You are on page 1of 8

Article

Cite This: Anal. Chem. 2018, 90, 1317−1324 pubs.acs.org/ac

Visualizing the Regulation of Hydroxyl Radical Level by Superoxide


Dismutase via a Specific Molecular Probe
Lingyu Zeng,† Tian Xia,§ Wei Hu,† Shiyu Chen,∥ Siyu Chi,† Yidi Lei,† and Zhihong Liu*,†

Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular
Sciences, Wuhan University, Wuhan 430072, China
§
College of Life Science, Wuhan University, Wuhan 430072, China

Department of Chemistry, New York University, New York, New York 10003, United States
*
S Supporting Information

ABSTRACT: Hydroxyl radical (·OH), as the most delete-


rious reactive oxygen species, is believed to be the etiological
agent for many diseases and aging. An altered ·OH level has
been confirmed in certain types of superoxide dismutase
(SOD) mutation, but the regulation of ·OH by SOD in situ is
still controversial or unclear because of the lack of effective
tools to detect ·OH in the biological environment. Herein we
report the first two-photon excitable molecular probe (P2) for
·OH, which is able to track the subtle fluctuation of ·OH level
both in vitro and in vivo with high sensitivity and specificity.
The probe was successfully applied to visualize ·OH variations
in a variety of SOD1-involved biological processes, confirming
that the inhibited enzymatic activity and down-regulated
expression of SOD1 both lead to elevated intracellular ·OH
level. This is the first report to visually reveal the relationship between SOD1 and ·OH level with a molecular tool.

fluorescent probes for bioimaging can be constructed from


S uperoxide dismutase (SOD) plays a pivotal role in
balancing the concentration of reactive oxygen species
(ROS) as the first enzymatic defense against superoxide, which
organic small molecules, nanomaterials, and metal complexes
(Table S1). Small-molecule probes, known for their favorable
acts as a sink for many other radicals generated intra- biocompatibility, are mainly based on ·OH-induced hydrox-
cellularly.1,2 Among them, hydroxyl radicals (·OH) are ylation of aromatic ring,17−22 hydrogen atom abstraction,23−25
considered as the leading cause of oxidative stress damage, or adduction with a radical trap.10,26−28 Although some efforts
which attacks virtually all types of biomacromolecules and have been made to image ·OH in living cells under
causes irreparable cellular damage.3 Previous studies have physiological or pathological processes like inflamma-
reported that such a highly toxic species can be accumulated by tion,10,23−27,29 there is still plenty of room for improving the
the mutation of Cu, Zn−SOD (SOD1).4 Although increasing performance of probes. (e.g., the problems of limited
explorations have confirmed the coordination between SOD1 selectivity, auto-oxidation, photobleaching, etc.13,30)
and ·OH, some important issues concerning the regulation of · To circumvent the existing problems for in vivo ·OH
OH by SOD1 still remain controversial or unclear.3,5−9 To detection and help to elucidate the connection between SOD1
gain in-depth understanding on the relationship between the enzyme and intracellular ·OH levels, herein we present a
enzymatic activity and ·OH level, more direct experimental rationally designed ·OH-specific two-photon fluorescent probe
evidence is required. with high sensitivity and pronounced specificity. The reason to
It is widely accepted that ·OH detection in living systems is incorporate a two-photon excitable fluorophore is to achieve
highly challenging because of its short lifetime, small diffusion better time/space resolution by two-photon microscopy
distance, extremely low concentration, and the interference (TPM),31 which is particularly favorable for species that are
from other ROS.10−13 Currently, the major method for ·OH difficult to investigate in vivo. With the assistance of this
detection in a biosystem is electron paramagnetic resonance molecular probe, we tracked ·OH levels in vitro and in vivo.
(EPR) method with a spin trapper.8,14,15 Fluorescent probes, Furthermore, we realized the visualization of ·OH in some
by contrast, have advantages and potential prospect for SOD1-involved biological processes for the first time,
monitoring species in situ in the body, such as the relatively
inexpensive equipment and easy operation, high selectivity and Received: October 12, 2017
sensitivity, and feasible real-time imaging with good space Accepted: December 14, 2017
resolution by microscopy.16 Basically, the ·OH-specific Published: December 14, 2017

© 2017 American Chemical Society 1317 DOI: 10.1021/acs.analchem.7b04191


Anal. Chem. 2018, 90, 1317−1324
Analytical Chemistry Article

confirming that the inhibited enzyme activity and down- internal PMTs were used to collect the signals in an 8 bit
regulated expression of SOD1 could contribute to escalated unsigned 1024 × 1024 pixels at 1.58 μs/pixel scan speed. Cells
intracellular ·OH level. were observed under EC Plan-Neofluar 100 × /1.3 oil

