You are on page 1of 9

Chemico-Biological Interactions 362 (2022) 109998

Contents lists available at ScienceDirect

Chemico-Biological Interactions
journal homepage: www.elsevier.com/locate/chembioint

Research paper

Structurally diverse steroids from an endophyte of Aspergillus tennesseensis


1022LEF attenuates LPS-induced inflammatory response through the
cholinergic anti-inflammatory pathway
Jun-Cheng Su a, Qianrong Pan b, Xingyuan Xu a, Xia Wei d, Xueping Lei b, **, Peng Zhang c, *
a
State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
b
Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory
Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
c
Tobacco Research Institute of Chinese Academy of Agricultural Sciences, Qingdao, 266101, China
d
Pharmaceutical College, Guangxi Medicinal University, Nanning, 530021, China

A R T I C L E I N F O A B S T R A C T

Keywords: The emerging cholinergic anti-inflammatory pathway plays a key role in regulating inflammation. Steroids are
Steroids known to possess remarkable anti-inflammatory activity. However, the links between steroids and the cholin­
Acetylcholinesterase ergic anti-inflammatory pathway remain unidentified. In this study, eight steroids (1–8) featuring five different
Inflammation
structural types were characterized from an endophytic fungus Aspergillus tennesseensis 1022LEF, and were
The cholinergic anti-inflammatory pathway
subsequently evaluated for their potential role in regulating the cholinergic anti-inflammatory pathway. As a
result, compound 8, with the best potency, showed remarkable anti-inflammatory activity at the nanomolar to
low micromolar level. Further pharmacological study indicated that 8 notably increased α7nAchR expression and
inhibited the activation of its down-stream signaling pathways. Collectively, the present study not only high­
lighted the potential correlation between steroids and the cholinergic anti-inflammatory pathway, but also
identified 8 as a dual-functional modulator via directly inhibition to acetylcholinesterase as well as up-regulation
of α7nAchR expression.

1. Introduction of tumor necrosis factor such as TNF-α and other cytokines [7].
Steroids represent a remarkable group of evolutionarily conserved
Acetylcholinesterase (ACchE) is a serine hydrolase that hydrolyzes lipid natural products. While most of the steroids feature a classical
neurotransmitter acetylcholine into acetic acid and choline [1]. Except cyclopenta[a]phenanthrene core, in some cases, these molecules may
for its “classical” role in synaptic transmission, AchE also possesses also act as biogenetic precursors to provide additional skeleton types via
several “non-classical” functions in non-neuronal cells, and is known to different biosynthetic steps [8]. Meanwhile, steroids are the major
regulate Alzheimer’s disease (AD), tumorigenesis, glaucoma, and sources of anti-inflammatory drugs, as representative by dexametha­
myasthenia gravis [2,3]. Therefore, it is not surprising that AChE has sone, a glucocorticoid receptor agonist [9]. However, despite of the fact
become an attractive target from the pharmaceutical community, which that the anti-inflammatory effects of steroids are well-documented, few
resulted in several FDA approved drugs, especially for the treatment of studies had correlated these phenotypes with the cholinergic
AD [4,5]. Recently, emerging evidences had demonstrated that AchE anti-inflammatory pathway. Therefore, it is of great significance to
dysfunction could lead to inflammatory disorders, indicating that validate whether this remarkable group of natural products possess an
acetylcholinesterase inhibitors (AchEIs) could be served as promising alternative anti-inflammatory mechanism.
anti-inflammatory agents [6]. This pathway, termed “the cholinergic As part of our ongoing efforts to discover bioactive secondary me­
anti-inflammatory pathway”, is mediated by the α7 subunit of the tabolites from endophytic fungi [10–13], eight steroids belonging to five
nicotinic acetylcholine (Ach) receptor (α7nAChR) to inhibit the release distinct skeletons were isolated from a tobacco-derived endophytic

* Corresponding author.
** Corresponding author.
E-mail addresses: xuepinglei@gzhmu.edu.cn (X. Lei), zhangpeng@caas.cn (P. Zhang).

https://doi.org/10.1016/j.cbi.2022.109998
Received 13 April 2022; Received in revised form 17 May 2022; Accepted 28 May 2022
Available online 29 May 2022
0009-2797/© 2022 Elsevier B.V. All rights reserved.
J.-C. Su et al. Chemico-Biological Interactions 362 (2022) 109998

