ArticlePDF Available

Adunctin E from Conamomum rubidum Induces Apoptosis in Lung Cancer via HSP90AA1 Modulation: A Network Pharmacology and In Vitro Study

MDPI
International Journal of Molecular Sciences (IJMS)
Authors:

Abstract

Lung cancer stands out as a leading cause of death among various cancer types, highlighting the urgent need for effective anticancer drugs and the discovery of new compounds with potent therapeutic properties. Natural sources, such as the Conamomum genus, offer various bioactive compounds. Adunctin E (AE), a dihydrochalcone derived from Conamomum rubidum, exhibited several pharmacological activities, and its potential as an anticancer agent remains largely unexplored. Thus, this study aimed to elucidate its apoptotic-inducing effect and identify its molecular targets. The network pharmacology analysis led to the identification of 71 potential targets of AE against lung cancer. Subsequent gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Reactome pathway enrichment analyses revealed the involvement of these targets in cancer-associated signaling pathways. Notably, HSP90AA1, MAPK1, and PIK3CA emerged as key players in apoptosis. In silico molecular docking and dynamic simulations suggested a strong and stable interaction between AE and HSP90AA1. In vitro experiments further confirmed a significant apoptotic-inducing effect of AE on lung cancer cell lines A549 and H460. Furthermore, immunoblot analysis exhibited a substantial decrease in HSP90AA1 levels in response to AE treatment. These findings support the potential anticancer activity of AE through the HSP90AA1 mechanism, underscoring its promise as a novel compound worthy of further research and development for anti-lung cancer therapy.
Citation: Iksen, I.; Singharajkomron,
N.; Nguyen, H.M.; Hoang, H.N.T.; Ho,
D.V.; Pongrakhananon, V. Adunctin E
from Conamomum rubidum Induces
Apoptosis in Lung Cancer via
HSP90AA1 Modulation: A Network
Pharmacology and In Vitro Study. Int.
J. Mol. Sci. 2024,25, 11368. https://
doi.org/10.3390/ijms252111368
Academic Editor: Moon Nyeo Park
Received: 17 September 2024
Revised: 17 October 2024
Accepted: 21 October 2024
Published: 22 October 2024
Copyright: © 2024 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
International Journal of
Molecular Sciences
Article
Adunctin E from Conamomum rubidum Induces Apoptosis in
Lung Cancer via HSP90AA1 Modulation: A Network
Pharmacology and In Vitro Study
Iksen Iksen
1
, Natsaranyatron Singharajkomron
1
, Hien Minh Nguyen
2
, Hanh Nhu Thi Hoang
3,4
, Duc Viet Ho
4
and Varisa Pongrakhananon 1,5, *
1
Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University,
Bangkok 10330, Thailand; ikseniksen08@gmail.com (I.I.); natsaranyatron.s@gmail.com (N.S.)
2Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam;
ngyenminhhien@tdtu.edu.vn
3Faculty of Engineering and Food Technology, Hue University of Agriculture and Forestry, Hue University,
Hue City 49000, Vietnam; htnhanh@hueuni.edu.vn
4Faculty of Pharmacy, Hue University of Medicine and Pharmacy, Hue University, Hue City 49000, Vietnam;
hvietduc@hueuni.edu.vn
5Preclinical Toxicity and Efficacy Assessment of Medicines and Chemicals Research Unit, Chulalongkorn
University, Bangkok 10330, Thailand
*Correspondence: varisa.p@pharm.chula.ac.th
Abstract: Lung cancer stands out as a leading cause of death among various cancer types, highlighting
the urgent need for effective anticancer drugs and the discovery of new compounds with potent
therapeutic properties. Natural sources, such as the Conamomum genus, offer various bioactive
compounds. Adunctin E (AE), a dihydrochalcone derived from Conamomum rubidum, exhibited
several pharmacological activities, and its potential as an anticancer agent remains largely unexplored.
Thus, this study aimed to elucidate its apoptotic-inducing effect and identify its molecular targets.
The network pharmacology analysis led to the identification of 71 potential targets of AE against lung
cancer. Subsequent gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and
Reactome pathway enrichment analyses revealed the involvement of these targets in cancer-associated
signaling pathways. Notably, HSP90AA1, MAPK1, and PIK3CA emerged as key players in apoptosis.
In silico molecular docking and dynamic simulations suggested a strong and stable interaction
between AE and HSP90AA1.
In vitro
experiments further confirmed a significant apoptotic-inducing
effect of AE on lung cancer cell lines A549 and H460. Furthermore, immunoblot analysis exhibited a
substantial decrease in HSP90AA1 levels in response to AE treatment. These findings support the
potential anticancer activity of AE through the HSP90AA1 mechanism, underscoring its promise as a
novel compound worthy of further research and development for anti-lung cancer therapy.
Keywords: adunctin E; apoptosis; Conamomum rubidum; lung cancer; network pharmacology;
molecular docking
1. Introduction
Lung cancer remains a significant global health concern, with high incidence and
mortality rates [
1
]. It is primarily categorized into two main subtypes: non-small cell lung
cancer (NSCLC), which constitutes approximately 85% of cases, and small-cell lung cancer
(SCLC), which accounts for the remaining 15% [
2
,
3
]. Unfortunately, nonspecific lung cancer-
associated symptoms often lead to its late diagnosis or diagnosis at an advanced stage [
4
].
Current standard therapies for lung cancer include surgery, radiotherapy, chemotherapy,
and targeted therapy [
5
]. Despite advances in lung cancer therapy, therapeutic resistance,
cancer recurrence, and metastasis often develop after multiple treatments, and the mortality
rate is gradually increasing [68], highlighting the urgent need for new anticancer drugs.
Int. J. Mol. Sci. 2024,25, 11368. https://doi.org/10.3390/ijms252111368 https://www.mdpi.com/journal/ijms
Int. J. Mol. Sci. 2024,25, 11368 2 of 16
Plants are promising sources of natural biological actives for drug development.
Compounds, particularly those extracted from the genus Conamomum, a synonym of
the genus Amomum, in the Zingiberaceae family, have demonstrated pharmacological
activities such as antimicrobial and antioxidant activities and have long been used in
traditional medicine for their anti-inflammatory and fever-reducing properties [
9
12
]. A
recent study has reported the potent cytotoxicity of compounds such as adunctin E (AE),
a dihydrochalcone isolated from Conamomum rubidum in lung cancers [
13
]. However, the
anticancer activity of AE and its underlying molecular mechanism remain unexplored.
Recently, bioinformatics has significantly advanced in drug research and development
by not only identifying potential therapeutic targets but also aiding in drug design [
14
,
15
].
Network pharmacology approaches have emerged as invaluable tools in this process and
facilitated the identification of potential molecular targets of novel compounds promptly
and precisely [
16
,
17
]. These approaches, through integration with multiple bioinformatic
databases, provide insights into the molecular targets of diseases and predict targets of new
compounds. Furthermore, the mechanism of action was investigated through the pathway
analysis. In silico molecular docking allows for the prediction of interactions between new lead
compounds and identified target molecules [
18
,
19
]. Importantly, targets can be validated using
in vitro
experimental models, providing essential preclinical data of new lead compounds for
further investigation and ultimately expediting the drug discovery [20].
In this study, we aimed to identify the molecular targets of AE by integrating network
pharmacology methodology, investigate its interaction with molecular targets using in silico
molecular docking, and validate these molecular targets through
in vitro
lung cancer cell-
based experiments. The findings of this study could offer insights into the anticancer activity
of AE and elucidate its molecular mechanisms of action for potential therapeutic development.
2. Results
2.1. Pharmacokinetic Parameters and Target Identification of AE in NSCLC1
The workflow is illustrated in Figure 1. Pharmacokinetic parameters were analyzed by
pkCSM, indicating that AE has high intestinal absorption, low blood–brain barrier permeabil-
ity, and less toxic (Table S1). Additionally, AE was predicted to inhibit CYP enzymes, which
may influence both its therapeutic efficacy and safety profile. Molecular targets of AE were
retrieved from the Swiss Target Prediction database and SEA, extracting 160 predicted targets
(Table S2). NSCLC-associated targets were obtained from GeneCards, OMIM, and DisGeNET,
yielding a total of 5693 targets after removing duplicates. A Venn diagram identified 71 com-
mon targets of AE and NSCLC (Figure 2A and Table S3). Subsequently, a compound–target
interaction network was constructed using Cytoscape 3.9.1 (Figure 2B). In the network, active
components were labeled in yellow, whereas the 71 common targets were highlighted in blue.
Int. J. Mol. Sci. 2024, 25, x FOR PEER REVIEW 3 of 16
Figure 1. Workflow for the investigation of the molecular targets of adunctin E (AE).
Figure 2. Molecular target identification of adunctin E. (A) This Venn diagram represents targets of
adunctin E (blue), targets in non-small cell lung cancer (NSCLC, yellow), and common targets be-
tween the compound and the disease. (B) A compound–target network was constructed by
Figure 1. Workflow for the investigation of the molecular targets of adunctin E (AE).
Int. J. Mol. Sci. 2024,25, 11368 3 of 16
Int. J. Mol. Sci. 2024, 25, x FOR PEER REVIEW 3 of 16
Figure 1. Workflow for the investigation of the molecular targets of adunctin E (AE).
Figure 2. Molecular target identification of adunctin E. (A) This Venn diagram represents targets of
adunctin E (blue), targets in non-small cell lung cancer (NSCLC, yellow), and common targets be-
tween the compound and the disease. (B) A compound–target network was constructed by
Figure 2. Molecular target identification of adunctin E. (A) This Venn diagram represents targets
of adunctin E (blue), targets in non-small cell lung cancer (NSCLC, yellow), and common targets
between the compound and the disease. (B) A compound–target network was constructed by
Cytoscape 3.9. Active components were labeled in yellow, whereas the 71 common targets were
highlighted in blue. (C) The protein–protein interaction (PPI) network of the common targets was
analyzed by importing 71 common targets to the search tool from the STRING.