■ EXPERIMENTAL SECTION
Chemical Synthesis. Briefly, the probes P1−P4 were
objective.
The mean fluorescence density was measured by Image-Pro
Plus (v. 6.0) and calculated via the equation (mean density =
synthesized by Pd-catalyzed C−N cross-coupling reactions. IODsum/areasum), where IOD and area were integral optical
For P1/P3 (R1 = Me), compound 1 was reacted with density and area of fluorescent region.
secondary amine catalyzed by RuPhos and Pd(OAc)2 in E. coli Infection. E. coli was grown in antibiotic-free Luria
anhydrous t-BuOH using CsCO3 for basic condition. For P2/ broth (LB) medium at 37 °C for 18 h. Then the bacteria
P4 (R2 = Me), BrettPhos was used instead of RuPhos. Then suspension was divided into several 1.5 mL Eppendorf tubes,
the nitro-group was reduced by N2H4·H2O and Pd/C (for P1/ and the number of E. coli was determined by O.D.600 nm (ca. 2
P4) or the Boc-group was deprotected by trifluoroacetic acid × 109 E. coli/tube). Next the bacteria were harvested by
(for P2/P3). The characterization of 1H NMR, 13C NMR, centrifugation and resuspended in 100 μL of sterile PBS by
HRMS spectra, and detailed procedures for each probe are rigorous vortex and pipetting, following heat treatment by 90
available in Supporting Information (Figures S15−S34). °C water bath for 1 h. When the RAW 264.7 cells had grown
Synthesis of 1-(9,9-Dimethyl-7-((4-(methylamino)- to 2 × 105 cells/well in the confocal dish, the culture medium
phenyl)amino)-9H-fluoren-2-yl)ethan-1-one (P2). Com- was replaced by 1 mL of fresh medium (DMEM, 1%
pound 5 (150 mg, 0.33 mmol) was dissolved in anhydrous penicillin/streptomycin, 10% FBS) containing 1 μL of E. coli
DCM. Under an ice−water bath, 1 mL of trifluoroacetic acid suspension (100 moi as final). After certain time of infection,
was added dropwise into the above mixture and reacted at
the cells were washed twice with PBS and then labeled with P2.
room temperature for 24 h. Then the reaction was quenched
SOD1-Overexpressing Cells. The SOD1 mammalian
by water, extracted with DCM, washed with brine, and dried
with anhydrous Na2SO4. The solution was concentrated and expression plasmid was prepared from cDNA clone by
then purified by silica gel column chromatography using PE/ standard molecular biology techniques. HeLa cells were
EA (6/1, v/v) and gave a yellow solid. (68 mg, 58% yield) 1H divided 24 h before experiment to generate 50% confluence
NMR (400 MHz, DMSO-d6) δ 8.01 (s, 1H), 7.93 (d, J = 17.3 at the time of transfection. The concentration of empty vector
Hz, 2H), 7.67 (dd, J = 20.4, 8.1 Hz, 2H), 7.03−6.91 (m, 3H), and plasmid were determined by a microplate reader and then
6.79 (d, J = 10.4 Hz, 1H), 6.57 (s, 2H), 5.48 (s, 1H), 2.68 (d, J were diluted to 0.2 mg/mL stocking solution. Then 0.4 μg
= 3.9 Hz, 3H), 2.60 (s, 3H), 1.41 (s, 6H). 13C NMR (100 vector and the constructed plasmids were separately mixed
MHz, DMSO-d6) δ 197.70, 156.96, 152.79, 148.44, 146.47, with 50 μL of Opti-MEM (Gibco) and 3 μL of Lipofectamine
144.95, 134.17, 131.05, 128.81, 126.99, 123.59, 122.74, 122.49, 2000 (Invitrogen), resulting in a total volume of 100 μL. The
118.42, 113.12, 112.83, 107.60, 46.61, 30.63, 27.43, 27.19. cell medium was replaced by 500 μL of Opti-MEM, and then
HRMS (DART): calcd for C24H25ON2 [M + H]+, 357.1961; the as-prepared mixture was added dropwise. The culture
found, 357.1961. medium was replaced with fresh medium (DMEM, 1%
General Procedure for In Vitro Imaging Experiment. penicillin/streptomycin, 10% FBS) after 6 h. After 24 h, the
HeLa cells and RAW 264.7 macrophage cells were cultured in two groups of cells were treated with 2 μg/mL PMA for 2 h.
Dulbecco’s modified Eagle’s medium (DMEM, Thermo Then the four groups were loaded with P2. After bioimaging,
Scientific) supplemented with 1% penicillin/streptomycin the RNA in above the samples was extracted by Trizol
and 10% fetal bovine serum (FBS) and incubated in an (Takara), and then the SOD1 mRNA level was determined by
atmosphere of 5/95 (v/v) of CO2/air at 37 °C. Human qRT-PCR.
umbilical vein endothelial cells (HUEVCs) were cultured in RNAi Cells. Chemically synthesized siRNA oligo was
RPMI Medium 1640 basic (Gibco), with 1% penicillin/ designed with minor reversion as literature reported
streptomycin and 10% FBS at the same condition. Before sequence,32 and manufactured by GenePharma. The siRNA
imaging, the cells were passed and plated into 35 mm glass- sequences were as follows, and the scramble control was a
bottomed confocal dish (NEST) and were allowed to grow to universal negative sequence.
the desired cell density. Sense sequence (5′-3′): GGATGAAGAGAGGCATGTTTT
TEMPOL, GSH, LPS (lot no. L2880), and PMA were
Antisense sequence (5′-3′): AACATGCCTCTCTT-
purchased from Sigma-Aldrich. ATN-244 (Bis(choline)-
CATCCTT
tetrathiomolybdate) was gained from MedChem Express.
Two groups of HUEVCs were divided 24 h prior to
Interferon-gamma (IFN-γ) was obtained from GenScript (lot
no. Z02916). MitoTracker Red was purchased from Life transfection to gain 50% confluence, and then the culture
Technology. medium was replaced by 500 μL of Opti-MEM before
Before introducing the reagents, the cells were washed once experiment. The siRNA oligo (1 OD) was dissolved in 125
by warm PBS and replenished by fresh medium containing μL of DEPC water, and then the cells were transfected by
certain drugs. Unless otherwise noted, all the probe-loading adding the mixture of 20 μL of siRNA solution, 200 μL of
steps used 10 μM P2 (medium containing 0.2% DMF) for 30 Opti-MEM, and 16 μL of Lipofectamine 2000. Then the
min incubation under the same condition for cell culture. culture medium was replaced by fresh medium (DMEM, 1%
Before imaging, the culture medium was removed and the cells penicillin/streptomycin, 10% FBS) after 6 h. After 36 h of
were washed twice by PBS. transfection, the two groups of cells were loaded with P2. After
The two-photon fluorescence microscopy images for living bioimaging, the RNA in the above samples was extracted by
cells were obtained by exciting the P2 with 740 nm excitation, Trizol (Takara), and then the SOD1 mRNA level was
and emission was collected from 430−680 nm range. The determined by qRT-PCR.
1318 DOI: 10.1021/acs.analchem.7b04191
Anal. Chem. 2018, 90, 1317−1324
Analytical Chemistry Article

