You are on page 1of 9

Article

Cite This: ACS Infect. Dis. 2019, 5, 1376−1384 pubs.acs.org/journal/aidcbc

Discovery of Simplified Sampangine Derivatives with Potent


Antifungal Activities against Cryptococcal Meningitis
Zhuang Li,†,‡,∥ Na Liu,†,∥ Jie Tu,† Changjin Ji,† Guiyan Han,† and Chunquan Sheng*,†,‡

School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, People’s Republic of China

School of Pharmacy, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou, Fujian 350122, People’s
Republic of China
*
S Supporting Information
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

ABSTRACT: Cryptococcal meningitis (CM) is associated


with high morbidity and mortality. Current antifungal drug
Downloaded via NOTTINGHAM TRENT UNIV on August 13, 2019 at 04:06:05 (UTC).

therapy for CM has the following challenges: limited efficacy,


significant side effects, emerging drug resistance, and
unavailability in highly needed countries. There is an urgent
need to develop novel CM therapeutic agents with a new
mode of action. On the basis of the antifungal natural product
sampangine, herein, novel simplified isoxazole derivatives were
identified to possess excellent inhibitory activity against
Cryptococcus neoformans (C. neoformans). Particularly, com-
pound 9a was highly active (the minimum inhibitory
concentration of 80% inhibition, MIC80 = 0.031 μg/mL)
and significantly inhibited biofilm formation, melanin, and urease production of C. neoformans. 9a had good blood−brain barrier
(BBB) permeability and effectively reduced the brain fungal burden in a murine model of cryptococcosis. The antifungal
mechanism of compound 9a was preliminarily investigated by transmission electron microscopy and flow cytometry. It was able
to cause necrocytosis of C. neoformans cells and cell cycle arrest in the G1/S phase. Isoxazole compound 9a represents a
promising lead compound for the development of novel CM therapeutic agents.
KEYWORDS: cryptococcal meningitis, sampangine derivatives, anticryptococcal activity, biofilm

L ife-threatening invasive fungal infections (IFIs) are mainly


caused by species of Candida, Cryptococcus, Aspergillus, and
Pneumocystis.1 Recently, the incidence and mortality of IFIs have
Screening of compound libraries13 and drug repurposing14
have been used to identify small molecules against the
C. neoformans infection. Alternatively, the discovery of novel
been increased rapidly due to the growing number of antifungal lead compounds from natural products represents an
immunosuppressive patients and limitations of available effective strategy.15 Two important classes of antifungal agents,
antifungal therapy.2,3 In the central nervous system (CNS), polyenes and echinocandins, were originally discovered from
Cryptococcus neoformans (C. neoformans) is the most common natural products.15 In our previous studies, a series of novel
cause of fungal infection among the HIV-1/AIDS populations, antifungal lead compounds were identified from the natural
leading to cryptococcal meningitis (CM). It is estimated that product sampangine by structural simplification and structural
there are about 1,000,000 cases of CM in AIDS patients each optimization.16−18 As compared with sampangine, the simplified
year with over 600,000 annual deaths.4−6 The high mortality of new compounds showed improved antifungal potency and more
CM is associated with the lack of effective antifungal favorable physicochemical properties.16−18 Interestingly, they
treatment.7,8 Clinically, only three drugs, namely, amphotericin revealed promising features to overcome drug resistance of
B (AMB), flucytosine, and fluconazole (FLC), are available for Candida albicans (C. albicans) in that compound ZG-20-7 was
the induction and maintenance regimens of CM.7,8 The fungicidal against FLC-resistant strains and significantly
combination of AMB and 5-flucytosine is the “gold standard” inhibited biofilm formation and yeast-to-hypha morphological
for the treatment of CM. However, AMB has limitations due to transitions.16−18
significant adverse effects (e.g., renal toxicity), and 5-flucytosine Herein, the potential applications of sampangine derivatives
has not been marketed in most African and Asian countries, for the treatment of CM were investigated. After structural
where the morbidity of CM is high.8 FLC acts by a fungistatic optimization of compound ZG-20-7, novel tricyclic isoxazole
mechanism and is limited by severe drug resistance and derivatives were identified to possess excellent activity against
hepatotoxicity. The newly developed echinocandin antifungal C. neoformans. Particularly, compound 9a was highly active
agents are poorly effective against Cryptococcus species.9,10 Thus,
there is an urgent need to develop novel anticryptococcal Received: March 2, 2019
agents.11,12 Published: May 9, 2019

© 2019 American Chemical Society 1376 DOI: 10.1021/acsinfecdis.9b00086


ACS Infect. Dis. 2019, 5, 1376−1384
ACS Infectious Diseases Article

Figure 1. Discovery of compound 9a as a novel antifungal agent.

Table 1. In Vitro Antifungal Activity of Tricyclic Isoxazole Derivatives (MIC80, μg/mL, 48 h)a

a
Abbreviations: C. neo., Cryptococcus neoformans; C. alb., Candida albicans.