fungus Aspergillus tennesseensis 1022LEF (Fig. 1). The structures of the Table 1
1
isolated compounds were established by comprehensive analysis of their H and 13C NMR spectroscopic data of 1 (CDCl3, J in Hz)a.
UV, HRMS, 1D and 2D NMR data, quantum chemical calculation, as well no. δC, type δH
as comparison with those reported in literatures. To uncover the buried
1 35.0, CH2 a 2.17, m
links between steroids, AchE, and the cholinergic anti-inflammatory b 2.00, m
pathway, a series of enzyme-based, cell-based, and in silico experi­ 2 38.1, CH2 a 2.54, m
ments were performed. The most potent compound 8, with a highly b 2.20, m
degraded C21 skeleton, was identified as a dual-functional cholinergic 3 209.5, C
4 43.8, CH2 a 2.44
anti-inflammatory pathway modulator via direct binding and inhibition b 2.04, m
to AchE as well as up-regulation of α7nAchR expression simultaneously. 5 42.7, CH 2.31, m
6 42.2, CH2 a 2.50, m
2. Results and discussion b 2.07, m
7 198.6, C
8 134.9, C
2.1. Structural elucidation 9 162.4, C
10 38.7, C
The molecular formula of tennessoid A (1) was deduced as C28H42O3 11 65.2, CH 4.46, dd (7.1, 6.7)
by its HRESIMS ion peak at 425.3048 [M − H]− (calcd for C28H41O3, 12 49.0, CH2 a 2.25, m
b 1.63, dd (13.7, 4.5)
425.3056), corresponding to 8 degrees of unsaturation (DOUs). The UV 13 46.3, C
spectrum of 1 displayed the absorption maxima at 192, 215, and 246 14 46.4, CH 2.43
nm, suggesting the occurrence of conjugated system in 1. The 1H and 13C 15 28.9, CH2 a 1.85, m
NMR spectra of 1 showed signals corresponding to two keto carbonyl b 1.23, m
16 24.8, CH2 a 2.15
groups (δC 209.5 and 198.6), one tetra-substituted double bond (δC
b 1.47, m
162.4 and 134.9), one terminal double bond [δH 4.71 (1H, s) and 4.65 17 54.8, CH 1.26, m
(1H, s); δC 156.2 and 106.9], one oxygenated methine [δH 4.46 (1H, dd, 18 14.1, CH3 0.53, s
J = 7.1 and 6.7 Hz); δC 65.2], and five methyl groups [δH 1.30 (3H, s), 19 16.6, CH3 1.30, s
1.00 (3H, d, J = 6.8 Hz), 0.99 (3H, d, J = 6.8 Hz), 0.95 (3H, d, J = 6.4 20 35.9, CH 1.35, m
21 18.8, CH3 0.95, d (6.4)
Hz), and 0.53 (3H, s); δC 22.2, 22.1, 18.8, 16.6, and 14.1]. The above-
22 34.4, CH2 a 1.51, m
mentioned functionalities accounted for 4 of 8 DOUs, which required b 1.12, m
1 to possess a tetracyclic skeleton. The full assignments of the 1H and 13C 23 30.9, CH2 a 2.08
NMR spectroscopic data of 1 were approached by 1H–1H COSY, HSQC, b 1.88, m
24 156.2, C
and HMBC experiments (Table 1).
25 33.6, CH 2.20
Five spin systems, namely, H2-1 to H2-2, H2-4 to H2-6, H-11 to H2-12, 26 22.2, CH3 1.00, d (6.8)
H-14 to H3-21/H2-23, and H3-26 to H3-27, could be deduced from the 27 22.1, CH3 0.99, d (6.8)
1
H–1H COSY spectrum of 1 (Fig. 2). In its HMBC spectrum, correlations 28 106.9, CH2 a 4.71, s
from H2-1 to C-3/C-9, from H2-2 to C-4, from H2-4 to C-10, from H-5 to b 4.65, s

C-7, from H2-6 to C-8, from H2-12 to C-9/C-14/C-17, from H-14 to C-7, a
Overlapped signals are reported without designating multiplicity.
from H2-28 to C-23/C-25, as well as from H3-26/H3-27 to C-24 led to the

Fig. 1. Chemical structures of compounds 1–8.

2
J.-C. Su et al. Chemico-Biological Interactions 362 (2022) 109998

Fig. 3. Experimental and calculated ECD spectra of 1.

