Abstract
Acute respiratory distress syndrome (ARDS) is a high-mortality disease
and lacks effective pharmacotherapy. A traditional Chinese medicine
(TCM) formula, Ning Fei Ping Xue (NFPX) decoction, was demonstrated to
play a critical role in alleviating inflammatory responses of the lung.
However, its therapeutic effectiveness in ARDS and active compounds,
targets, and molecular mechanisms remain to be elucidated. The present
study investigates the effects of NFPX decoction on ARDS mice induced
by lipopolysaccharides (LPS). The results revealed that NFPX alleviated
lung edema evaluated by lung ultrasound, decreased lung wet/Dry ratio,
the total cell numbers of bronchoalveolar lavage fluid (BALF), and
IL-1β, IL-6, and TNF-α levels in BALF and serum, and ameliorated lung
pathology in a dose-dependent manner. Subsequently, UPLC-HRMS was
performed to establish the compounds of NFPX. A total of 150 compounds
in NFPX were characterized. Moreover, integrating network pharmacology
approach and transcriptional profiling of lung tissues were performed
to predict the underlying mechanism. 37 active components and 77
targets were screened out, and a herbs-compounds-targets network was
constructed. Differentially expressed genes (DEGs) were identified from
LPS-treated mice compared with LPS combined with NFPX mice. GO, KEGG,
and artificial intelligence analysis indicated that NFPX might act on
various drug targets. At last, potential targets, HRAS, SMAD4, and
AMPK, were validated by qRT-PCR in ARDS murine model. In conclusion, we
prove the efficacy of NFPX decoction in the treatment of ARDS.
Furthermore, integrating network pharmacology, transcriptome, and
artificial intelligence analysis contributes to illustrating the
molecular mechanism of NFPX decoction on ARDS.
Keywords: Ning Fei Ping Xue decoction, acute respiratory distress
syndrome, network pharmacology, transcriptome analysis, artificial
intelligence analysis, inflammatory responses
Introduction
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a
devastating clinical syndrome characterized by increased non-fluid
extravascular pulmonary water, decreased pulmonary compliance, and
acute hypoxic respiratory failure ([47]Thompson et al., 2017; [48]Cao
et al., 2020). The pathophysiological changes of ALI/ARDS are
represented by alveolar interstitial edema, pulmonary hemorrhage, lung
consolidation, and inflammatory cells infiltration. These processes are
thought to be related to many target inflammatory cells and effector
cytokines ([49]Brooks et al., 2020). As a treasury of medicine in
China, traditional Chinese medicine (TCM) plays an important role in
attenuating inflammation and improving immune function ([50]Zhang and
Wei, 2020).
In recent years, more and more attention has been paid to the roles of
TCM in ARDS treatment. One study has shown that hydroxysafflor yellow A
alleviates LPS-induced ARDS in mice by blocking TLR4/NF-κB signaling
pathway ([51]Zhang et al., 2017). In a rat model, silymarin can
attenuate LPS-induced lung injury by inhibiting the MAPK signaling
pathway ([52]Zhu and Sun, 2018). Chen et al. have found that honokiol
could protect the pulmonary microvascular endothelial barrier from
damage by LPS in ARDS models by promoting the SIRT3/AMPK signaling
pathway and suppressing Ang-2 expression ([53]Chen et al., 2018). It
was also reported that celastrol might reduce ARDS-related lung injury
caused by LPS in rats by inactivating NF-κB signaling pathways ([54]Wei
and Wang, 2017). Although there are a lot of achievements achieved from
the studies of TCM in ARDS, the detailed molecular mechanisms of TCM
are rarely known due to the complexity and diversity of TCM ingredients
and the synergistic or antagonistic effects among the ingredients.
Different from the pattern of “one target, one drug” in modern
medicine, TCM theory emphasizes a holistic view of the human body.
Conventional experimental pharmacological techniques may not be
applicable to the research field of TCM on account of the complexity of
its components, targets, and mechanisms, posing challenges for the
development of TCM.
The development of transcriptomics, proteomics, and metabolomics marked
the beginning of the post-genomic era, which promoted the birth of
network pharmacology ([55]Pan et al., 2020). Network pharmacology is a
sophisticated tool system that deciphers the mechanisms of complex herb
formulas from the component level to gene level based on multiple large
databases ([56]Boezio et al., 2017). One of the most important
characteristics of TCM is “holistic philosophy,” which coincides with
systemic analysis of “network pharmacology.” As an advanced research
method, the network pharmacology of TCM transforms the research
paradigm from “one target, one drug” into the novel “network target,
multi-components.” This helps assess the compatibility and
cooperativity of TCM and elaborate the relationships of targets and
signaling pathways in the network ([57]Chen et al., 2016).
Ning Fei Ping Xue (NFPX) decoction is a kind of TCM formula. It is
comprised of twenty herbs: Paeonia lactiflora Pall. [Paeoniaceae;
Paeoniae Radix Alba, 7 g], Atractylodes macrocephala Koidz.
[Asteraceae; Atractylodis macrocephalae rhizoma, 10 g], Conioselinum
anthriscoides “Chuanxiong” [Apiaceae; Chuanxiong Rhizoma, 7 g],
Angelica sinensis (Oliv.) Diels [Apiaceae; Angelicae Sinensis Radix,
7 g], Poria cocos (Schw.) Wolf [Polyporaceae; Poria, 10 g], Carthamus
tinctorius L. [Asteraceae; Carthami Flos, 7 g], Phellodendron chinense
C.K.Schneid. [Rutaceae; Phellodendri Chinrnsis Cortex, 10 g], Coptis
chinensis Franch. [Ranunculaceae; Coptidis Rhizoma, 10 g], Astragalus
mongholicus Bunge [Fabaceae; Astragali radix, 80 g], Scutellaria
baicalensis Georgi [Lamiaceae; Scutellariae Radix, 10 g], Phragmites
australis (Cav.) Trin. ex Steud. [Poaceae; Phragmitis Rhizoma, 10 g],
Gardenia jasminoides J.Ellis [Rubiaceae; Gardeniae Fructus, 10 g],
Rehmannia glutinosa (Gaertn.) DC. [Orobanchaceae; Rehmanniae Radix
Praeparata, 10 g], Prunus persica (L.) Batsch [Rosaceae; Persicae
Semen, 7 g], Descurainia sophia (L.) Webb ex Prantl [Brassicaceae;
Descurainiae semen lepidii semen, 5 g], Coix lacryma-jobi var. ma-yuen
(Rom.Caill.) Stapf [Poaceae; Coicis Semen, 10 g], Alisma
plantago-aquatica subsp. orientale (Sam.) Sam. [Alismataceae; Alismatis
rhizoma, 15 g], Polyporus umbellatus (Pers) Fr. [Polyporaceae;
Polyporus, 10 g], Neolitsea cassia (L.) Kosterm. [Lauraceae; Cinnamomi
cortex, 7 g], and Pheretima, 7 g. NFPX decoction has been found to
mitigate the inflammatory response of acute and chronic respiratory
diseases in clinical practice. Improved oxygen saturation, increased
number of ventilator-free days, and shortened ICU and hospital lengths
of stay were observed in patients with respiratory failure after the
administration of NFPX decoction. However, its efficacy in ARDS and
specific molecule target and mechanism still need to be investigated.
In the present study, we have first investigated the effects of NFPX
decoction on ameliorating lung edema and inflammatory response of ARDS
mice induced by lipopolysaccharide (LPS). Additionally, we have
explored the mechanisms by screening specific molecular targets using
network pharmacology, transcriptome analysis, and artificial
intelligence analysis to provide the theoretical basis for the clinical
application of NFPX decoction on ARDS patients. The detailed schematic
of the workflow in the current study is shown in [58]Figure 1.
FIGURE 1.
[59]FIGURE 1
[60]Open in a new tab
The schematic of the workflow.
Materials and Methods
Acute Respiratory Distress Syndrome (ARDS) Murine Model
Eight-week-old male C57BL/6N mice were purchased from Vital River
Animal Institute (Beijing, China) and were maintained under specific
pathogen-free (SPF) conditions. The mice were randomly divided into six
groups (five mice per group): Control, LPS+PBS, LPS+2.6 g/kg NFPX
(LNFPX), LPS+5.2 g/kg NFPX (MNFPX), LPS+10.4 g/kg NFPX (HNFPX), and
10.4 g/kg NFPX. Doses of LPS (2 mg/kg) and NFPX (2.6, 5.2, and
10.4 g/kg) were chosen according to previous reports and our pilot
studies ([61]Lang et al., 2017). NFPX granules were kindly provided by
Prof. Jianxin Chen (Beijing University of Chinese Medicine, Beijing,
China), which were extracted by ethanol ([62]Bu et al., 2020). The
extraction procedures are as follows: water and ground NFPX material
were placed in a glass tube (12:1); the solution was kept boiling for
1 h; then, water (8:1) was added for second water extraction step. The
water supernatant was filtered and dried using a rotary evaporator
under vacuum followed by freeze-drying to obtain the water extract. 55%
ethanol was added to water extract in the glass tube, and the mixture
was sonicated for 1 h. The ethanol extract was filtered through a
0.45 µm syringe filter; then, the extract was made into granules. Mice
were anesthetized with gaseous isoflurane, followed by instillation of
40 μl LPS (Escherichia coli 055: B5, L8880; Solarbio, Beijing, China)
into the tracheas using 22G needles to establish ARDS model or 40 μl
PBS as control. For NFPX pretreatment, various doses of NFPX granules
dissolved in water were administered intragastrically daily for
7 consecutive days before LPS administration. On day 8, the animals
were anesthetized with gaseous isoflurane; retroorbital venous blood,
BALF, and lung tissues were collected for the subsequent analysis.
Lung Ultrasound of Mice
Lung ultrasound was performed using a high-resolution Vevo2100
Ultrasound System (Visualsonics Inc., Toronto, Canada) with an
ultrahigh-frequency (40 MHz) transducer probe to obtain a maximum
resolution of 30 µm and imaging depth of 10.0 mm. The hair of the
anterior chest in mice was removed by depilatory cream after 24 h
exposure to LPS. Lung ultrasound videos were recorded and analyzed by
two expert technicians (Shanshan Zhang and Xiaoming Dong).