(C. neoformans with a minimum inhibitory concentration of 80%


inhibition, MIC80, of 0.031 μg/mL, Figure 1) and showed potent
■ RESULTS
In Vitro Antifungal Activities and Structure−Activity
efficacy in the murine model of CM, representing a promising Relationship of the Tricyclic Isoxazole Derivatives.
lead compound for the development of novel CM therapeutic Scaffold hopping of ZG-20-7 by isoxazole/pyridine replacement
agents. was proven to be an effective strategy to improve the antifungal
1377 DOI: 10.1021/acsinfecdis.9b00086
ACS Infect. Dis. 2019, 5, 1376−1384
ACS Infectious Diseases Article

Table 2. In Vitro Antifungal Activity of Part of the Target Compounds (MIC80, μg/mL)a
48 h 72 h
compds C. gla. 8535 C. alb. 4108 C. alb. 0304103 C. tro. 5008 C. neo. ATCC 34877 C. gat. ATCC 14116 C. neo. H99
9a 1 1 1 1 0.031 0.25 0.031
9c 2 2 2 2 0.12 0.25 0.062
9d 4 4 4 8 0.5 0.5 0.25
9f 2 2 0.5 16 1 2 0.5
9h 16 8 16 16 0.5 1 1
FLC 8 >64 >64 >64 4 2 2
VRC 0.25 >64 >64 >64 0.062 0.031 0.12
a
Abbreviations: C. gla., Candida glabrata; C. tro., Candida tropicalis; C. gat., Cryptococcus gattii.

activity against C. neoformans.19 However, our previously Table 3. In Vitro Antifungal Activity of Compound 9a on
reported isoxazole derivatives were inactive in the in vivo CM Mycete and Dermatophytes (MIC80, μg/mL)a
model (Figure S1). To further explore the structure−activity
compds A. f um. 7544 T. rub. TIA M. gyp. MZC
relationship (SAR) and identify new antifungal lead compounds
with potent in vivo activity toward CM, derivatives 9a−h were 9a 1 0.12 0.25
designed and synthesized (see the Supporting Information for FLC >64 2 16
chemical synthesis and structural characterization). Their in VRC 0.031 0.031 0.062
a
vitro antifungal activities were assayed against C. albicans Abbreviations: A. f um., Aspergillus f umigutus; T. rub., Trichophyton
SC5314 and C. neoformans H99. FLC and voriconazole rubrum; M. gyp., Microsporum gypseum.
(VRC) were used as the positive controls. Most compounds
showed better inhibitory activity against C. neoformans than group (4 and 16 μg/mL) were used as controls. As shown in
C. albicans (Table 1). Compounds 9a and 9c showed the best Figure 2B, compound 9a had excellent fungicidal activity against
antifungal activity against C. neoformans H99 (MIC80 = 0.031 C. neoformans, which could completely kill fungi cells at 1 μg/mL
μg/mL), which was significantly more potent than FLC and within 48 h. In contrast, FLC did not show fungicidal activity at
VRC. 16 μg/mL. Compound 9a also showed excellent fungicidal
SAR revealed that the isoxazole/pyridine replacement led to activity to drug resistant C. albicans (Figure S2).
substantial improvement of the anti-C. neoformans activity. The Inhibitory Effect of Compound 9a on the Virulence
change of the molecular scaffold also resulted in different SAR Factors of C. neoformans. Biofilm is an important virulence
on the substitutions. The nitro group on the thiophene ring was factor contributing to antifungal drug resistance, which often
not essential for the antifungal activity, which was different from leads to repeated fungal infections in the clinic and represents a
that observed in the tricyclic pyridine derivatives.16−18 The significant therapeutic challenge.20 Thus, the effect of
bromo substitution (9b) was found to have a negative impact on compound 9a on biofilm formation of C. neoformans was
the antifungal activity. When the quinone group was reduced evaluated using AMB as the positive control. The results showed
(9e) or replaced with pyranone (9g), an obvious decrease of the that it significantly inhibited the biofilm formation with an
antifungal activity was observed. The replacement of thiophene inhibition rate of more than 80% at the concentration of 0.062
with furan, benzene, or a pyridine ring was also unfavorable for μg/mL (Figure 3), which was more potent than AMB.
the antifungal activity. Melanin and urease are two important virulence factors
On the basis of the antifungal activity, compounds 9a, 9c, 9d, during C. neoformans infection. Melanin contributes to virulence
9f, and 9h were further assayed for the inhibitory activity against by protecting fungal cells against attack by multiple toxic insults
drug resistant Candida species, such as C. glabrata, C. tropicalis, and immune effector cells.21 Urease in C. neoformans plays an
and C. albicans, and other Cryptococcus species, such as important role in the survival of C. neoformans in phagolyso-
C. neoformans var. grubii and C. gattii (Table 2). These somes and the dissemination to the brain causing meningoen-
compounds showed an MIC80 value ranging from 0.031 to 16 cephalitis.22 Thus, various induction mediums were used to
μg/mL. Among them, compound 9a was more potent than FLC determine the effects of compound 9a on the production of
and comparable to VRC (Table 2). To further investigate melanin and urease. As shown in Figure 4A, the melanin
whether compound 9a has broad-spectrum antifungal activity, production could be determined; colonies containing melanin
A. f umigates, T. rubrum, and M. gypseum strains were assayed. turn black when exposed to melanin induction medium. The
The result showed that compound 9a could inhibit a wide range results revealed that compound 9a inhibited the production of
of fungal pathogens (Table 3). Due to its excellent melanin at the concentration of 8 μg/mL. Similarly, if the cells
anticryptococcal activity, compound 9a was selected for further produced urease, the medium would appear red. Compound 9a
biological evaluations. also inhibited the production of urease at the concentration of
Time-growth curves were assayed to further evaluate the 32 μg/mL (Figure 4B).
fungistatic activity of compound 9a. As shown in Figure 2A, Compound 9a Induced Morphological Changes in
different concentrations of compound 9a (0.0078−0.062 μg/ C. neoformans Cells. In order to investigate the fungicidal
mL) were used in the time-growth curve assay. The results mechanism of compound 9a, its influence on the morphology of
showed that compound 9a could inhibit C. neoformans H99 cell C. neoformans H99 was monitored by transmission electron
growth in a concentration-dependent manner and completely microscopy (TEM). C. neoformans H99 cells were treated with
inhibited cell growth at 0.12 μg/mL. The time-kill curve assay compound 9a at the concentrations of 0.062 and 2 μg/mL for 8
was used to test whether compound 9a had fungicidal activity. h, and cells treated with dimethyl sulfoxide (DMSO) were used
The drug-free group, FLC group (4 and 16 μg/mL), and AMB as a control group. As shown in Figure 5A, normal C. neoformans
1378 DOI: 10.1021/acsinfecdis.9b00086
ACS Infect. Dis. 2019, 5, 1376−1384
ACS Infectious Diseases Article