2.2. Construction of PPI Network and Enrichment Analyses of GO, KEGG and
Reactome Pathways
To evaluate potential associations among targets, the PPI network of the 71 common
targets was analyzed using the STRING (Figure 2C). In the network, nodes represented
common targets, and edges indicated an association between nodes, including neighboring,
fusion, and co-occurring genes. Sixty-four common targets were connected, whereas seven
nodes did not show interactions. Subsequently, the common targets were subjected to
GO functional annotation analyses which were performed by mapping the target genes
to Gene Ontology categories and conducting enrichment analysis to identify significantly
enriched terms. The top 20 enriched biological processes associated with cancer included
responses to chemical, metabolic process regulation, regulation of cell death and apoptotic
process, and protein phosphorylation, according to the degree of significance (Figure 3A).
In cellular component associations with lung cancer, the targets were mainly found in the
cell projection membrane, cell periphery, plasma membrane, catalytic complex, and cyclin
E1-cyclin-dependent kinase 2 (CDK2) complex (Figure 3B). For molecular functions, the
targets were involved in ribonucleotide binding, protein kinase activity, catalytic activity,
carbohydrate derivative binding, and ion binding (Figure 3C).
Int. J. Mol. Sci. 2024,25, 11368 4 of 16
Int. J. Mol. Sci. 2024, 25, x FOR PEER REVIEW 5 of 16
Figure 3. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway
enrichment analyses were conducted on the potential targets of adunctin E in non-small cell lung
cancer (NSCLC). Data analyzed in STRING were imported to RStudio with the ggplot2 package.
The GO terms examined include (A) biological process, (B) cellular component, and (C) molecular
function. (D) The KEGG pathway associated with these common targets was analyzed.
2.3. Potential Target Identification
The common target network obtained from STRING was imported into Cytoscape
(Figure 4A), and the topology analysis highlighted the top 16 molecules based on their
degree of connectivity (Figure 4B and Table S4). These molecules, appearing dark to light
green according to their degree scores, are potential targets of AE in NSCLC. Among the
identified targets, the key regulators implicated in NSCLC pathogenesis included
HSP90AA1, MAPK1, CDK2, cyclin-dependent kinase 1 (CDK1), phosphatidylinositol-4,5-
bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), heat shock protein 90 alpha fam-
ily class B member 1 (HSP90AB1), toll-like receptor 4 (TLR4), aurora kinase A (AURKA),
cyclin B1 (CCNB1), polo-like kinase 1 (PLK1), glycogen synthase kinase 3 beta (GSK3B),
cyclin E2 (CCNE2), cyclin E1 (CCNE1), histone deacetylase 2 (HDAC2), nitric oxide syn-
thase 2 (NOS2), and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit
delta (PIK3CD).
Because of the crucial role of apoptosis induction in anticancer therapy, we focused
primarily on potential targets classified as apoptosis regulators. Among these targets,
HSP90AA1, MAPK1, and PIK3CA emerged as promising candidates based on their de-
gree scores in the network analysis.
Figure 3. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway
enrichment analyses were conducted on the potential targets of adunctin E in non-small cell lung
cancer (NSCLC). Data analyzed in STRING were imported to RStudio with the ggplot2 package.
The GO terms examined include (A) biological process, (B) cellular component, and (C) molecular
function. (D) The KEGG pathway associated with these common targets was analyzed.
In the KEGG pathway analysis, these targets participated in cancer-related pathways
such as viral carcinogenesis, the PI3K–AKT signaling pathway, microRNAs in cancer, cellular
senescence, cell cycle, autophagy, and apoptosis (Figure 3D). Furthermore, the Reactome
pathway analysis identified several significantly enriched pathways, highlighting key bio-
logical processes related to the input gene set. The analysis used p-adjust values to ensure
statistical significance. The data suggested that the molecular targets of AE in NSCLC were
associated with signal transduction, receptor tyrosine kinase signaling, extracellular matrix
degradation, regulation of transcription of cell cycle genes by p53, MAPK family signaling
cascades, apoptosis, cell cycle, and PI3K/AKT signaling in cancer (Table 1).
Table 1. Pathways of adunctin E’s targets in non-small cell lung cancer by the Reactome pathway analysis.
Pathways Targets
Signal transduction
HDAC7, DD4, MAPK1, HDAC5, CTSD, MMP7, ITGAV, CCNE1, PIK3CA, CDK2,
PAK1, NOS3, PLK1, HDAC3, ADORA2B, F2, PAK2, GSK3B, PDE2A, HSP90AA1,
PDE5A, RET, RAC1, DRD2, HSP90AB1, PIK3CD, CDK1, MAPK8, RXRA, HDAC2,
NTRK1, JAK3, CCKBR, ITGB3, ADORA2A, JAK1
Int. J. Mol. Sci. 2024,25, 11368 5 of 16
Table 1. Cont.
Pathways Targets
Signaling by receptor tyrosine kinases
MAPK1, CTSD, ITGAV, PIK3CA, PAK1, NOS3, HDAC3, PAK2, HSP90AA1, RAC1,
HDAC2, NTRK1, JAK3, ITGB3, ADORA2A
Degradation of the extracellular matrix CTSD, MMP7, MMP13, CAPN2, PRSS1, MMP1, CTSB, CAPN1, CAPNS1
TP53 regulates transcription of cell cycle
genes AURKA, CCNB1, CCNE1, CDK2, CDK1, CCNE2
MAPK family signaling cascades MAPK1, PIK3CA, PAK1, PAK2, RET, RAC1, CDK1, JAK3, ITGB3, JAK1
Apoptosis HSP90AA1, MAPK1, PAK2, TLR4, MAPK8, DAPK1, DAPK3, DAPK2
Cell cycle
MAPK1, AURKA, CCNB1, CCNE1, CDK2, PLK1, GSK3B, HSP90AA1, HSP90AB1,
CDK1, CCNE2
PI3K/AKT signaling in cancer PIK3CA, GSK3B, RAC1, PIK3CD, NTRK1
2.3. Potential Target Identification
The common target network obtained from STRING was imported into Cytoscape
(Figure 4A), and the topology analysis highlighted the top 16 molecules based on their
degree of connectivity (Figure 4B and Table S4). These molecules, appearing dark to light
green according to their degree scores, are potential targets of AE in NSCLC. Among
the identified targets, the key regulators implicated in NSCLC pathogenesis included
HSP90AA1, MAPK1, CDK2, cyclin-dependent kinase 1 (CDK1), phosphatidylinositol-4,5-
bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), heat shock protein 90 alpha family
class B member 1 (HSP90AB1), toll-like receptor 4 (TLR4), aurora kinase A (AURKA),
cyclin B1 (CCNB1), polo-like kinase 1 (PLK1), glycogen synthase kinase 3 beta (GSK3B),
cyclin E2 (CCNE2), cyclin E1 (CCNE1), histone deacetylase 2 (HDAC2), nitric oxide syn-
thase 2 (NOS2), and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit delta
(PIK3CD).
Int. J. Mol. Sci. 2024, 25, x FOR PEER REVIEW 6 of 16
Figure 4. (A) The association of common molecular targets of adunctin E and non-small cell lung
cancer was constructed by Cytoscape 3.9.1. The top 16 common targets with the highest degree
scores were generated using the cytoHubba plugin. Colors ranging from dark blue to light green
indicate a higher to lower score of degree. (B) Plot of the degree values of the top 16 common targets.
2.4. Molecular Docking and Molecular Dynamic Analysis of AE Target Interactions
Molecular docking experiments were performed to analyzed potential interactions
between AE and the identified apoptosis regulators. The results revealed that AE binds to
these targets through a combination of hydrogen bonding, van der Waals forces, hydro-
phobic interaction, and electrostatic interactions (Figure 5A–C). Specifically, the binding
energies of AE with HSP90AA1, MAPK1, and PIK3CA were 10.1, 7.7 and 8.1 kcal/mol,
respectively (Table 2). The ligand efficiency between AE and HSP90AA1 was notably
high, indicating significant contributions from each heavy atom in the ligand to the bind-
ing interaction with the target protein. Conversely, the interactions between AE and either
MAPK1 or PIK3CA exhibited moderate ligand efficiency.
Furthermore, molecular dynamics simulations were performed to investigate the sta-
bility and conformational changes of the ligand–target protein complexes over time (Fig-
ure 5D,E). The ligand movements for AE with HSP90AA1, MAPK1, and PIK3CA were
4.53 ± 0.07 Å, 4.44 ± 0.06 Å, and 6.58 ± 0.1 Å. The conformational changes in the ligands
were 1.64 ± 0.01 Å, 1.64 ± 0.03 Å, and 2.33 ± 0.03 Å for HSP90AA1, MAPK1, and PIK3CA,
respectively. Notably, the RMSD of the ligand’s movement and conformation with
HSP90AA1 remained relatively stable up to the 25 ns mark. In contrast, greater fluctua-
tions were observed in the interactions between AE and both MAPK1 and PIK3CA. These
findings suggest that AE forms particularly stable complexes with HSP90AAA1, indicat-
ing the strongest interaction among the tested proteins. This highlights the potential of
AE as a therapeutic agent in NSCLC by targeting key signaling pathways involved in
apoptosis regulation.
Table 2. Interaction strength between 22-(40py)-JA adunctin E and potential targets.