■ RESULTS AND DISCUSSION


Design and Screening of ·OH-Specific Two-Photon
Fluorescent Probe. To design a reaction-based fluorescent
probe for ·OH, utilizing the susceptibility of the recognition
domain to oxidation is a common strategy. The pioneering
work of Nagano on ·OH probe utilized the oxidative O-
dearylation mechanism,33 in which the aryloxy group was
removed by an ipso-substitution reaction, while other highly
reactive species can cause a response as well (Scheme 1a).13
Figure 1. (a) Fluorescence enhancement of 10 μM P1−P4 in the
presence of increasing amount (0.1−10 equiv) of ·OH. (b)
Scheme 1. (a) Reported Sensing Mechanism of hROS Probe Fluorescence spectra of the ·OH-probe reaction products (with 1
Based on the Oxidative O-Dearylation Mechanism.33 (b) equiv of ·OH), 10, and 11. Inset: the photos of reaction systems
Our Design of the Two-Photon Fluorescent Probes for under 365 nm UV lamp. -: probe alone; + : the reaction system of 1
Specific Detection of ·OH. (c) Proposed Fluorescent equiv of ·OH and probe. Fluorescence intensity (F) for P1/P3 was
Products after ·OH-Probe Reaction gained from the emission peak at 560 nm under 360 nm excitation,
and F of P2/P4 were obtained from emission at 550 nm under 350
nm excitation.