Figure 2. Compound 9a inhibited and killed C. neoformans H99 cells. (A) Time-growth curves of C. neoformans H99 obtained by using initial
inoculums of 2 × 106 colony-forming units (CFU)/mL. The concentration of cells was determined after 0, 4, 8, 12, 24, and 48 h of incubation.
C. neoformans H99 cells were treated with different concentrations of compound 9a. (B) Time-kill curves of C. neoformans H99 obtained by using an
initial inoculum of 2 × 106 CFU/mL. The number of viable cells was measured after 12, 24, 36, and 48 h of incubation. Cells were treated with
compound 9a (0.062 and 1 μg/mL), FLC (4 and 16 μg/mL), and AMB (4 and 16 μg/mL). The number of fungal cells was counted at different times
and to the base 10 logarithm.

Figure 3. Effects of different concentrations of compound 9a and AMB on biofilm formation. C. neoformans H99 cells were treated with compound 9a
and AMB (0.062−8 μg/mL) for 24 h. The results of biofilm formation were represented as the means ± standard deviations for three independent
experiments. Statistical differences among groups were determined by ANOVA. The comparison between the two components was completed by the
t-test. *P < 0.05 compared to the control group; **P < 0.01 compared to the control group; ***P < 0.001 compared to the control group; ****P <
0.0001 compared to the control group.

cells revealed an intact cell wall and membrane, dense capsule, indicated that compound 9a could significantly increase the
uniform central density, and regular cell morphology. For the subpopulation of cells in the G1 phase (P < 0.001) and reduce
cells treated with compound 9a at 0.062 μg/mL, a sparse capsule the number of cells in the G2 phase (P < 0.001) (Figure 7B),
and decomposed organelles could be observed (Figure 5B). At resulting in cell cycle arrest at the G1/S phase. Similarly, FLC
the same time, the cell membrane is separated from the cell wall. was proven to cause the accumulation of G1 and S phase cells in
After the cells were treated with compound 9a at 2 μg/mL, large our previous studies.20
vacuoles could be observed (Figure 5C), which is characteristic Compound 9a Showed Potent Activity in a Murine
of necrocytosis. TEM results indicated that compound 9a was Model of CM. The blood−brain barrier (BBB) permeability of
able to cause significant damage to C. neoformans cells, both at compound 9a was tested by the parallel artificial membrane
low and high concentrations. permeation assay (PAMPA) prior to the investigation of in vivo
Compound 9a Caused Necrocytosis of C. neoformans antifungal activity. The results indicated that compound 9a had
Cells and Induced G1/S Phase Arrest. Necrocytosis of good BBB permeability and was superior to clonidine (Table 4).
C. neoformans H99 treated with compound 9a was further Moreover, compound 9a showed good metabolic stability in
investigated by flow cytometry. The percentage of necrotic cells mouse liver microsomes (T1/2 > 120 min). Due to the excellent
in the control group was 2.83%. After treating with compound in vitro antifungal activity, good ability to cross the BBB, and
9a at 0.12, 0.5, and 2 μg/mL for 24 h, the percentages of necrotic metabolic stability, compound 9a was chosen for the evaluation
cells were 10.1%, 34.9%, and 46.9%, respectively (Figure 6). The of in vivo anti-cryptococcus potency. Mice CM models were
results suggested that compound 9a caused a remarkable established by injecting C. neoformans H99 cells into the veins of
necrotic effect instead of apoptosis, which was different from the the tails. Mice were assigned into four groups: the control group,
results of early apoptosis caused by FLC (percentage of the FLC treatment group, and two 9a treatment groups (5 and
apoptosis cells: 11.5%). 10 mg/kg). FLC and 9a treatments were initiated at 2 h
The effect of compound 9a on the cell cycle was investigated postinfection. Such treatments were continued for 5 consecutive
using flow cytometric analysis to further clarify the antifungal days. The treatment with compound 9a significantly reduced the
mechanism. The results showed that 10.04% of cells treated with fungal burden in the brain at 5 mg/kg (P < 0.05) and 10 mg/kg
DMSO were in the G1 phase. The ratios in the G1 phase were (P < 0.01). The results highlighted the promising features of
49.19%, 37.68%, and 45.92% after being treated with compound compound 9a as a lead compound for the treatment of CM
9a at 0.5, 1, and 2 μg/mL, respectively (Figure 7A). The results (Figure 8, Table S1).
1379 DOI: 10.1021/acsinfecdis.9b00086
ACS Infect. Dis. 2019, 5, 1376−1384
ACS Infectious Diseases Article