AchE inhibitors for developing potential anti-inflammatory agents. To


approach this goal, we firstly evaluated the inhibitory activity of these
Fig. 2. Key 1H–1H COSY, HMBC, and NOE correlations of 1. compounds against AchE at the enzyme level by using the Ellman assay.
As shown in Fig. 4A, at the concentration of 50 μM, compounds 5 and 8,
establishment of a 6/6/6/5 tetra-carbocyclic core, with a C9 side chain with a rearranged or a highly degraded ergostane skeleton, showed
located at C-17. Next, based on the HMBC cross peaks from H3-18 to C- significant inhibitory effect against AchE, followed by the commonly
14/C-17, from H3-19 to C-1/C-5, Me-18 and Me-19 were deduced to be reported ergosteroid 2. Although these compounds belong to steroids of
located at C-13 and C-10, respectively. Moreover, on the basis of the different structural subtypes, the presence of an α,β-unsaturated ketone
characteristic down-fielded chemical shift of C-11 (δC 65.2), as well as moiety at the southwestward of the molecule seem to be critical for the
the molecular formula information, a hydroxyl group was placed at C- inhibitory effect. Among them, compound 8 was identified as the most
11. Hence, the planar structure of 1 was established to be a new potent inhibitor with IC50 value of 11.16 ± 2.79 μM (Fig. 4C), followed
ergosterol-type steroid as shown in Fig. 2. by compound 5 (IC50 = 27.12 ± 1.27 μM).
The stereochemistry of 1 was established by combined analysis of its
NOESY spectrum as well as quantum chemical calculations. In detail, 2.3. Molecular docking
NOE correlations between H3-19 and H-1a/H-6a/H-11, between H-11/
H3-18, and between H3-18 and H-15b/H-20 indicated that these protons To further understand the possible modes-of-action of these steroid
were located at the same side of the 6/6/6/5 tetracyclic skeleton, while AchE inhibitors, the binding modes of compounds 8, 5, and 2 with AchE
NOE correlations between H-1b and H-5, between H-14 and H-17 sug­ were obtained by molecular docking. As disclosed from Fig. 5, all these
gested that these atoms were located at the opposite orientation. The steroids fitted well with the same peripheral anionic site of AchE, with
above conclusions were further confirmed by comparing the NMR data the C-17 aliphatic side chains extended into the deep of the pocket to
of 1 with those reported in literatures, which was in consistance with the generate hydrophobic interactions with Tyr72, Tyr124, Val294, Phe295,
reported ergosteroids [14,15]. Furthermore, the absolute configuration and Phe297, whereas the α,β-unsaturated ketone moieties faced the
of 1 was established by TDDFT ECD calculation at PCM/B3LYP/6-31+G solvent region. Compound 8, the most potent inhibitor, is anchored at
(d) level in methanol. With a good agreement between the calculated the pocket entrance through two hydrogen bonds. The imidazole side­
ECD spectrum of 5R,10S,11R,13R,14R,17R,20R-1 and the experimental chain of His287 forms a hydrogen bond with the carbonyl group of
curve, the aboslute configuration of 1 was assigned as 5R,10S,11R,13R, compound 8, and the phenol sidechain of Tyr341 is hydrogen-bonded to
14R,17R,20R (Fig. 3). the aliphatic alcohol of compound 8 through a water bridge (Fig. 5A). By
Compounds 2–8 were identified as ergosta-4,6,8(14),22-tetraen-3- contrast, compound 5 showed only one hydrogen-bonding interaction
one (2) [16,17], 5α,8α-epidioxyergosta-6,22-dien-3β-ol (3) [18], dan­ with Tyr341 (Fig. 5B), whereas compound 2 had no obvious interactions
kasterone B (4) [19], dankasterone A (5) [20,21], herbarulide (6) [22], with either His287 or Tyr341 (Fig. 5C). These observations were in
7-nor-ergosterolide (7) [15], and demethylincisterol A3 (8) [23] by accordance with the result of the AchE inhibition assay, and may pro­
comparing of their NMR data with those reported in the literatures. vide an alternative view to understand why compound 8 possessed the
Among these compounds, 2 and 3 shared a classical cyclopenta[a] best inhibitory activity. Thus, compound 8 was selected as representa­
phenanthrene core with an intact C28 skeleton. 4 and 5 are rearranged tive compound for further pharmacological research.
steroids with a rare 13(14 → 8)abeo-8-ergostane skeleton. Compound 6
belongs to an unusual class of 5,6-epoxy-5,6-seco-ergosteroid and com­
2.4. Compound 8 inhibits for nitric oxide (NO) production and
pound 7 is a C27 7-oxa-ergosteroid. Compound 8 features a highly
proinflammatory cytokine expression
degraded steroid scaffold with a C21 skeleton. Except for 2 and 3, all
these known steroids are rarely found in nature, despite of their clear
As described above, compound 8 inhibits AchE via direct binding to
biogenetic relationship with the classical ergosteroids.
the active site. To validate the anti-inflammatory effect of compound 8
at the cellular level, we measured the LPS-induced NO production and
2.2. Compound 8 is an effective AchE inhibitor proinflammatory cytokine expression in RAW264.7 cells with the
treatment of compound 8. As shown in Fig. 6A, compound 8 had a strong
Based on the simultaneous discovery of eight steroids featuring five inhibitory effect against LPS-induced NO production, a critical signaling
distinct skeletons, we wondered whether steroids could serve as novel molecule during inflammation process. The suppressive effect of

3
J.-C. Su et al. Chemico-Biological Interactions 362 (2022) 109998

Fig. 4. The effect of compounds on AchE activity. (A) The effect of compounds 1–8 on AchE activity at 50 μM. Tacrine (TA) was set as positive control with final
concentration of 0.333 μM. (B) The AchE inhibition curves of compound 5 and (C) compound 8. The data are presented as mean ± SD.

Fig. 5. In silico analysis of the interactions of compounds 8 (A), 5 (B), and 2 (C) with AchE protein (PDB: 6F25).

compound 8 was in a dose dependent manner, with inhibitory ratio of significantly reduced TNF-α and IL-6 production in a dose dependent
40%, 60%, and 91% at 0.625 μM, 1.25 μM and 2.5 μM, respectively. manner (Fig. 6B). Similarly, RT-qPCR assay also confirmed that com­
Notably, no obvious cytotoxicity was observed at these concentrations. pound 8 treatments obviously reduced the mRNA of TNF-α and IL-6 in a
The secretions of IL-6 and TNF-α, two crucial proinflammatory media­ dose dependent manner (Fig. 6C). All these results showed that com­
tors, were also measured in after co-incubation with 8 (Fig. 6B). LPS pound 8 was a potential anti-inflammatory agent.
stimulation dramatically (P < 0.001) increased TNF-α and IL-6 secretion
in comparison with the control group, whereas compound 8 treatment

4
J.-C. Su et al. Chemico-Biological Interactions 362 (2022) 109998

Fig. 6. Compound 8 suppressed LPS-induced NO production and inflammatory factors expression. (A) Compound 8 reduced LPS-induced NO production in RAW
246.7 cells. The RAW 246.7 cells were stimulated with 20 ng/mL and different concentration of compound 8 for 24 h. The condition medium was collected and
subjected for NO production. (B&C) Compound 8 downregulated LPS-induced inflammatory cytokines in RAW 246.7 cells. The cells were treated with different
concentration of compound 8, the condition medium was collected for Elisa assay (B). The cells were collected and subjected for RT-qPCR assay (C). The data are
presented as mean ± SD, ***P < 0.001 compared with the control group, #P < 0.05 and ###P < 0.001 compared with LPS group.