Targets PDB
Binding Energy
(kcal/mol)
Ligand Efficiency
(kcal/mol per Heavy Atom)
Number of Interactions
Hydrogen van der Waals Hydrophobic Electrostatic
HSP90AA1 4BQG 10.1 0.32 - 12 9 -
MAPK1 (ERK2) 1WZY 7.7 0.24 1 11 11 -
PIK3CA 6DGT 8.1 0.25 2 7 3 1
Figure 4. (A) The association of common molecular targets of adunctin E and non-small cell lung
cancer was constructed by Cytoscape 3.9.1. The top 16 common targets with the highest degree scores
were generated using the cytoHubba plugin. Colors ranging from dark blue to light green indicate a
higher to lower score of degree. (B) Plot of the degree values of the top 16 common targets.
Because of the crucial role of apoptosis induction in anticancer therapy, we focused
primarily on potential targets classified as apoptosis regulators. Among these targets,
HSP90AA1, MAPK1, and PIK3CA emerged as promising candidates based on their degree
scores in the network analysis.
Int. J. Mol. Sci. 2024,25, 11368 6 of 16
2.4. Molecular Docking and Molecular Dynamic Analysis of AE Target Interactions
Molecular docking experiments were performed to analyzed potential interactions
between AE and the identified apoptosis regulators. The results revealed that AE binds
to these targets through a combination of hydrogen bonding, van der Waals forces, hy-
drophobic interaction, and electrostatic interactions (Figure 5A–C). Specifically, the binding
energies of AE with HSP90AA1, MAPK1, and PIK3CA were
10.1,
7.7 and
8.1 kcal/mol,
respectively (Table 2). The ligand efficiency between AE and HSP90AA1 was notably high,
indicating significant contributions from each heavy atom in the ligand to the binding
interaction with the target protein. Conversely, the interactions between AE and either
MAPK1 or PIK3CA exhibited moderate ligand efficiency.
Int. J. Mol. Sci. 2024, 25, x FOR PEER REVIEW 7 of 16
Figure 5. Molecular docking and molecular dynamics between adunctin E and protein targets: 2D
and 3D interactions between adunctin E and HSP90AA1 (A), MAPK1 (B), and PIK3CA (C). Root
mean square deviation for ligand movement (D) and ligand conformation (E). Simulation between
22 adunctin E and HSP90AA1 (red), MAPK1 (green), and PIK3CA (blue) for 25 ns were plotted.
Figure 5. Molecular docking and molecular dynamics between adunctin E and protein targets: 2D
and 3D interactions between adunctin E and HSP90AA1 (A), MAPK1 (B), and PIK3CA (C). Root
mean square deviation for ligand movement (D) and ligand conformation (E). Simulation between
22 adunctin E and HSP90AA1 (red), MAPK1 (green), and PIK3CA (blue) for 25 ns were plotted.
Int. J. Mol. Sci. 2024,25, 11368 7 of 16
Table 2. Interaction strength between 22-(40py)-JA adunctin E and potential targets.
Targets PDB Binding Energy
(kcal/mol)
Ligand Efficiency
(kcal/mol per Heavy Atom)
Number of Interactions
Hydrogen van der
Waals
Hydrophobic Electrostatic
HSP90AA1
4BQG 10.1 0.32 - 12 9 -
MAPK1
(ERK2)
1WZY
7.7 0.24 1 11 11 -
PIK3CA 6DGT 8.1 0.25 2 7 3 1
Furthermore, molecular dynamics simulations were performed to investigate the
stability and conformational changes of the ligand–target protein complexes over time
(Figure 5D,E). The ligand movements for AE with HSP90AA1, MAPK1, and PIK3CA
were 4.53
±
0.07 Å, 4.44
±
0.06 Å, and 6.58
±
0.1 Å. The conformational changes in the
ligands were 1.64
±
0.01 Å, 1.64
±
0.03 Å, and 2.33
±
0.03 Å for HSP90AA1, MAPK1, and
PIK3CA, respectively. Notably, the RMSD of the ligand’s movement and conformation
with HSP90AA1 remained relatively stable up to the 25 ns mark. In contrast, greater
fluctuations were observed in the interactions between AE and both MAPK1 and PIK3CA.
These findings suggest that AE forms particularly stable complexes with HSP90AAA1,
indicating the strongest interaction among the tested proteins. This highlights the potential
of AE as a therapeutic agent in NSCLC by targeting key signaling pathways involved in
apoptosis regulation.
2.5. In Vitro Apoptosis-Inducing Effect of AE on Nsclc
To assess the
in vitro
cytotoxic effects of AE on NSCLC, A549 and H460 cells were
treated with varying concentrations of AE (0–100
µ
M) for 48 h, and a cytotoxic assay using
MTT was performed. The results demonstrated a significant reduction in the viability
of NSCLC, with an IC
50
of 15.72
±
3.37 and 15.71
±
3.43
µ
M in A549 and H460 cells,
respectively (Figure 6A). Furthermore, AE-induced cell apoptosis was evaluated by treating
cells with similar conditions, followed by analysis with annexin-V/propidium iodide (PI)
staining. The result revealed that the number of early (annexin-V
+
, PI
) and late (annexin-
V
+
, PI
+
) apoptotic cells gradually increased in a dose-dependent manner, with 48% and
64% observed in A549 cells and 34% and 68% in H460 cells treated with 10 and 20
µ
M of
AE, respectively (Figure 6B,C).
2.6. AE Downregulates HSP90AA1 Expression
Based on the above results, HSP90AA1 emerged as a top potential candidate targeted
by AE in NSCLC. To underscore the clinical relevance of HSP90AA1 in lung cancer, an
analysis of its differential expression and overall survival was performed. Data revealed a
significant upregulation of HSP90AA1 mRNA expression in lung cancer tissues compared
with lung normal tissues in all datasets (Figure 7A). Furthermore, lung cancer patients with
high HSP90AA1 levels exhibited lower overall survival than those with low HSP90AA1
expression (Figure 7B). These finding suggests the prognostic significance of HSP90AA1 in
lung cancer, highlighting it as a potential therapeutic target.
Further, to investigate the effect of AE on HSP90AA1 levels, cells were treated with
various concentrations of AE. Immunoblot analysis demonstrated a gradual decrease in
HSP90AA1 levels in a dose-dependent manner (Figure 8). At a concentration of 20
µ
M,
AE substantially reduced HSP90AA1 expression levels to 0.15- and 0.12-fold in A549 and
H460 cells, respectively, compared with the untreated control cells. This finding confirms
that HSP90AA1 serves as a molecular target of AE in facilitating lung cancer cell apoptosis.
Int. J. Mol. Sci. 2024,25, 11368 8 of 16
Figure 6.
In vitro
cytotoxicity and apoptosis induction of adunctin E. (A) A549 and H460 cells
were treated with adunctin E (0–100
µ
M) for 48 h. Cell viability was determined by MTT assay.
Plots are presented as a percentage of cell viability. (B) Apoptosis cells were evaluated by annexin-
V/propidium iodide (PI) staining. Representative histograms from the flow cytometry analysis are
shown. (C) The number of early (annexin-V
+
, PI
) and late (annexin-V
+
, PI
+
) apoptotic cells were
plotted. Data are presented as the mean ±SEM (n = 3). * p< 0.05 vs. untreated control cells.
Int. J. Mol. Sci. 2024,25, 11368 9 of 16
Int. J. Mol. Sci. 2024, 25, x FOR PEER REVIEW 9 of 16
(C) The number of early (annexin-V
+
, PI
) and late (annexin-V
+
, PI
+
) apoptotic cells were plotted.
Data are presented as the mean ± SEM (n = 3). * p < 0.05 vs. untreated control cells.
2.6. AE Downregulates HSP90AA1 Expression
Based on the above results, HSP90AA1 emerged as a top potential candidate targeted
by AE in NSCLC. To underscore the clinical relevance of HSP90AA1 in lung cancer, an
analysis of its differential expression and overall survival was performed. Data revealed
a significant upregulation of HSP90AA1 mRNA expression in lung cancer tissues com-
pared with lung normal tissues in all datasets (Figure 7A). Furthermore, lung cancer pa-
tients with high HSP90AA1 levels exhibited lower overall survival than those with low
HSP90AA1 expression (Figure 7B). These finding suggests the prognostic significance of
HSP90AA1 in lung cancer, highlighting it as a potential therapeutic target.
Figure 7. HSP90AA1 is a molecular target of adunctin E. (A) HSP90AA1 expression was upregulated
in lung cancer. HSP90AA1 expressions in both normal lung (blue circles) and lung tumor (red cir-
cles) tissues were assessed utilizing GEO data. (B) Kaplan–Meier survival analysis of lung cancer
patients who had high and low HSP90AA1 expressions from the GEO cohort. HR, hazard ratio.
Further, to investigate the effect of AE on HSP90AA1 levels, cells were treated with
various concentrations of AE. Immunoblot analysis demonstrated a gradual decrease in
HSP90AA1 levels in a dose-dependent manner (Figure 8). At a concentration of 20 μM,
AE substantially reduced HSP90AA1 expression levels to 0.15- and 0.12-fold in A549 and
H460 cells, respectively, compared with the untreated control cells. This finding confirms
that HSP90AA1 serves as a molecular target of AE in facilitating lung cancer cell apopto-
sis.
Figure 7. HSP90AA1 is a molecular target of adunctin E. (A)HSP90AA1 expression was upregulated
in lung cancer. HSP90AA1 expressions in both normal lung (blue circles) and lung tumor (red circles)
tissues were assessed utilizing GEO data. (B) Kaplan–Meier survival analysis of lung cancer patients
who had high and low HSP90AA1 expressions from the GEO cohort. HR, hazard ratio.
Int. J. Mol. Sci. 2024, 25, x FOR PEER REVIEW 10 of 16
Figure 8. (A) A549 and H460 cells were treated with adunctin E (0–20 μM) for 48 h. The expression
of HSP90AA1 was analyzed by immunoblotting. Blots were reprobed with anti-GAPDH antibody
to ensure equal loading. Representative blots from triplicate independent experiments are shown.