antifungal agents (e.g., triazole derivative VT-159823), the


discovery of anticryptococcal molecules with new chemical
scaffolds and a new mode of action is a challenging task.
Natural products are an important source of antifungal agent
(e.g., AMB and echinocandins). Compound 9a was derived
from structural simplification and structural optimization of the
antifungal natural product sampangine. Although sampangine
showed potent in vitro antifungal activity, its further develop-
ment was terminated due to the lack of in vivo potency.16−18 As
compared with sampangine, the anticryptococcal activity of
compound 9a was greatly improved, which was also more potent
than triazole antifungal agents FLC and VRC. Moreover, an
antifungal agent with fungicidal activity is preferred more than a
drug only inhibiting fungal growth. Unlike FLC, compound 9a
revealed excellent fungicidal activity and completely killed
C. neoformans cells at 1 μg/mL. Interestingly, there is a
significant increase in dead cells from 36 to 48 h. Obvious
cellular damage can be observed in the TEM images after
treatment with compound 9a. The sustained damage caused by
a high concentration of drugs may lead to massive death of
fungal cells at a certain point in time.
Virulence factors that contribute to promoting disease or
damaging the host are produced by fungal pathogens.
Antivirulence agents are expected to decrease evolutionary
Figure 4. Effects of compound 9a on melanin and urease production by pressure for the development of drug resistance.24 Interestingly,
C. neoformans H99 cells. (A) C. neoformans H99 cells grown on L- compound 9a could significantly inhibit several important
DOPA medium were treated with different concentrations of the
virulence factors of C. neoformans. The formation of biofilms is a
compounds for 48 h to determine melanin production. (B) Strains
grown on urea medium for 72 h were used to determine the production kind of survival mechanism of fungi, which is associated with
of urease. high-level drug resistance.25,26 Compound 9a showed excellent
activity to inhibit C. neoformans biofilm formation (>80%
inhibition at 0.062 μg/mL), highlighting its potential to treat
biofilm-related infections. Moreover, compound 9a also
effectively inhibited the production of melanin and urease in
C. neoformans cells. Melanin has antioxidant properties and
protects fungal cells against the oxidative stress responses, which
is generally understood to be the first line of defense against
reactive oxygen species (ROS) formation.27,28 Several com-
pounds (e.g., o-vanillin) with fungicidal activity were reported to
cause mitochondrial dysfunction by triggering oxidative stress
responses.29 Thus, it is suggested that compound 9a might
induce the oxidative stress in fungal cells, leading to the
inhibition of melanin production and fungicidal effect.
Although several new anticryptococcal molecules have been
identified, it is still a big challenge to translate in vitro activity into
therapeutic efficacy due to unfavorable physicochemical proper-
ties and pharmacokinetic profiles.11,12,14 For example, our
Figure 5. TEM of C. neoformans H99 under different concentrations of previously reported alkyl analogues of compound 9a lacked in
compound 9a for 8 h. (A) Cells treated with DMSO. (B) Cells treated vivo anti-CM potency (Figure S1).19 In contrast, compound 9a
with compound 9a at 0.062 μg/mL. (C) Cells treated with compound had good BBB permeability and metabolic stability and
9a at 2 μg/mL. The organelles indicated by the arrows are as follows: 1,
capsules present in the outer layers of the cell wall; 2, cell well; 3, cell
significantly reduced the brain fungal burden in a murine
membrane; 4, decomposed organelles; 5, wrinkled cell membrane; 6, model of cryptococcosis. Thus, it represents a promising lead
significantly increased vacuoles. compound for the development of novel therapeutic agents for
CM. However, the water solubility of compound 9a (7.72 μg/

■ DISCUSSION
CM is a serious disease with high incidence and mortality in
mL, Table S2) was relatively poor, which remains to be
improved to achieve better pharmacokinetic profiles as well as
the in vivo antifungal potency.
HIV-positive individuals.7,8 However, current antifungal treat- As an important step in drug discovery and development,
ment of CM remains a big challenge mainly due to poor efficacy clarifying the mode of action of compound 9a is highly required.
of existing drugs, significant side effects, emerging drug Herein, preliminary antifungal mechanisms of compound 9a
resistance, and unavailability in highly needed countries.7,8 were investigated. The results revealed that compound 9a
Thus, novel anti-CM agents with excellent anticryptococcal caused necrocytosis of C. neoformans cells and cell cycle arrest in
activity, favorable BBB penetration, and good safety profiles are the G1/S phase, which is totally different from FLC and acts by a
highly desirable. As compared with the optimization of known new mode of action. It has been reported that large doses of
1380 DOI: 10.1021/acsinfecdis.9b00086
ACS Infect. Dis. 2019, 5, 1376−1384
ACS Infectious Diseases Article

Figure 6. Compound 9a caused C. neoformans H99 cells necrosis. Cells treated with compound 9a at 0.12, 0.5, and 2 μg/mL and FLC at 16 μg/mL for
24 h. Necrosis was detected by flow cytometry (n = 3). Representative images from three independent experiments are displayed.

