA, oleic acid (OA), phosphatidylcholine (PC; 18:1/
18:1), and PC (20:4/20:4) were purchased from Sigma-Aldrich (St.
Louis, MO, USA).
2.6. Ethics committee approval
The institutional review board of Saitama Medical University Inter-
national Medical Center (No. 16223) and Hamamatsu University
School of Medicine (No. 17288) approved all aspects of the study.
Informed consent was obtained from all the patients.
3. Results
3.1. Distribution of FFAs in the IA walls on DESI-IMS
We rst analyzed the DESI-IMS mass spectra acquired from ruptured
and unruptured IA walls. Five MS peaks with m/z of 255.23, 279.23,
281.25, 283.26, and 303.23 were identied in the samples, corre-
sponding to palmitic acid (PA; C16:0), linoleic acid (LA; C18:2), OA
(C18:2), stearic acid (SA; C18:0), and AA (C20:4), respectively (sup-
plementary Fig. 1 and Supplementary Table 3) according to previous
reports [5,6]. AA was further conrmed by MS/MS analysis (Supple-
mentary Fig. 2). Using standard lipids, we have also conrmed that all
these FAs were detected as FFAs in this study (Supplementary Fig. 3).
The distribution patterns of PA, LA, OA, SA, and AA were analyzed in
both the ruptured and unruptured IA tissues (Fig. 1). AA was detected
only in the ruptured but not in the unruptured IA walls (Fig. 1A and B).
Moreover, accumulations of LA and AA in the same tissue region as the
ruptured IA walls were also observed (Fig. 1A and B). No signicant
change in the distribution of PA and SA was found between ruptured and
unruptured IA walls.
The average intensity of the distribution of FFAs were also analyzed
in this study. The distribution of LA and OA were increased in the
ruptured IA walls by 3.48-fold (P =0.003) and 2.85-fold (P =0.013),
respectively, as compared with the unruptured IA walls (Fig. 2A). In the
unruptured IAs, the amount of AA in the sample was less than the
detection sensitivity of the device, so it was not detected as MS peak
corresponding to AA. Compared with the noise levels in the unruptured
IA walls, an increase in AA level of approximately 300 times was
detected in the ruptured IA walls (Fig. 2B).
3.2. Distribution of AA-containing phospholipids in the IA walls on DESI-
IMS
We next analyzed the distribution of AA-containing phospholipids in
the IA walls using DESI-IMS. Four MS peaks with m/z of 766.53, 794.55,
810.53, and 885.55 were detected in the ruptured IA walls and assigned
to phosphatidylethanolamine (PE; 18:0/20:4), PE (20:0/20:4), phos-
phatidylserine (PS; 18:0/20:4), and phosphatidylinositol (PI; 18:0/
20:4), respectively, on the basis of their mass accuracy (Supplementary
Table 3), biological distributions, and data from previous reports [3,7].
Among these phospholipids, PE (20:0/20:4) and PI (18:0/20:4) were
observed in abundance especially in the walls of the ruptured IAs
(Fig. 3A). In the unruptured IAs, the amounts of AA containing phos-
pholipids in the sample were less than the detection sensitivity of the
device, so it was not detected as a peak corresponding to the phospho-
lipids (Fig. 3B). Compared with the noise levels of DESI-IMS data ac-
quired from unruptured IA walls, the intensity level of the distributions
of PE (18:0/20:4), PE (20:0/20:4), PS (18:0/20:4), and PI (18:0/20:4) in
the ruptured IA walls were increased by approximately 2.0-fold,
3.7-fold, 3.5-fold, and 5.5-fold, respectively (Fig. 3C).
The changes in the distribution of FFAs and phospholipids between
ruptured and unruptured IA walls were given by Supplementary Table 4.
3.3. Inspections of the IA walls by the immunohistology
We tried to examine the IA walls by immunohistochemical staining
method. The sectioned slides of the samples were stained by
hematoxylin-eosin (HE) staining and immunostaining including COX2.
It was difcult to interpret the results of the immunostaining because the
R. Takeda et al.
Clinical Neurology and Neurosurgery 208 (2021) 106881
3
condition of the slides was inappropriate for the immunostaining. Under
the HE staining, many blood cells were detected on ruptured IA walls
different from unruptured IA walls (Fig. 1).
4. Discussion
This study showed for the rst time that AA was not detected in the
unruptured human IA walls. Our ndings suggest that the stability of the
turnover of AA in human unruptured IA walls is sustained. Previous
studies have reported the elevated COX2 expression in the IA wall [7,8].
Additionally, IA is considered a chronic inammatory disease and is
conceptually the same as other chronic inammatory diseases such as
cancer, atherosclerosis, and impaired glucose tolerance [1]. Our nd-
ings may suggest that the inammatory responses in human IAs,
Fig. 1. Distribution of free fatty acids (FFAs) in the walls of ruptured (R) and unruptured (U) human intracranial aneurysms (IAs). A
,
Molecular ion images of FFAs in
the ruptured IA human walls. B
,
Molecular ion images of the FFAs in the unruptured human IA walls. ND indicates not detected. Scale bar: 0.5 mm. H&E:
hematoxylin-eosin. Palmitic acid (PA), linoleic acid (LA), oleic acid (OA) and stearic acid (SA) were detected in both ruptured and unruptured human IA walls, but
arachidonic acid (AA) was only detected in ruptured human IA walls.