(B) HSP90AA1 protein levels were quantified and normalized with those of GAPDH. Relative
HSP90AA1 protein levels were plotted. Data are presented as mean ± SEM (n = 3). * p < 0.05 vs.
untreated control cells.
3. Discussion
The landscape of anticancer drug discovery and research, particularly in the context
of lung cancer, presents ongoing challenges. Despite advances in current therapeutic in-
terventions, the OS rates have gradually increased [1]. Our findings shed light on the re-
markable anticancer properties of AE against lung cancer. Using the network pharmacol-
ogy approach, HSP90AA1 was identified as a significant molecular target of AE. In silico
assays revealed a potent and stable interaction between AE and HSP90AA1. Furthermore,
AE induced apoptosis of lung cancer cells through this mechanism. This underscores AE
as a promising candidate for further anticancer drug research and development.
Network pharmacology offers various advantages in drug research and discovery.
This high-throughput approach has significantly accelerated the drug discovery process
while minimizing costs [21,22]. Enabling the identification of potential molecular targets
of various biologically active compounds has garnered significant interest in the field of
drug discovery [16]. In this study, we identified 71 possible AE targets in lung cancer.
Pathway analysis using GO, KEGG, and Reactome further narrowed down potential mo-
lecular targets, showing notable associations with apoptosis signaling pathways in cancer,
particularly for HSP90AA1, MAPK1, and PIK3CA.
Apoptosis dysregulation is a recognized hallmark of cancer [23]. Apoptosis, or pro-
grammed cell death, is crucially involved in normal physiologies, including embryonic
development and tissue homeostasis, without mediating inflammatory responses or be-
ing harmful to neighboring cells [24,25]. An abnormal apoptosis mechanism contributes
to the pathogenesis of various diseases including cancers [26]. Cancer cells often acquire
deregulated apoptotic signaling either by the upregulation of antiapoptotic and/or pro-
survival proteins or the downregulation of proapoptotic signaling [27–29]. Therefore, can-
cer therapeutics target the editing of these apoptotic signaling pathways. This study also
demonstrated the potent cytotoxic effect of AE on lung cancer cells, mediated through
apoptosis mechanisms.
Notably, HSP90AA1, MAPK1, and PIK3CA emerged as potential molecular targets
for AE in inducing apoptosis. Notably, in silico experiments revealed that AE formed the
strongest and most stable interaction with HSP90AA1, suggesting that this protein may
play a critical role in the mediating of apoptotic effects by AE. Despite strong support
from molecular docking and dynamic simulation studies, there is still a possibility of
Figure 8. (A) A549 and H460 cells were treated with adunctin E (0–20
µ
M) for 48 h. The expression
of HSP90AA1 was analyzed by immunoblotting. Blots were reprobed with anti-GAPDH antibody
to ensure equal loading. Representative blots from triplicate independent experiments are shown.
(B)HSP90AA1 protein levels were quantified and normalized with those of GAPDH. Relative
HSP90AA1 protein levels were plotted. Data are presented as mean
±
SEM (n = 3). * p< 0.05 vs.
untreated control cells.
Int. J. Mol. Sci. 2024,25, 11368 10 of 16
3. Discussion
The landscape of anticancer drug discovery and research, particularly in the context of
lung cancer, presents ongoing challenges. Despite advances in current therapeutic interven-
tions, the OS rates have gradually increased [
1
]. Our findings shed light on the remarkable
anticancer properties of AE against lung cancer. Using the network pharmacology ap-
proach, HSP90AA1 was identified as a significant molecular target of AE. In silico assays
revealed a potent and stable interaction between AE and HSP90AA1. Furthermore, AE
induced apoptosis of lung cancer cells through this mechanism. This underscores AE as a
promising candidate for further anticancer drug research and development.
Network pharmacology offers various advantages in drug research and discovery.
This high-throughput approach has significantly accelerated the drug discovery process
while minimizing costs [
21
,
22
]. Enabling the identification of potential molecular targets
of various biologically active compounds has garnered significant interest in the field of
drug discovery [
16
]. In this study, we identified 71 possible AE targets in lung cancer.
Pathway analysis using GO, KEGG, and Reactome further narrowed down potential
molecular targets, showing notable associations with apoptosis signaling pathways in
cancer, particularly for HSP90AA1, MAPK1, and PIK3CA.
Apoptosis dysregulation is a recognized hallmark of cancer [
23
]. Apoptosis, or pro-
grammed cell death, is crucially involved in normal physiologies, including embryonic
development and tissue homeostasis, without mediating inflammatory responses or being
harmful to neighboring cells [
24
,
25
]. An abnormal apoptosis mechanism contributes to
the pathogenesis of various diseases including cancers [
26
]. Cancer cells often acquire
deregulated apoptotic signaling either by the upregulation of antiapoptotic and/or prosur-
vival proteins or the downregulation of proapoptotic signaling [
27
29
]. Therefore, cancer
therapeutics target the editing of these apoptotic signaling pathways. This study also
demonstrated the potent cytotoxic effect of AE on lung cancer cells, mediated through
apoptosis mechanisms.
Notably, HSP90AA1, MAPK1, and PIK3CA emerged as potential molecular targets
for AE in inducing apoptosis. Notably, in silico experiments revealed that AE formed the
strongest and most stable interaction with HSP90AA1, suggesting that this protein may
play a critical role in the mediating of apoptotic effects by AE. Despite strong support
from molecular docking and dynamic simulation studies, there is still a possibility of
indirect effects. Additional experimental validation is required to confirm the specificity
and mechanism of this interaction. Moreover, it is plausible that AE exerts its effects
through multiple pathways, and indirect modulation of other proteins cannot be excluded.
HSP90AA1, a member of the molecular chaperone network, is upregulated in response
to cellular stress [
30
] and has been implicated in cancer aggressiveness and poor survival
outcomes in various cancers [
31
33
]. It interacts with key signaling targets such as MAPK
and AKT that regulate the stability and functions of the targets, contributing to cancer
aggressiveness including apoptosis resistance and metastasis [
34
37
]. An inhibitor of
HSP90AA1 was reported to remarkably induce apoptosis and suppress cell proliferation in
lung cancer through an ERK/AKT-dependent mechanism [
34
] and to induce autophagic
cell death in osteosarcoma by suppressing AKT/mTOR signaling [
37
], highlighting its
importance in cancer biology.
In this study, we demonstrated that AE downregulates HSP90AA1, a critical protein
involved in cancer cell survival. Our findings indicate that AE binds to the N-terminal
domain of HSP90AA1, specifically interacting with THR184 (Figure S1). This interaction is
likely to disrupt the ATP-binding activity of HSP90AA1, which is essential for its chaperone
function. By interfering with the N-terminal ATPase activity, AE may induce structural
instability in HSP90AA1, impairing its proper folding and function. Consequently, the
misfolded HSP90AA1 is likely targeted for degradation through the proteasome pathway,
which plays a significant role in the clearance of misfolded proteins [
38
]. While our
current study primarily focuses on protein-level effects, we acknowledge the need for
further mechanistic investigations, such as RNA-based assays, to fully elucidate how AE
Int. J. Mol. Sci. 2024,25, 11368 11 of 16
modulates protein levels specifically, whether this modulation occurs via the inhibition of
translation or through targeted degradation.
MAPK1 (also known as ERK2) is a crucial target of HSP90AA1, whereas PIK3CA serves
as an upstream kinase for AKT signaling [
37
,
39
]. This suggests that HSP90AA1 inhibition
holds significant promise as a therapeutic approach for cancers, given its central role in
regulating key signaling pathways involved in cancer progression. However, it is important
to acknowledge that HSP90AA1 operates within broader, interconnected signaling networks
that include the MAPK and PI3K/AKT pathways [
34
,
37
]. These proteins are frequently
co-regulated in various cancer contexts, and their overlapping roles may limit the specificity
of targeting HSP90AA1 alone. Future investigations will focus on elucidating the effects of
AE on the MAPK, PI3K, and AKT pathways, thereby enhancing the therapeutic potential
of AE by demonstrating its influence on multiple cancer-related mechanisms. This multi-
target approach could provide a more comprehensive strategy for addressing the complex
signaling networks in lung cancer. In addition, further studies, particularly in animal
models, are warranted to validate the anticancer efficacy of AE and elucidate its mechanism
of action.
4. Materials and Methods
4.1. Chemicals and Reagents
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) was purchased
from Sigma-Aldrich (St. Louis, MO, USA). AE (Figure 1) was isolated from Conamomum
rubidum, and its 1H and 13C nuclear magnetic resonance (NMR) spectra were reported
previously [
13
]. Briefly, the isolation of AE from Conamomum rubidum involved extracting
with methanol and partitioning with n-hexane. In addition, the n-hexane fraction was
separated using silica gel chromatography by using an n-hexane-acetone solvent system.
The pharmacokinetic parameters were analyzed by pkCSM [40].
4.2. Identification of the Targets of AE and Nsclc-Related Genes
The possible targets of AE were retrieved from the Swiss Target Prediction database [
41
]
and the Similarity ensemble approach (SEA) [
42
]. Key NSCLC-associated molecular targets
were obtained from GeneCards [
43
], Online Mendelian Inheritance in Man (OMIM) [
44
],
and DisGeNET [
45
]. The compound–target network was visualized using Cytoscape ver-
sion 3.9.1 [
46
]. The common targets of AE and NSCLC were identified by Venny version
2.1.0 [47] and presented in a Venn diagram.
4.3. Construction of the Protein–Protein Interaction Network
The protein–protein interaction network was constructed using STRING version
11.5 [
48
] by incorporating the common targets of AE and NSCLC. The protein type was
specified as Homo sapiens with a confidence level set to 0.7, and other parameters were
set to default values. To create the interaction network, protein interaction relationships
were imported into Cytoscape version 3.9.1. Subsequently, top targets were analyzed using
the cytoHubba plugin [
49
]. Core proteins with the highest degree values were subjected to
further analyses.