acetic acid (120 mM) could cause necrosis of C. albicans and RPMI 1640 medium to a concentration of 2 × 106 cells/mL.
oxygen consumption in necrotic cells was negligible.30 The Various concentrations of compound 9a were added. The
morphological changes associated with ROS-induced death samples were cultured at 30 °C under constant shaking (200
caused by acetic acid in C. albicans also suggested that the rpm). A portion of the cell suspension was removed and counted
necrosis induced by compound 9a might be related to oxidative at the indicated time points (2, 4, 6, 9, 12, 24, and 48 h). DMSO
stress. The detailed antifungal mechanisms and molecular was added into the control group. Three independent
targets of compound 9a remain to be further explored. Also, the experiments were performed.
SAR and structural optimizations of compound 9a are ongoing Fungicidal Activity Assay. Time-kill curves were used to
to find more potent compounds for the treatment of CM. evaluate the fungicidal activity of compound 9a. The


experimental operation was carried out according to the
METHODS reported method.32 Exponentially growing C. neoformans H99
cells were washed with PBS (3 × 1 mL) and then resuspended
In vitro Antifungal Activity Assay. In vitro antifungal with RPMI 1640 medium to 2 × 106 cells/mL and divided into
activity was measured by the serial dilution method in 96-well different bottles. Different concentrations of the compound
microtest plates. C. albicans strain SC5314 and C. neoformans were added into the bottles, and the suspension was cultured at
strain H99 were used in this study. MIC was analyzed according 30 °C under constant shaking (200 rpm). A portion of the cell
to the recommendations of the Clinical and Laboratory suspension was removed at the indicated time points (12, 24, 36,
Standards Institute (CLSI, M27). RPMI 1640 (Gibco BRL) and 48 h) and diluted to appropriate folds. The diluent cell
was buffered with 0.165 M MOPS (Sigma), NaHCO3 (0.2%), suspension was plated on SDA agar medium to determine the
and NaOH (0.27%) as the test medium. All strains were concentration after incubation for 48 h at 30 °C.
routinely grown in YPD with 1% yeast extract (BD), 2% peptone In vitro Biofilm Formation Assay. The experimental
(BD), and 2% D-glucose (Sangon Biotech) liquid medium at 30 procedure was performed according to the reported method
°C in a shaking incubator. Briefly, the initial concentration of with a few modifications.32 Suspensions of C. neoformans H99
fungal suspensions in RPMI 1640 medium was 103 CFU/mL, cells (1.0 × 106 CFU/mL in RPMI 1640 medium) were added
and tested compounds were dissolved in DMSO serially diluted into a 96-well tissue culture plate (Corning, USA) and allowed
in growth medium. The yeast strains were incubated at 35 °C for to incubate at 37 °C for 90 min. After that, the upper culture
48 or 76 h. medium and the nonadherent cells were removed. The adherent
Growth Curve Assay. The growth curve assay was cells were washed with PBS three times. Fresh RPMI 1640
performed according to our reported method with a few medium with or without drugs was then added to adherent cells.
modifications.31 Briefly, exponentially growing C. neoformans The concentration for AMB was 64 μg/mL, and a drug-free
H99 cells were harvested and resuspended in 5 mL of fresh sample served as a control. Concentrations for compound 9a
1381 DOI: 10.1021/acsinfecdis.9b00086
ACS Infect. Dis. 2019, 5, 1376−1384
ACS Infectious Diseases Article

Figure 8. Therapeutic efficacy of compound 9a and FLC in mouse


models of cryptococcosis. C. neoformans brain burdens (in CFU/g of
brain) are shown for animals treated with the indicated dose of 9a and
FLC for 5 days after tail vein inoculation with 2 × 105 CFU/animal. P
values are from ANOVA. *P < 0.05, **P < 0.01, and ***P < 0.001.