Fig. 2. Differences in the intensity level of the distribution of free fatty acids (FFAs) between the ruptured and unruptured human intracranial aneurysm (IA) walls.
A, Average intensity of FFAs in the walls of ruptured and unruptured IAs walls (n =6 for ruptured IA and n =5 for unruptured IA). All data are presented as mean
±SEM. * *P =0.003 and *P =0.013 (two-tailed t-test). Signicantly increased distribution of oleic acid (OA) and linoleic acid (LA) in ruptured human IA walls
compared to that of unruptured human IA walls was noted in this study. No signicance change was found in the distribution of palmitic acid (PA) and stearic acid
(SA) between ruptured and unruptured IA walls. B
,
Ratio of the intensity of palmitic acid (PA) and arachidonic acid (AA) between the ruptured and unruptured IA
walls. The noise level in the unruptured IA walls was used as the intensity of AA in the unruptured IA walls. Compared to the unruptured IA walls, about 300-fold
increased AA was observed in ruptured IA walls.
R. Takeda et al.
Clinical Neurology and Neurosurgery 208 (2021) 106881
4
including the COX2 expression, is not induced from the stimulation that
AA regulates independent of its metabolites. Furthermore, considering
that AA has been detected enough and suggested to play an important
role in other inammatory disease [914], our ndings might suggest
that unruptured human IAs have a different inammatory association
from other inammatory diseases regarding the response of AA and
COX2.
Unlike the unruptured IA walls, high levels of AA were detected on
the ruptured IA walls. We also found high level of LA, precursor in AA
synthesis, in the ruptured IA walls, and accumulations of LA and AA in
the same tissue region as the ruptured IA walls were also observed.
Considering that not only AA but also the precursor of AA increased in
the ruptured IA, it may be possible that AA is associated to the pro-
gression and rupture of IA walls. However, according to the result that
many blood cells were also detected on ruptured IA walls by
hematoxylin-eosin staining, it seems reasonable to think that much of
the AA in the ruptured IAs is free AA caused by the platelets activated by
the rupture of the aneurysms. If that is the case, this nding suggests that
DESI-IMS is a useful tool for semi-quantication of the activation of
platelets in excised specimens. In this study, we also found high level of
OA. We cant have interpreted this result yet. The signicance of OA was
smaller than that of AA and LA, and more cases are necessary to interpret
this result.
We have previously demonstrated that PI (18:0/20.4) accumulated
at high levels in the thickened aneurysmal walls in experimentally
induced IA with synthetic dedifferentiated smooth muscle cells [3]. It is
uncertain why the current study demonstrated AA-containing phos-
pholipids including PI (18:0/20.4) at the noise levels in the human
unruptured IA walls. It might be due to the difference of the wall of the
aneurysms in each sample; the difference of the species and experi-
mentally induced or not. On the other hand, high levels of
AA-containing phospholipids including PI (18:0/20.4) were detected on
ruptured IA walls. PI (18:0/20:4) play key roles in a wide range of
cellular processes, including cell migration, invasion, and proliferation
[3]. PE also play important roles in the regulation of cell proliferation,
metabolism, organelle function, endocytosis, autophagy, stress
response, and apoptosis [15]. Therefore, our results might be a clue for
exploring the mechanism of rupture and hemostasis in IAs.
The sample size in this study was too small, and the human IA
samples, especially those from ruptured IAs, might have been already
modied by several factors, which might have affected the results of the
present study. Therefore, more cases and further analysis are necessary
to interpret our present results. However, our ndings might offer new
insights into the associations of AA metabolites, including COX2, with IA
walls in humans.
Fig. 3. Distribution of arachidonic acid (AA)-containing phospholipids in the walls of human intracranial aneurysm (IA)s. A, Molecular ion images of AA-containing
phospholipids in ruptured human IA walls. B, Molecular ion images of AA-containing phospholipids in unruptured human IA walls. ND indicates not detected. AA
containing phospholipids were only detected I ruptured IA walls. Scale bar: 0.5 mm. C, Ratio of the intensity of the AA-containing phospholipids between the
ruptured and unruptured IA walls. The noise level in the unruptured IA walls was used as the intensity of AA-containing phospholipids in the unruptured IA walls.
About 26 folds increased AA-containing phospholipids were detected in ruptured IA walls compared to that of unruptured IA walls. PE: Phosphatidylethanolamine;
PS: Phosphatidylserine; PI: Phosphatidylinositol.