4.4. Bioinformatic Analyses of Gene Ontology (Go), and Kyoto Encyclopedia of Genes and Genomes
(Kegg), and Reactome Pathways
GO and KEGG were retrieved from the STRING version 11.5 database by importing the
common targets of AE and NSCLC. GO analysis was performed to explore the functionality
of genes, including biological processes, cellular components, and molecular functions [
50
].
In NSCLC, the putative molecular mechanisms of AE were elucidated through KEGG
pathway enrichment analyses [
51
]. Data were visualized by R software version 1.4.1717
with ggplot2 [
52
]. Results were visualized as a bubble plot, with the X-axis representing
the gene ratio, the Y-axis denoting the GO terms, and bubble size and color indicating the
number of associated genes and their statistical significance.
Int. J. Mol. Sci. 2024,25, 11368 12 of 16
Reactome pathway analysis was also employed to investigate pathways associated
with the identified targets [
53
]. Targets were mapped to corresponding pathways in the
Reactome database. A statistical method was applied to assess pathway enrichment,
using an adjusted p-value threshold (e.g., <0.05) to determine significance. The enriched
pathways were then visualized and interpreted to understand potential alterations in
biological processes and signaling pathways relevant to the study.
4.5. Molecular Docking and Dynamics
The X-ray crystal structures of the potential targets were retrieved from the Protein
Data Bank (PDB). The structure of AE was drawn by ChemDraw Ultra version 15.0 (Perkin
Elmer, Waltham, MA, USA). Molecular docking studies of AE with the protein targets
were conducted using the PyRx Virtual Screening Tool version 0.8. Ligand conformations
that exhibited the highest clusters were analyzed to determine their free binding energies
(
G). The binding interactions between the ligands and target proteins were evaluated
using PyMOL version 2.4 (Schrödinger, Portland, OR, USA) and BIOVIA Discovery Studio
Visualizer 2022 (Biovia, San Diego, CA, USA). Molecular dynamics simulations were
performed using Yasara software (https://www.yasara.org/) with the AMBER14 force
field. The simulations were conducted at a temperature of 298 K and pH of 7.4, lasting for
25 ns. The default macro md_run.mcr and md_analyse.mcr. were employed for analyses.
The root mean square deviation (RMSD) graphic was generated using RStudio software
version 1.4.1717 [54].
4.6. Gene Expression Datasets and Differential Expression Analysis
mRNA expression data were obtained from the Gene Expression Omnibus (GEO)
database [
54
]. Two GEO datasets, namely, GSE30219 (tumors, n = 239; normal lung tissues,
n = 14) and GSE31210 (tumors, n = 226; normal lung tissues, n = 19), were analyzed. The
expression levels of HSP90AA1 in normal and tumor lung tissues were compared.
4.7. Survival Analysis
mRNA expression data of HSP90AA1 and lung cancer survival information were
obtained from the GEO databases (GSE30219, n = 239; GSE31210, n = 226) [
55
]. According
to the median expression level, patients were categorized into high and low HSP90AA1
expression groups. The overall survival rates in these two groups were compared using
Kaplan–Meier plots generated by Prism 10 version 10.2.3 (GraphPad Software, Boston, MA,
USA). A log-rank p-value < 0.05 was considered statistically significant.
4.8. Cell Culture
Human NSCLC H460 and A549 cells were purchased from the American Type Culture
Collection (ATCC, Manassas, VA, USA). H460 were cultured in Roswell Park Memorial
Institute (RPMI) Medium, while A549 cells were maintained in Dulbecco’s Modified Eagle
Medium (DMEM). Both media were supplemented with 10% fetal bovine serum (FBS),
100 U/mL penicillin–streptomycin antibiotic solution, and 2 mM L-glutamine. The cell
cultures were incubated in a humidified incubator at 37
C with 5% CO
2
. All media and
supplements were sourced from Gibco (Waltham, MA, USA).
4.9. Cytotoxicity Assay
Cells at a density of 5
×
10
3
cells/well were seeded onto a 96-well plate. After
overnight incubation to allow for cell attachment, the cells were treated with various
concentrations of AE for 48 h. Following treatment, 100
µ
L of the MTT solution (0.5 mg/mL)
was added to each well and incubated for another 4 h. The formazan crystals formed were
then solubilized using dimethylsulfoxide, and the optical intensity was measured at a
wavelength of 570 nm using a microplate reader (VICTOR3/Wallac 1420, Perkin Elmer,
Waltham, MA, USA). The percentage of viable cells was calculated as a percentage relative
Int. J. Mol. Sci. 2024,25, 11368 13 of 16
to the control cells. The inhibitory concentration at 50% (IC
50
) was determined using Prism
10 version 10.2.3 (GraphPad Software, Boston, CA, USA).
4.10. Apoptosis Assay
For the apoptosis evaluation by annexin-V/PI staining, apoptotic cells were assessed
using an apoptosis detection kit (Invitrogen, Waltham, MA, USA). Cells were treated with
various concentrations (0–20
µ
M) of AE for 48 h, washed with cold phosphate-buffered saline,
and resuspended in a binding buffer. The cells were then incubated with annexin-V-FITC/PI
solution for 15 min at room temperature. The fluorescence intensity of each cell was then
analyzed using an EPICS-XL flow cytometer (Beckman Coulter, Indianapolis, IN, USA).
4.11. Immunoblot Analysis
Cells were lysed with a lysis buffer composed of 20 mM Tris-HCl (pH 7.5), 1 mM
MgCl
2
, 150 mM NaCl, 20 mM NaF, 0.5% sodium metavanadate, 1% nonidet-P40, 0.1 mM
phenylmethylsulfonyl fluoride, and protease inhibitor cocktail. Lysis was carried out for
45 min at 4
C. Following lysis, protein concentrations were measured using a BCA Pro-
tein Assay Reagent Kit (Thermo Fisher Scientific, Waltham, MA, USA). The lysates were
subsequently separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis
(SDS-PAGE) and transferred to polyvinylidene difluoride (PVDF) membranes (Bio-Rad
Laboratories, Hercules, CA, USA). The membranes were blocked with 5% skim milk in
Tris-buffer saline containing 0.075% Tween-20 and incubated with specific primary anti-
bodies overnight at 4
C, followed by incubation with corresponding secondary antibodies
at room temperature for 2 h. Rabbit HSP90AA1 (1:1000, CST#8165), mouse anti-GAPDH
(1:1000, CST# 97166), HRP-conjugated anti-rabbit (1:1000, CST#7074), and HRP-conjugated
anti-mouse (1:1000, CST#7076) were the antibodies used. Protein expression was visualized
using an enhanced chemiluminescence system with Immobilon Western chemilumines-
cent HRP substrate (Merck Millipore, Burlington, MA, USA). Densitometry analysis was
performed using Image J software (https://imagej.net/ij/).
4.12. Statistical Analysis
Data are presented as the mean
±
standard deviation, derived from three independent
experiments. Statistical analysis was conducted using Prism 10 version 10.2.3 (GraphPad
Software, Boston, CA, USA). One-way analysis of variance (ANOVA) followed by Tukey’s
multiple comparison test were employed to evaluate statistical significance, with p-value < 0.05.
5. Conclusions
This study highlights the potential of AE as a promising candidate for anticancer drug
development, particularly in lung cancer. AE significantly induced lung cancer cell death
via apoptosis mechanisms. Through network pharmacology and in silico molecular docking
and dynamic analyses, HSP90AA1 was identified as a potential molecular target of AE.
Subsequent
in vitro
experiments confirmed that AE induces apoptosis via an HSP90AA1-
dependent mechanism. These findings provide valuable scientific insights into the potential
of AE for further anticancer drug research and development targeting lung cancer.
Supplementary Materials: The following supporting information can be downloaded at:
https://www.mdpi.com/article/10.3390/ijms252111368/s1.
Author Contributions: Conceptualization, V.P.; methodology, V.P.; validation, V.P. and I.I.; formal
analysis, V.P., I.I. and N.S.; investigation, V.P., I.I., N.S., H.M.N., H.N.T.H. and D.V.H.; resources,
V.P.; data curation, V.P. and I.I.; writing—original draft preparation, V.P. and I.I.; writing—review
and editing, V.P.; visualization, V.P. and I.I.; supervision, V.P.; project administration, V.P.; funding
acquisition, V.P. All authors have read and agreed to the published version of the manuscript.
Funding: This research was funded by Thailand Science research and Innovation Fund Chula-
longkorn University for the
in vitro
and in silico experiments (HEAF67330003 to V.P.), the Second
Century Fund, Chulalongkorn University (C2F to I.I.), Hue University for adunctin E isolation
Int. J. Mol. Sci. 2024,25, 11368 14 of 16
(DHH2023-04-197), and the Postdoctoral Scholarship Programme of Vingroup Innovation Foundation
(VINIF, VINIF.2023.STS.30 to H.N.T.H.).
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: All data supporting the findings of this study as well as Supplementary
Materials are available within the paper and published online.
Acknowledgments: We thank the Pharmaceutical Research Instrument Center (Faculty of Pharma-
ceutical Sciences, Chulalongkorn University, Bangkok, Thailand) for the research facility.
Conflicts of Interest: The authors declare no conflicts of interest.
References
1. Siegel, R.L.; Giaquinto, A.N.; Jemal, A. Cancer statistics, 2024. CA Cancer J. Clin. 2024,74, 12–49. [PubMed]
2.
Nicholson, A.G.; Tsao, M.S.; Beasley, M.B.; Borczuk, A.C.; Brambilla, E.; Cooper, W.A.; Dacic, S.; Jain, D.; Kerr, K.M.;
Lantuejoul, S.; et al
. The 2021 WHO classification of lung tumors: Impact of advances since 2015. J. Thorac. Oncol. 2022,
17, 362–387. [CrossRef] [PubMed]
3.