which further verifies the proposed mechanism depicted in


Scheme S2.
We next examined the specificity of probes P1−P4, judging
by their response toward hypochlorous acid (HClO) because it
often has strong interference to the detection of ·OH. As
shown in Figure S2, P2 and P4 were inert to HClO but P1 and
P3 showed obvious response (∼100-fold fluorescence
enhancement). Then we selected P2 and P3 and traced their
reactions with HClO by LC-MS. It was found that P3 could
react with HClO to produce the fluorescent compound 11
(Figure S1h), while P2 did not react (Figure S1d). We
We thus thought to change the O-linkage to N-linkage, using attributed the HClO-responsibility of P1/P3 to N-chlorination
an amine-derived fluorophore with electron-rich aromatics as onto the nitrogen atom linked with R1 (proposed mechanism
the recognition group. Moreover, we infer that the released shown in Scheme S3). Taking the sensitivity and specificity
fluorophore after the probe-target reaction, which has a typical into consideration, it is obvious that P2 possesses the best
electronic structure, will possess good two-photon fluorescence comprehensive properties. We further checked the pH
properties.31 Bearing these considerations in mind, we dependence of P2 in the range of pH 4.0−8.0. The probe
designed a series of probes P1−P4 (Scheme 1b), using the had very low fluorescence in the whole pH range studied, while
p-phenylenediamine derivative as the recognition group and its reaction product with ·OH, compound 10, displayed strong
fluorene moiety as the fluorophore. In order to modulate the and stable fluorescence in the physiological pH range (Figure
reactivity toward ·OH, we also fabricated different N-methyl S3). Finally, P2 was chosen as the ·OH-specific probe for
substitution on R1 and R2. The four probes (P1−P4) were subsequent studies.
synthesized via palladium-catalyzed C−N cross-coupling Sensing Performance of P2 toward ·OH. In aqueous
reactions (Scheme S1). buffer (0.1 M phosphate buffer, pH 7.4, containing 5% DMF),
Evaluating the Sensitivity and Specificity of Probe the fluorescence of P2 was nearly undetectable (Φ = 0.001).
P1−P4 to ·OH. With the probes P1−P4 in hand, we first Upon the reaction with varying amounts of ·OH, a
tested their reactivity toward ·OH (generated through the concentration-dependent fluorescence enhancement at 550
Fenton reaction between FeSO4 and H2O234−36) in aqueous nm was detected under excitation at 350 nm (Figure 2a). As
solutions. Before the reaction with target, all the probes shown in Figure 2b, the titration curve of P2 by ·OH shows a
exhibited extremely low fluorescence quantum yield in aqueous fast response at low concentrations and then reaches a plateau
buffer (Φ< 0.01, Table S2). After varying amounts of Fenton at 0.5 equiv of ·OH. The maximal fluorescence enhancement
reagents (FeSO4/H2O2 = 1/6, molar ratio, regarding [Fe2+]= (F/F0) was up to ca. 200-fold, which stands among the highest
[·OH]37) were introduced, enhanced fluorescence was levels of signal-to-background ratio among the reported
observed for all probes (Figure 1a). The products displayed fluorescent probes for ·OH. The detection limit was calculated
different fluorescence (orange for P1 and P3, yellow for P2 as 0.04 μM according to the 3sb/m criterion, where m is the
and P4, Figure 1b), implying that two different kinds of slope for the range of linearity and sb is the standard deviation
fluorescent substances were generated because of different of the blank (n = 11). This result shows that probe P2 has
substitution on R1 site. To clarify the reactions, we synthesized good sensitivity to ·OH, compared with most of the small-
the two proposed products (10 and 11 in Scheme 1c) and molecule-based ·OH probes. (Table S1) We also tested the
compared their fluorescence spectra with the reaction time-dependent response of P2 toward 1 equiv of ·OH, which
products. As shown in Figure 1b, the emission spectra of 10 shows that the maximal fluorescence enhancement can be
and 11 perfectly match that of the ·OH-probe reaction achieved within 20 min (Figure S4).
products. More evidence was shown in LC-MS analysis by Next, we evaluated the possibility of using P2 to detect ·OH
comparing the retention time of the compounds (Figure S1), under two-photon excitation mode. First, we determined the
1319 DOI: 10.1021/acs.analchem.7b04191
Anal. Chem. 2018, 90, 1317−1324
Analytical Chemistry Article

Figure 2. Response of P2 to ·OH under one-photon excitation mode.


(a) Fluorescence spectra and (b) titration curve of 10 μM P2 in the
presence of increasing amount of ·OH (0, 0.005, 0.01, 0.02, 0.04, 0.06,
0.08, 0.1, 0.15, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1 equiv; inset:
response in the range of 0.005−0.1 equiv). Fluorescence intensity (F)
was gained from the emission peak at 550 nm under 350 nm
excitation.
Figure 4. Responses of 10 μM P2 to 1, 5, 10 equiv of interfering
active absorption cross section (the product of absorption species (shown as dark gray, light gray, and white bar, respectively)
and 1 equiv of ·OH (red bar). (a: P2 only; b: Fe2+; c: ascorbic acid; d:
cross section and fluorescence quantum yield, δΦ, which
malondialdehyde; e: formaldehyde; f: acetaldehyde; g: glutaraldehyde;
determines the brightness of two-photon fluorophores) of both h: acetylactone; i: glutathione; j: cysteine; k: homocysteine; l: Na2S4;
P2 and 10. The maximal δΦ value of 10 in aqueous buffer (0.1 m: Na2S; n: NO; o: NO2−; p: NO3−; q: ONOO−; r: H2O2; s:
M phosphate buffer, pH 7.4, containing 5% DMF) was tBuOOH; t: 1O2; u: O2·‑; v: ROO·; w: tBuO·; x: ClO−; y: ·OH)
detected to be 83 GM at 740 nm, whereas the δΦ value of P2 Fluorescence intensity (F) was gained from the emission peak at 550
was nearly undetectable (Figure 3a). Such a significant nm under 350 nm excitation.