Hematoxylin and Eosin (HE) and TUNEL Staining
For histological examination, the left lung lobes were perfused with 4%
paraformaldehyde and embedded in paraffin. Four-micron thick slides
were stained with HE and were reviewed by two skilled pathologists. To
quantify the lung injury and inflammation response, a semiquantitative
histology score method was adopted ([63]Dai et al., 2018). Briefly,
alveolar edema, pulmonary hemorrhage, atelectasis, and inflammatory
cells infiltration were each scored on a 0–4 scale. The total score was
then calculated by adding the scores of all four histological indexes.
The apoptotic cells of mouse samples were detected by the TUNEL kit
(Roche, Indianapolis, United States) according to the manufacturer’s
instructions. Controls were set with PBS instead of the primary
antibody.
Lung Wet/Dry (W/D) Weight Ratio
The wet/Dry ratio is an indicator of pulmonary edema by calculating
extravascular lung water. Lung lobes were harvested and weighted as
soon as possible to get the “wet weight.” Then, lung tissues are placed
in an incubator at 65°C for 48 h and re-weighed to get the “dry
weight.” Lung wet/Dry ratio = wet weight divided by dry weight.
Enzyme-Linked Immunosorbent Assay (ELISA)
Retroorbital venous blood was collected into 2 ml Eppendorf tubes. The
tubes were left at room temperature until the blood had clotted. Serum
was separated by centrifugation at 1000 × g for 15 min. Moreover, BALF
was collected by intratracheally administering 1 ml of PBS. The
concentrations of IL-1β, IL-6, and TNF-α in serum and BALF were
determined by ELISA kits (Cusabio, Wuhan, China) according to the
manufacturer’s protocols.
Ultra-Performance Liquid Chromatography–High-Resolution Mass Spectrometry
(UPLC-HRMS) Analysis
The UPLC system was performed on an Agilent 1290 LC system (Agilent
Technologies Inc., Palo Alto, CA, United States) equipped with a binary
pump, a thermostat-controlled column compartment, an autosampler, and a
DAD detector. Waters ACQUITY UPLC CSH C18 (2.1 × 100 mm, 1.7 μm) was
employed at 30°C with sample injection volume of 3 μl. The mobile phase
consisted of 0.1% formic acid in water (A) and 0.1% formic acid in
acetonitrile (B) using gradient program at a flow rate of 0.3 ml/min
and was eluted with gradient elution program as follows: 0–5 min (5%
B), 5–8 min (5–10% B), 8–18 min (10% B), 18–23 min (10–17% B),
23–26 min (17–20% B), 26–44 min (20–28%% B), 44–46 min (28–40%% B),
46–56 min (40–60% B), 56–60 min (60–95% B), 60–63 min (95% B),
63–63.1 min (95–5% B), and 63.1–65 min (5% B) protocol. The Mass
Spectrometer AB Sciex TripleTOF 4600 (AB SCIEX, Foster City, CA, United
States), equipped with an electrospray ionization (ESI) source, was
controlled by Analyst TF 1.7.1. software (AB SCIEX, Foster City, CA,
United States). The spectrometer was operated in full-scan TOF-MS at
m/z 50–1700 and information-dependent acquisition (IDA)MS/MS modes,
with negative and positive ionization modes. The optimized parameters
of mass spectrometry as follows: Ion Source Temperature was 500°C;
Curtain Gas was 35 psi; Ion Source Gas 1 and 2 were 50 psi; Ion Spray
Voltage was 5000 V (positive)/4500 V (negative); Declustering Potential
was 100 V (MS and MS/MS); Collision Energy was 40 eV; Collision Energy
Spread was 20 eV (MS/MS); mass range was 50–1700 m/z (MS)/50–1250 m/z
(MS/MS); Ion Release Delay was 30 ms; Ion Release Width was 15 ms.
Data analysis was performed by PeakView 1.2 software (AB SCIEX, Foster
City, CA, United States). The phytochemical compounds were tentatively
characterized based on their retention time, mass accuracy of precursor
ions, MS/MS spectra, and fragmentation pathways, referring to the
Natural Products HR-MS/MS Spectra Library and literature report.
Identification of Bioactive Components and Targets in NFPX Decoction
All candidate components and targets of the twenty traditional
medicinal herbs in NFPX (Paeonia lactiflora Pall. [Paeoniaceae;
Paeoniae Radix Alba, 7 g], Atractylodes macrocephala Koidz.
[Asteraceae; Atractylodis macrocephalae rhizoma, 10 g], Conioselinum
anthriscoides “Chuanxiong” [Apiaceae; Chuanxiong Rhizoma, 7 g],
Angelica sinensis (Oliv.) Diels [Apiaceae; Angelicae Sinensis Radix,
7 g], Poria cocos (Schw.) Wolf [Polyporaceae; Poria, 10 g], Carthamus
tinctorius L. [Asteraceae; Carthami Flos, 7 g], Phellodendron chinense
C.K.Schneid. [Rutaceae; Phellodendri Chinrnsis Cortex, 10 g], Coptis
chinensis Franch. [Ranunculaceae; Coptidis Rhizoma, 10 g], Astragalus
mongholicus Bunge [Fabaceae; Astragali radix, 80 g], Scutellaria
baicalensis Georgi [Lamiaceae; Scutellariae Radix, 10 g], Phragmites
australis (Cav.) Trin. ex Steud. [Poaceae; Phragmitis Rhizoma, 10 g],
Gardenia jasminoides J.Ellis [Rubiaceae; Gardeniae Fructus, 10 g],
Rehmannia glutinosa (Gaertn.) DC. [Orobanchaceae; Rehmanniae Radix
Praeparata, 10 g], Prunus persica (L.) Batsch [Rosaceae; Persicae
Semen, 7 g], Descurainia sophia (L.) Webb ex Prantl [Brassicaceae;
Descurainiae semen lepidii semen, 5 g], Coix lacryma-jobi var. ma-yuen
(Rom.Caill.) Stapf [Poaceae; Coicis Semen, 10 g], Alisma
plantago-aquatica subsp. orientale (Sam.) Sam. [Alismataceae; Alismatis
rhizoma, 15 g], Polyporus umbellatus (Pers) Fr [Polyporaceae;
Polyporus, 10 g], Neolitsea cassia (L.) Kosterm. [Lauraceae; Cinnamomi
cortex, 7 g], and Pheretima, 7 g) were retrieved from the traditional
Chinese medicine systems pharmacology (TCMSP) database
([64]http://tcmspw.com/tcmsp.php) ([65]Ru et al., 2014) and SymMap
database ([66]https://www.symmap.org) ([67]Wu et al., 2019). Oral
bioavailability (OB) is usually an essential pharmacokinetic parameter
([68]Xu et al., 2012). As a qualitative parameter, drug-likeness (DL)
plays a role in evaluating the druggability of a component ([69]Tao et
al., 2013). In the current study, we set up the compounds in NFPX with
OB ≥30% and DL index ≥0.18 as bioactive ingredients, as shown in
previous reports ([70]Guo et al., 2019; [71]Yu et al., 2020).
Collection of Gene Symbols for ARDS and Construction of Protein–Protein
Interaction (PPI) Networks
Underlying gene symbols of ARDS were obtained from two databases,
namely, GeneCards database ([72]https://www.genecards.org/) and OMIM
database ([73]http://www.omim.org/). Then, the protein targets of NFPX
were mapped with ARDS using the comparative toxicogenomics database
(CTD) ([74]http://ctdbase.org/) ([75]Davis et al., 2021). The obtained
intersection genes were uploaded onto STRING 11.0
([76]http://string-db.org/) ([77]Szklarczyk et al., 2019) to obtain the
protein–protein interactions (PPI) network of NFPX treatment in ARDS.
Construction of Networks and Analysis of Target Pathways
To further characterize the molecular mechanism of NFPX on ARDS, the
herbs-compounds-targets network was established using Cytoscape 3.7.2
software (Bethesda, MD, United States). The potential pathways were
identified by Gene Ontology (GO) enrichment analysis and the Kyoto
Encyclopedia of Genes and Genomes (KEGG) pathway analysis.
RNA-seq and Pathway Enrichment Analysis
Lung tissue samples were sent to the Beijing Genomics Institute (BGI,
Shenzhen, China) for RNA extraction, cDNA library construction,
qualification, further RNA-seq detection by Illumina HiSeqTM sequencing
platform, and final bioinformatic analysis. Total RNA was extracted
from the tissues using Trizol (Invitrogen, Carlsbad, CA, United States)
according to manual instructions. Subsequently, total RNA was qualified
and quantified using a NanoDrop and Agilent 2100 bioanalyzer (Thermo
Fisher Scientific, MA, United States). Oligo(dT)-attached magnetic
beads were used to purify mRNA. Purified mRNA was fragmented into small
pieces with fragment buffer at an appropriate temperature. Then,
first-strand cDNA was generated using random hexamer-primed reverse
transcription, followed by second-strand cDNA synthesis. Afterward,
A-Tailing Mix and RNA Index Adapters were added by incubating to end
repair. The cDNA fragments obtained from the previous step were
amplified by PCR, and products were purified by Ampure XP Beads and
then dissolved in EB solution. The product was validated on the Agilent
Technologies 2100 bioanalyzer for quality control. The double-stranded
PCR products from the previous step were heated, denatured, and
circularized by the splint oligo sequence to get the final library. The
single-strand circle DNA (ssCir DNA) was formatted as the final
library. The final library was amplified with phi29 to make DNA
nanoball (DNB), which had more than 300 copies of one molecular, DNBs
were loaded into the patterned nanoarray, and single-end 50 bases reads
were generated on the BGIseq500 platform (BGI-Shenzhen, China). The
quantitative analysis for DEGs was performed based on the GO functional
and KEGG pathway analysis. log2(Fold Change) ≥ 1 and FDR ≤ 0.05 were
used as the threshold for significant DEGs ([78]Li et al., 2019;
[79]Cao et al., 2020).
Specific Gene Module–Based Target Identification
Gene module pair–based target identification (GMPTI) approach was
utilized to predict novel compound–target interactions based on a
drug-induced gene expression profile ([80]http://www.bcxnfz.top/TMP/).