R. Takeda et al.
Clinical Neurology and Neurosurgery 208 (2021) 106881
5
5. Conclusion
In this study, DESI-IMS revealed the changes in the distribution of
FFAs and lipids in the ruptured and unruptured IA walls in human
samples. We found that AA and AA-containing phospholipids were not
detected in the unruptured IA while they were detected in a signicantly
larger amount in the ruptured IA. These ndings may offer new insights
on the association of AA metabolites, including COX2, with IA walls in
humans.
Funding
This work was supported by JSPS KAKENHI Grant Numbers
17K10849 (to Dr. Takeda), 21K09188 (to Dr. Takeda), JP15H05898B1
(to Dr. Setou), and AMED Grant Number JP19gm0910004 (to Dr.
Setou).
CRediT authorship contribution statement
Ririko Takeda: Conceptualization, Methodology, Resources,
Writing original draft, Writing review & editing, Project adminis-
tration. Ariful Islam: Investigation, Formal analysis, Data curation,
Writing review & editing. Tomohito Sato: Validation, Formal anal-
ysis, Investigation, Data curation. Hiroki Kurita: Resources, Supervi-
sion. Tomoaki Kahyo: Validation, Investigation. Tetsumei Urano:
Supervision. Mitsutoshi Setou: Methodology, Project administration.
Declarations of interest
None.
Appendix A. Supporting information
Supplementary data associated with this article can be found in the
online version at doi:10.1016/j.clineuro.2021.106881.
References
[1] T. Aoki, J. Frȍsen, M. Fukuda, K. Bando, G. Shioi, K. Tsuji, E. Ollikainen, K. Nozaki,
J. Laakkonen, S. Narumiya, Prostaglandin E2-EP2-NF-κB signaling in macrophages
as a potential therapeutic target for intracranial aneurysms, Sci. Signal. 10 (2017)
eaah6037, https://doi.org/10.1126/scisignal.aah6037.
[2] R.J. Gewirtz, H.S. Dhillon, S.E. Goes, S.M. DeAtley, S.W. Scheff, Lactate and free
fatty acids after subarachnoid hemorrhage, Brain Res. 840 (1999) 8491, https://
doi.org/10.1016/s0006-8993(99)01752-7.
[3] T. Ikedo, M. Minami, H. Kataoka, K. Hayashi, M. Nagata, R. Fujikawa, F. Yamazaki,
M. Setou, M. Yokode, S. Miyamoto, Imaging mass spectroscopy delineates the
thinned and thickened walls of intracranial aneurysms, Biochem Biophys. Res
Commun. 495 (2018) 332338, https://doi.org/10.1016/j.bbrc.2017.10.133.
[4] J. Kast, M. Gentzel, M. Wilm, K. Richardson, Noise ltering techniques for
electrospray quadrupole time of ight mass spectra, J. Am. Soc. Mass Spectrom. 14
(7) (2003) 766776, https://doi.org/10.1016/S1044-0305(03)00264-2.
[5] M. Mamun, S. Sato, E. Naru, O. Sakata, E. Hoshikawa, A. Suzuki, A. Islam,
T. Kahyo, T. Sato, T.K. Ito, M. Horikawa, R. Fukui, K. Izumi, M. Setou, Higher
Accumulation of Docosahexaenoic Acid in the Vermilion of the Human Lip than in
the Skin, Int. J. Mol. Sci. 21 (2020) 2807, https://doi.org/10.3390/ijms21082807.
[6] E. Takeyama, A. Islam, N. Watanabe, H. Tsubaki, M. Fukushima, M. Mamun,
S. Sato, T. Sato, F. Eto, I. Yao, T.K. Ito, M. Horikawa, M. Setou, Dietary intake of
green nut oil or DHA ameliorates DHA distribution in the brain of a mouse model of
dementia accompanied by memory recovery, Nutrients 11 (2019) 2371, https://
doi.org/10.3390/nu11102371.
[7] J.S. Hudson, A.J. Marincovich, J.A. Roa, M. Zanaty, E.A. Samaniego, D.M. Hasan,
Aspirin and intracranial aneurysms: a critical review of the current evidence,
Stroke 50 (2019) 25912596, https://doi.org/10.1161/STROKEAHA.119.026094.
[8] J. Rodemerk, A. Junker, B. Chen, D. Pierscianek, P. Dammann, M. Darkwah
Oppong, A. Radbruch, M. Forsting, S. Maderwald, H.H. Quick, Y. Zhu, R. Jabbarli,
U. Sure, K.H. Wrede, Pathophysiology of intracranial aneurysms: COX-2
expression, iron deposition in aneurysm wall, and correlation with magnetic
resonance imaging, Stroke 51 (2020) 25052513, https://doi.org/10.1161/
STROKEAHA.120.030590.
[9] T. Hiraide, K. Ikegami, T. Sakaguchi, Y. Morita, T. Hayasaka, N. Masaki, M. Waki,
E. Sugiyama, S. Shinriki, M. Takeda, Y. Shibasaki, S. Miyazaki, H. Kikuchi,
H. Okuyama, M. Inoue, M. Setou, H. Konno, Accumulation of arachidonic acid-
containing phosphatidylinositol at the outer edge of colorectal cancer, Sci. Rep. 6
(2016) 29935, https://doi.org/10.1038/srep29935.