200-fold fluorescence enhancement. These results have


indicated the pronounced selectivity of P2 for ·OH detection,
which lays a solid foundation of using P2 to specifically
monitor ·OH in complicated biological environments.
Two-photon Fluorescence Imaging of Exogenous
and Endogenous ·OH in Biological Systems. Before
using P2 in bioimaging, we checked its cytotoxicity by
recording the survival rate of HeLa cells after 24 h incubation
with P2 using the tetrazolium-based colorimetric assay (MTT
Figure 3. (a) Two-photon active cross section (δΦ) of P2 and 10 assay), which demonstrated a low cytotoxicity of the probe
under 710−850 nm excitation. (b) Two-photon titration curve of 10 (Figure S5). As the first step of evaluating the sensing ability of
μM P2 by increasing amount of ·OH (0, 0.005, 0.01, 0.02, 0.04, 0.06, P2 in living cells, we introduced exogenous ·OH via in situ
0.08, 0.1, 0.15, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1 equiv; inset: Fenton reaction, which mimics the process in mitochondria to
response in the range of 0.005−0.1 equiv). Fluorescence intensity (F) generate ·OH flux under oxidative stress.39 As shown in Figure
was gained from the emission peak at 550 nm under 740 nm 5, upon the addition of catalyst Fe2+, the intracellular
excitation.
fluorescence intensity was immediately promoted and then
increased quickly. Since the foregoing experiments have
difference of δΦ would guarantee a sufficient contrast in precluded the response of P2 to H2O2 or Fe2+ alone, this
bioimaging, presenting an obvious change of brightness before remarkable fluorescence enhancement indicates a sensitive
and after the reaction of probe with ·OH. In a fluorescence response of P2 toward ·OH generated in situ. Meanwhile, we
titration experiment under two-photon excitation (740 nm), noticed a faint background (Figure 5, P2 only). To figure out
the [·OH]-dependent turn-on fluorescence signals were also the origin of this weak fluorescence, we pretreated two groups
obtained (Figure 3b). These results suggest that P2 can be an of HeLa cells with ·OH scavengers, 2,2,6,6-tetramethylpiper-
effective two-photon excitable fluorescent probe for ·OH idine-N-oxyl (TEMPOL) and glutathione (GSH). (The ·OH-
detection. depleting abilities were verified in solution assay, as shown in
Subsequently, we investigated the selectivity of P2 over a Figure S6.) Compared with the control group, the fluorescence
variety of potential interfering substances, including common of scavenger-treated groups was obviously weakened (Figure
reactive species in biological system and some agents expected S7), and a similar phenomenon was also observed in RAW
to react.13,38 We tested the following categories of interfer- 264.7 cells (Figure S8). These results demonstrate that P2 is
ences: reductive reagents (e−: Fe2+, ascorbic acid), reactive sensitive enough to illuminate basal-level ·OH in living cells.
carbonyl species (malondialdehyde, formaldehyde, acetalde- Next, we proceeded to elucidate the intracellular location of
hyde, glutaraldehyde, acetylactone), reactive sulfur species the fluorescence. Given the close relationship between ROS
(glutathione, cysteine, homocysteine, Na2S4, Na2S), and highly and mitochondria, we costained the HeLa cell with
reactive oxygen-containing species (NO, NO2−, NO3−, MitoTracker Red and P2. As shown in Figure S9, a broad
ONOO−, H2O2, tBuOOH, 1O2, O2·‑, ROO·, tBuO·, ClO−, · fluorescence overlap emitted from mitochondria and basal-
OH). As Figure 4 shows, all these potential interfering species level ·OH can be observed. The calculation for Pearson’s
(with 1, 5, and 10 equiv) caused negligible change of correlation coefficient (Rr) and overlap coefficient (R) were ca.
fluorescence of P2, but ·OH with 1 equiv can promote nearly 0.63 and 0.66, respectively, indicating a certain correlation40
1320 DOI: 10.1021/acs.analchem.7b04191
Anal. Chem. 2018, 90, 1317−1324
Analytical Chemistry Article

Figure 6. Monitoring the ·OH generation within RAW 264.7 cells


surviving the E. coli infection (100 moi) after 0, 3, 6, 18 h. Scale bar:
10 μm. Fluorescence intensity (F) was displayed as yellow bars.

mouse by topical application of PMA, which is a widely used


acute inflammatory model. As shown in Figure S13, much
stronger accumulated fluorescence was detected from the
PMA-treated ear compared with the contralateral control
(more clear profile of endogenous ·OH generation at different
depth are shown in Figure S14 and S15). To summarize, all the
Figure 5. Change of intracellular two-photon fluorescence with above results have collectively demonstrated that P2 is able to
adding exogenous Fenton reagent. The HeLa cells were preloaded trace the fluctuation of ·OH endogenously generated upon
with 10 μM P2 for 30 min, followed by sequential treatment with 1 different kinds of stimuli both in vitro and in vivo.
mM H2O2 and 1 mM FeSO4. The images of both two-photon
fluorescence and bright field channel were collected with 2 min
Probing the ·OH Alteration Upon Regulation of SOD1
intervals. Scale bar: 10 μm. at Activity and Expression Level. The success of visualizing
·OH by P2 in biological environments inspired us to explore
the relationship between the variation of ·OH level and SOD
between mitochondria and ·OH generation in living cancer status. Biological studies have suggested that both the activity
cells. Moreover, we also tested the photostability of fluorescent and expression level of SOD may contribute to variation of
product by continuously illuminating compound 10-labeled ROS level.1,43,44 At first, we tried to modulate the activity of
cells for 30 min under the above imaging conditions. SOD and determine if P2 can detect the possible change of
Negligible fluorescence change was observed (Figure S10), intracellular ·OH level. We utilized ATN-244 (choline
which guarantees the long-time observation of ·OH under two- tetrathiomolybdate), a preclinical antitumor drug that targets
photon excitation. onto the copper ion and inhibits copper/zinc-dependent
Having proven the ability of P2 to recognize ·OH in living enzyme including SOD1,45,46 as the SOD1 activity inhibitor.
cells, we proceeded to explore its capability of capturing ·OH As shown in Figure 7, the intracellular fluorescence increased
generated endogenously in vitro. We thus incubated P2-loaded by 2.3-fold after 2 h treatment with 100 μM inhibitor and
RAW 264.7 cells with heat-killed E. coli, serving as a strong showed a ca. 3.2-fold enhancement after 18 h treatment. This
inflammatory stimulus to yield excess ROS including ·OH.41 A result indicates a higher level of ·OH production in the cells
distinct time-dependent intracellular fluorescence enhance- with lowered SOD1 activity, as the catalytic role of copper ion
ment (up to 4-fold) with obvious morphological changes can was blocked by ATN-244. As further support, when the cells
be seen, showing the accumulating ·OH generation during the were treated with a lower concentration of ATN-244 (10 μM),
inflammation process (Figure 6). We then treated RAW 264.7 a relatively weaker but also time-dependent fluorescence
cells with another moderate immune stimulus, the combina- enhancement was also observed (Figure S16).
tion of lipopolysaccharide (LPS) and interferon-gamma (IFN- Next, we moved on to manipulate the SOD1 mRNA, trying
γ), and a 1.6-fold intracellular fluorescence enhancement was to illustrate the relationship between SOD1 gene expression
seen (Figure S11). To verify the universality of these findings, level and intracellular ·OH content. To this end, we
we also tested on HeLa cells using phorbol 12-myristate 13- constructed the SOD1 overexpressing cells by transfecting
acetate (PMA) as stimulus. Again, we observed a time- the recombinant plasmid, which contains the fragment of
dependent fluorescence elevation (maximal 1.7-fold) after 1 h SOD1 gene, into two groups of HeLa cells (entries 2 and 4).
stimulation (Figure S12). In order to further highlight the As controls, two more groups were transfected with empty
applicability of the probe in bioimaging in vivo, we tried using vector (entries 1 and 3). After 24 h transfection, two groups
P2 to illuminate ·OH in a living body. As known, the detection (entries 3 and 4) were stimulated with PMA prior to P2
of ·OH in vivo is a highly challenging task, and very few loading. We can see the SOD1 overexpressed cells (entry 2)
luminescent probes have been reported for this subject.42 We exhibited lower fluorescence than the control group (entry 1),
constructed the swelling ear model on living BALB/cA-nu which suggests a decreased basal level of ·OH resulting from
1321 DOI: 10.1021/acs.analchem.7b04191
Anal. Chem. 2018, 90, 1317−1324
Analytical Chemistry Article