GMPTI considers experiments with gene expression profiles from a
collection of samples belonging to two classes, for example,
drug-treated vs. control. The genes can be ordered in a ranked list L,
according to their differential expression between the classes. Given
the defined gene module pair (GMP) for each target, the goal of GMPTI
is to compare L to each target-specific gene module pair using a
similarity metric slightly adjusted from that used in Gene Set
Enrichment Analysis ([81]Subramanian et al., 2005). We defined the raw
similarity score as follows:
[MATH:
TCSLt=ESLup−ESL
mi>down,
:MATH]
where
[MATH:
ESLup
:MATH]
is the enrichment of tup for L and
[MATH:
ESLdo
wn :MATH]
is the enrichment of tdown for L.
[MATH:
TCSLt :MATH]
denotes the Total Correlation Score of the GMP (tup, tdown) of one
target, with respect to the signatures L. TCS ranges between −2 and 2.
It measures the degree of similarity between query L and target-induced
gene expression profiles.
We assess the significance of an actual TCS value by comparing it with
the set of scores TCSNULL computed with random permutations of both top
and bottom gene modules for each target. A nominal p value for the TCSi
of target i is estimated using the portion of the TCSNULL distribution
above the actual TCSi, as follows:
[MATH:
P=N(
abs(TCSNULL<
/msub>)≥abs<
mrow>(TCSi))100
0, :MATH]
where
[MATH:
abs(TCSNULL
mrow>) :MATH]
is the absolute values of all correlation scores for random GMPs with
respect to a query gene list L.
[MATH:
abs(TCSi) :MATH]
is the absolute value of the similarity score of target i with respect
to L.
Structural Docking of NFPX Ingredients and Potential Targets
To test interactions of NFPX ingredients and the eight potential
targets, the target-structure-based docking method was utilized. Among
the eight targets, we collected the known three-dimensional structures
for the five targets, AMPK (PDB ID: 4cfe), HRAS (PDB ID: 6mqt), SOD1
(PDB ID: 5o40), AKT2 (PDB ID: 3d0e), and RAC1 (PDB ID: 3th5) from the
PDB database ([82]https://www.rcsb.org/). The protein structures of the
other three targets, SMAD4, P53, and HIF-1, were collected from the
AlphaFold Protein Structure Database
([83]https://alphafold.ebi.ac.uk/), which includes the highly accurate
protein structures predicted using AlphaFold v2.0. Then, these targets
were docked by the NFPX ingredients with a three-dimensional structure
on the representative conformations using the SYBYL−Surflex docking in
standard precision mode.
qRT-PCR
Total RNA was extracted from lung tissues using the RNA extraction kit
(Qiagen, Hilden, Germany). qRT-PCR was performed utilizing the qRT-PCR
kit (Thermo Fisher, United States) in the ABI StepOnePlus PCR system
according to the manufacturer’s protocol. The ACTB mRNA expression
level was employed as an internal control. The primers were designed as
follows: SMAD4, forward, 5′-GTCATCCTGCTCACCAGATGTC-3′ and
reverse, 5′-TGCTCAGACAGGCATCGTTAC-3′; HIF-1, forward,
5′-AGCAAGATCTCGGCGAAGC-3′ and reverse,
5′-ACCACCGGCATCCAGAAGT-3′; MAPK, forward,
5′-ACAGGCAGCGGAGACACCTA-3′ and reverse,
5′-GGGGAGGATGATCGAGACAC-3′; HRAS, forward,
5′-ATCCAGCTGATCCAGAACCAC-3′ and reverse,
5′-TCCCGCATGGCACTATACTC-3′; SOD1, forward,
5′-CAGAAGGCAAGCGGTGAAC-3′ and reverse,
5′-GAGGTCCTGCACTGGTACAGC-3′; AKT2, forward,
5′-TGCTGCCGCCAGTTCATA-3′ and reverse,
5′-GCAGGAGGCTCCTCGGATAC-3′; RAC1, forward,
5′-CAGATGCAGGCCATCAAGTG-3′ and reverse,
5′-GTCAAAGACGGTGGGGATGT-3′; P53, forward,
5′-CTCCCTCTGAGCCAGGAGAC-3′ and reverse,
5′-GACACTCGGAGGGCTTCACT-3′; ACTB, forward,
5′-TTCATGGATGCCACAGGATT-3′ and reverse,
5′-TGACGGCCAGGTCATCACTA-3′. The qRT-PCR results were analyzed and
expressed as the relative mRNA expression of the CT (threshold cycle)
value, which was then converted to fold changes.
Statistical Analysis
Values were represented as the mean ± SD, and two-tailed t-test was
used for two preselected groups by GraphPad Prism 7.0 (GraphPad
Software Inc, CA, United States). p value < 0.05 was considered
statistically significant.
Results
NFPX Attenuates the Ultrasound Imaging Lesions of ARDS
The typical ultrasonographic artifacts of normal lung tissues are
characteristic of lung sliding with horizontal, parallel lines below
the pleural line, referred to as A-lines. In contrast, lung
ultrasonograms of ARDS usually show B-lines and pleural thickening and
ground-glass areas ([84]Picano et al., 2006). B-lines are defined as
comet tail-like hyperechoic reverberation artifacts arising from and
perpendicular to the pleural line, which is representative of thickened
interlobular septa. To elucidate the imaging characteristics of
different disposing groups, we performed lung ultrasound after 24 h
treatment of LPS. As shown in [85]Figure 2A, lung tissues in healthy
mice showed A-lines (white arrow) and uniformly continuous pleural line
(black arrow). In contrast, multiple well-defined B-lines and thickened
pleural and ground-glass areas can be seen in the LPS-induced mouse
model ([86]Figure 2B). As anticipated, NFPX lightened the ultrasound
abnormalities caused by LPS. It can be observed that fewer B-lines and
ground-glass areas exist in LPS+2.6 g/kg NFPX or LPS+5.2 g/kg NFPX
group ([87]Figures 2C,D) than those in the LPS treatment group. What is
more, a high concentration of NFPX treatment with or without LPS
appears the same as that in the ultrasound images of normal mice
([88]Figures 2E,F). These data reasonably suggested that NFPX may
relieve the alveolar interstitial edema and thickened interlobular
septa.
FIGURE 2.
[89]FIGURE 2
[90]Open in a new tab
The ultrasound imaging lesions of ARDS were evaluated by lung
ultrasound. (A) Control group: (B) LPS+PBS group; (C) LPS+2.6 g/kg NFPX
group; (D) LPS+5.2 g/kg NFPX group; (E) LPS+10.4 g/kg NFPX group; (F)
10.4 g/kg NFPX group. White arrow: A-lines; red arrow: B-lines; black
arrow: pleural line.
NFPX Mitigates LPS-Induced ARDS by Inhibiting Cell Apoptosis and Inflammatory
Reaction
The previous data provide intuitive evidence for NFPX exerting
protective effects against ARDS. We further validated the protective
effects of NFPX during experimental ARDS. HE staining was performed to
assess the pathological changes of the lung. As shown in [91]Figures
3A,B, alveolar edema, pulmonary hemorrhage, atelectasis, and
inflammatory cells infiltration were the most severe in the LPS group
and had the highest lung injury score correspondingly. NFPX treatment
effectively alleviated these LPS-induced pathological changes in a
dose-dependent manner. Compared to naïve mice, NFPX alone treatment did
not exhibit significant pathological changes in tissues. Besides,
several indicators associated with lung microvascular permeability and
extravascular lung water were quantified, including lung wet/dry weight
ratio and cell number in bronchoalveolar lavage fluid (BALF). As
expected, administration with NFPX prominently reduced the lung wet/dry
weight ratio and cell number in BALF induced by LPS in a dose-dependent
manner (p < 0.05, [92]Figures 3C,D). These results sufficiently
supported that NFPX remarkably abrogated LPS-induced pathological
changes without exerting side effects.
FIGURE 3.
[93]FIGURE 3
[94]Open in a new tab
NFPX mitigates LPS-induced ARDS by inhibiting cell apoptosis and
inflammatory reaction. (A,B) The lung histopathology analysis was
examined by HE staining and scored by two independent pathologists.
Scale bar, 50 μm. (C) Lung wet/dry ratios were calculated by weighting
the initial weight and the dry weight. (D) Cell numbers in BALF were
observed by cell counter. (E,F) Cell apoptosis was estimated by the
TUNEL staining assay. Scale bar, 50 μm. (G–I) The levels of IL-1β,
IL-6, and TNF-α in BALF were detected by the ELISA assay. (J–L) The
levels of IL-1β, IL-6, and TNF-α in serum were detected by the ELISA
assay. (a) Control group: (b) LPS+PBS group; (c) LPS+2.6 g/kg NFPX
group; (d) LPS+5.2 g/kg NFPX group; (e) LPS+10.4 g/kg NFPX group; (f)
10.4 g/kg NFPX group. *p < 0.05.
Next, we further explore how the NFPX plays a role in deterring the
development of ARDS induced by LPS. Cell apoptosis and inflammation
reaction are the core pathophysiologic mechanisms of ARDS. The
proinflammatory cytokines, IL-1β, IL-6, and TNF-α, contribute to the
infiltration of inflammatory cells during ARDS development. In
accordance with the above data, NFPX also hampers the cell apoptosis
and the level of cytokines in the lung during ARDS in a
concentration-dependent manner ([95]Figures 3E–I). Considering the
characterization of ARDS as the systemic inflammatory reaction, the
levels of three cytokines in peripheral serum were also examined.
Results have shown that NFPX reduced the secretion of cytokines in
serum, although the levels of IL-1β, IL-6, and TNF-α are lower than
those in BALF ([96]Figures 3J–L). These findings indicate that NFPX
mitigates LPS-induced ARDS by inhibiting cell apoptosis and
inflammatory reaction.
Identification of the Major Chemical Compounds in NFPX
To achieve good resolution, selectivity and peak shape within a short
analysis time, various mobile phase systems, and linear gradients were
investigated. Finally, aqueous acetonitrile with 0.1% formic acid on
the optimized gradient was selected as the mobile phase. The MS
parameters were optimized by adjusting the ion intensity and
appropriate ionization, and the optimal parameters were finally
selected.