[10] M. Hughes-Fulford, C.F. Li, J. Boonyaratanakornkit, S. Sayyah, Arachidonic acid
activates phosphatidylinositol 3-kinase signaling and induces gene expression in
prostate cancer, Cancer Res. 66 (2006) 14271433, https://doi.org/10.1158/
0008-5472.CAN-05-0914.
[11] J. Qin, W. Wang, R. Zhang, Novel natural product therapeutics targeting both
inammation and cancer, Chin. J. Nat. Med. 15 (2017) 401416, https://doi.org/
10.1016/S1875-5364(17)30062-6.
[12] N.K.V. Gundala, V.G.M. Naidu, U.N. Das, Amelioration of streptozotocin-induced
type 2 diabetes mellitus in Wistar rats by arachidonic acid, Biochem Biophys. Res.
Commun. 496 (2018) 105113, https://doi.org/10.1016/j.bbrc.2018.01.007.
[13] F. Cipollone, M.L. Fazia, COX-2 and atherosclerosis, J. Cardiovasc Pharmacol. 47
(Suppl 1) (2006) S26S36, https://doi.org/10.1097/00005344-200605001-00006.
[14] T. Sato, M. Horikawa, S. Takei, F. Yamazaki, T.K. Ito, T. Kondo, T. Sakurai,
T. Kahyo, K. Ikegami, S. Sato, R. Sato, Y. Jinno, H. Kawano, S. Naoe, M. Arita,
Y. Kashiwagi, M. Setou, Preferential incorporation of administered
eicosapentaenoic acid into thin-cap atherosclerotic plaques, Arterioscler. Thromb.
Vasc. Biol. 39 (2019) 18021816, https://doi.org/10.1161/
ATVBAHA.119.313093.
[15] Y. Chen, Z. Ma, X. Shen, L. Li, J. Zhong, L.S. Min, L. Xu, H. Li, J. Zhang, L. Dai,
Serum lipidomics proling to identify biomarkers for non-small cell lung cancer,
Biomed. Res. Int. 2018 (2018), 5276240, https://doi.org/10.1155/2018/5276240
eCollection 2018.
R. Takeda et al.
... First, arachidonic acid (AA) is a polyunsaturated fatty acid that can transform into a range of inflammatory agents. However, a study identified that AA metabolism in the unruptured IA wall remains comparatively stable, possibly due to the relatively diminished oxidative stress and inflammation [66]. Next, the build-up of lipids plays a significant role in IA rupture. ...
Article
Full-text available
Many vascular diseases are linked to lipid metabolism disorders, which cause lipid accumulation and peroxidation in the vascular wall. These processes lead to degenerative changes in the vessel, such as phenotypic transformation of smooth muscle cells and dysfunction and apoptosis of endothelial cells. In intracranial aneurysms, the coexistence of lipid plaques is often observed, indicating localized lipid metabolism disorders. These disorders may impair the function of the vascular wall or result from it. We summarize the literature on the relationship between lipid metabolism disorders and intracranial aneurysms below.
... The Land cycle has the potential for rapid large-scale reactions. Previous reports have shown that phospholipase A2 releases substantial amounts of arachidonic acid and lysolecithin from phospholipids within seconds [18,19]. In our study, we also examine the levels of lysophospholipids in the kidneys after irradiation, which serve as a source for phospholipid synthesis via the Land cycle. ...
Article
Full-text available
To understand the ultra-early reaction of normal organ lipids during irradiation, we investigated the response of lipids, including polyunsaturated fatty acid (PUFA) chains, which are particularly susceptible to damage by ROS, in mice’s kidneys, lungs, brains, and livers within 5 min of single high-dose irradiation. In this study, we set up three groups of C56BL/6 male mice and conducted whole-body irradiation with 0 Gy, 10 Gy, and 20 Gy single doses. Kidney, lung, brain, and liver tissues were collected within 5 min of irradiation. PUFA-targeted and whole lipidomic analyses were conducted using liquid chromatography–tandem mass spectrometry (LC-MS/MS). The results showed that PUFA chains of kidney phosphatidylcholine (PC), phosphatidylethanolamine (PE), and triacylglycerol (TG) significantly increased within 5 min of 10 Gy and 20 Gy irradiation. The main components of increased PUFA chains in PC and PE were C18:2, C20:4, and C22:6, and in TG the main component was C18:2. The kidney lipidomes also showed significant changes from the perspective of lipid species, mainly dominated by an increase in PC, PE, TG, and signal lipids, while lipidomes of the lung, brain, and liver were slightly changed. Our results revealed that acute PUFA chains increase and other lipidomic changes in the kidney upon whole-body irradiation within 5 min of irradiation. The significantly increased lipids also showed a consistent preference for possessing PUFA chains. The lipidomic changes varied from organ to organ, which indicates that the response upon irradiation within a short time is tissue-specific.
