ArticlePDF AvailableLiterature Review

Recent advances in identifying protein targets of bioactive natural products

Authors:

Abstract and Figures

Background Natural products exhibit structural complexity, diversity, and historical therapeutic significance, boasting attractive functions and biological activities that have significantly influenced drug discovery endeavors. The identification of target proteins of active natural compounds is crucial for advancing novel drug innovation. Currently, methods for identifying targets of natural products can be categorized into labeling and label-free approaches based on whether the natural bioactive constituents are modified into active probes. In addition, there is a new avenue for rapidly exploring the targets of natural products based on their innate functions. Aim This review aimed to summarize recent advancements in both labeling and label-free approaches to the identification of targets for natural products, as well as the novel target identification method based on the natural functions of natural products. Methods We systematically collected relevant articles published in recent years from PubMed, Web of Science, and ScienceDirect, focusing on methods employed for identifying protein targets of bioactive natural products. Furthermore, we systematically summarized the principles, procedures, and successful cases, as well as the advantages and limitations of each method. Results Labeling methods allow for the direct labeling of target proteins and the exclusion of indirectly targeted proteins. However, these methods are not suitable for studying post-modified compounds with abolished activity, chemically challenging synthesis, or trace amounts of natural active compounds. Label-free methods can be employed to identify targets of any natural active compounds, including trace amounts and multicomponent mixtures, but their reliability is not as high as labeling methods. The structural complementarity between natural products and their innate receptors significantly increase the opportunities for finding more promising structural analogues of the natural products, and natural products may interact with several structural analogues of receptors in humans. Conclusion Each approach presents benefits and drawbacks. In practice, a combination of methods is employed to identify targets of natural products. And natural products' innate functions-based approach is a rapid and selective strategy for target identification. This review provides valuable references for future research in this field, offering insights into techniques and methodologies.
Content may be subject to copyright.
Heliyon 10 (2024) e33917
Available online 29 June 2024
2405-8440/© 2024 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC license
(http://creativecommons.org/licenses/by-nc/4.0/).
Review article
Recent advances in identifying protein targets of bioactive
natural products
Xuan Jiang
a
, Kinyu Shon
d
, Xiaofeng Li
e
, Guoliang Cui
d
, Yuanyuan Wu
a
,
Zhonghong Wei
a
, Aiyun Wang
a
,
b
,
c
, Xiaoman Li
a
,
*
, Yin Lu
a
,
b
,
c
,
**
a
Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese
Medicine, Nanjing, 210023, China
b
Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing,
210023, China
c
Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese
Medicine, Nanjing, 210023, China
d
Department of Gastroenterology, The Second Afliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, China
e
Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
ARTICLE INFO
Keywords:
Protein targets
Natural products
Labeling methods
Label-free approaches
Natural products innate functions-based
approach
ABSTRACT
Background: Natural products exhibit structural complexity, diversity, and historical therapeutic
signicance, boasting attractive functions and biological activities that have signicantly inu-
enced drug discovery endeavors. The identication of target proteins of active natural compounds
is crucial for advancing novel drug innovation. Currently, methods for identifying targets of
natural products can be categorized into labeling and label-free approaches based on whether the
natural bioactive constituents are modied into active probes. In addition, there is a new avenue
for rapidly exploring the targets of natural products based on their innate functions.
Aim: This review aimed to summarize recent advancements in both labeling and label-free ap-
proaches to the identication of targets for natural products, as well as the novel target identi-
cation method based on the natural functions of natural products.
Methods: We systematically collected relevant articles published in recent years from PubMed,
Web of Science, and ScienceDirect, focusing on methods employed for identifying protein targets
of bioactive natural products. Furthermore, we systematically summarized the principles, pro-
cedures, and successful cases, as well as the advantages and limitations of each method.
Results: Labeling methods allow for the direct labeling of target proteins and the exclusion of
indirectly targeted proteins. However, these methods are not suitable for studying post-modied
compounds with abolished activity, chemically challenging synthesis, or trace amounts of natural
active compounds. Label-free methods can be employed to identify targets of any natural active
compounds, including trace amounts and multicomponent mixtures, but their reliability is not as
high as labeling methods. The structural complementarity between natural products and their
innate receptors signicantly increase the opportunities for nding more promising structural
* Corresponding author. Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy,
Nanjing University of Chinese Medicine, Nanjing 210023, China.
** Corresponding author. Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy,
Nanjing University of Chinese Medicine, Nanjing, 210023, China.
E-mail addresses: lixm@njucm.edu.cn (X. Li), luyingreen@njucm.edu.cn (Y. Lu).
Contents lists available at ScienceDirect
Heliyon
journal homepage: www.cell.com/heliyon
https://doi.org/10.1016/j.heliyon.2024.e33917
Received 2 January 2024; Received in revised form 28 June 2024; Accepted 28 June 2024
Heliyon 10 (2024) e33917
2
analogues of the natural products, and natural products may interact with several structural
analogues of receptors in humans.