The UPLC-HRMS method in both positive and negative ion modes was
employed to characterize the major constituents in NFPX rapidly. A
total of 150 compounds in NFPX were unambiguously or tentatively
characterized by comparing their retention times and MS data with the
Natural Products HR-MS/MS Spectral Library database or with data
reported in the literature. The detailed compound information was
summarized in [97]Table 1 and the relevant chromatograms are shown in
[98]Figures 4–[99]6, and the detailed structural formula of 150
compounds in NFPX is summarized in [100]Supplementary Table S1.
TABLE 1.
Identification of the major chemical compounds in NFPX.
No RT(min) Adduct ions Measured m/z Respected m/z ppm Formula M.W.
Identification Source
1 1.86 [M+H]^+ 268.1026 268.104 −5.3 C[10]H[13]N[5]O[4] 267.10
Adenosine DL
2 2.02 [M+FA−H]^− 407.1204 407.1195 2.2 C[15]H[22]O[10] 362.12 Catalpol
SD
3 2.19 [M+H]^+ 284.098 284.0989 −3.3 C[10]H[13]N[5]O[5] 283.09
Guanosine DL
4 3.16 [M+H]^+ 328.1384 328.1391 −2.1 C[15]H[21]NO[7] 327.13
Fructose-phenylalanine DL
5 3.21 [M+FA−H]^− 731.2226 731.2251 −3.5 C[27]H[42]O[20] 686.23
Rhmannioside D SD
6 5.11 [M−H]^− 359.1 359.0984 4.5 C[15]H[20]O[10] 360.11 Erigeside C HH
7 5.13 [M+Na]^+ 498.1568 498.1582 −2.8 C[20]H[29]NO[12] 475.17
O-β-D-Gentiobiosyl-D-(-)-mandelamide TR
8 5.49 [M+H]^+ 205.0961 205.0972 −5.1 C[11]H[12]N[2]O[2] 204.09
L-Tryptophan DL/TR
9 5.59 [M−H]^− 391.1229 391.1246 −4.3 C[16]H[24]O[11] 392.13
Shanzhiside SZ
10 6.72 [M+FA−H]^− 449.1292 449.1301 −1.9 C[17]H[24]O[11] 404.13
Feretoside SZ
11 7.06 [M+FA−H]^− 407.1561 407.1559 0.5 C[16]H[26]O[9] 362.16
5-Deoxylamiol SD
12 7.33 [M]^+ 314.1742 314.1751 −2.8 C[19]H[24]NO[3] 314.18
Magnocurarine HB
13 8.11 [M+FA−H]^− 449.13 449.1301 −0.1 C[17]H[24]O[11] 404.13
Gardenoside SZ
14 8.48 [M+H]^+ 384.1151 384.115 0.3 C[14]H[17]N[5]O[8] 383.11
Succinyladenosine DL
15 9.28 [M−H]^− 475.1467 475.1457 2.1 C[20]H[28]O[13] 476.15
L-(+)-mandelic acid-O-β-D-Gentiobioside TR
16 9.71 [M+FA−H]^− 449.1305 449.1301 1 C[17]H[24]O[11] 404.13 Deacetyl
asperulosidic acid methyl ester SZ
17 9.98 [M−H]^− 345.1555 345.1555 −1.1 C[16]H[26]O[8] 346.16
Jasminoside B SZ
18 10.12 [M]^+ 342.1695 342.17 −1.4 C[20]H[24]NO[4] 342.17
Phellodendrine HB
19 10.27 [M−H]^− 475.1443 475.1457 −3 C[20]H[28]O[13] 476.15
D-(+)-mandelic acid-O-β-D-Gentiobioside TR
20 10.35 [M+H]^+ 506.1994 506.2021 −5.3 C[25]H[31]NO[10] 505.19
L-Phenylalaninosecologanin B HQs
21 10.4 [M+FA−H]^− 493.2272 493.2291 −3.8 C[21]H[36]O[10] 448.23
Atractyloside A BZ
22 10.53 [M+FA−H]^− 391.1616 391.161 1.6 C[16]H[26]O[8] 346.16
Jasminoside D SZ
23 10.94 [M−H]^− 353.0865 353.0878 −3.7 C[16]H[18]O[9] 354.10
Neochlorogenic acid SZ/HB/HH/DG/SZ
24 10.99 [M+H]^+ 448.1958 448.1966 −1.8 C[23]H[29]NO[8] 447.19
N-Methylhigenamine 7-glucopyranoside HB
25 10.61 [M]^+ 344.1843 344.1856 −3.9 C[20]H[26]NO[4] 344.19
Tembetarine HB
26 11.22 [M]^+ 342.1673 342.17 −7.8 C[20]H[24]NO[4] 342.17 Magnoflorine
HB/HL
27 11.24 [M+FA−H]^− 477.1601 477.1614 −2.7 C[19]H[28]O[11] 432.16
8-O-Acetylmussaenoside SZ
28 11.68 [M−H]^− 431.155 431.1559 −2.1 C[19]H[28]O[11] 432.16
Cuchiloside RG
29 12.16 [M−H]^− 787.1941 787.1938 −0.1 C[33]H[40]O[22] 788.20
Quercetin3-O-β-D-glucopyranosyl-7-O-β-gentiobioside TLZ
30 12.44 [M+FA−H]^− 502.1558 502.1566 −1.6 C[20]H[27]NO[11] 457.16
L-Amygdalin TR
31 12.52 [M]^+ 314.1735 314.1751 −5 C[19]H[24]NO[3] 314.18 Oblongine HB
32 12.62 [M+FA−H]^− 502.1582 502.1566 0.5 C[20]H[27]NO[11] 457.16
D-Amygdalin TR
33 12.64 [M+FA−H]^− 595.1871 595.188 −1.5 C[23]H[34]O[15] 550.19
Genipin 1-gentiobioside SZ/SD
34 13.92 [M+H]^+ 308.091 308.0917 −2.4 C[18]H[13]NO[4] 307.08
Lycoranine B HL
35 14.7 [M]^+ 356.1836 356.1856 −5.7 C[21]H[26]NO[4] 356.19 Menisperine
HB
36 14.96 [M−H]^− 353.0871 353.0878 −2 C[16]H[18]O[9] 354.10 Chlorogenic
acid SZ/HB/HH/DG/SZ
37 15.15 [M+FA−H]^− 433.1366 433.1352 3.3 C[17]H[24]O[10] 388.14
Geniposide SZ/SD
38 15.6 [M+H]^+ 504.2224 504.2228 −0.8 C[26]H[33]NO[9] 503.22
(13aS)-5,8,13,13a-Tetrahydro-3,9,10-trimethoxy-6H-dibenzo[a,g]quinolizi
n-2-yl β-D-glucopyranoside HL
39 16.14 [M−H]^− 367.103 367.1035 5.3 C[17]H[20]O[9] 368.11
5-O-Feruloylquinic acid HB
40 16.81 [M−H]^− 353.0864 353.0878 −4 C[16]H[18]O[9] 354.10
Cryptochlorogenic acid SZ/HB/HH/DG/SZ
41 17.45 [M+H]^+ 377.1452 377.1456 −1 C[17]H[20]N[4]O[6] 376.14 Vitamin
B2 LG
42 17.47 [M+FA−H]^− 525.1624 525.1614 2 C[23]H[28]O[11] 480.16
Albiflorin BS
43 17.63 [M]^+ 356.1849 356.1856 −2.1 C[21]H[26]NO[4] 356.19
5,6,6a,7-Tetrahydro-11-hydroxy-1,2,10-trimethoxy-6,6-dimethyl-4H-dibenz
o[de,g]quinolinium HB
44 18.65 [M+FA−H]^− 375.1649 375.1661 −3.1 C[16]H[26]O[7] 330.17
Epijasminoside A SZ
45 19.53 [M+FA−H]^− 375.1656 375.1661 −1.2 C[16]H[26]O[7] 330.17
Picrocrocin SZ
46 21.14 [M+FA−H]^− 525.1647 525.1614 6.4 C[23]H[28]O[11] 480.16
Paeoniflorin BS
47 21.39 [M+H]^+ 356.1843 356.1856 −3.7 C[21]H[25]NO[4] 355.18
Tetrahydropalmatine HL/HB
48 22.14 [M+H]^+ 352.1059 352.1179 −5.5 C[20]H[17]NO[5] 351.11
8-Oxoepiberberine HL/HB
49 22.2 [M]^+ 322.1057 322.1074 −5.2 C[19]H[16]NO[4] 322.11
Groenlandicine HL
50 22.81 [M]^+ 324.1227 324.123 −1 C[19]H[18]NO[4] 324.12
Demethyleneberberine HL/HB
51 23.63 [M−H]^− 611.1627 611.1618 1.5 C[27]H[32]O[16] 612.17
Hydroxysafflor Yellow A HH
52 23.78 [M−H]^− 367.1027 367.1035 −2.1 C[17]H[20]O[9] 368.11
3-O-Feruloylquinic acid HB
53 24.05 [M+H]^+ 260.1272 260.1281 −3.5 C[15]H[17]NO[3] 259.12
Platydesmine HB
54 24.21 [M−H]^− 543.1181 543.1144 5.5 C[26]H[24]O[13] 544.12
Hyemaloside B BS
55 24.23 [M]^+ 356.1842 356.1856 −4 C[21]H[26]NO[4] 356.19
N-Methylcorydine HL/HB
56 24.32 [M−H]^− 771.2008 771.1989 2.4 C[33]H[40]O[21] 772.21 Quercetin
3-O-glucosyl-rutinoside TLZ
57 24.43 [M]^+ 354.1687 354.17 −3.6 C[21]H[24]NO[4] 354.17
N-Methylcanadine HL/HB
58 24.48 [M−H]^− 625.141 625.141 0 C[27]H[30]O[17] 626.15
Quercetin7-O-β-gentiobioside TLZ
59 24.57 [M−H]^− 785.2522 785.251 1.6 C[35]H[46]O[20] 786.26
Purpureaside C SD
60 25.1 [M]^+ 320.0911 320.0917 −2 C[19]H[14]NO[4] 320.09 Coptisine HL
61 25.58 [M−H]^− 367.1027 367.1035 −2.1 C[17]H[20]O[9] 368.11
4-O-Feruloylquinic acid HB
62 25.86 [M]^+ 336.1212 336.123 −5.5 C[20]H[18]NO[4] 336.12
Epiberberine HL/HB
63 25.99 [M−H]^− 385.1152 385.114 3.1 C[17]H[22]O[10] 386.12
2-Hydroxyethyl,
6-[(2E)-3-(3,4-dihydroxyphenyl)-2-propenoate]-β-D-glucopyranoside BZ
64 26.03 [M−H]^− 193.0521 193.0506 7.6 C[10]H[10]O[4] 194.06 Ferulic
Acid CX/DG/LG
65 26.09 [M]^+ 338.1373 338.1387 −4.1 C[20]H[20]NO[4] 338.14
Columbamine HL/HB
66 26.3 [M+FA−H]^− 491.1208 491.1195 2.6 C[22]H[22]O[10] 446.12
Calycosin-7-glucoside HQ
67 26.37 [M−H]^− 799.2677 799.2666 1.4 C[36]H[48]O[20] 800.27 Jionoside
A1 SD
68 26.4 [M−H]^− 547.1475 547.1457 3.3 C[26]H[28]O[13] 548.15
Chrysin-6-C-hexoside -8-C- pentoside HQs