Optical density at 490 nm (OD490) was determined using a


microtiter plate reader.
Melanin and Urease Production Assay. Exponentially
growing C. neoformans H99 cells were harvested, washed three
times with PBS, and resuspended in RPMI 1640 medium at 1 ×
106 cells/mL. Different concentrations of compound 9a and
FLC were added into the suspension. Cell suspensions
containing different concentrations of drugs were dropped
onto the L-DOPA or urea medium and incubated at 37 °C to
detect the production of melanin and urease.
Transmission Electron Microscopy. Transmission elec-
tron microscopy images of C. neoformans H99 cells were
obtained according to the previous protocol with a few
modifications.32 In the presence of compound 9a (0.062 and
2 μg/mL), C. neoformans H99 cells were collected after 8 h of
growth in liquid YPD medium, washed three times with PBS
Figure 7. Effect of compound 9a on cell cycle in C. neoformans H99 solution, and fixed at 4 °C for 24 h in 1 mL of 4%
cells. (A) Cell cycle analysis by flow cytometry. C. neoformans H99 cells paraformaldehyde solution. The samples were then washed
were treated for 24 h with different concentrations of compound 9a. with saline and fixed for 90 min with 1% phosphotungstic acid.
(B) Mean percentages of cells in G1 and G2. The percentages in
The fixed cells were dehydrated through a graded series of
different phases were calculated by flow cytometry software. Statistical
significance among groups was determined by analyses of variance. ethanol and embedded with EPON-812. Ultrathin sections were
Comparison between the drug-free treatment group and the compound prepared and observed after double staining with uranium under
9a treatment group was performed by the Student’s t-test. The data a transmission electron microscope (HITACHI H-800, Japan)
were shown as means ± SDs from three independent experiments. **P with 1 × 104 magnification.
< 0.01, compared with control; ***P < 0.001, compared with control. Cell Necrosis Detection by Flow Cytometry. The
experimental procedure was performed according to the
Table 4. Permeability (Pe × 10−6 cm/s) and Prediction of reported method with a few modifications.33 Exponentially
BBB Penetration of Compound 9a growing C. neoformans H99 cells were harvested, washed twice
with PBS, and resuspended in RPMI 1640 medium in 20 mL at 5
compound Pea (10−6 cm/s) predictionb
× 105 cells/mL. Different concentrations of compound 9a were
clonidine 3.71 ± 0.33 CNS+ added into the suspension. Each sample was centrifuged and
9a 7.91 ± 0.01 CNS+ washed with PBS three times. Each 50 mg of cells (wet weight)
a
Values are expressed as the mean ± SD of at least three independent was resuspended with 100 μL of PBS (containing 0.24%
experiments. bCompounds with Pe > 3.08 × 10−6 cm/s could cross snailase) and then incubated for 1 h at 30 °C to break the cell
the BBB by passive diffusion (CNS+). Compounds with Pe < 1.13 × wall. The sample was washed with PBS twice and fixed with 70%
10−6 cm/s could not cross the BBB (CNS−), and compounds with ethanol overnight. Then, the ethanol was removed. After the
1.13 × 10−6 cm/s < Pe < 3.08 × 10−6 cm/s show uncertain BBB
sample was resuspended in 400 μL of binding buffer, it was then
permeation (CNS±).
added to 5 μL of annexin V-FITC and incubated at room
temperature for 15 min. Then, 10 μL of propidium iodide (PI)
was added into the suspensions and incubated at room
were 0.062−8 μg/mL. Then, the plates were incubated for a temperature for another 15 min in the dark. The stained cells
further 24 h at 37 °C. A semiquantitative measure of biofilm were analyzed by a flow cytometer (Becton, Dickinson, San Jose,
formation was calculated by using an XTT reduction assay. CA).
1382 DOI: 10.1021/acsinfecdis.9b00086
ACS Infect. Dis. 2019, 5, 1376−1384
ACS Infectious Diseases


Article

Cell Cycle Analysis by Flow Cytometry. The preparation ACKNOWLEDGMENTS


procedure of the sample treated with different concentrations of This work was supported by the National Natural Science
compound 9a is similar to the step of the cell necrosis assay. The Foundation of China (Grant 81725020 to C.S.), the Innovation
cells were then stained with 50 μg/mL of PI at 4 °C for 30 min. Program of Shanghai Municipal Education Commission (Grant
The samples were sonicated to obtain separate cells. Data were 2019-01-07-00-07-E00073 to C.S.), and the Science and
collected using a FACSCalibur cytometer (Becton, Dickinson, Technology Commission of Shanghai Municipality (Grant
San Jose, CA) and analyzed with Cell Quest 3.0 software. 17XD1404700 to C.S.).


In Vitro Blood−Brain Barrier Permeation Assay. BBB
penetration of compound 9a was performed using a parallel REFERENCES
artificial membrane permeation assay (PAMPA) according to a
(1) Spitzer, M., Robbins, N., and Wright, G. D. (2017) Combinatorial
previous protocol.34 The acceptor 96-well microplate was filled strategies for combating invasive fungal infections. Virulence. 8 (2),
with 300 μL of a PBS/EtOH (7/3) solution, and the filter 169−185.
membrane was impregnated with 4 μL of porcine brain lipid (2) Brown, G. D., Denning, D. W., and Levitz, S. M. (2012) Tackling
(PBL) in dodecane (20 mg/mL). The compound was diluted to human fungal infections. Science 336 (6082), 647.
100 μg/mL with PBS/EtOH (7/3), and then, 200 μL was added (3) Denning, D. W., and Bromley, M. J. (2015) Infectious Disease.
to a donor well (PVDF membrane, pore size 0.45 mm). The How to bolster the antifungal pipeline. Science 347 (6229), 1414−1416.
acceptor 96-well microplate filled with 300 μL of PBS/EtOH (4) Liu, T. B., Perlin, D. S., and Xue, C. (2012) Molecular mechanisms
(7/3) was placed on the donor plate to form an interlayer, which of cryptococcal meningitis. Virulence. 3 (2), 173−181.
was allowed to stand at 25 °C for 12 h. After the incubation was (5) Park, B. J., Wannemuehler, K. A., Marston, B. J., Govender, N.,
complete, the donor plate was removed and the concentration of Pappas, P. G., and Chiller, T. M. (2009) Estimation of the current
global burden of cryptococcal meningitis among persons living with
compounds in the acceptor wells was determined by using a UV HIV/AIDS. AIDS 23 (4), 525−530.
plate reader (SpectraMax i3). Three replicates were performed (6) Sloan, D. J., and Parris, V. (2014) Cryptococcal meningitis:
for each group. epidemiology and therapeutic options. Clin. Epidemiol. 6, 169−182.
In Vivo Antifungal Potency Assay. Female ICR mice were (7) Rajasingham, R., Smith, R. M., Park, B. J., Jarvis, J. N., Govender,
purchased from JOINN Laboratories and were 20 to 25 g at the N. P., Chiller, T. M., Denning, D. W., Loyse, A., and Boulware, D. R.
time of experimentation. An inoculum of 2 × 105 CFU of (2017) Global burden of disease of HIV-associated cryptococcal
C. neoformans in 0.2 mL of normal saline (NS) was used for each meningitis: an updated analysis. Lancet Infect. Dis. 17 (8), 873−881.
mouse. FLC (2 mg/kg in NS) and compound 9a (10 mg/kg (8) Williamson, P. R., Jarvis, J. N., Panackal, A. A., Fisher, M. C.,
suspended in NS with 1.5% glycerin and 0.5% Tween 80) were Molloy, S. F., Loyse, A., and Harrison, T. S. (2017) Nat. Rev. Neurol. 13
injected intraperitoneally 2 h after inoculation. The compound (1), 13−24.
(9) Coelho, C., and Casadevall, A. (2016) Cryptococcal therapies and
was administered continuously for 5 days. Groups of mice were drug targets: the old, the new and the promising. Cell. Microbiol. 18 (6),
sacrificed 5 days after continuous administration. The brains 792−799.
were removed and homogenized. Serial 10 000-fold dilutions (10) Perfect, J. R. (2017) The antifungal pipeline: a reality check. Nat.
were plated on YPD agar supplemented with chloramphenicol Rev. Drug Discovery 16 (9), 603−616.
and incubated in air at 30 °C for at least 48 h to enumerate the (11) Liu, N., Tu, J., Dong, G., Wang, Y., and Sheng, C. (2018)
total fungal burden. The control group consisted of mice treated Emerging New Targets for the Treatment of Resistant Fungal
with NS. The concentration (CFU/mL) of each brain was Infections. J. Med. Chem. 61 (13), 5484−5511.
calculated, and the differences between groups were analyzed by (12) Liu, N., Wang, C., Su, H., Zhang, W., and Sheng, C. (2016)
analysis of variance (ANOVA). Strategies in the discovery of novel antifungal scaffolds. Future Med.