2.5. Compound 8 suppresses LPS-induced activation NF-κB and Stat3 stimulation obviously reduced α7nAchR expression, but compound 8
signaling pathway in RAW 246.7 cells significantly increased α7nAchR expression in a dose dependent manner
(Fig. 8A). Moreover, we found that methyllycaconitine pretreatment, an
Since NF-κB transcription factors and the corresponding signaling inhibitor of α7nAchR [30], obviously attenuated compound 8 mediated
pathways are central regulator for inflammation response [24,25], we inhibitory effect on the secretion of TNF-α and IL-6 (Fig. 8B). Taken
evaluated the effect of compound 8 on the critical members of NF-κB these together, our result indicated that compound 8 might suppress
signaling pathway using Western blotting assay, including IKKα, IKKβ, LPS-induced inflammation by increasing α7nAchR expression.
p-IKKα/βSer176/180, IκBα, p-IκBαSer32, NF-κB p65, p–NF–κB p65Ser536. We
found that LPS stimulation significantly increased the phosphorylation 3. Conclusions
of IKKa/β, IκBα, NF-κB p65, which indicates the activation of NF-κB
signaling pathway [26]. Whereas, compound 8 treatments obviously Although steroids are known to possess remarkable anti-
reduced LPS-induced up-regulation of p-IKKα/βSer176/180, p-IκBαSer32, inflammatory activity, either as endogenous molecules or exogenous
p–NF–κB p65Ser536 without affecting the total IKKa/β, IκBα, and NF-κB drugs, the targets and pharmacological mechanisms of these small
p65 expression (Fig. 7). We also found that compound 8 treatments molecules are not fully understood. In this study, eight steroids featuring
significantly suppressed LPS-induced phosphorylation of Stat3, another five different structural types were isolated from a tabacco-derived
regulator of inflammation [27]. These results suggested that compound fungus A. tennesseensis. To seek whether steroids possess an alternative
8 suppressed LPS-induced activation NF-κB and Stat3 signaling anti-inflammatory mechanism beyond glucocorticoid receptor activa­
pathway. tion, compound 8 was selected as a chemical probe for further mecha­
nism investigation. As a result, compound 8 strongly suppressed LPS-
2.6. Compound 8 increases alpha7 nicotinic acetylcholine receptor induced NO production and inflammatory factors expression,
(α7nAchR) expression including TNF-α and IL-6. In addition, treatment with compound 8
notably increased α7nAchR expression and inhibited the phosphoryla­
Considering the fact that compound 8 exhibited much greater ac­ tion of its down-stream NF-κB and Stat3 signaling pathway. Moreover,
tivity in cells than at the enzyme level, we speculated that compound 8 in the competitive experiments, the inhibitory effect on TNF-α and IL-6
may possess an alternative anti-inflammatory mechanism apart from secretion mediated by compound 8 was obviously attenuated by the
direct binding and inhibition of AchE. It had been demonstrated that the α7nAchR antagonist. Taken together, we speculate that the anti-
activation of α7nAChR, a major receptor of Ach, had obvious inhibitory inflammatory effect of compound 8 may due to its AchE suppressive
effects on LPS-mediated NF-κB and Stat3 signaling pathways [28,29]. effect by direct binding to the enzyme and up-regulation of α7nAchR
Thus, we detected the effect of compound 8 on α7nAchR expression expression.
using Western blotting assay. Surprisingly, the results showed that LPS

5
J.-C. Su et al. Chemico-Biological Interactions 362 (2022) 109998

Fig. 7. Compound 8 inhibited LPS-induced activation


of NF-kB and Stat3 signaling pathway. The RAW
246.7 cells were exposed with 20 ng/mL LPS for 24 h,
and then treated with or without various concentra­
tions of compound 8 for 24 h. And then, the cells were
collected and subjected for Western blotting assay to
detect several critical regulators of NF-κB and Stat3
signaling pathway. β-actin was set as loading control.
The representative blots were presented in (A), the
quantitative data of blots was measured using Image J
and showed in (B). The data are presented as mean ±
SD, ***P < 0.001 compared with the control group,
#
P < 0.05, ##P < 0.01 and ###P < 0.001 compared
with LPS group.

4. Experimental section 4.2. Fungal material

4.1. General experimental procedures The candidate fungal strain Aspergillus tennesseensis 1022LEF was
previously isolated from healthy leaves of cultivated tobacco, Nicotiana
Optical rotations were acquired using a Jasco P-1020 digital polar­ tabacum L., which was collected from Hubei, China
imeter (Jasco, Tokyo, Japan) with MeOH as solvent. The UV measure­ (108◦ 23′ 12′′ –110◦ 38′ 08′′ E, 29◦ 07′ 10′′ –31◦ 24′ 13′′ N). This fungal strain
ments were performed on a Shimadzu UV-2700 spectrophotometer was taxonomically identified based on the molecular protocol by
(Shimadzu, Kyoto, Japan). The ECD spectrum was obtained by a sequencing of the ITS region of the rDNA. A BLAST search result
Chirascan-plus Circular Dichroism Spectrometers (Applied Photo­ demonstrated that the submitted sequence was most similar (99%) to
physics, Leatherhead, UK). 1D (1H and 13C) and 2D (HSQC, HMBC, that of A. tennesseensis. This fungus was then assigned the GenBank
COSY, NOESY) NMR spectra were recorded on an Agilent DD2 NMR number of MW898422 and was deposited in the China General Micro­
spectrometer (Agilent Technologies, Waldbronn, Germany) operating at biological Culture Collection Center (CGMCC) with a deposition number
500 MHz for the 1H channel and 125 MHz for the 13C channel. Chemical of 21469.
shifts were referenced to the residual solvent values of 7.26/77.16 ppm
(1H/13C) for CDCl3 and 2.50/39.52 ppm (1H/13C) for DMSO‑d6, 4.3. Fermentation, extraction, and purification
respectively. The HRESIMS data were obtained with a Waters ACQUITY
UPLC I-Class-Vion IMS Q-TOF mass spectrometer (Waters Corp., Mas­ Large-scale fermentation of A. tennesseensis 1022LEF was statically
sachusetts, America). Silica gel (200–300 mesh, Haiyang Chemical performed at 28 ◦ C in a total of 150 of 1 L Erlenmeyer flasks, with 300
Factory, Qingdao, China), Lobar LiChroprep RP-18 (40–60 μm, Merck, mL of commercially available potato dextrose broth medium (Solarbio
Darmstadt, Germany), and Sephadex LH-20 (Merck) were applied for Life Sciences CO., LTD., Beijing, China) in each flask. Since the myce­
column chromatography. Preparative thin-layer chromatography was lium was grown slowly in medium, the cultivation was extended to 50
performed with precoated TLC plates (GF254, Haiyang Chemical Fac­ days. Subsequently, the mycelium and the broth were filtered and
tory). All solvents used for extraction and purification were of either extracted exhaustively with EtOAc to afford 20.2 g of crude extracts. The
analytical or HPLC purity. obtained extracts were partitioned between petroleum ether (PE) and
MeOH. The MeOH fractionation was subjected to silica gel vacuum
liquid chromatography with mixtures of PE–EtOAc (from 10:1 to 1:1, v/
v) and CH2Cl2–MeOH (from 20:1 to 5:1) gradient systems. As a result,