69 26.42 [M]^+ 338.1375 338.1387 −3.5 C[20]H[20]NO[4] 338.14
Jateorhizine HL/HB
70 26.82 [M−H]^− 547.1451 547.1457 −1.1 C[26]H[28]O[13] 548.15
Chrysin-6-C-glucoside-8-C-arabinoside HQs
71 27.17 [M−H]^− 547.1492 547.1457 6.4 C[26]H[28]O[13] 548.15
Chrysin-6-C-hexoside -8-C- pentoside HQs
72 27.47 [M+H]^+ 352.1163 352.1179 −4.7 C[20]H[17]NO[5] 351.11
Oxoberberine HL/HB
73 27.73 [M−H]^− 547.1472 547.1457 2.7 C[26]H[28]O[13] 548.15
Chrysin-6-C-pentoside-8-C-hexoside HQs
74 27.92 [M−H]^− 631.1714 631.1668 7.2 C[30]H[32]O[15] 632.17
Galloylpaeoniflorin BS
75 27.99 [M−H]^− 547.148 547.1457 4.2 C[26]H[28]O[13] 548.15
Chrysin-6-C-arabinoside-8-C-glucoside HQs
76 28.31 [M−H]^− 609.1492 609.1461 5.1 C[27]H[30]O[16] 610.15 Rutin HH
77 28.44 [M−H]^− 623.198 623.1981 −0.2 C[29]H[36]O[15] 624.21
Aceteoside SD
78 28.52 [M]^+ 336.1212 336.123 −1 C[20]H[18]NO[4] 336.12 Berberine
HL/HB
79 28.67 [M−H]^− 547.147 547.1457 2.3 C[26]H[28]O[13] 548.15
Chrysin-6-C-pentoside-8-C-hexoside HQs
80 28.82 [M]^+ 352.1529 352.1543 −4.1 C[21]H[22]NO[4] 352.15 Palmatine
HB
81 28.98 [M+FA−H]^− 579.1723 579.1719 0.6 C[26]H[30]O[12] 534.17
Amurensin HB
82 29.76 [M+FA−H]^− 671.2206 671.2193 2 C[29]H[38]O[15] 626.22
Isomucronulatol-7,2′-di-O-glucoside HQ
83 29.86 [M−H]^− 623.2002 623.1981 3.3 C[29]H[36]O[15] 624.21
Isoaceteoside SD
84 30.02 [M−H]^− 461.0709 461.0725 −3.6 C[21]H[18]O[12] 462.08
Scutellarin HQs
85 30.15 [M+FA−H]^− 537.2182 537.2189 −1.3 C[22]H[36]O[12] 492.22
Jasminoside I/H/S SZ
86 30.21 [M−H]^− 491.1226 491.1254 −5.6 C[23]H[24]O[12] 492.13
Eupatolin HH
87 30.03 [M+H]^+ 207.101 207.1016 −2.8 C[12]H[14]O[3] 206.09
Senkyunolide F CX
88 30.36 [M+FA−H]^− 507.1116 507.1144 −5.6 C[22]H[22]O[11] 462.12
Pratensein-7-O-glucoside HQ
89 30.61 [M−H]^− 593.1523 593.1512 1.9 C[27]H[30]O[15] 594.16
Kaempferol-3-O-rutinoside TLZ/HH
90 30.92 [M]^+ 350.1367 350.1387 −5.7 C[21]H[20]NO[4] 350.14 Fagaronine
HB
91 31.94 [M+FA−H]^− 507.1505 507.1508 0.2 C[23]H[26]O[10] 462.15
Lactiflorin BS
92 32.31 [M−H]^− 755.2388 755.2404 −2.1 C[34]H[44]O[19] 756.25
6''-O-[trans-Sinapoyl] -genipin gentiobioside SZ
93 32.87 [M−H]^− 725.2311 725.2298 1.7 C[33]H[42]O[18] 726.24
6''-O-[trans-Feruloyl] genipin gentiobioside SZ
94 33.48 [M+FA−H]^− 475.1248 475.1246 0.5 C[22]H[22]O[9] 430.13 Ononin
HQ
95 33.72 [M+FA−H]^− 1021.3796 1021.377 2.6 C[44]H[64]O[24] 976.38
Crocin I SZ
96 34.02 [M−H]^− 551.2156 551.2134 4 C[27]H[36]O[12] 552.22
6′-O-trans-Sinapoyljasminoside L SZ
97 34.3 [M−H]^− 431.0987 431.0984 0.8 C[21]H[20]O[10] 432.11
Apigenin-7-O- β-D-glucoside HH
98 35.35 [M−H]^− 559.1479 559.1457 3.9 C[27]H[28]O[13] 560.15
3-O-Sinapoyl-5-O-caffeoylquinic acid SZ
99 35.38 [M−H]^− 551.2161 551.2134 4.9 C[27]H[36]O[12] 552.22
6′-O-trans-Sinapoyljasminoside L Isomer SZ
100 35.38 [M−H]^− 345.0607 345.0616 −2.6 C[17]H[14]O[8] 346.07
Viscidulin III HQs
101 35.63 [M−H]^− 593.1883 593.1876 1.2 C[28]H[34]O[14] 594.19
6′-O-sinapoylgeniposide SZ
102 36.08 [M−H]^− 475.0876 475.0882 −1.3 C[22]H[20]O[12] 476.10
5,7,2′-Trihydroxy-6-methoxy flavone-7-O-glucuronide HQs
103 36.21 [M+FA−H]^− 507.1509 507.1508 0.2 C[23]H[26]O[10] 462.15
Methylnissolin 3-O-glucoside HQ
104 36.48 [M−H]^− 445.0777 445.0776 0.8 C[21]H[18]O[11] 446.08 Baicalin
HQs
105 38.23 [M−H]^− 447.094 447.0933 1.6 C[21]H[20]O[11] 448.10
Dihydrobaicalin HQs
106 38.32 [M−H]^− 559.1444 559.1457 −2.4 C[27]H[28]O[13] 560.15
4-O-sinapoyl-5-O-caffeoylquinic acid SZ
107 38.53 [M−H]^− 463.1628 463.161 3.9 C[23]H[28]O[10] 464.17
Isomucronulatol-7-O-glucoside HQ
108 38.9 [M−H]^− 447.0936 447.0933 0.7 C[21]H[20]O[11] 448.10
Naringenin-7-O-glucuronide HQs
109 39.03 [M−H]^− 659.1601 659.1618 −2.5 C[31]H[32]O[16] 660.17
3,5-Di-O-caffeoyl-4-O-(3-hydroxy-3-methyl) glutaroylquinic acid SZ
110 39.11 [M−H]^− 559.148 559.1457 0.7 C[27]H[28]O[13] 560.15
3-O-Sinapoyl-4-O-caffeoylquinic acid SZ
111 39.41 [M−H]^− 283.0622 283.0612 3.5 C[16]H[12]O[5] 284.07 Calycosin
HQ
112 39.79 [M−H]^− 445.0788 445.0776 2.6 C[21]H[18]O[11] 446.08
Norwogonin 7-O-β-D-glucuronide HQs
113 40.57 [M−H]^− 475.0893 475.0882 2.3 C[22]H[20]O[12] 476.10
Diosmetin 7-O-β-D-glucuronide SD
114 40.79 [M−H]^− 445.0792 445.0776 3.5 C[21]H[18]O[11] 446.08
Baicalein 6-O-β-D-glucuronide HQs
115 41.42 [M−H]^− 429.0837 429.0827 2.3 C[21]H[18]O[10] 430.09
Chrysin7-O-β-D-glucuronide HQs
116 41.59 [M−H]^− 459.0926 459.0933 −1.5 C[22]H[20]O[11] 460.10
Oroxylin A 7-O-glucuronide HQs
117 42.35 [M−H]^− 475.088 475.0882 −0.4 C[22]H[20]O[12] 476.10
5,6,7-Trihydroxy-8-methoxyflavone-7-O-glucuronopyranoside HQs
118 43.65 [M−H]^− 459.0921 459.0933 −2.6 C[22]H[20]O[11] 460.10
Wogonoside HQs
119 47.39 [M+H]^+ 947.5185 947.521 −2.6 C[47]H[78]O[19] 946.51
Astragaloside VI HQ
120 47.63 [M−H]^− 299.0566 299.0561 1.6 C[16]H[12]O[6] 300.06
3′,5,7-Trihydroxy-4′-methoxyflavone SD
121 47.8 [M+H]^+ 191.1056 191.1067 −5.5 C[12]H[14]O[2] 190.10
3-N-butylphthalide CX/DG
122 48.07 [M+FA−H]^− 1021.3796 1021.377 2.6 C[44]H[64]O[24] 976.38
Crocin I SZ
123 48.19 [M−H]^− 269.0465 269.0455 3.5 C[15]H[10]O[5] 270.05 Baicalein
HQs
124 48.58 [M+H]^+ 947.5211 947.521 0.1 C[47]H[78]O[19] 946.51
Astragaloside VI isomer HQ
125 48.87 [M−H]^− 329.2344 329.2333 3.2 C[18]H[34]O[5] 330.24 Pinellic
acid /
126 49.18 [M+FA−H]^− 829.4589 829.4591 −0.3 C[41]H[68]O[14] 784.46
Astragaloside IV HQ
127 49.34 [M+FA−H]^− 549.3416 549.3433 −8.6 C[30]H[48]O[6] 504.35
16-Oxoalisol A ZX
128 49.62 [M+FA−H]^− 697.2698 697.2713 −2.2 C[32]H[44]O[14] 652.27
Crocin Ⅲ SZ
129 50.07 [M+FA−H]^− 515.1925 515.1923 0.4 C[26]H[30]O[8] 470.19
Limonin HH
130 50.38 [M+H]^+ 827.4476 827.4787 −3.4 C[43]H[70]O[15] 826.47
Astragaloside Ⅱ HQ
131 50.88 [M−H]^− 651.2668 651.2658 1.5 C[32]H[44]O[14] 652.27 Crocin Ⅲ
isomer SZ
132 51.01 [M+H]^+ 547.3624 547.3629 −1 C[32]H[50]O[7] 546.36 23-Acetyl
16-oxoalisol A ZX
133 51.24 [M+H]^+ 827.4781 827.4787 −0.8 C[43]H[70]O[15] 826.47
Isoastragaloside Ⅱ HQ
134 51.68 [M−H]^− 283.0616 283.0612 1.4 C[16]H[12]O[5] 284.07 Wogonin
HQs
135 51.86 [M+H]^+ 827.4789 827.4787 0.2 C[43]H[70]O[15] 826.47
Cyclosiversioside D HQ
136 52 [M−H]^− 373.0908 373.0929 −5.6 C[19]H[18]O[8] 374.10
Skullcapflavone II HQs
137 52.09 [M+H]^+ 193.1217 193.1223 −3.1 C[12]H[16]O[2] 192.12
Senkyunolide A CX/DG
138 52.49 [M−H]^− 283.0623 283.0612 3.9 C[16]H[12]O[5] 284.07 Oroxylin
A HQs
139 52.64 [M+H]^+ 231.1377 231.138 −1.1 C[15]H[18]O[2] 230.13
Atractylenolide III BZ
140 52.83 [M+H]^+ 487.34 487.3418 −3.7 C[30]H[46]O[5] 486.33 Alisol C
ZX
141 53.08 [M+H]^+ 869.4859 869.4893 −3.9 C[45]H[72]O[16] 868.48
Astragaloside I HQ
142 53.6 [M−H]^− 311.2233 311.2228 1.7 C[18]H[32]O[4] 312.23
12,13-Dihydroxy-9Z,15Z-octadecadienoic acid /
143 53.86 [M+H]^+ 869.4908 869.4893 3.1 C[45]H[72]O[16] 868.48
Isoastragaloside I HQ