Conclusion: Each approach presents benets and drawbacks. In practice, a combination of
methods is employed to identify targets of natural products. And natural products innate
functions-based approach is a rapid and selective strategy for target identication. This review
provides valuable references for future research in this eld, offering insights into techniques and
methodologies.
1. Introduction
Compared to small-molecule drugs synthesized chemically, natural products offer signicant advantages in terms of structural
novelty, diversity, biocompatibility, and functional versatility. Moreover, they have undergone natural selection and optimization
through long-term evolutionary processes [1]. Approximately 70 % of all approved therapeutic agents originate from natural products
or their derivatives [2]. Thus, natural products are crucial sources of new drugs [3]. Notably, the 2015 Nobel Prize in Physiology or
Medicine awarded to Youyou Tu for discovering the plant natural product artemisinin, which revolutionized malaria treatment,
brought immense pride and optimism to the natural product community worldwide. However, unclear modes of action and challenges
in identifying target proteins hinder the development of natural products in drug discovery. Thus, exploring and identifying targets of
natural products is essential for understanding their mechanisms of action, identifying potential toxicities, and guiding
structure-activity relationship studies. Furthermore, it contributes to the discovery of novel disease-relevant targets, playing a critical
role in innovative drug research based on natural products.
Currently, with the advancement in chemical biology and biophysics, an increasing array of methods has emerged for identifying
targets of natural bioactive molecules. These methods can be broadly classied into labeling and label-free approaches based on
whether natural bioactive constituents are modied into active probes. In probe-based methods, natural bioactive molecules are
transformed into active probes to explore their interactions with targets, whereas label-free methods rely on changes in the biophysical
properties of the target protein upon their binding with the natural bioactive molecule for the identication of the target protein. In
addition, a novel avenue for rapidly exploring the targets of natural products based on their innate functions has also been developed.
This review primarily summarizes these two methodological categories as well as the new approach based on the innate functions of
natural products, providing a systematic elucidation of their principles, workows, and successful cases, as well as the advantages and
disadvantages associated with each approach. This review aims to inspire and provide guidance to researchers in this eld.
Fig. 1. The typical process of afnity chromatography. Initially, bioactive natural products are immobilized onto a solid support matrix (such as
agarose beads or resin), serving as baits. Subsequently, cell lysates or tissue homogenates are either incubated with the active compound-loaded
solid support matrix (when agarose beads serve as the solid support matrix) or passed through the solid support matrix (when resin materials
serve as the solid support matrix). Afterward, multiple washes with inert solvents are performed to remove unbound proteins. Finally, the retained
proteins are eluted either by heating, utilizing high-ionic-strength solvents, or adding an excess of a free drug to compete with the target protein
binding. Subsequently, these proteins are identied through quantitative proteomic analysis.
X. Jiang et al.
Heliyon 10 (2024) e33917
3
Table 1
Recent studies that conducted afnity chromatography-based target identication of natural products.
No. Natural product Structure Category Bioactivity Target protein Reference
1 Artesunate
Quaternary
ammonium salts
Regeneration of pancreatic β cell mass
from
α
cells
Gephyrin [4]
2 Arzanol
Terpenoids Anti-inammatory activity Brain glycogen
phosphorylase
[5]
3 Baicalein Flavonoids Elimination of liver tumor-initiating
stem cell-like cells resistant to mTORC1
inhibition
SAR1B guanosine
triphosphatase
[6]
4 20(S)-
Protopanaxadiol
Saponins Reduction of fatigue CK-MM [7]
5 IJ-5 Terpenoids Anti-inammatory activity Ubiquitin-
conjugating enzyme
UbcH5
[8]
6 Sappanone A Ketones Anti-neuroinammatory activity IMPDH2 [10]
7 Celastrol Terpenoids Intervention of the progression of
hepatocellular carcinoma
ROCK2 [11]
8 Ainsliadimer A Terpenoids Anticancer and anti-inammatory
activity
IKKβ [12]
(continued on next page)
X. Jiang et al.
Heliyon 10 (2024) e33917
4
2. Target identication of natural bioactive components using labeling techniques
Labeling techniques entail chemically modifying natural bioactive components to generate probes with specic markers, such as
uorescent dyes or biotin conjugates. These labeled probes enable the identication of target proteins through corresponding uo-
rescence imaging or enrichment techniques.
2.1. Afnity chromatography
Afnity chromatography stands out as the most traditional method of labeling. As shown in Fig. 1, the typical procedure involves
initially identifying the active functional groups of natural bioactive components and then immobilizing them onto a solid support
matrix, such as agarose beads or resin. Once immobilized, these probes act as baits. Subsequently, the cell or tissue lysate is either
incubated with the solid support matrix loaded with active compounds (when agarose beads are used as the solid support matrix) or
passed through the solid support matrix (when resin materials are used as the solid support matrix). Following this, multiple washes
with inert solvents are performed to remove non-specically bound proteins, leaving the proteins that exhibit afnity binding with the
immobilized natural bioactive components attached to the solid support matrix. Finally, the bound proteins are eluted through various
means, such as heating, using high ionic strength solvents, or adding an excess of free drug to compete with the target protein binding.