6
J.-C. Su et al. Chemico-Biological Interactions 362 (2022) 109998

Fig. 8. Compound 8 mediated inhibitory effects on


LPS-induced inflammation may partly dependent on
α7nAchR activation. (A) Compound 8 increased the
expression of α7nAchR. (B) Methyllycaconitine
treatment (100 nM) significantly weaken the inhibi­
tory effect of compound 8 (1.25 μM) on LPS-induced
inflammation. After indicated treatment, the culture
supernatant was collected and applied for Elisa assay
to detect TNF-α and IL-6 secretion. The data are
presented as mean ± SD, ***P < 0.001 compared
with the control group, #P < 0.05, ##P < 0.01 and
###
P < 0.001 compared with LPS group, &&P < 0.01
compared with compound 8 group.

three major fractions, Fr.1–Fr.3, were obtained and combined by TLC 4.5. Molecular docking
and HPLC analysis. Fr.1 (4.8 g), eluted with PE–EtOAc 2:1 and 1:1, was
refractionated by reversed-phase column chromatography (CC) eluting The ligand structures used for molecular docking were generated
with a stepped MeOH–H2O gradient system (from 1:9 to 10:0) to afford with ChemDraw and prepared by LigPrep module of Glide in
six subfractions, Fr.1.1–Fr.1.6. Fr.1.2 was chromatographed on silica gel Schrödinger. The receptor structure (PDB ID: 6F25) was downloaded
CC (CH2Cl2–MeOH as the mobile phase, from 30:1 to 20:1) to afford 4 from the PDB Bank (http://www.rcsb.org) and prepared by hydroge­
(5.8 mg) and 8 (13.6 mg). The new compound 1 (7.0 mg) was isolated nation and dewatered through Protein Preparation Wizard module. The
from Fr.1.3 through prep.-TLC (plate: 20 × 20 cm; developing solvents: binding site used for docking was generated by Receptor Grid Genera­
PE–acetone, 5:1), while 7 (8.8 mg) was obtained from Fr.1.6 by tion module and the site size was controlled at 15 Å × 15 Å × 15 Å. The
Sephadex LH-20 with CH2Cl2–MeOH 1:1 as the mobile phase. Fr.2 (2.4 dockings were carried out with Ligand Docking module. The in­
g) eluting with CH2Cl2–MeOH 20:1 was subjected to silica gel CC teractions between protein and ligands were analyzed by Pymol (version
(CH2Cl2–MeOH, from 50:1 to 20:1), and then followed by prep.-TLC 2.2.2).
(CH2Cl2-acetone, 20:1) and Sephadex LH-20 to yield compounds 5
(9.0 mg), 6 (3.9 mg), and 2 (7.7 mg), respectively. Finally, Fr.3 (1.8 g, 4.6. Anti-inflammatory activity
eluted with CH2Cl2–MeOH 10:1) was purified by silica gel CC
(PE–EtOAc, from 10:1 to 2:1) to yield 3 (26.8 mg). 4.6.1. Reagents
Tennessoid A (1): Colorless oil; [α] +25.8 (c = 0.10, MeOH); UV The Lipopolysaccharide (LPS) was purchased from Sigma-Aldrich (St
(MeOH) λmax (log ε) 192 (3.22), 215 (3.69), 246 (3.98) nm; CD (MeOH): Louis, MO, USA). Radioimmunoprecipitation assay (RIPA) buffer and
λmax (Δε) = 200 (13.75), 237 (− 19.48), 282 (2.72); (+)-HRESIMS m/z Griess reagent for NO assay kit were supplied by Yeasen Biotech
427.3209 [M + H]+ (calcd for C28H43O3, 427.3212), (− )-HRESIMS m/z (Shanghai, China). Quantikine Elisa kit of TNF-a and IL-6 were obtained
425.3048 [M − H]− (calcd for C28H41O3, 425.3056); For 1H and 13C from NeoBioscience (Shenzhen, China). Antibodies against IKKα, IKKβ,
NMR data, see Table 1. p-IKKa/βSer176/180, IκBα, p-IκBαSer32, NF-κB p65, p–NF–κB p65Ser536,
Stat3, p-Stat3Tyr705 and HRP-conjugated anti-rabbit IgG antibody were
4.4. Quantum chemical calculations provided by Cell Signaling Technology (Boston, MA, United States), and
the antibody of a7nAchR was purchased from Abcam (Cambridge, Mass,
The random conformational search of 1 was performed under UK). S-Acetylthiocholine iodide, S-butyrylthiocholine iodide, 5, 5′ -
MMFF94s molecular force field using SYBYL X 2.1.1, with an energy dithio-bis-(2-nitrobenzoic) acid (DTNB, Ellman’s reagent) and AchE
cutoff of 10 kcal mol− 1 to the global minima. The obtained conformers derived from human erythrocytes were purchased from Sigma-Aldrich
were optimized using Gaussion09 at B3LYP/6-31+G(d) level in gas (St. Louis, MO, USA). Methyllycaconitine was purchased from Target­
phase followed by ECD calculations at B3LYP/6-31+G(d) level in Mol, USA.
methanol. The overall ECD spectrum was weighted according to Boltz­
mann distribution, and the predicted curve was subsequently compared 4.6.2. Cell culture and treatment
with the experimental ones, respectively [31,32]. Murine macrophage RAW246.7 cells were obtained from American
Type Culture Collection (ATCC, Manassas, VA, USA). The cells were
maintained with RIPM-1640 medium supplemented with 10% fetal