144 54.84 [M−H]^− 519.333 519.3327 0.5 C[30]H[48]O[7] 520.34 Alisol P
ZX
145 55.05 [M+H]^+ 869.4896 869.4893 0.3 C[45]H[72]O[16] 868.48
Neoastragaloside I HQ
146 55.13 [M+H]^+ 191.1065 191.1067 −0.8 C[12]H[14]O[2] 190.10
Ligustilide CX/DG
147 55.32 [M+FA−H]^− 573.3455 573.3433 3.8 C[32]H[48]O[6] 528.35
23-Acetyl alisol C ZX
148 56.41 [M+H]^+ 233.1532 233.1536 −1.7 C[15]H[20]O[2] 232.15
Atractylenolide II BZ
149 57.41 [M+FA−H]^− 535.3641 535.364 0.1 C[30]H[50]O[5] 490.37 Alisol
A ZX
150 61.84 [M+H]^+ 515.3707 515.3731 −4.7 C[32]H[50]O[5] 514.37
23-Acetyl alisol B ZX
[101]Open in a new tab
FIGURE 4.
[102]FIGURE 4
[103]Open in a new tab
The base peak intensity chromatogram of NFPX by UPLC-HRMS in negative
ion mode.
FIGURE 6.
[104]FIGURE 6
[105]Open in a new tab
The UV chromatogram of NFPX in 254 nm.
FIGURE 5.
[106]FIGURE 5
[107]Open in a new tab
The base peak intensity chromatogram of NFPX by UPLC-HRMS in positive
ion mode.
Screening of Bioactive Components and Targets in NFPX on ARDS
A total of 1610 components in NFPX were obtained from the TCMSP
database and SymMap database ([108]Supplementary Table S2). OB ≥ 30%
and DL index ≥0.18 served as the criteria of bioactive components.
Among the 1610 components in NFPX, 821 components (51.0%) met the
criterion of OB ≥ 30%, 663 components (41.2%) met the criterion of DL
index ≥0.18, and 254 components (15.8%) met both criteria of OB ≥ 30%
and DL index ≥0.18. Therefore, these 254 components were selected as
candidate bioactive components for further analyses ([109]Supplementary
Table S3). Among the 254 candidate bioactive components, 13754 protein
targets were retrieved from the TCMSP database and SymMap database
([110]Supplementary Table S4). 3381 gene symbols for ARDS were
collected from the GeneCards database and OMIM database
([111]Supplementary Table S5). Then, gene intersections were generated
by mapping the targets of NFPX with ARDS using the CTD database.
Consequently, 77 targets of 37 components in NFPX associated with ARDS
were obtained, and the detailed information of the 77 targets of NFPX
on ARDS is shown in [112]Table 2. PPI network was constructed to reveal
the intersections of 77 target symbols using the STRING software
([113]Figure 7).
TABLE 2.
Targets of NFPX on ARDS were screened by network pharmacology analysis.
Herb name Symbol Description Score
Atractylodis macrocephalae rhizoma AR Androgen receptor 39.29
Atractylodis macrocephalae rhizoma NCOA2 Nuclear receptor coactivator 2
5.4
Carthami Flos ADA Adenosine deaminase 25.31
Carthami Flos ALOX5 Arachidonate 5-lipoxygenase 35.92
Carthami Flos APOD Apolipoprotein D 2.28
Carthami Flos CD40LG CD40 Ligand 72.98
Carthami Flos CRAT Carnitine O-acetyltransferase 9.6
Carthami Flos CRP C-reactive protein 49.97
Carthami Flos CTSD Cathepsin D 20.47
Carthami Flos EGFR Epidermal growth factor receptor 42.83
Carthami Flos EIF6 Eukaryotic translation initiation factor 6 8.88
Carthami Flos EPHX1 Epoxide hydrolase 1 12.06
Carthami Flos GFAP Glial fibrillary acidic protein 28.46
Carthami Flos GPHN Gephyrin 21.29
Carthami Flos GSTM1 Glutathione S-transferase Mu 1 15.39
Carthami Flos ICAM1 Intercellular adhesion molecule 1 44.2
Carthami Flos IFNG Interferon gamma 61.83
Carthami Flos INS Insulin 60.63
Carthami Flos INSR Insulin receptor 19.9
Carthami Flos IRF1 Interferon regulatory factor 1 29.12
Carthami Flos MAPK8 Mitogen-activated protein kinase 8 20.63
Carthami Flos NR1I2 Nuclear receptor subfamily 1 group I member 2 6.09
Carthami Flos NR1I3 Nuclear receptor subfamily 1 group I member 3 8.89
Carthami Flos REN Renin 39.96
Carthami Flos SLC22A5 Solute carrier family 22 member 5 18.11
Carthami Flos STAT3 Signal transducer and activator of transcription 3
51.28
Carthami Flos THBD Thrombomodulin 53.46
Chuanxiong Rhizoma ABI1 Abl interactor 1 4.84
Chuanxiong Rhizoma ADORA2A Adenosine A2a receptor 17.54
Chuanxiong Rhizoma CCK Cholecystokinin 15.54
Chuanxiong Rhizoma CHAT Choline O-acetyltransferase 38.56
Chuanxiong Rhizoma GAD2 Glutamate decarboxylase 2 9.58
Chuanxiong Rhizoma GAMT Guanidinoacetate N-methyltransferase 23.69
Chuanxiong Rhizoma GCG Glucagon 13.57
Chuanxiong Rhizoma HTR3A 5-Hydroxytryptamine receptor 3A 15.77
Chuanxiong Rhizoma LPL Lipoprotein lipase 17.68
Chuanxiong Rhizoma MAPK14 Mitogen-activated protein kinase 14 13.1
Chuanxiong Rhizoma PYY Peptide YY 10.3
Cinnamomi Cortex IRF3 Interferon regulatory factor 3 29.91
Cinnamomi Cortex PRL Prolactin 20.72
Cinnamomi Cortex TRPV4 Transient Receptor Potential Cation Channel
Subfamily V Member 4 26.23
Gardeniae Fructus KCNH2 Potassium voltage-gated channel subfamily H
member 2 40.48
Gardeniae Fructus SMPD2 Sphingomyelin phosphodiesterase 2 5.43
Gardeniae Fructus SOAT1 Sterol O-acyltransferase 1 2.07
Gardeniae Fructus TYR Tyrosinase 27.98
Astragali radix FASN Fatty acid synthase 3.77
Descurainiae semen lepidii semen PPARG Peroxisome
proliferator-activated receptor gamma 38.7
Descurainiae semen lepidii semen TRPA1 Transient receptor potential
cation channel subfamily A member 1 15.59
Descurainiae semen lepidii semen TRPV1 Transient receptor potential
cation channel subfamily V member 1 14.15
Descurainiae semen lepidii semen VEGFA Vascular endothelial growth
factor A 55.21
Paeoniae Radix Alba ABAT 4-Aminobutyrate aminotransferase 4.17
Paeoniae Radix Alba APRT Adenine phosphoribosyltransferase 20.39
Paeoniae Radix Alba ASL Argininosuccinate lyase 16.65
Paeoniae Radix Alba CAT Catalase 31.01
Paeoniae Radix Alba CBS Cystathionine beta-synthase 10.92
Paeoniae Radix Alba GAPDH Glyceraldehyde-3-phosphate dehydrogenase
20.87
Paeoniae Radix Alba GYS1 Glycogen synthase 1 5.42
Paeoniae Radix Alba HAO1 Hydroxyacid oxidase 1 0.94
Paeoniae Radix Alba HDAC8 Histone deacetylase 8 8.09
Paeoniae Radix Alba HDC Histidine decarboxylase 15.18
Paeoniae Radix Alba HMOX1 Heme oxygenase 1 33.65
Paeoniae Radix Alba HP Haptoglobin 40.54
Paeoniae Radix Alba KYNU Kynureninase 9.46
Paeoniae Radix Alba LTF Lactotransferrin 16.34
Paeoniae Radix Alba LYZ Lysozyme 10.24
Paeoniae Radix Alba MAPK1 Mitogen-activated protein kinase 1 38.57
Paeoniae Radix Alba MMUT Methylmalonyl-CoA mutase 16.2
Paeoniae Radix Alba MPO Myeloperoxidase 65.09
Paeoniae Radix Alba PYCR1 Pyrroline-5-carboxylate reductase 1 10.26
Paeoniae Radix Alba SPR Sepiapterin reductase 2.82
Paeoniae Radix Alba TPO Thyroid peroxidase 13.14
Phellodendri Chinrnsis Cortex TNF Tumor necrosis factor 102.74
Phellodendri Chinrnsis Cortex TRPV3 Transient receptor potential cation
channel subfamily V member 3 7.41
Polyporus DHCR7 7-Dehydrocholesterol reductase 17.47
Rehmanniae Radix Praeparata P3H1 Prolyl 3-hydroxylase 1 9.44
Rehmanniae Radix Praeparata PGR Progesterone receptor 11.52
Rehmanniae Radix Praeparata PLG Plasminogen 36.75
[114]Open in a new tab
FIGURE 7.