Schwendenwein, A.; Megyesfalvi, Z.; Barany, N.; Valko, Z.; Bugyik, E.; Lang, C.; Ferencz, B.; Paku, S.; Lantos, A.; Fillinger, J.;
et al. Molecular profiles of small cell lung cancer subtypes: Therapeutic implications. Mol. Ther. Oncolytics 2021,20, 470–483.
[CrossRef] [PubMed]
4.
Walters, S.; Maringe, C.; Coleman, M.P.; Peake, M.D.; Butler, J.; Young, N.; Bergström, S.; Hanna, L.; Jakobsen, E.; Kölbeck, K.; et al.
Lung cancer survival and stage at diagnosis in Australia, Canada, Denmark, Norway, Sweden and the UK: A population-based
study, 2004–2007. Thorax 2013,68, 551–564. [CrossRef] [PubMed]
5. Bironzo, P.; Di Maio, M. A review of guidelines for lung cancer. J. Thorac. Dis. 2018,10, S1556–S1563. [CrossRef] [PubMed]
6.
Schoenfeld, A.J.; Antonia, S.J.; Awad, M.M.; Felip, E.; Gainor, J.; Gettinger, S.N.; Hodi, F.S.; Johnson, M.L.; Leighl, N.B.; Lovly,
C.M.; et al. Clinical definition of acquired resistance to immunotherapy in patients with metastatic non-small-cell lung cancer.
Ann. Oncol. 2021,32, 1597–1607.
7.
Liu, W.J.; Du, Y.; Wen, R.; Yang, M.; Xu, J. Drug resistance to targeted therapeutic strategies in non-small cell lung cancer.
Pharmacol. Ther. 2020,206, 107438.
8.
Suda, K.; Mitsudomi, T.; Shintani, Y.; Okami, J.; Ito, H.; Ohtsuka, T.; Toyooka, S.; Mori, T.; Watanabe, S.I.; Asamura, H.; et al.
Clinical Impacts of EGFR Mutation Status: Analysis of 5780 Surgically Resected Lung Cancer Cases. Ann. Thorac. Surg. 2021,
111, 269–276.
9.
Nguyen, D.D.; Nguyen-Ngoc, H.; Tran-Trung, H.; Nguyen, D.K.; Nguyen, L.T.T. Limonene and eucalyptol rich essential oils
with their antimicrobial activity from the leaves and rhizomes of Conamomum vietnamense N.S. Lý& T.S. Hoang (Zingiberaceae).
Pharmacia 2023,70, 91–96.
10.
Cai, R.; Yue, X.; Wang, Y.; Yang, Y.; Sun, D.; Li, H.; Chen, L. Chemistry and bioactivity of plants from the genus Amomum.J.
Ethnopharmacol. 2021,281, 114563. [CrossRef]
11.
Van, H. Chemical constituents and biological activities of essential oils of Amomum genus (Zingiberaceae). Asian Pac. J. Trop.
Biomed. 2021,11, 519–526. [CrossRef]
12.
Huong, L.T.; Huong, T.T.; Bich, N.T.; Ogunwande, I.A. Chemical compositions, larvicidal and antimicrobial activities of Zingiber
castaneum (Škorniˇck. & Q.B. Nguyn) and Zingiber nitens (M.F. Newman) essential oils. Braz. J. Pharm. Sci. 2022,58, e200204.
13.
Hoang, H.N.T.; Vo, H.Q.; Nguyen, L.T.K.; Thi-Tran, L.T.; Nguyen, H.T.; Pham, T.V.; Le, H.T.; Canh Le, C.V.; Nguyen, B.C.; Ho,
D.V. Conamonin A and dihydrochalcones from the whole plants of Conamomum rubidum (Lamxay & N.S.Lý) Škorniˇck. & A.D.
Poulsen showing anti-inflammatory and cytotoxic activities. Nat. Prod. Res. 2024, 1–6. [CrossRef]
14.
Singharajkomron, N.; Yodsurang, V.; Seephan, S.; Kungsukool, S.; Petchjorm, S.; Maneeganjanasing, N.; Promboon, W.; Dang-
wilailuck, W.; Pongrakhananon, V. Evaluating the expression and prognostic value of genes encoding microtubule-associated
proteins in lung cancer. Int. J. Mol. Sci. 2022,23, 14724. [CrossRef]
15. Li, K.; Du, Y.; Li, L.; Wei, D.Q. Bioinformatics approaches for anti-cancer drug discovery. Curr. Drug Targets 2019,21, 3–17.
16. Poornima, P.; Kumar, J.D.; Zhao, Q.; Blunder, M.; Efferth, T. Network pharmacology of cancer: From understanding of complex
interactomes to the design of multi-target specific therapeutics from nature. Pharmacol. Res. 2016,111, 290–302. [CrossRef]
17.
Iksen, I.; Witayateeraporn, W.; Wirojwongchai, T.; Suraphan, C.; Pornputtapong, N.; Singharajkomron, N.; Nguyen, H.M.;
Pongrakhananon, V. Identifying molecular targets of Aspiletrein-derived steroidal saponins in lung cancer using network
pharmacology and molecular docking-based assessments. Sci. Rep. 2023,13, 1545. [PubMed]
18.
Krzywik, J.; Mozga, W.; Aminpour, M.; Janczak, J.; Maj, E.; Wietrzyk, J.; Tuszy´nski, J.A.; Huczy´nski, A. Synthesis, biological
evaluation and molecular docking studies of new amides of 4-chlorothiocolchicine as anticancer agents. Bioorg. Chem. 2020,
97, 103664.
19.
Arjmand, B.; Hamidpour, S.K.; Alavi-Moghadam, S.; Yavari, H.; Shahbazbadr, A.; Tavirani, M.R.; Gilany, K.; Larijani, B. Molecular
docking as a therapeutic approach for targeting cancer stem cell metabolic processes. Front. Pharmacol. 2022,13, 768556.
Int. J. Mol. Sci. 2024,25, 11368 15 of 16
20. Wilding, J.L.; Bodmer, W.F. Cancer cell lines for drug discovery and development. Cancer Res. 2014,74, 2377–2384. [CrossRef]
21.
Hopkins, A.L. Network pharmacology: The next paradigm in drug discovery. Nat. Chem. Biol. 2008,4, 682–690. [CrossRef]
[PubMed]
22.
Li, L.; Yang, L.; Yang, L.; He, C.; He, Y.; Chen, L.; Dong, Q.; Zhang, H.; Chen, S.; Li, P. Network pharmacology: A bright guiding
light on the way to explore the personalized precise medication of traditional Chinese medicine. Chin. Med. 2023,18, 146.
[PubMed]
23. Hanahan, D. Hallmarks of cancer: New dimensions. Cancer Discov. 2022,12, 31–46.
24. Elmore, S. Apoptosis: A review of programmed cell death. Toxicol. Pathol. 2007,35, 495–516. [CrossRef]
25. Voss, A.K.; Strasser, A. The essentials of developmental apoptosis. F1000Research 2020,9, 148. [CrossRef]
26.
Chaudhry, G.E.S.; Md Akim, A.; Sung, Y.Y.; Sifzizul, T.M.T. Cancer and apoptosis: The apoptotic activity of plant and marine
natural products and their potential as targeted cancer therapeutics. Front. Pharmacol. 2022,13, 842376.
27.
Neophytou, C.M.; Trougakos, I.P.; Erin, N.; Papageorgis, P. Apoptosis deregulation and the development of cancer multi-drug
resistance. Cancers 2021,13, 4363. [CrossRef]
28.
Huang, J.Q.; Li, H.F.; Zhu, J.; Song, J.W.; Zhang, X.B.; Gong, P.; Liu, Q.Y.; Zhou, C.H.; Wang, L.; Gong, L.Y. SRPK1/AKT axis
promotes oxaliplatin-induced anti-apoptosis via NF-κB activation in colon cancer. J. Transl. Med. 2021,19, 280. [CrossRef]
29.
Chen, S.; Bie, M.; Wang, X.; Fan, M.; Chen, B.; Shi, Q.; Jiang, Y. PGRN exacerbates the progression of non-small cell lung cancer
via PI3K/AKT/Bcl-2 antiapoptotic signaling. Genes Dis. 2022,9, 1650–1661. [CrossRef]
30.
Peng, C.; Zhao, F.; Li, H.; Li, L.; Yang, Y.; Liu, F. HSP90 mediates the connection of multiple programmed cell death in diseases.
Cell Death Dis. 2022,13, 929. [CrossRef]
31.
Wang, J.; Cui, S.; Zhang, X.; Wu, Y.; Tang, H. High expression of heat shock protein 90 is associated with tumor aggressiveness
and poor prognosis in patients with advanced gastric cancer. PLoS ONE 2013,8, e62876. [CrossRef] [PubMed]
32.
Liu, H.; Zhang, Z.; Huang, Y.; Wei, W.; Ning, S.; Li, J.; Liang, X.; Liu, K.; Zhang, L. Plasma HSP90AA1 predicts the risk of breast
cancer onset and distant metastasis. Front. Cell Dev. Biol. 2021,9, 639596. [CrossRef] [PubMed]
33.
Liu, K.; Kang, M.; Li, J.; Qin, W.; Wang, R. Prognostic value of the mRNA expression of members of the HSP90 family in non-small
cell lung cancer. Exp. Ther. Med. 2019,17, 2657. [CrossRef] [PubMed]
34.
Niu, M.; Zhang, B.; Li, L.; Su, Z.; Pu, W.; Zhao, C.; Wei, L.; Lian, P.; Lu, R.; Wang, R. Targeting HSP90 inhibits proliferation and
induces apoptosis through AKT1/ERK pathway in lung cancer. Front. Pharmacol. 2022,12, 724192. [CrossRef]
35.