7
J.-C. Su et al. Chemico-Biological Interactions 362 (2022) 109998

bovine serum and 1% penicillin/streptomycin solution in humidified membranes were incubated with primary anti-bodies overnight at 4 ◦ C,
atmosphere at 37 ◦ C with 5% CO2. For inducing inflammation, the cells following by incubating with HRP-conjugated with secondary anti­
were treated with 20 ng/mL LPS for 24 h following with or without bodies at room temperature for 1.5 h. After that, the bands were visu­
various concentration of compound 8 treatment for indicated times. alized using the enhanced chemiluminescence detection system. The
quantitative data of blots were measured with ImageJ software (NIH,
4.6.3. AchE activity assay NY). And three independent experiments were conducted.
The enzymatic activity was detected based on Ellman assay as pre­
vious described [33]. In brief, compounds were dissolved in DMSO. The 4.6.8. Statistical analysis
reaction mixture (totally 200 μL) containing phosphate buffer (pH 8.0), The data were analyzed using GraphPad Prism 8.0 software (San
test compound, and acetyl cholinesterase (0.02 U/mL), was incubated Diego, USA) and presented as the mean ± standard deviation (SD).
for 20 min (37 ◦ C). Then, the reaction was initiated by the addition of 40 Comparisons of data among three or more groups were analyzed by one-
μL of solution containing DTNB (0.625 mM) and acetylthiocholine io­ way ANOVA multiple comparisons. Comparisons of data between two
dide (0.625 mM) for AchE inhibitory activity assay, respectively. The groups were evaluated using a two-tailed unpaired t-test. P < 0.05 was
hydrolysis of acetylthiocholine was monitored at 405 nm every 30 s for considered significant difference.
1 h. Tacrine was used as a positive control with final concentration of
0.333 μM. All the reactions were performed in triplicate. The percentage Author statement
inhibition was calculated as follows: % inhibition = (E − S)/E × 100 (E
is the activity of the enzyme without test compound and S is the activity Conceptualization, Jun-Cheng Su, Xueping Lei, and Peng Zhang;
of enzyme with test compound). Experiment implementation, Jun-Cheng Su and Xueping Lei; Data pro­
cessing, Qianrong Pan, Xingyuan Xu, and Xia Wei; Writing—original
4.6.4. Measurement of NO production draft preparation, Jun-Cheng Su; Writing—review and editing, Xueping
The NO level in culture media was determined using a Griess reagent Lei and Peng Zhang. All authors have read and approved the final
for NO kit. Briefly, the cytotoxicity of compound 8 was evaluated by manuscript.
using a CCK8 assay kit (TargetMol, USA) as described previously. Then,
the cells were treated with or without various concentrations of com­ Declaration of competing interest
pound 8 in the presence and absence of LPS (20 ng/mL) for 24 h. The
supernatant medium was collected and mixed with same volume of The authors declare that they have no known competing financial
Griess reagent, and the absorbance was determined by a SynergyMx interests or personal relationships that could have appeared to influence
Multi-Mode Microplate Reader (Biotek, Winooski, VT) at 550 nm. The the work reported in this paper.
experiments were performed three times independently.
Acknowledgments
4.6.5. RNA extraction and quantitative real-time polymerase chain
reaction (RT-qPCR) This work was financially supported by the National Natural Science
The total RNA was isolated from the cell using an RNA Extraction Kit Foundation of China (nos. 82003605 and 32070391), Science and
(Solarbio Science & Technology, Beijing, China) in accordance with the Technology Program of Guangzhou (202002030026), and Central
manufacturer’s procedure. Complementary DNA (cDNA) was synthe­ Public-interest Scientific Institution Basal Research Fund (Y2022QC32