by PMA stimulation, thus decreasing ·OH level by the Fenton/


Haber−Weiss mechanism. To further reveal the relationship
between intracellular ·OH and SOD1 expression, we harvested
the above four groups of cells and performed quantitative real
time polymerase chain reaction (qRT-PCR) to determine the
SOD1 mRNA level. As shown with the gray bar graph in
Figure 8, significantly enhanced expression levels of SOD1
gene were determined in the two groups transfected with
recombinant plasmid (entry 2 and 4), in accordance with their
reduced fluorescence intensities. Interestingly, we found a
slightly elevated mRNA expression of SOD1 in HeLa cells
stimulated with PMA (entry 3), which is a recurring result of
previous literature.47 These results confirmed that the
intracellular ·OH content has a close correlation with the
expression level of SOD1, and our probe P2 displayed its
capability to light up the subtle ·OH fluctuation in such a
regulation process.
Figure 7. Illumination of the ·OH release along the deactivation of Finally, we tried applying our probe in a ·OH-participating
SOD1 by enzyme inhibitor ATN-244. The RAW 264.7 cells were pathological process involving SOD1 mutation. As a
treated by 100 μM ATN-244 for 0, 2, 6, 18 h, and then incubated with preliminary attempt in this regard, we managed to knockdown
10 μM P2 for 30 min. Scale bar: 10 μm. Fluorescence intensity (F) the SOD1 gene in human umbilical vein endothelial cells
was displayed as yellow bars. (HUVECs) by introducing short interfering RNA (siRNA).
Like some of the mutation of SOD1 gene in ALS, such a silent
higher expression level of SOD1 (Figure 8). In the groups gene will probably deprive the normal radical-clearance
treated with empty vector (entries 1 and 3), an obvious function of SOD1 and thus result in an elevated ·OH level.
Compared with the control, the cells transfected with siRNA
exhibited moderate increase of intracellular fluorescence after
36 h transfection, and the qRT-PCR result indicated that ca.
55% gene was successfully suppressed (Figure 9). Such results

Figure 8. Visualization of the ·OH content in PMA-treated HeLa cells


under different SOD1 gene expression level. The cells in entries 2 and
Figure 9. Images of HUEVC cells separately transfected with
4 were transfected with SOD1 gene plasmids, and empty vectors were
scrambled RNA duplex (control) and SOD1 siRNA (SOD1
used for entries 1 and 3 as control. Cells in entries 3 and 4 were
knockdown), and finally incubated with 10 μM P2 for 30 min.
further treated with 2 μg/mL PMA for 2 h. All the groups were finally
Scale bar: 10 μm. Fluorescence intensity (F) was displayed as yellow
incubated with 10 μM P2 for 30 min before imaging. Scale bar: 10
bars. The mRNA level of SOD1 was determined by qRT-PCR, shown
μm. Fluorescence intensity (F) was displayed as yellow bars. The
as the relative expression in gray bars.
mRNA level of SOD1 was determined by qRT-PCR, shown as the
relative expression in gray bars.
convinced us that the down-regulation of SOD1 gene
fluorescence enhancement (ca. 1.7-fold) can be observed in contributes to the elevated endogenous ·OH generation. It
the PMA-treated cells, which is consistent with the above result also confirms again that the probe P2 could be a useful tool for
in HeLa cells (Figure S10). However, for the SOD1- uncovering some unsolved puzzles in ·OH and SOD related
events.