... In this paper, we identified two molecules, PS(35:5) (m/z 766.47) and PE-NMe(38:5) (m/z 778.54), to illustrate the proposed method of identifying relevant molecules via entropy heat maps. The results are consistent with the findings in our previous report [32]. In the present study, the 31-month kidney had more PE-NMe(38:5) than the 3-month kidney. ...
Article
Full-text available
Mass spectrometry imaging (MSI) allows us to visualize the spatial distribution of molecular components in a sample. A large amount of mass spectrometry data comprehensively provides molecular distributions. In this study, we focus on the information in the obtained data and use the Shannon entropy as a quantity to analyze MSI data. By calculating the Shannon entropy at each pixel on a sample, the spatial distribution of the Shannon entropy is obtained from MSI data. We found that low-entropy pixels in entropy heat maps for kidneys of mice had different structures between two ages (3 months and 31 months). Such changes cannot be visualized by conventional imaging techniques. We further propose a method to find informative molecules. As a demonstration of the proposed scheme, we identified two molecules by setting a region of interest which contained low-entropy pixels and by exploring changes of peaks in the region.
... Due to not using matrix molecules such as MALDI-MSI, it is also free from ion suppression due to matrix-derived peaks at lower m/z regions [21]. It can detect analytes as intact molecular ions due to not using ion beams or laser power [22]. After optimizing DESI-MSI parameters, we acquired DESI-MSI data at the usual speed (2 pixels/s) [5] and a little higher speed (8 pixels/s) from standard imipramine and chloroquine. ...
Article
Full-text available
Mass spectrometry imaging (MSI) is well-known for the non-labeling visualization of analytes, including drugs and their metabolites in biological samples. In this study, we applied three different tools of MSI, desorption electrospray ionization (DESI)-MSI, matrix-assisted laser desorption ionization (MALDI)-MSI, and a newly developed atmospheric pressure (AP)-MALDI-MSI known as iMScopeTM QT for rapid mapping of imipramine, chloroquine, and their metabolites in C57BL/6 male wild-type mice. Among three MSI tools, better detection capability for targeted drugs at higher speed (up to 32 pixels/s) was observed in iMScope QT. It revealed that imipramine and its metabolites were significantly accumulated in the renal cortex of mice, but chloroquine and its metabolites were highly accumulated in the renal pelvis and renal medulla of mice. Additionally, a higher accumulation of imipramine was noted in the thalamus, hypothalamus, septum, and hindbrain of mice brains. However, chloroquine and its metabolites showed notable accumulation in the lateral ventricle, fourth ventricle, and fornix of the mice brains. These findings of our study can be helpful in understanding clinically relevant properties, efficacy, and potential side effects of these drugs. Our study also showed the potentiality of iMScope QT for rapid mapping of small drugs and their metabolites in biological samples.
... DESI-IMS data were acquired by using Xevo G2-XS quadrupole time-of-flight (Q-TOF) mass spectrometer (Waters, Milford, MA, USA) equipped with a 2D DESI source in negative ion mode following previously described method with slight modification. 12 Prior to measurement, DESI-IMS mass spectra were calibrated using 500 μM sodium formate solution prepared in 90% 2-propanol. As spray solvent, 98% methanol was delivered at a flow rate of 3 μL/min using ACQUITY UPLC binary solvent manager (Waters, Milford, MA, USA). ...
Article
The characteristic patterns of mass spectra in imaging mass spectrometry (IMS) strongly reflect the tissue environment. However, the boundaries formed where different tissue environments collide have not been visually assessed. In this study, IMS and convolutional neural network (CNN), one of the deep learning methods, were applied to the extraction of characteristic mass spectra patterns from training brain regions on rodents' brain sections. CNN produced classification models with high accuracy and low loss rate in any test data sets of mouse coronal sections measured by desorption electrospray ionization (DESI)-IMS and of mouse and rat sagittal sections by matrix-assisted laser desorption (MALDI)-IMS. On the basis of the extracted mass spectra pattern features, the histologically plausible segmentation and classification score imaging of the brain sections were obtained. The boundary imaging generated from classification scores showed the extreme changes of mass spectra patterns between the tissue environments, with no significant buffer zones for the intermediate state. The CNN-based analysis of IMS data is a useful tool for visually assessing the changes of mass spectra patterns on a tissue section, and it will contribute to a comprehensive view of the tissue environment.
... To reveal the effects of stress and GNO supplementation on 2AG levels in the different brain regions of SAMP8 mice for the first time, we applied desorption electrospray ionization-mass spectrometry imaging (DESI-MSI). DESI-MSI is a recently developed tool of MSI widely used for the detection and visualization of drugs [16], small metabolites [17], neurotransmitters [18], lipids [19], and proteins [20] under ambient conditions [21]. Recently, Nielsen et al. also reported the capability of DESI-MSI to detect 2AG with higher sensitivity [22]. ...