sized from 1 μg total RNA using SuperScript® VILO cDNA Synthesis Kit and Y2021XK25).
(Invitrogen). The RT-qPCR amplification was conducted with One-Step
SYBR PrimeScript Plus RT-PCR kit on LightCycler®480 Real-Time PCR
Appendix A. Supplementary data
System (Roche, Swiss). The primer sequences for qRT-PCR were as fol­
lows: TNF-α-Forwards 5′ -CCCTCACACTCAGATCATCTTCT-3′ , Reverse
Supplementary data to this article can be found online at https://doi.
5′ -TGAGGCCATAGTAGAGTGTCCT-3′ , IL-6-Forwards 5′ - CCAA­
org/10.1016/j.cbi.2022.109998.
GAGGTGAGTGCTTCCC-3′ , Reverse 5′ - CTGTTGTTCAGACTCTCTCCCT-
3’. α7nAChR-Forwards 5′ - AATGCTGCAAAGAGCCATACC-3′ , Reverse
References
5′ -TGAGGCCATAGTAGAGTGTCCT-3′ , GAPDH-Forwards 5′ - CTTCATT­
GACCTCAACTACATGGTCTA-3′ , Reverse 5′ - GATGA CAAGCTTCCC [1] I. Silman, The multiple biological roles of the cholinesterases, Prog. Biophys. Mol.
ATTCTCAG-3’. The fold change of mRNA was calculated using the Biol. 162 (2021) 41–56.
2− ΔΔCT method, and GAPDH was set as internal reference. The experi­ [2] S. Zhou, G. Huang, Synthesis and activities of acetylcholinesterase inhibitors,
Chem. Biol. Drug Des. 98 (6) (2021) 997–1006.
ments were conducted in triplicate. [3] S.D. Richbart, J.C. Merritt, N.A. Nolan, P. Dasgupta, Acetylcholinesterase and
human cancers, Adv. Cancer Res. 152 (2021) 1–66.
4.6.6. ELISA assay for TNF-α and IL-6 [4] N. Argueta, E. Notari, K. Szigeti, Role of pharmacogenomics in individualizing
treatment for alzheimer’s disease, CNS Drugs 36 (2022) 365–376.
The RAW 246.7 cells were treated with or without different con­ [5] G. Marucci, M. Buccioni, D.D. Ben, C. Lambertucci, R. Volpini, F. Amenta, Efficacy
centration of compound 8 in the presence or absence of LPS for 24 h. The of acetylcholinesterase inhibitors in Alzheimer’s disease, Neuropharmacology 190
culture supernatant was collected and used to detect TNF-α and IL-6 (2021), 108352.
[6] L.V. Borovikova, S. Ivanova, M. Zhang, H. Yang, G.I. Botchkina, L.R. Watkins,
secretion following with the instruction provided by the manufacturer.
H. Wang, N. Abumrad, J.W. Eaton, K.J. Tracey, Vagus nerve stimulation attenuates
The absorbance was measured using a SynergyMx Multi-Mode Micro­ the systemic inflammatory response to endotoxin, Nature 405 (6785) (2000)
plate Reader (Biotek, Winooski, VT) at 450 nm. Three experiments were 458–462.
[7] K.J. Tracey, The inflammatory reflex, Nature 420 (6917) (2002) 853–859.
conducted independently.
[8] M.V. Donova, O.V. Egorova, Microbial steroid transformations: current state and
prospects, Appl. Microbiol. Biotechnol. 94 (6) (2012) 1423–1447.
4.6.7. Western blotting assay [9] M. Vohra, A.R. Sharma, K. Satyamoorthy, P.S. Rai, Pharmacogenomic
The cells with or without indicated treatment were collected and considerations for repurposing of dexamethasone as a potential drug against SARS-
CoV-2 infection, Pers. Med. 18 (4) (2021) 389–398.
lysed RIPA buffer containing phenylmethylsulfonyl fluoride and prote­ [10] X.D. Li, J.C. Su, B.Z. Jiang, Y.L. Li, Y.Q. Guo, P. Zhang, A. Janthinoid, An
ase inhibitor cocktail. Equal amount of protein was electrophoresed in unprecedented tri-nor-meroterpenoid with highly modified bridged 4a,1-
10% sodium dodecyl sulfate–polyacrylamide gel electrophoresis gels (epoxymethano)phenanthrene scaffold, produced by the endophyte of Penicillium
janthinellum TE-43, Org. Chem. Front. 8 (22) (2021) 6196–6202.
(SDS-PAGE) and then the separated proteins were transferred to PVDF [11] C. Xu, K. Xu, X.L. Yuan, G.W. Ren, X.Q. Wang, W. Li, N. Deng, X.F. Wang, P. Zhang,
membrane (Millipore, USA). After blocking with 5% BSA, the Characterization of diketopiperazine heterodimers as potential chemical markers