Chem. 8 (12), 1435−1454.
(13) Samantaray, S., Correia, J. N., Garelnabi, M., Voelz, K., May, R.
ASSOCIATED CONTENT C., and Hall, R. A. (2016) Novel cell-based in vitro screen to identify
*
S Supporting Information small-molecule inhibitors against intracellular replication of Crypto-
The Supporting Information is available free of charge on the coccus neoformans in macrophages. Int. J. Antimicrob. Agents 48 (1),
ACS Publications website at DOI: 10.1021/acsinfec- 69−77.
(14) Nixon, G. L., McEntee, L., Johnson, A., Farrington, N., Whalley,
dis.9b00086. S., Livermore, J., Natal, C., Washbourn, G., Bibby, J., Berry, N., Lestner,
Potency of compound 8a; time-kill curves and water J., Truong, M., Owen, A., Lalloo, D., Charles, I., and Hope, W. (2018)
solubility of compound 9a; supplementary in vivo results; Repurposing and Reformulation of the Antiparasitic Agent Flubenda-
detailed procedure for chemical synthesis and spectral zole for Treatment of Cryptococcal Meningoencephalitis, a Neglected
data of targets compounds; HPLC spectra of representa- Fungal Disease. Antimicrob. Agents Chemother. 62 (4), No. e01909-17.
(15) Di Santo, R. (2010) Natural products as antifungal agents against
tive compounds (PDF) clinically relevant pathogens. Nat. Prod. Rep. 27 (7), 1084−1098.


(16) Jiang, Z., Liu, N., Dong, G., Jiang, Y., Liu, Y., He, X., Huang, Y.,
AUTHOR INFORMATION He, S., Chen, W., Li, Z., Yao, J., Miao, Z., Zhang, W., and Sheng, C.
(2014) Scaffold hopping of sampangine: discovery of potent antifungal
Corresponding Author lead compound against Aspergillus fumigatus and Cryptococcus
*E-mail: shengcq@smmu.edu.cn (C.S.). neoformans. Bioorg. Med. Chem. Lett. 24 (17), 4090−4094.
(17) Jiang, Z., Liu, N., Hu, D., Dong, G., Miao, Z., Yao, J., He, H., Jiang,
ORCID
Y., Zhang, W., Wang, Y., and Sheng, C. (2015) The discovery of novel
Chunquan Sheng: 0000-0001-9489-804X antifungal scaffolds by structural simplification of the natural product
Author Contributions sampangine. Chem. Commun. 51 (78), 14648−14651.
∥ (18) Liu, N., Zhong, H., Tu, J., Jiang, Z., Jiang, Y., Jiang, Y., Jiang, Y., Li,
Z.L. and N.L. contributed equally to this work.
J., Zhang, W., Wang, Y., and Sheng, C. (2018) Discovery of simplified
Notes sampangine derivatives as novel fungal biofilm inhibitors. Eur. J. Med.
The authors declare no competing financial interest. Chem. 143, 1510−1523.