[115]FIGURE 7
[116]Open in a new tab
The PPI network of targets between NFPX and ARDS. Each node represents
a protein and edges represent protein–protein associations.
Herbs-Compounds-targets Network Analysis
To investigate the underlying mechanisms of NFPX on ARDS,
Herbs-compounds-targets network of NFPX on ARDS was constructed, which
included 125 nodes and 552 edges, displaying that multiple compounds
and targets are involved in the effects of NFPX treating ARDS
([117]Figure 8). Among these bioactive components, the top five degree
components associated with multiple ARDS targets include histidine
decarboxylase (MOL4480, degree = 18), androgen receptor (MOL422, degree
= 11), telomerase protein component 1 (MOL675, degree = 10), amine
oxidase B (MOL 1801, degree = 9), nitric-oxide synthase (MOL 1893,
degree = 6). In addition, the top five-degree targets related to
multiple bioactive compounds include INS (degree = 47), GAPDH (degree =
40), TNF (degree = 36), VEGFA (degree = 34), CAT (degree = 33).
FIGURE 8.
[118]FIGURE 8
[119]Open in a new tab
Herbs-compounds-targets network of NFPX on ARDS. The red arrow
represents herbs in NFPX; the orange diamond represents the bioactive
compounds of NFPX; the blue ellipse represents the target genes.
Analysis of GO and KEGG Enrichment Pathway
To clarify the biological characteristics of putative targets of NFPX
on ARDS in detail, the GO and KEGG pathway analyses of involved targets
were conducted. The enrichment results included 1366 BP terms, 346 MF
terms, and 188 CC terms. The top 20 significantly enriched terms in
biological process (BP), molecular function (MF), and cellular
component (CC) categories are shown in [120]Figures 9A–C, which
indicated that NFPX may regulate inflammatory action via identical
protein binding, nuclear receptor activity and enzyme binding in
extracellular space, extracellular region, and cell surface to exert
its therapeutic effects on ARDS. 233 relevant pathways of NFPX were
obtained by KEGG pathway enrichment. The key KEGG pathways of NFPX on
ARDS are shown in [121]Figure 9D, including the HIF-1 signaling
pathway, AGE-RAGE signaling pathway, and FOXO signaling pathway, which
are involved in the processes of oxidative stress, inflammatory
response, cell metabolism, and cell cycle.
FIGURE 9.
[122]FIGURE 9
[123]Open in a new tab
The 20 most significance therapy target genes of GO and KEGG pathway
enrichment analysis of NFPX on ARDS. (A) GO analysis in biological
process (BP); (B) GO analysis in molecular function (MF); (C) GO
analysis in cellular component (CC). (D) KEGG pathway enrichment
analysis.
RNA-Seq Analysis
To further verify the target genes, six groups of mice lung tissues
were analyzed for RNA-seq detection. Overall, more than 1965 million
reads were acquired and the percentages of uniquely mapped paired reads
were 87.19–89.38%. Hierarchical clustering heatmap illustrated 11629
significantly DEGs. It is clear that such a cluster thermogram
successfully separates the control group from the LPS group. In
contrast, the gene expression profile of LPS+NFPX group lies between
the control group and LPS group and the gene expression profile of the
LPS+LNFPX group was more similar to that of the LPS group compared with
that of the LPS+MNFPX group and LPS+HNFPX group ([124]Figure 10).
FIGURE 10.
FIGURE 10
[125]Open in a new tab
Hierarchical clustering analysis of genes that were differentially
expressed in lung tissue samples using heatmap; each group contains
four to five individuals. Blue-white indicates lower expression, and
red indicates high expression.
Then, these DEGs were further subjected to annotation with volcano maps
by DESeq2 software. Compared with the LPS group, 21 significantly
upregulated genes and 48 downregulated genes in the LPS+LNFPX group;
one upregulated gene and two downregulated genes in the LPS+MNFPX
group; 96 upregulated gene and 403 downregulated genes in the LPS+HNFPX
group were screened ([126]Figure 11). The summary of upregulated and
downregulated genes is presented in [127]Supplementary Table S6.
FIGURE 11.
FIGURE 11
[128]Open in a new tab
The DEGs with statistical significance from lung tissues between ARDS
mice and ARDS mice were pretreated by different concentrations of NFPX
screened using a volcano plot. Red notes indicate upregulated genes,
and blue notes indicate downregulated genes. (A) LPS group vs.
LPS+LNFPX group; (B) LPS group vs. LPS+MNFPX group; (C) LPS group vs.
LPS+HNFPX group.
At last, we performed GO and KEGG pathway analysis to highlight the up-
and downregulation of four groupings of genes. As depicted in
[129]Figure 12, top 20 generally changed GO terms and KEGG pathways
were ranked by enrichment score. The immune-inflammation response
pathway had the largest number of DEGs. The most enriched GO terms of
LPS vs. LPS+LNFPX included immune system process, lymphocyte
activation, and T cell activation ([130]Figure 12A). The mainly
enriched GO terms of LPS vs. LPS+MNFPX included response to hyperoxia,
energy coupled proton transport, and ATP synthesis ([131]Figure 12C).
The represented enriched GO terms of LPS vs. LPS+HNFPX included immune
system process, immune response, and leukocyte activation ([132]Figure
12E). Analogously, KEGG enrichment analysis also displayed that the
mainly enriched pathways were connected with the immune-inflammation
response. The most enriched KEGG pathways of LPS vs. LPS+LNFPX included
primary immunodeficiency, T cell receptor signaling pathway, and
hematopoietic cell lineage ([133]Figure 12B). The most enriched KEGG
pathways of LPS vs. LPS+MNFPX included oxidative phosphorylation,
ribosome, and Parkinson’s disease ([134]Figure 12D). The most enriched
KEGG pathways of LPS vs. LPS+HNFPX included Staphylococcus aureus
infection, allograft rejection, and Leishmaniasis ([135]Figure 12F).
FIGURE 12.
[136]FIGURE 12
[137]Open in a new tab
GO analysis (A,C,E) and KEGG pathway analysis (B,D,F) of the biological
function of differentially regulated genes. (A,B) LPS group vs.
LPS+LNFPX group; (C,D) LPS group vs. LPS+MNFPX group; (E,F) LPS group
vs. LPS+HNFPX group.
Specific Gene Module–Based Target Identification for NFPX Based on the
Transcriptional Data
We here utilized a gene module pair–based target identification (GMPTI)
approach ([138]http://www.bcxnfz.top/TMP/) to predict biological
targets based on NFPX-induced gene expression profiles. GMPTI was
proposed based on the assumption that similar drugs induced similar
gene expression responses. Firstly, a specific transcriptional gene
module pair (GMP) was automatically extracted for each target-induced
transcriptional profile and can be used as a gene signature to
represent the target. Then, for NFPX, we can calculate correlation
scores for the GMPs of each target with the NFPX-induced gene
expression profiles (see Methods). The correlation analysis among
groups shown in [139]Supplementary Table S7 suggests that the data are
reliable. 3275 potential targets are listed in [140]Supplementary Table
S8 by comparing the p value of the LPS group with LPS+NFPX groups. With
comprehensive analysis of network pharmacology, transcriptomics, and
artificial intelligence, eight ARDS-related targets were selected:
SMAD4, HIF-1, AMPK, HRAS, SOD1, AKT2, RAC1, and P53.
Then, these targets were docked by the NFPX ingredients with a
three-dimensional structure on the representative conformations using
the SYBYL − Surflex docking in standard precision mode. The docking
results were ranked based on the CScore ranking ([141]Supplementary
Table S9). We observed many compound–target interactions with high
docking scores. For example, with the docking score of 5 as the
threshold, we can find that 63, 1, 105, 87, 84, 78, 99, and 17
ingredients interact with AKT2, AMPK, HARS, HIF-1, P35, RAC1, SMAD4,
and SOD1, respectively. More specifically, with the docking score
ranking, some potential active components of NFPX can be screened from
these compounds. For example, the compound astragaloside IV interacts
with SMAD4, P35, HIF-1, AKT2, RAC1, HARS, AMPK, and SOD1 with a docking
score of 11.56, 10.86, 10.6, 9.75, 9.15, 8.94, 5.57, and 5.05,
respectively, indicating that astragaloside IV may function by a
multi-target mode. Similarly, neochlorogenic acid interacts with P35,
HIF-1, RAC1, SMAD4, HARS, SOD1, AKT2, and AMPK with a docking score of
10.82, 9.92, 9.64, 8.87, 8.38, 7.71, 7.32, and 4.57, respectively.