8
J.-C. Su et al. Chemico-Biological Interactions 362 (2022) 109998

for discrimination of two dominant black aspergilli, Aspergillus niger and Aspergillus from the endophytic fungus Pleospora herbarum1, J. Nat. Prod. 62 (4) (1999)
tubingensis, Phytochemistry 176 (2020), 112399. 629–630.
[12] K. Xu, Q. Zhou, X.Q. Li, T. Luo, X.L. Yuan, Z.F. Zhang, P. Zhang, Cadinane- and [23] T.A. Mansoor, J. Hong, C.O. Lee, S.J. Bae, K.S. Im, J.H. Jung, Cytotoxic sterol
drimane-type sesquiterpenoids produced by Paecilomyces sp. TE-540, an endophyte derivatives from a marine sponge Homaxinella sp, J. Nat. Prod. 68 (3) (2005)
from Nicotiana tabacum L., are acetylcholinesterase inhibitors, Bioorg. Chem. 104 331–336.
(2020), 104252. [24] S.C. Sun, The non-canonical NF-kappaB pathway in immunity and inflammation,
[13] X.L. Yuan, X.F. Wang, K. Xu, W. Li, D. Chen, P. Zhang, Characterization of a new Nat. Rev. Immunol. 17 (9) (2017) 545–558.
insecticidal anthraquinone derivative from an endophyte of Acremonium vitellinum [25] J.C. Su, W. Cheng, J.G. Song, Y.L. Zhong, X.J. Huang, R.W. Jiang, Y.L. Li, M.M. Li,
against Helicoverpa armigera, J. Agric. Food Chem. 68 (41) (2020) 11480–11487. W.C. Ye, Y. Wang, Macrocyclic diterpenoids from Euphorbia helioscopia and their
[14] A.A. Gunatilaka, G. Samaranayake, D.G. Kingston, G. Hoffmann, R.K. Johnson, potential anti-inflammatory activity, J. Nat. Prod. 82 (10) (2019) 2818–2827.
Bioactive ergost-5-ene-3 beta, 7 alpha-diol derivatives from Pseudobersama [26] K. Taniguchi, M. Karin, NF-kappaB, inflammation, immunity and cancer: coming of
mossambicensis, J. Nat. Prod. 55 (11) (1992) 1648–1654. age, Nat. Rev. Immunol. 18 (5) (2018) 309–324.
[15] C.M. Cui, X.M. Li, L. Meng, C.S. Li, C.G. Huang, B.G. Wang, 7-Nor-ergosterolide, a [27] Y. Fan, R. Mao, J. Yang, NF-kappaB and STAT3 signaling pathways collaboratively
pentalactone-containing norsteroid and related steroids from the marine-derived link inflammation to cancer, Protein Cell 4 (3) (2013) 176–185.
endophytic Aspergillus ochraceus EN-31, J. Nat. Prod. 73 (11) (2010) 1780–1784. [28] Q.Q. Han, M.Y. Deng, H. Liu, U. Ali, X.Y. Li, Y.X. Wang, Cynandione A and PHA-
[16] G. Jinming, H. Lin, L. Jikai, A novel sterol from Chinese truffles Tuber indicum, 543613 inhibit inflammation and stimulate macrophageal IL-10 expression
Steroids 66 (10) (2001) 771–775. following alpha7nAChR activation, Biochem. Pharmacol. 190 (2021), 114600.
[17] H. Fujimoto, E. Nakamura, E. Okuyama, M. Ishibashi, Six immunosuppressive [29] Z. Lu, P. Xie, D. Zhang, P. Sun, H. Yang, J. Ye, H. Cao, C. Huo, H. Zhou, Y. Chen,
features from an ascomycete, Zopfiella longicaudata, found in a screening study W. Ye, L. Yu, J. Liu, 3-Dehydroandrographolide protects against
monitored by immunomodulatory activity, Chem. Pharm. Bull. (Tokyo) 52 (8) lipopolysaccharide-induced inflammation through the cholinergic anti-
(2004) 1005–1008. inflammatory pathway, Biochem. Pharmacol. 158 (2018) 305–317.
[18] S. Hybelbauerova, J. Sejbal, M. Dracinsky, A. Hahnova, B. Koutek, Chemical [30] N.N. Yang, J.W. Yang, Y. Ye, J. Huang, L. Wang, Y. Wang, X.T. Su, Y. Lin, F.T. Yu,
constituents of Stereum subtomentosum and two other birch-associated S.M. Ma, L.Y. Qi, L.L. Lin, L.Q. Wang, G.X. Shi, H.P. Li, C.Z. Liu,
basidiomycetes: an interspecies comparative study, Chem. Biodivers. 5 (5) (2008) Electroacupuncture ameliorates intestinal inflammation by activating
743–750. alpha7nAChR-mediated JAK2/STAT3 signaling pathway in postoperative ileus,
[19] T. Amagata, M. Tanaka, T. Yamada, M. Doi, K. Minoura, H. Ohishi, T. Yamori, Theranostics 11 (9) (2021) 4078–4089.
A. Numata, Variation in cytostatic constituents of a sponge-derived Gymnascella [31] J.C. Su, S. Wang, W. Cheng, X.J. Huang, M.M. Li, R.W. Jiang, Y.L. Li, L. Wang, W.
dankaliensis by manipulating the carbon source, J. Nat. Prod. 70 (11) (2007) C. Ye, Y. Wang, Phloroglucinol derivatives with unusual skeletons from Cleistocalyx
1731–1740. operculatus and their in vitro antiviral activity, J. Org. Chem. 83 (15) (2018)
[20] D. Kumla, T. Shine Aung, S. Buttachon, T. Dethoup, L. Gales, J.A. Pereira, A. Inacio, 8522–8532.
P.M. Costa, M. Lee, N. Sekeroglu, A.M.S. Silva, M.M.M. Pinto, A. Kijjoa, A new [32] J.G. Song, J.C. Su, Q.Y. Song, R.L. Huang, W. Tang, L.J. Hu, X.J. Huang, R.
dihydrochromone dimer and other secondary metabolites from cultures of the W. Jiang, Y.L. Li, W.C. Ye, Y. Wang, Cleistocaltones A and B, antiviral
marine sponge-associated fungi Neosartorya fennelliae KUFA 0811 and Neosartorya phloroglucinol-terpenoid adducts from Cleistocalyx operculatus, Org. Lett. 21 (23)
tsunodae KUFC 9213, Mar. Drugs 15 (12) (2017) 375. (2019) 9579–9583.
[21] T. Amagata, M. Doi, M. Tohgo, K. Minoura, A. Numata, Dankasterone, a new class [33] G.Y. Huang, H. Cui, X.Y. Lu, L.D. Zhang, X.Y. Ding, J.J. Wu, L.X. Duan, S.J. Zhang,
of cytotoxic steroid produced by a Gymnascella species from a marine sponge, Z. Liu, R.R. Zhang, (+/-)-Dievodialetins A-G: seven pairs of enantiomeric coumarin
Chem. Commun. 14 (1999) 1321–1322. dimers with anti-acetylcholinesterase activity from the roots of Evodia lepta Merr,
[22] K. Krohn, C. Biele, H.J. Aust, S. Draeger, B. Schulz, Herbarulide, a Phytochemistry 182 (2021), 112597.
ketodivinyllactone steroid with an unprecedented homo-6-oxaergostane skeleton

You might also like