Basso, A.D.; Solit, D.B.; Chiosis, G.; Giri, B.; Tsichlis, P.; Rosen, N. Akt forms an intracellular complex with heat shock protein
90 (Hsp90) and Cdc37 and is destabilized by inhibitors of Hsp90 function. J. Biol. Chem. 2002,277, 39858–39866. [CrossRef]
[PubMed]
36.
Bhattacharyya, N.; Gupta, S.; Sharma, S.; Soni, A.; Bagabir, S.A.; Bhattacharyya, M.; Mukherjee, A.; Almalki, A.H.; Alkhanani,
M.F.; Haque, S.; et al. CDK1 and HSP90AA1 appear as the novel regulatory genes in non-small cell lung cancer: A bioinformatics
approach. J. Pers. Med. 2022,12, 393. [CrossRef]
37.
Mori, M.; Hitora, T.; Nakamura, O.; Yamagami, Y.; Horie, R.; Nishimura, H.; Yamamoto, T. Hsp90 inhibitor induces autophagy
and apoptosis in osteosarcoma cells. Int. J. Oncol. 2015,46, 47. [CrossRef]
38.
Xie, M.; Yu, T.; Jing, X.; Ma, L.; Fan, Y.; Yang, F.; Ma, P.; Jiang, H.; Wu, X.; Shu, Y.; et al. Exosomal circSHKBP1 promotes gastric
cancer progression via regulating the miR-582-3p/HUR/VEGF axis and suppressing HSP90 degradation. Mol. Cancer 2020,
19, 112. [CrossRef]
39.
Miricescu, D.; Totan, A.; Stanescu-Spinu, I.I.; Badoiu, S.C.; Stefani, C.; Greabu, M. PI3K/AKT/mTOR signaling pathway in breast
cancer: From molecular landscape to clinical aspects. Int. J. Mol. Sci. 2021,22, 173.
40.
Pires, D.E.V.; Blundell, T.L.; Ascher, D.B. pkCSM: Predicting small-molecule pharmacokinetic and toxicity properties using
graph-based signatures. J. Med. Chem. 2015,58, 4066–4072.
41.
Daina, A.; Michielin, O.; Zoete, V. SwissTargetPrediction: Updated data and new features for efficient prediction of protein targets
of small molecules. Nucleic Acids Res. 2019,47, W357–W3664. [CrossRef] [PubMed]
42.
Keiser, M.J.; Roth, B.L.; Armbruster, B.N.; Ernsberger, P.; Irwin, J.J.; Shoichet, B.K. Relating protein pharmacology by ligand
chemistry. Nat. Biotechnol. 2007,25, 197–206. [CrossRef] [PubMed]
43.
Safran, M.; Dalah, I.; Alexander, J.; Rosen, N.; Iny Stein, T.; Shmoish, M.; Nativ, N.; Bahir, I.; Doniger, T.; Krug, H.; et al. GeneCards
Version 3: The human gene integrator. Database 2010,2010, baq020. [CrossRef] [PubMed]
44.
Amberger, J.S.; Bocchini, C.A.; Scott, A.F.; Hamosh, A. OMIM.org: Leveraging knowledge across phenotype-gene relationships.
Nucleic Acids Res. 2019,47, D1038–D1043.
45.
Queralt-Rosinach, N.; Piñero, J.; Bravo, À.; Sanz, F.; Furlong, L.I. DisGeNET-RDF: Harnessing the innovative power of the
Semantic Web to explore the genetic basis of diseases. Bioinformatics 2016,32, 2236–2238. [CrossRef]
46.
Shannon, P.; Markiel, A.; Ozier, O.; Baliga, N.S.; Wang, J.T.; Ramage, D.; Amin, N.; Schwikowski, B.; Ideker, T. Cytoscape: A
software environment for integrated models of biomolecular interaction networks. Genome Res. 2003,13, 2498–2504. [CrossRef]
47.
Venny. An Interactive Tool for Comparing Lists with Venn’s Diagrams. Available online: https://bioinfogp.cnb.csic.es/tools/
venny/index.html (accessed on 8 April 2024).
48.
Szklarczyk, D.; Gable, A.L.; Lyon, D.; Junge, A.; Wyder, S.; Huerta-Cepas, J.; Simonovic, M.; Doncheva, N.T.; Morris, J.H.;
Bork, P.; et al. STRING v11: Protein–protein association networks with increased coverage, supporting functional discovery in
genome-wide experimental datasets. Nucleic Acids Res. 2019,47, D607–D613.
Int. J. Mol. Sci. 2024,25, 11368 16 of 16
49.
Chin, C.H.; Chen, S.H.; Wu, H.H.; Ho, C.W.; Ko, M.T.; Lin, C.Y. cytoHubba: Identifying hub objects and sub-networks from
complex interactome. BMC Syst. Biol. 2014,8(Suppl. S4), S11. [CrossRef]
50.
Ashburner, M.; Ball, C.A.; Blake, J.A.; Botstein, D.; Butler, H.; Cherry, J.M.; Davis, A.P.; Dolinski, K.; Dwight, S.S.; Eppig, J.T.; et al.
Gene ontology: Tool for the unification of biology. The Gene Ontology Consortium. Nat. Genet. 2000,25, 25–29. [CrossRef]
51. Kanehisa, M.; Goto, S. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000,28, 27–30. [CrossRef]
52.
Ito, K.; Murphy, D. Application of ggplot2 to pharmacometric graphics. CPT Pharmacomet. Syst. Pharmacol. 2013,2, e79. [CrossRef]
[PubMed]
53.
Milacic, M.; Beavers, D.; Conley, P.; Gong, C.; Gillespie, M.; Griss, J.; Haw, R.; Jassal, B.; Matthews, L.; May, B.; et al. The reactome
pathway knowledgebase 2024. Nucleic Acids Res. 2024,52, D672–D678. [CrossRef] [PubMed]
54. RStudio. RStudio: Integrated Development for R. Available online: http://www.rstudio.com (accessed on 8 April 2024).
55.
Edgar, R.; Domrachev, M.; Lash, A.E. Gene expression omnibus: NCBI gene expression and hybridization array data repository.
Nucleic Acids Res. 2002,30, 207–210. [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths in the United States and compiles the most recent data on population‐based cancer occurrence and outcomes using incidence data collected by central cancer registries (through 2020) and mortality data collected by the National Center for Health Statistics (through 2021). In 2024, 2,001,140 new cancer cases and 611,720 cancer deaths are projected to occur in the United States. Cancer mortality continued to decline through 2021, averting over 4 million deaths since 1991 because of reductions in smoking, earlier detection for some cancers, and improved treatment options in both the adjuvant and metastatic settings. However, these gains are threatened by increasing incidence for 6 of the top 10 cancers. Incidence rates increased during 2015–2019 by 0.6%–1% annually for breast, pancreas, and uterine corpus cancers and by 2%–3% annually for prostate, liver (female), kidney, and human papillomavirus‐associated oral cancers and for melanoma. Incidence rates also increased by 1%–2% annually for cervical (ages 30–44 years) and colorectal cancers (ages <55 years) in young adults. Colorectal cancer was the fourth‐leading cause of cancer death in both men and women younger than 50 years in the late‐1990s but is now first in men and second in women. Progress is also hampered by wide persistent cancer disparities; compared to White people, mortality rates are two‐fold higher for prostate, stomach and uterine corpus cancers in Black people and for liver, stomach, and kidney cancers in Native American people. Continued national progress will require increased investment in cancer prevention and access to equitable treatment, especially among American Indian and Alaska Native and Black individuals.
Article
Full-text available
The Reactome Knowledgebase (https://reactome.org), an Elixir and GCBR core biological data resource, provides manually curated molecular details of a broad range of normal and disease-related biological processes. Processes are annotated as an ordered network of molecular transformations in a single consistent data model. Reactome thus functions both as a digital archive of manually curated human biological processes and as a tool for discovering functional relationships in data such as gene expression profiles or somatic mutation catalogs from tumor cells. Here we review progress towards annotation of the entire human proteome, targeted annotation of disease-causing genetic variants of proteins and of small-molecule drugs in a pathway context, and towards supporting explicit annotation of cell- and tissue-specific pathways. Finally, we briefly discuss issues involved in making Reactome more fully interoperable with other related resources such as the Gene Ontology and maintaining the resulting community resource network.
Article
Full-text available
Network pharmacology can ascertain the therapeutic mechanism of drugs for treating diseases at the level of biological targets and pathways. The effective mechanism study of traditional Chinese medicine (TCM) characterized by multi-component, multi-targeted, and integrative efficacy, perfectly corresponds to the application of network pharmacology. Currently, network pharmacology has been widely utilized to clarify the mechanism of the physiological activity of TCM. In this review, we comprehensively summarize the application of network pharmacology in TCM to reveal its potential of verifying the phenotype and underlying causes of diseases, realizing the personalized and accurate application of TCM. We searched the literature using “TCM network pharmacology” and “network pharmacology” as keywords from Web of Science, PubMed, Google Scholar, as well as Chinese National Knowledge Infrastructure in the last decade. The origins, development, and application of network pharmacology are closely correlated with the study of TCM which has been applied in China for thousands of years. Network pharmacology and TCM have the same core idea and promote each other. A well-defined research strategy for network pharmacology has been utilized in several aspects of TCM research, including the elucidation of the biological basis of diseases and syndromes, the prediction of TCM targets, the screening of TCM active compounds, and the decipherment of mechanisms of TCM in treating diseases. However, several factors limit its application, such as the selection of databases and algorithms, the unstable quality of the research results, and the lack of standardization. This review aims to provide references and ideas for the research of TCM and to encourage the personalized and precise use of Chinese medicine.