Confirmation of the Targets in ARDS Mice
At last, the potential targets mentioned above were verified by
qRT-PCR. As illustrated in [142]Figure 13, SMAD4 expression was
significantly downregulated in the MNFPX treated group and HNFPX
treated group compared with LPS-treated group (p < 0.05) ([143]Figure
13A). Moreover, the NFPX-treated group significantly decreased HIF-1
and AMPK expression in in a dose-dependent manner (p < 0.05)
([144]Figures 13B,C). In contrast, there was no statistical difference
in the expression of HRAS, SOD1, AKT2, RAC1, and P53 in NFPX+LPS groups
when compared with LPS group ([145]Figures 13D–H).
FIGURE 13.
[146]FIGURE 13
[147]Open in a new tab
mRNA expression of potential targets of NFPX by qRT-PCR. *p < 0.05.
Discussion
There has been a long history of using TCM in treating pulmonary
diseases. However, the complexity of components of formula and
ambiguity of mechanisms prevent their widespread use. In the previous
literature, almost all studies about ARDS treatment by TCM have focused
on the single component or bioactive molecules extracted from TCM
([148]Li et al., 2018; [149]Long et al., 2020). The appearance of
network pharmacology analysis and high throughput sequencing break the
barriers and greatly promote the development of TCM theory. Here, we
demonstrated that NFPX can block the occurrence and development of ARDS
for the first time. NFPX can alleviate lung impairment and prevent
airway mucus overproduction via inhibiting cell apoptosis and
inflammation, which coincide with the current treatment strategies of
ARDS that modify the inflammatory process or promote the
re-establishment of functional lung tissue. Furthermore, we explored
the potential molecular mechanisms of NFPX against the ARDS by
integrating network pharmacology, transcriptome analysis, and
artificial intelligence analysis.
ARDS is a group of clinical disorders characterized by noncardiogenic
pulmonary edema. Lung ultrasound examination has been widely used to
evaluate pulmonary edema in intensive care units due to several
advantages, including high sensitivity, bedside examination, no
radiation, and real-time assessment. Nevertheless, it has been rarely
reported that lung ultrasound was applied in the ARDS mice model
because of their small size ([150]Rubin et al., 2016). Here, an
ultrahigh-frequency transducer probe was adopted to obtain
high-resolution images. Our data gave preliminary evidences that NFPX
relieved the alveolar interstitial syndrome and pleural thickening.
Given the spatial heterogeneity of lung lesions in ARDS, both normal
and abnormal artifacts can be observed in the same image. How to
compare the scope, extent, types of lung lesions remains problematic.
Therefore, further studies are needed for quantitative analysis of lung
injury.
It is widely believed that inflammation response and oxidative stress
are the most prominent initial causes of ARDS. We not only illustrated
the therapeutic effect of NFPX in ARDS from the macroperspective by
lung ultrasound but also investigated the influence of NFPX on
pathomorphological changes, apoptosis, release of cytokines from local
lung tissues, and blood circulation in the ARDS mouse model. The LPS
intratracheal instillation mouse model is a reliable and reproducible
mouse model of ARDS ([151]Quijada et al., 2020). It has been widely
used for fundamental research due to its similar pathophysiology to
human ARDS. In our study, we observed that the degree of lung injury,
lung W/D weight ratio, inflammatory cells infiltration, cell apoptosis,
and cytokines release induced by LPS were significantly improved under
the intervention of NFPX, and these effects manifested an apparent
dose-dependent manner. Besides, apparent side effects were not observed
in mice after 1 wk of HNFPX administration. Reportedly, as the main
components of NFPX, Carthami Clos and Scutellariae Radix play an
essential role in treating LPS-induced ARDS ([152]Zhang et al., 2017;
[153]Long et al., 2020; [154]Davis et al., 2021).
The above results have authenticated that NFPX played a critical role
in treating ARDS. Nevertheless, the underlying mechanism is still
indeterminate. In this study, we integrated the data based on network
pharmacology, transcriptome, and artificial intelligence analysis.
Bioactive components were identified meeting the criteria of OB ≥ 30%
and DL index ≥0.18, which were regarded as pharmacokinetically active.
Moreover, the herbs-compounds-targets network indicated that 77 target
genes were closely associated with 37 bioactive components of NFPX. GO
analysis indicated that NFPX may regulate inflammatory action via
identical protein binding, nuclear receptor activity, and enzyme
binding in extracellular space, extracellular region, and cell surface
to exert its therapeutic effects on ARDS. A study by John et al. has
confirmed that the extracellular location and cell surface of essential
genes are quite significant ([155]Kuchtey and Kuchtey, 2014). Besides,
considering that most of the essential genes contribute to protein
binding, nuclear receptor activity, and enzyme binding, it is quite
reasonable to predict that the potential mechanisms of NFPX may involve
multiple biological processes and molecular functions. Furthermore,
KEGG enrichment shows that the principal signaling pathways
participated in the process of treating ARDS by NFPX, including HIF-1
signaling pathway, AGE-RAGE signaling pathway, and FOXO signaling
pathway. Numerous researches have proved that HIF-1, as a promoter of
inflammation storm, could aggravate the inflammation and lung injury of
ARDS ([156]Suresh et al., 2019; [157]Jahani et al., 2020;
[158]Serebrovska et al., 2020). Advanced glycation end products (AGE)
could activate its receptor RAGE and promote oxidative stress leading
to cell damage and inflammation ([159]Shen et al., 2020). The roles of
the AGE-RAGE signaling pathway are involved in lung diseases such as
ARDS, lung cancer, and idiopathic pulmonary fibrosis and have been
demonstrated in previous reports ([160]Machahua et al., 2016;
[161]Ahmad et al., 2018; [162]Zhu et al., 2021). Besides, Sandeep et
al. have announced that Forkhead box-O (FOXO) is essential in the
exudative phase of ARDS ([163]Artham et al., 2019). In sum, the data of
network pharmacology provide preliminary insights into the action
mechanism of NFPX against ARDS.
In addition, we explored the underlying mechanism via transcriptome
analysis. Consistent with the phenomena that NFPX mitigates lung edema,
cell apoptosis, and inflammatory reaction induced by LPS in a
dose-dependent manner, the gene clusters profile shown in the heatmap
also manifested the same trend. Unexpectedly, only one upregulated gene
and two downregulated genes in the LPS vs. LPS+MNFPX group were
screened probably due to sequencing error affecting the data
reliability of the LPS+MNFPX group in the following analysis. Despite
that, the GO and KEGG enrichment pathway analysis in the LPS vs.
LPS+LNFPX group and LPS+HNFPX group revealed that pathways associated
with immune-inflammation response were the core regulation mechanism,
which was in accordance with the previous data. What is more, SMAD4,
HIF-1, and AMPK were screened by comprehensive analysis of network
pharmacology, transcriptomics, and artificial intelligence. SMAD4, a
member of the SMAD family of signal transduction proteins, is expressed
in alveolar epithelial cells and has an inhibitory effect on tumors by
reducing angiogenesis and increasing blood vessel hyperpermeability.
However, Zhang and his colleagues have found that ARDS-associated
pulmonary fibrosis might ameliorate through the SMAD4 signaling pathway
([164]Zhang et al., 2015). It is well known that hypoxia is a key
feature of ARDS accompanied by multiple important cellular processes,
including cell apoptosis, inflammatory response, and angiogenesis
regulated by HIF-1. Data from previous studies have confirmed the
important effects of HIF-1 in ARDS ([165]Harris et al., 2019;
[166]Suresh et al., 2019; [167]Wang et al., 2020). Ample amounts of
evidence support the idea that the AMPK pathway exerts its effects in
LPS-induced ARDS ([168]Wang et al., 2016; [169]Bone et al., 2017;
[170]Chen et al., 2018). Therefore, the above evidence suggested that
NFPX can effectively treat ARDS by regulating the gene expression level
of SMAD4, HIF-1, and AMPK to interfere with the immune-inflammation
response. Moreover, further validation experiments are required in the
future.
Conclusion
In conclusion, we prove the efficacy of NFPX decoction in the treatment
of ARDS, thus rationalizing its potential as a novel therapeutic regime
for ARDS treatment. Additionally, integrating network pharmacology,
transcriptome, and artificial intelligence analysis illustrates the
molecular mechanism of NFPX decoction on ARDS.
Data Availability Statement
The raw data supporting the conclusions of this article will be made
available by the authors, without undue reservation.
Ethics Statement
The animal study was reviewed and approved by the Animal Ethics
Committee of The First Affiliated Hospital of Zhengzhou University
(Permit #: KY-2021-0144).
Author Contributions
XL and MY conceived and designed the experiments. XL, WM, PL, JG, QL,
CH, and YL performed the experiments. PL, WM, HN, and LX analyzed the
data. XL and MY prepared all the figures and wrote the manuscript. BF
provided technical support. All authors read and approved the final
manuscript.
Funding
This study was supported by the project of the National Science
Foundation of China (Nos. 82174175 and 82074212); the project of Health
Commission of Henan Province (Nos. SB201901036 201403060); the project
of Science and Technology Department of Henan Province (Nos.
142300410327 and 202102310381); the project of Education Department of
Henan Province (No. 19A320015).
Conflict of Interest
The authors declare that the research was conducted in the absence of
any commercial or financial relationships that could be construed as a
potential conflict of interest.
Publisher’s Note
All claims expressed in this article are solely those of the authors
and do not necessarily represent those of their affiliated
organizations, or those of the publisher, the editors and the
reviewers. Any product that may be evaluated in this article, or claim
that may be made by its manufacturer, is not guaranteed or endorsed by
the publisher.
Supplementary Material
The Supplementary Material for this article can be found online at:
[171]https://www.frontiersin.org/articles/10.3389/fphar.2021.731377/ful
l#supplementary-material
[172]Click here for additional data file.^ (154.6KB, XLSX)
[173]Click here for additional data file.^ (32.7KB, XLSX)
[174]Click here for additional data file.^ (1.9MB, DOCX)
[175]Click here for additional data file.^ (3.8MB, XLSX)
[176]Click here for additional data file.^ (455.5KB, XLSX)
[177]Click here for additional data file.^ (198.9KB, XLSX)
[178]Click here for additional data file.^ (359.1KB, XLSX)
[179]Click here for additional data file.^ (28.6KB, XLSX)
[180]Click here for additional data file.^ (427B, CSV)
References