Article
Full-text available
The endocannabinoid 2-arachidonoylglycerol (2AG) is an important modulator of stress responses. Its level in the brain increases in response to stress, but region-specific effects of stress on brain 2AG are not well known yet. Moreover, green nut oil (GNO), oil extracted from the seeds of Plukenetia volubilis has several health benefits, but its effects on brain 2AG levels are unknown. Therefore, we conducted this study to explore the effects of stress and GNO supplementation on 2AG levels in specific brain regions of senescence-accelerated mouse prone 8 (SAMP8). In this study, desorption electrospray ionization-mass spectrometry imaging (DESI-MSI) revealed that water-immersion stress for three days significantly increased 2AG levels in several brain regions of SAMP8 mice, including the hypothalamus, midbrain, and hindbrain. No significant change was observed in the relative abundance of brain 2AG in stress given SAMP8 mice after eighteen days of removing stress load compared to control SAMP8 mice. GNO supplementation also increased brain 2AG in SAMP8 mice without stress load. Additionally, GNO supplementation sustained the increased brain 2AG levels in stress given SAMP8 mice after eighteen days of removing stress load. Among all brain regions, a relatively higher accumulation of 2AG was noted in the hypothalamus, midbrain, and hindbrain of GNO-fed SAMP8. Our data explored the potentiality of GNO supplementation to improve brain 2AG levels which might be used to treat anxiety and depressive behaviors.
Article
Full-text available
Accepted version: https://rodemerk.com/publications/original-research/aneurysm_cox-2/ Background and purpose: The pathophysiology of development, growth, and rupture of intracranial aneurysms (IAs) is only partly understood. Cyclooxygenase 2 (COX-2) converts arachidonic acid to prostaglandin H2, which, in turn, is isomerized to prostaglandin E2. In the human body, COX-2 plays an essential role in inflammatory pathways. This explorative study aimed to investigate COX-2 expression in the wall of IAs and its correlation to image features in clinical (1.0T, 1.5T, and 3.0T) magnetic resonance imaging (MRI) and ultra-high-field 7T MRI. Methods: The study group comprised 40 patients with partly thrombosed saccular IAs. The cohort included 17 ruptured- and 24 unruptured IAs, which had all been treated microsurgically. Formaldehyde-fixed paraffin-embedded samples were immunohistochemically stained with a monoclonal antibody against COX-2 (Dako, Santa Clara, CA; Clone: CX-294). We correlated Perls Prussian blue staining, MRI, and clinical data with immunohistochemistry, analyzed using the Trainable Weka Segmentation algorithm. Results: Aneurysm dome size ranged between 2 and 67 mm. The proportion of COX-2 positive cells ranged between 3.54% to 85.09%. An upregulated COX-2 expression correlated with increasing IA dome size (P=0.047). Furthermore, there was a tendency of higher COX-2 expression in most ruptured IAs (P=0.064). At all field strengths, MRI shows wall hypointensities due to iron deposition correlating with COX-2 expression (P=0.022). Conclusions: Iron deposition and COX-2 expression in IAs walls correlate with signal hypointensity in MRI, which might, therefore, serve as a biomarker for IA instability. Furthermore, as COX-2 was also expressed in small unruptured IAs, it could be a potential target for specific medical treatment.
Article
Full-text available
The vermilion of the human lip is a unique facial area because of certain distinguishing features from the adjacent tissues such as the white lip (skin) and oral mucosa. However, the distinction in terms of molecular distribution between the vermilion and skin has remained unexplored. Therefore, we aimed to map the human lip by mass spectrometry imaging to gain understanding of the free fatty acid distribution in the vermilion. The lip specimens trimmed off during cheiloplasty were analyzed using desorption electrospray ionization–mass spectrometry imaging. Distributions of two monounsaturated fatty acids and three polyunsaturated fatty acids were observed in the human lip tissue: palmitoleic acid (POA) and oleic acid (OA) and linoleic acid (LA), arachidonic acid (AA), and docosahexaenoic acid (DHA), respectively. Although POA, OA, LA, and AA were differentially distributed across the vermilion and skin, DHA showed a higher accumulation in the epithelium of the vermilion compared to that in the skin. Our results clearly demonstrated the difference in fatty acid distributions between the vermilion and skin. The highly abundant DHA in the epithelium of the vermilion may have an antioxidant role and may thus protect the lip from aging. Our findings can provide a novel strategy for treating lip disorders.
Article
Full-text available
Docosahexaenoic acid (DHA), an omega-3 polyunsaturated fatty acid, has significant healthbenefits. Previous studies reported decreased levels of DHA and DHA-containing phosphatidylcholines inthe brain of animals suffering from Alzheimer’s disease, the most common type of dementia; furthermore,DHA supplementation has been found to improve brain DHA levels and memory efficiency in dementia. Oilextracted from the seeds of Plukenetia volubilis (green nut oil; GNO) is also expected to have DHA like effectsas it contains approximately 50% α-linolenic acid, a precursor of DHA. Despite this, changes in the spatialdistribution of DHA in the brain of animals with dementia following GNO or DHA supplementation remainunexplored. In this study, desorption electrospray ionization imaging mass spectrometry (DESI-IMS) wasapplied to observe the effects of GNO or DHA supplementation upon the distribution of DHA in the brain ofmale senescence-accelerated mouse-prone 8 (SAMP8) mice, a mouse model of dementia. DESI-IMS revealedthat brain DHA distribution increased 1.85-fold and 3.67-fold in GNO-fed and DHA-fed SAMP8 mice,respectively, compared to corn oil-fed SAMP8 mice. Memory efficiency in SAMP8 mice was also improvedby GNO or DHA supplementation. In summary, this study suggests the possibility of GNO or DHAsupplementation for the prevention of dementia.