Article
Full-text available
Lung cancer is one of the leading cancers and causes of cancer-related deaths worldwide. Due to its high prevalence and mortality rate, its clinical management remains a significant challenge. Previously, the in vitro anticancer activity of Aspiletrein A, a steroid and a saponin from Aspidistra letreae, against non-small cell lung cancer (NSCLC) cells was reported. However, the anticancer molecular mechanism of other Aspiletreins from A. letreae remains unknown. Using in silico network pharmacology approaches, the targets of Aspiletreins were predicted using the Swiss Target Prediction database. In addition, key mediators in NSCLC were obtained from the Genetic databases. The compound-target interacting networks were constructed using the STRING database and Cytoscape, uncovering potential targets, including STAT3, VEGFA, HSP90AA1, FGF2, and IL2. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis demonstrated that several pathways were highly relevant to cancer pathogenesis. Additionally, molecular docking and molecular dynamic analyses revealed the interaction between key identified targets and Aspiletreins, including hydrogen bonding and Van der Waals interaction. This study provides potential targets of Aspiletreins in NSCLC, and its approach of integrating network pharmacology, bioinformatics, and molecular docking is a powerful tool for investigating the mechanism of new drug targets on a specific disease.
Article
Full-text available
Conamomum vietnamense , a new species of Zingiberaceae family, has been discovered and described from Tay Nguyen (Central Highlands, Vietnam) in 2022. The present study described the preparation of essential oil from leaves and rhizomes of the plant by hydrodistillation process. Then, the chemical composition of these essential oils was analyzed by Gas Chromatography-Mass Spectrometry (GC-MS), which indicated that limonene (18.74 and 26.20%) and eucalyptol (40.47 and 49.49%) were the main components, respectively. The essential oils also showed moderate antimicrobial activities against Gram-positive bacterial strains ( Enterococcus faecalis ATCC 299212, Staphylococcus aureus ATCC 25923, Bacillus cereus ATCC 14579), Gram-negative bacterial strains ( Escherichia coli ATCC 25922, Pseudomonas aeruginosa ATCC 27853, Salmonella enterica ATCC 13076), and a pathogenic yeast ( Candida albicans ATCC 10231) in the MIC range of 32–256 μg/mL, which was comparable to those of positive controls, streptomycin and cycloheximide. For the first time, the chemical composition and antimicrobial activity of the essential oil of C. vietnamense were studied.
Article
Full-text available
Abstract In this paper, the chemical constituents, larvicidal and antimicrobial activities of hydrodistilled essential oils from Zingiber castaneum Škorničk. & Q.B. Nguyễn and Zingiber nitens M.F. Newman were reported. The main constituents of Z. castaneum leaf were bicyclogermacrene (24.8%), germacrene D (12.9%), cis-β-elemene (11.2%) and β-pinene (10.3%), while sabinene (22.9%) and camphene (21.2%) were the significant compounds in the rhizome. However, the dominant compounds in the leaf of Z. nitens includes β-pinene (45.8%) and α-pinene (10.7%). Terpinen-4-ol (77.9%) was the most abundant compound of the rhizome. Z. castaneum rhizome oil displayed larvicidal activity against Aedes aegypti and Culex quinquefasciatus with LC50 values of 121.43 and 88.86 µg/mL, respectively, at 24 h. The leaf oil exhibited activity with LC50 values of 39.30 µg/mL and 84.97 µg/mL, respectively. Also, the leaf and rhizome oils of Z. nitens displayed greater larvicidal action towards Ae. aegypti with LC50 values of 17.58 µg/mL and 29.60 µg/mL, respectively. Only the rhizome oil displayed toxicity against Cx. quinquefasciatus with LC50 value of 64.18 µg/mL. All the studied essential oils inhibited the growth of Pseudomonas aeruginosa ATCC25923 with minimum inhibitory concentration (MIC) value of 50.0 µg/mL. This paper provides information on the larvicidal and antimicrobial potentials of Z. castaneum and Z. nitens essential oils.
Article
Full-text available
Microtubule-associated proteins (MAPs) play essential roles in cancer development. This study aimed to identify transcriptomic biomarkers among MAP genes for the diagnosis and prognosis of lung cancer by analyzing differential gene expressions and correlations with tumor progression. Gene expression data of patients with lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) from the Cancer Genome Atlas (TCGA) database were used to identify differentially expressed MAP genes (DEMGs). Their prognostic value was evaluated by Kaplan–Meier and Cox regression analysis. Moreover, the relationships between alterations in lung cancer hallmark genes and the expression levels of DEMGs were investigated. The candidate biomarker genes were validated using three independent datasets from the Gene Expression Omnibus (GEO) database and by quantitative reverse transcription polymerase chain reaction (qRT-PCR) on clinical samples. A total of 88 DEMGs were identified from TCGA data. The 20 that showed the highest differential expression were subjected to association analysis with hallmark genes. Genetic alterations in TP53, EGFR, PTEN, NTRK1, and PIK3CA correlated with the expression of most of these DEMGs. Of these, six candidates—NUF2, KIF4A, KIF18B, DLGAP5, NEK2, and LRRK2—were significantly differentially expressed and correlated with the overall survival (OS) of the patients. The mRNA expression profiles of these candidates were consistently verified using three GEO datasets and qRT-PCR on patient lung tissues. The expression levels of NUF2, KIF4A, KIF18B, DLGAP5, NEK2, and LRRK2 can serve as diagnostic biomarkers for LUAD and LUSC. Moreover, the first five can serve as prognostic biomarkers for LUAD, while LRRK2 can be a prognostic biomarker for LUSC. Our research describes the novel role and potential application of MAP-encoding genes in clinical practice.
Article
Full-text available
Heat shock protein (HSP) 90, an important component of the molecular chaperone network, is closely concerned with cellular signaling pathways and stress response by participating in the process of maturation and activation of client proteins, playing a crucial role both in the normal and abnormal operation of the organism. In functionally defective tissues, programmed cell death (PCD) is one of the regulable fundamental mechanisms mediated by HSP90, including apoptosis, autophagy, necroptosis, ferroptosis, and others. Here, we show the complex relationship between HSP90 and different types of PCD in various diseases, and discuss the possibility of HSP90 as the common regulatory nodal in multiple PCD, which would provide a new perspective for the therapeutic approaches in disease.
Article
Full-text available
Cancer is a multifactorial, multi-stage disease, including complex cascades of signaling pathways—the cell growth governed by dysregulated and abrupt cell division. Due to the complexity and multi-regulatory cancer progression, cancer is still a challenging disease to treat and survive. The screening of extracts and fractions from plants and marine species might lead to the discovery of more effective compounds for cancer therapeutics. The isolated compounds and reformed analogs were known as future prospective contenders for anti-cancer chemotherapy. For example, Taxol, a potent mitotic inhibitor discovered from Taxus brevifolia, suppresses cell growth and arrest, induces apoptosis, and inhibits proliferation. Similarly, marine sponges show remarkable tumor chemo preventive and chemotherapeutic potential. However, there is limited research to date. Several plants and marine-derived anti-cancer compounds having the property to induce apoptosis have been approved for clinical trials. The anti-cancer activity kills the cell and slows the growth of cancer cells. Among cell death mechanisms, apoptosis induction is a more profound mechanism of cell death triggered by naturally isolated anti-cancer agents. Evading apoptosis is the major hurdle in killing cancer cells, a mechanism mainly regulated as intrinsic and extrinsic. However, it is possible to modify the apoptosis-resistant phenotype of the cell by altering many of these mechanisms. Various extracts and fractions successfully induce apoptosis, cell-cycle modulation, apoptosis, and anti-proliferative activity. Therefore, there is a pressing need to develop new anti-cancer drugs of natural origins to reduce the effects on normal cells. Here, we’ve emphasized the most critical elements: i) A better understanding of cancer progression and development and its origins, ii) Molecular strategies to inhibit the cell proliferation/Carcino-genesis, iii) Critical regulators of cancer cell proliferation and development, iv) Signaling Pathways in Apoptosis: Potential Targets for targeted therapeutics, v) Why Apoptosis induction is mandatory for effective chemotherapy, vi) Plants extracts/fractions as potential apoptotic inducers, vii) Marine extracts as Apoptotic inducers, viii) Marine isolated Targeted compounds as Apoptotic inducers (FDA Approved/treatment Phase). This study provides a potential therapeutic option for cancer, although more clinical studies are needed to verify its efficacy in cancer chemotherapy.
Article
Full-text available
Lung cancer is one of the most invasive cancers affecting over a million of the population. Non-small cell lung cancer (NSCLC) constitutes up to 85% of all lung cancer cases, and therefore, it is essential to identify predictive biomarkers of NSCLC for therapeutic purposes. Here we use a network theoretical approach to investigate the complex behavior of the NSCLC gene-regulatory interactions. We have used eight NSCLC microarray datasets GSE19188, GSE118370, GSE10072, GSE101929, GSE7670, GSE33532, GSE31547, and GSE31210 and meta-analyzed them to find differentially expressed genes (DEGs) and further constructed a protein–protein interaction (PPI) network. We analyzed its topological properties and identified significant modules of the PPI network using cytoscape network analyzer and MCODE plug-in. From the PPI network, top ten genes of each of the six topological properties like closeness centrality, maximal clique centrality (MCC), Maximum Neighborhood Component (MNC), radiality, EPC (Edge Percolated Component) and bottleneck were considered for key regulator identification. We further compared them with top ten hub genes (those with the highest degrees) to find key regulator (KR) genes. We found that two genes, CDK1 and HSP90AA1, were common in the analysis suggesting a significant regulatory role of CDK1 and HSP90AA1 in non-small cell lung cancer. Our study using a network theoretical approach, as a summary, suggests CDK1 and HSP90AA1 as key regulator genes in complex NSCLC network.