1383 DOI: 10.1021/acsinfecdis.9b00086


ACS Infect. Dis. 2019, 5, 1376−1384
ACS Infectious Diseases Article

(19) Li, Z., Liu, N., Tu, J., Ji, C., Han, G., Wang, Y., and Sheng, C.
(2019) Discovery of novel simplified isoxazole derivatives of
sampangine as potent anti-cryptococcal agents. Bioorg. Med. Chem. 27
(5), 832−840.
(20) Tu, J., Li, Z., Jiang, Y., Ji, C., Han, G., Wang, Y., Liu, N., and
Sheng, C. (2019) Discovery of Carboline Derivatives as Potent
Antifungal Agents for the Treatment of Cryptococcal Meningitis. J.
Med. Chem. 62 (5), 2376−2389.
(21) Cenci, E., Bistoni, F., Mencacci, A., Perito, S., Magliani, W.,
Conti, S., Polonelli, L., and Vecchiarelli, A. (2004) A synthetic peptide
as a novel anticryptococcal agent. Cell. Microbiol. 6 (10), 953−961.
(22) Pini, G., Faggi, E., and Campisi, E. (2017) Enzymatic
characterization of clinical and environmental Cryptococcus neofor-
mans strains isolated in Italy. Rev. Iberoam. Micol. 34 (2), 77−82.
(23) Garvey, E. P., Sharp, A. D., Warn, P. A., Yates, C. M., and
Schotzinger, R. J. (2018) The novel fungal CYP51 inhibitor VT-1598 is
efficacious alone and in combination with liposomal amphotericin B in
a murine model of cryptococcal meningitis. J. Antimicrob. Chemother. 73
(10), 2815−2822.
(24) Dickey, S. W., Cheung, G. Y. C., and Otto, M. (2017) Different
drugs for bad bugs: antivirulence strategies in the age of antibiotic
resistance. Nat. Rev. Drug Discovery 16 (7), 457−471.
(25) Pierce, C. G., and Lopez-Ribot, J. L. (2013) Candidiasis drug
discovery and development: new approaches targeting virulence for
discovering and identifying new drugs. Expert Opin. Drug Discovery 8
(9), 1117−1126.
(26) Ramage, G., Rajendran, R., Sherry, L., and Williams, C. (2012)
Fungal biofilm resistance. Int. J. Microbiol. 2012, 528521.
(27) Brilhante, R. S. N., Espana, J. D. A., de Alencar, L. P., Pereira, V.
S., Castelo-Branco, D., Pereira-Neto, W. A., Cordeiro, R. A., Sidrim, J. J.
C., and Rocha, M. F. G. (2017) An alternative method for the analysis of
melanin production in Cryptococcus neoformans sensu lato and
Cryptococcus gattii sensu lato. Mycoses. 60 (10), 697−702.
(28) Kothamasi, D., Wannijn, J., Van Hees, M., Nauts, R., Van
Gompel, A., Vanhoudt, N., and Vandenhove, H. (2019) Exposure to
ionizing radiation affects the growth of ectomycorrhizal fungi and
induces increased melanin production and increased capacities of
reactive oxygen species scavenging enzymes. J. Environ. Radioact. 197,
16−22.
(29) Kim, J. H., Lee, H. O., Cho, Y. J., Kim, J., Chun, J., Choi, J., Lee, Y.,
and Jung, W. H. (2014) A vanillin derivative causes mitochondrial
dysfunction and triggers oxidative stress in Cryptococcus neoformans.
PLoS One 9 (2), No. e89122.
(30) Phillips, A. J., Sudbery, I., and Ramsdale, M. (2003) Apoptosis
induced by environmental stresses and amphotericin B in Candida
albicans. Proc. Natl. Acad. Sci. U. S. A. 100 (24), 14327−14332.
(31) Huang, Y., Dong, G., Li, H., Liu, N., Zhang, W., and Sheng, C.
(2018) Discovery of Janus Kinase 2 (JAK2) and Histone Deacetylase
(HDAC) Dual Inhibitors as a Novel Strategy for the Combinational
Treatment of Leukemia and Invasive Fungal Infections. J. Med. Chem.
61 (14), 6056−6074.
(32) Wang, S., Wang, Y., Liu, W., Liu, N., Zhang, Y., Dong, G., Liu, Y.,
Li, Z., He, X., Miao, Z., Yao, J., Li, J., Zhang, W., and Sheng, C. (2014)
Novel carboline derivatives as potent antifungal lead compounds:
design, synthesis, and biological evaluation. ACS Med. Chem. Lett. 5 (5),
506−511.
(33) Li, D. D., Xu, Y., Zhang, D. Z., Quan, H., Mylonakis, E., Hu, D. D.,
Li, M. B., Zhao, L. X., Zhu, L. H., Wang, Y., and Jiang, Y. Y. (2013)
Fluconazole assists berberine to kill fluconazole-resistant Candida
albicans. Antimicrob. Agents Chemother. 57 (12), 6016−6027.
(34) Xu, Y. X., Wang, H., Li, X. K., Dong, S. N., Liu, W. W., Gong, Q.,
Wang, T. D., Tang, Y., Zhu, J., Li, J., Zhang, H. Y., and Mao, F. (2018)
Discovery of novel propargylamine-modified 4-aminoalkyl imidazole
substituted pyrimidinylthiourea derivatives as multifunctional agents
for the treatment of Alzheimer’s disease. Eur. J. Med. Chem. 143, 33−47.

1384 DOI: 10.1021/acsinfecdis.9b00086


ACS Infect. Dis. 2019, 5, 1376−1384

You might also like