Article
Full-text available
Objective: n-3 polyunsaturated fatty acids, especially eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), have beneficial effects on atherosclerosis. Although specific salutary actions have been reported, the detailed distribution of n-3 polyunsaturated fatty acids in plaque and their relevance in disease progression are unclear. Our aim was to assess the pharmacodynamics of EPA and DHA and their metabolites in atherosclerotic plaques. Approach and Results: Apolipoprotein E-deficient (Apoe-/-) mice were fed a western diet supplemented with EPA (1%, w/w) or DHA (1%, w/w) for 3 weeks. Imaging mass spectrometry analyses were performed in the aortic root and arch of the Apoe-/- mice to evaluate the distribution of EPA, DHA, their metabolites and the lipids containing EPA or DHA in the plaques. Liquid chromatography-mass spectrometry and histological analysis were also performed. The intima-media thickness of atherosclerotic plaque decreased in plaques containing freer EPA and EPAs attached with several lipids. EPA was distributed more densely in the thin-cap plaques than in the thick-cap plaques, while DHA was more evenly distributed. In the aortic root, the distribution of total EPA level and cholesteryl esters containing EPA followed a concentration gradient from the vascular endothelium to the media. In the aortic arch, free EPA and 12-hydroxy-EPA colocalized with M2 macrophage. Conclusions: Administered EPA tends to be incorporated from the vascular lumen side and preferentially taken into the thin-cap plaque.
Article
Full-text available
Non-small cell lung cancer (NSCLC) is the leading cause of cancer death worldwide, which ranks top in both incidence and mortality. To broaden our understanding of the lipid metabolic alterations in NSCLC and to identify potential biomarkers for early diagnosis, we performed nontargeted lipidomics analysis in serum from 66 early-stage NSCLC, 40 lung benign disease patients (LBD), and 40 healthy controls (HC) using Ultrahigh Performance Liquid Chromatography-Quadrupole Time-of-Flight Mass Spectrometry (UHPLC-Q-TOF/MS). The identified biomarker candidates of phosphatidylcholines (PCs) and phosphatidylethanolamines (PEs) were further externally validated in a cohort including 30 early-stage NSCLC, 30 LBD, and 30 HC by a targeted lipidomic analysis. We observed a significantly altered lipid metabolic profile in early-stage NSCLC and identified panels of PCs and PEs to distinguish NSCLC patients and HC. The levels of PCs and PEs were found to be dysregulated in glycerophospholipid metabolism, which was the top altered pathway in early-stage NSCLC. Receiver operating characteristic (ROC) curve analysis revealed that panels of PCs and PEs exhibited good performance in differentiating early-stage NSCLC and HC. The levels of PE(16:0/16:1), PE(16:0/18:3), PE(16:0/18:2), PE(18:0/16:0), PE(17:0/18:2), PE(18:0/17:1), PE(17:0/18:1), PE(20:5/16:0), PE(18:0/18:1), PE(18:1/20:4), PE(18:0/20:3), PC(15:0/18:1), PC(16:1/20:5), and PC(18:0/20:1) in early-stage NSCLC were significantly increased compared with HC (p<0.05). Overall, our study has thus highlighted the power of using comprehensive lipidomic approaches to identify biomarkers and underlying mechanisms in NSCLC.
Article
Traditionally arachidonic acid (AA, 20:4 n-6) is considered as a pro-inflammatory molecule since it forms precursor to prostaglandins (PGs), leukotrienes (LTs) and thromboxanes (TXs) that have pro-inflammatory actions. Type 2 diabetes mellitus (type 2 DM) is considered as a low-grade systemic inflammatory condition in which circulating PGs and LTs are increased. Streptozotocin (STZ)-induced type 2 DM is used as a model of human type 2 DM in which peripheral insulin resistance, increased plasma interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) and hyperglycemia occurs. In the present study, we observed that oral supplementation of AA prevented STZ-induced type 2 DM in Wistar rats by restoring hyperglycemia, plasma levels of TNF-α and IL-6; adipose tissue NF-kB and lipocalin 2 (LPCLN2) and pancreatic tissue NF-kB and 5- and 12- lipoxygenase enzymes to normal. AA treatment enhanced insulin sensitivity and plasma lipoxin A4 (LXA4) levels, a potent anti-inflammatory molecule derived from AA. These results are supported by our previous studies wherein it was noted that plasma phospholipid content of AA and circulating LXA4 levels are low in those with type 2 DM. In a preliminary study, we also noted that high-fat-diet (HFD)-induced type 2 DM in Wistar rats can be prevented by oral supplementation of AA. These results suggest AA has anti-inflammatory and anti-diabetic actions by enhancing the production of its anti-inflammatory metabolite LXA4.