overexpressing cells, the group with PMA stimulation (entry
4) shows only a slightly higher fluorescence than that without
stimulation (entry 2), while it was still weaker than normal CONCLUSIONS
cells (entry 1) producing basal-level ·OH. This means the In summary, we have developed a two-photon excitable
overexpressed SOD1 successfully consumes the O2·‑, boosted molecular probe for specific detection of ·OH. Taking
1322 DOI: 10.1021/acs.analchem.7b04191
Anal. Chem. 2018, 90, 1317−1324
Analytical Chemistry Article

advantage of the N-dearylation reaction mechanism, the probe (11) Fulford, J.; Nikjoo, H.; Goodhead, D. T.; O’Neill, P. Int. J.
P2 shows a highly sensitive response (∼200-fold fluorescence Radiat. Biol. 2001, 77, 1053−1066.
enhancement to 1 equiv of ·OH) and pronounced selectivity (12) William, H. G.; Kang, J. J. Am. Water Works Assoc. 1988, 80,
over a series of biological reactive species. Equipped with the 57−63.
two-photon fluorophore (with 83 GM active cross-section), P2 (13) You, Y.; Nam, W. Chem. Sci. 2014, 5, 4123−4135.
is competent in detecting ·OH both in vitro and in vivo. P2 (14) Popovic-Bijelic, A.; Mojovic, M.; Stamenkovic, S.; Jovanovic,
M.; Selakovic, V.; Andjus, P.; Bacic, G. Free Radical Biol. Med. 2016,
enables the visualization of subtle variation of ·OH in various
96, 313−322.
SOD-modulated biological events. Our results show that
(15) Obata, T. Toxicol. Lett. 2002, 132, 83−93.
inhibited enzymatic activity and down-regulated expression of (16) Gomes, A.; Fernandes, E.; Lima, J. L. J. Biochem. Biophys.
SOD1 both lead to elevated generation of intracellular ·OH. Methods 2005, 65, 45−80.
This work is the first report to reveal the regulation of (17) Soh, N.; Makihara, K.; Ariyoshi, T.; Seto, D.; Maki, T.;
intracellular ·OH level by SOD enzyme with a visuable Nakajima, H.; Nakano, K.; Imato, T. Anal. Sci. 2008, 24, 293−296.
molecular tool. The favorable performance of P2 in bioimaging (18) Page, S. E.; Wilke, K. T.; Pierre, V. C. Chem. Commun. 2010,
also suggests that the probe (and further improved analogues) 46, 2423−2425.
can be useful tool for elucidating the functions and roles of · (19) Cui, G.; Ye, Z.; Chen, J.; Wang, G.; Yuan, J. Talanta 2011, 84,
OH in future studies. 971−976.


*
ASSOCIATED CONTENT
S Supporting Information
(20) Ganea, G. M.; Kolic, P. E.; El-Zahab, B.; Warner, I. M. Anal.
Chem. 2011, 83, 2576−2581.
(21) Perry, C. C.; Tang, V. J.; Konigsfeld, K. M.; Aguilera, J. A.;
The Supporting Information is available free of charge on the Milligan, J. R. J. Phys. Chem. B 2011, 115, 9889−9897.
(22) Zhuang, M.; Ding, C.; Zhu, A.; Tian, Y. Anal. Chem. 2014, 86,
ACS Publications website at DOI: 10.1021/acs.anal-
1829−1836.
chem.7b04191. (23) Yuan, L.; Lin, W.; Song, J. Chem. Commun. 2010, 46, 7930−
Organic synthesis and characterization, spectroscopic 7932.
measurements, cytotoxicity assay, cell imaging, mouse (24) Kim, M.; Ko, S. K.; Kim, H.; Shin, I.; Tae, J. Chem. Commun.
imaging, and additional figures and tables (PDF) 2013, 49, 7959−7961.


(25) Asano, M.; Doi, M.; Baba, K.; Taniguchi, M.; Shibano, M.;
Tanaka, S.; Sakaguchi, M.; Takaoka, M.; Hirata, M.; Yanagihara, R.;
AUTHOR INFORMATION
Nakahara, R.; Hayashi, Y.; Yamaguchi, T.; Matsumura, H.; Fujita, Y. J.
Corresponding Author Biosci. Bioeng. 2014, 118, 98−100.
*E-mail: zhhliu@whu.edu.cn. (26) Yapici, N.; Jockusch, S.; Moscatelli, A.; Mandalapu, S. R.;
ORCID Itagaki, Y.; Bates, D. K.; Wiseman, S.; Gibson, K. M.; Turro, N. J.; Bi,
Zhihong Liu: 0000-0003-1500-9342 L. Org. Lett. 2012, 14, 50−53.
(27) Cao, L.; Wu, Q.; Li, Q.; Shao, S.; Guo, Y. J. Fluoresc. 2014, 24,
Notes
313−318.
The authors declare no competing financial interest.


(28) Zhou, W.; Cao, Y.; Sui, D.; Lu, C. Angew. Chem., Int. Ed. 2016,
55, 4236−4241.
ACKNOWLEDGMENTS (29) Meng, L.; Wu, Y.; Yi, T. Chem. Commun. 2014, 50, 4843.
The authors acknowledge the financial support from the (30) Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M. T. D.; Mazur,
National Natural Science Foundation of China (nos. M.; Telser, J. Int. J. Biochem. Cell Biol. 2007, 39, 44.
21625503, 21535005), and the support of China Scholarship (31) Kim, H. M.; Cho, B. R. Chem. Rev. 2015, 115, 5014−5055.
Council. We also thank Fang Zhou of the Institute of (32) Lowndes, S. A.; Sheldon, H. V.; Cai, S.; Taylor, J. M.; Harris, A.
Hydrobiology, Chinese Academy of Sciences, for the assistance L. Microvasc. Res. 2009, 77, 314−326.
of confocal microscopy. (33) Setsukinai, K.; Urano, Y.; Kakinuma, K.; Majima, H. J.; Nagano,


T. J. Biol. Chem. 2003, 278, 3170−3175.
REFERENCES (34) Minero, C.; Lucchiari, M.; Maurino, V.; Vione, D. RSC Adv.
2013, 3, 26443−26450.
(1) Miao, L.; St. Clair, D. K. Free Radical Biol. Med. 2009, 47, 344− (35) Vermilyea, A. W.; Voelker, B. M. Environ. Sci. Technol. 2009,
356. 43, 6927−6933.
(2) Hu, J. J.; Wong, N.; Ye, S.; Chen, X.; Lu, M.; Zhao, A. Q.; Guo, (36) Kang, Y. W.; Hwang, K. Water Res. 2000, 34, 2786−2790.
Y.; Ma, A. C.; Leung, A. Y.; Shen, J.; Yang, D. J. Am. Chem. Soc. 2015, (37) Halliwell, B.; Gutteridge, J. M. C. Arch. Biochem. Biophys. 1986,
137, 6837−6843.
246, 501−514.
(3) Sayre, L. M.; Perry, G.; Smith, M. A. Chem. Res. Toxicol. 2008,
(38) Wardman, P. Free Radical Biol. Med. 2007, 43, 995−1022.
21, 172−188.
(39) Thomas, C.; Mackey, M. M.; Diaz, A. A.; Cox, D. P. Redox Rep.
(4) Hayashi, Y.; Homma, K.; Ichijo, H. Adv. Biol. Regul 2016, 60,
95−104. 2009, 14, 102−108.
(5) Liu, D.; Wen, J.; Liu, J.; Li, L. FASEB J. 1999, 13, 2318−2328. (40) Zinchuk, V.; Zinchuk, O. Curr. Protoc. Cell. Biol. 2008, 4.19.1−
(6) Sankarapandi, S.; Zweier, J. L. J. Biol. Chem. 1999, 274, 34576− 4.19.6.
34583. (41) Streit, W. J.; Kincaid-Colton, C. A. Sci. Am. 1995, 273, 54−61.
(7) Raha, S.; Robinson, B. H. Trends Biochem. Sci. 2000, 25, 502− (42) Li, Z.; Liang, T.; Lv, S.; Zhuang, Q.; Liu, Z. J. Am. Chem. Soc.
508. 2015, 137, 11179−11185.
(8) Bruijn, L. I.; Miller, T. M.; Cleveland, D. W. Annu. Rev. Neurosci. (43) Winterbourn, C. C. Toxicol. Lett. 1995, 82, 969−974.
2004, 27, 723−749. (44) Jomova, K.; Valko, M. Toxicology 2011, 283, 65−87.
(9) Mondola, P.; Damiano, S.; Sasso, A.; Santillo, M. Front. Physiol. (45) Lin, J.; Zahurak, M.; Beer, T. M.; Ryan, C. J.; Wilding, G.;
2016, 7, 594−601. Mathew, P.; Morris, M.; Callahan, J. A.; Gordon, G.; Reich, S. D.;
(10) Li, P.; Xie, T.; Duan, X.; Yu, F.; Wang, X.; Tang, B. Chem. - Eur. Carducci, M. A.; Antonarakis, E. S. Urol. Oncol.: Semin. Orig. Invest
J. 2010, 16, 1834−1840. 2013, 31, 581−588.

1323 DOI: 10.1021/acs.analchem.7b04191


Anal. Chem. 2018, 90, 1317−1324
Analytical Chemistry Article

(46) Lee, K.; Briehl, M. M.; Mazar, A. P.; Batinic-Haberle, I.;


Reboucas, J. S.; Glinsmann-Gibson, B.; Rimsza, L. M.; Tome, M. E.
Free Radical Biol. Med. 2013, 60, 157−167.
(47) Minc, E.; de Coppet, P.; Masson, P.; Thiery, L.; Dutertre, S.;
Amor-Gueret, M.; Jaulin, C. J. Biol. Chem. 1999, 274, 503−509.

1324 DOI: 10.1021/acs.analchem.7b04191


Anal. Chem. 2018, 90, 1317−1324

You might also like