Article
Object: The wall thickness of intracranial aneurysms (IAs) is heterogeneous. Although thinning of the IA wall is thought to contribute to IA rupture, the underlying mechanism remains poorly understood. Recently, imaging mass spectroscopy (IMS) has been used to reveal the distribution of phospholipids in vascular diseases. To investigate the feature of phospholipid composition of IA walls, we conducted IMS in a rat model of experimentally induced IA. Material and methods: IAs were surgically induced in 7-week-old male rats and analyzed by IMS in negative-ion mode. Results: A molecule at m/z 885.5 was more abundant in the thickened wall than in the thinned wall (P = 0.03). Multiple-stage mass spectroscopy revealed the molecule to be phosphatidylinositol containing stearic acid and arachidonic acid (PI 18:0/20:4). Immunohistochemistry indicated that vascular smooth muscle cells (SMCs) in the thickened wall had dedifferentiated phenotypes. To investigate the relationship between accumulation of PI (18:0/20:4) and phenotypic changes in SMCs, we subjected primary mouse aortic SMCs to liquid chromatography-tandem mass spectrometry. Notably, dedifferentiated SMCs had 1.3-fold more PI (18:0/20:4) than partly differentiated SMCs. Conclusions: Our study demonstrated the heterogeneity in phospholipid composition of the aneurysmal walls using experimentally induced IAs. PI (18:0/20:4) accumulated at high levels in the thickened aneurysmal wall where synthetic dedifferentiated SMCs exist, suggesting that this phospholipid may be involved in the phenotypic switching of medial SMCs in the IA wall.
Article
Inflammation is recently recognized as one of the hallmarks of human cancer. Chronic inflammatory response plays a critical role in cancer development, progression, metastasis, and resistance to chemotherapy. Conversely, the oncogenic aberrations also generate an inflammatory microenvironment, enabling the development and progression of cancer. The molecular mechanisms of action that are responsible for inflammatory cancer and cancer-associated inflammation are not fully understood due to the complex crosstalk between oncogenic and pro-inflammatory genes. However, molecular mediators that regulate both inflammation and cancer, such as NF-κB and STAT have been considered as promising targets for preventing and treating these diseases. Recent works have further demonstrated an important role of oncogenes (e.g., NFAT1, MDM2) and tumor suppressor genes (e.g., p53) in cancer-related inflammation. Natural products that target these molecular mediators have shown anticancer and anti-inflammatory activities in preclinical and clinical studies. Sesquiterpenoids (STs), a class of novel plant-derived secondary metabolites have attracted great interest in recent years because of their diversity in chemical structures and pharmacological activities. At present, we and other investigators have found that dimeric sesquiterpenoids (DSTs) may exert enhanced activity and binding affinity to molecular targets due to the increased number of alkylating centers and improved conformational flexibility and lipophilicity. Here, we focus our discussion on the activities and mechanisms of action of STs and DSTs in treating inflammation and cancer as well as their structure-activity relationships.
Article
Intracranial aneurysms are common but are generally untreated, and their rupture can lead to subarachnoid hemorrhage. Because of the poor prognosis associated with subarachnoid hemorrhage, preventing the progression of intracranial aneurysms is critically important. Intracranial aneurysms are caused by chronic inflammation of the arterial wall due to macrophage infiltration triggered by monocyte chemoattractant protein-1 (MCP-1), macrophage activation mediated by the transcription factor nuclear factor κB (NF-κB), and inflammatory signaling involving prostaglandin E2 (PGE2) and prostaglandin E receptor subtype 2 (EP2). We correlated EP2 and cyclooxygenase-2 (COX-2) with macrophage infiltration in human intracranial aneurysm lesions. Monitoring the spatiotemporal pattern of NF-κB activation during intracranial aneurysm development in mice showed that NF-κB was first activated in macrophages in the adventitia and in endothelial cells and, subsequently, in the entire arterial wall. Mice with a macrophage-specific deletion of Ptger2 (which encodes EP2) or macrophage-specific expression of an IκBα mutant that restricts NF-κB activation had fewer intracranial aneurysms with reduced macrophage infiltration and NF-κB activation. In cultured cells, EP2 signaling cooperated with tumor necrosis factor–α (TNF-α) to activate NF-κB and synergistically induce the expression of proinflammatory genes, including Ptgs2 (encoding COX-2). EP2 signaling also stabilized Ccl2 (encoding MCP-1) by activating the RNA-stabilizing protein HuR. Rats administered an EP2 antagonist had reduced macrophage infiltration and intracranial aneurysm formation and progression. This signaling pathway in macrophages thus facilitates intracranial aneurysm development by amplifying inflammation in intracranial arteries. These results indicate that EP2 antagonists may therefore be a therapeutic alternative to surgery.