The eluted proteins are then subjected to quantitative proteomic analysis for identication. This method has been utilized for the
discovery of target proteins for numerous well-known compounds. For instance, afnity chromatography assays have enabled the
identication of the targets of artesunate [4], arzanol [5], baicalein [6], 20(S)-protopanaxadiol [7], and the herb-derived sesquiter-
pene lactone compound IJ-5 [8] (Table 1). A notable example is the study by Ito et al. [9], where immobilized probes of thalidomide
were employed to identify its target protein, cereblon (CRBN). CRBN forms an E3 ubiquitin ligase complex with damaged DNA binding
protein 1 and Cul4A, thereby inhibiting ubiquitin ligase activity crucial for limb outgrowth in zebrash and chicks, thus contributing to
thalidomide teratogenicity. This research was published in the journal Science.
Furthermore, natural bioactive components can be biotinylated. By exploiting the strong afnity interaction between streptavidin
and biotin, the biotinylated natural bioactive components can be captured, enriched, and puried using streptavidin-coated agarose
solid support carriers. This facilitates the capture of target proteins associated with natural bioactive components, which can subse-
quently be identied via mass spectrometry (MS). Tu and colleagues transformed sappanone A, an anti-neuroinammatory active
component from the traditional Chinese medicine Su-Mu, into a chemical probe conjugated with biotin. This probe was employed to
sh out the direct target proteins from glial cells, leading to the identication of inosine-5
-monophosphate dehydrogenase 2
(IMPDH2) as a key target of sappanone A [10]. This breakthrough was recognized as one of the Top 10 Medical Advances in Chinain
2017. In addition, biotin afnity purication systems have been employed to identify the targets of celastrol, an active component from
the medicinal plant Tripterygium wilfordii Hook F [11], the natural anti-inammatory active molecule ainsliadimer A [12], the marine
natural product pateamine A [13], and adenanthin, a diterpenoid derived from the leaves of Isodon adenanthus [14] (Table 1).
Afnity chromatography facilitates rapid and large-scale enrichment of target proteins. Nevertheless, it faces three primary lim-
itations [15]. Firstly, discerning between high-afnity target proteins and abundant proteins with low afnity poses a challenge.
Secondly, the washing process relies on empirical conditions, wherein overly stringent conditions may signicantly decrease the
number of identied targets, while weak washing conditions could yield false-positive results. Lastly, it fails to capture the interaction
Table 1 (continued )
No. Natural product Structure Category Bioactivity Target protein Reference
9 Pateamine A
Alkaloids Inhibition of eukaryotic translation
initiation
Eukaryotic initiation
factor 4A
[13]
10 Adenanthin Terpenoids Induction of leukemic cell
differentiation
Peroxiredoxins I and
II
[14]
Note: The red-marked portion in the structure refers to the labeling binding site.
X. Jiang et al.
Heliyon 10 (2024) e33917
5
between drugs and proteins within live cells under physiological conditions. Additionally, the biotin afnity tag introduces steric
hindrance, potentially diminishing or even abolishing the activity of natural bioactive components and hindering the entry of probe
molecules into cells.
2.2. Activity-based protein proling (ABPP)
Given the limitations of afnity chromatography, chemists and biologists have focused on exploring target identication methods
suitable for targets exhibiting low abundance and live cells. Currently, the most widely employed and rapidly advancing technique is
ABPP, which was introduced by Dr. Cravatts research team at the Scripps Research Institute in the United States in 1999 [16].
Activity-based probes (ABPs) form the cornerstone of ABPP technology. Unlike immobilized probes, ABPs do not necessitate pre-
xation to a matrix material and can be directly applied to live cells, elucidating drugtarget interactions under physiological con-
ditions [17]. ABPs comprise three components: a reactive group, a reporter group, and a linker connecting the reactive and reporter
groups (Fig. 2). The reactive group serves as the core of ABPs. Depending on the reactive group, ABPs can be classied as either
covalent or non-covalent probes for natural bioactive components. For example, natural products containing electrophilic moieties,
such as Michael acceptors, β-lactones, β-lactams, epoxides, haloalkyl carbonyl compounds, and isothiocyanates, can form covalent
bonds with the nucleophilic groups of target proteins, facilitating the covalent binding of the probe molecule to the target protein [18].
The design of such probes merely entails incorporating a reporter group via a linker in a region that does not affect the activity of the
natural products.
Fluorescent dyes or biotin are commonly employed as reporter groups in ABPs. However, these labeling molecules suffer from
issues such as large size and poor solubility, signicantly hampering the ability of ABPs to penetrate the cell membrane [15]. To
overcome this limitation, Speers et al. [19] integrated click chemistry with ABPs. In this method, inert reactive groups, such as alkynes
and azides, replace the original reporter groups. These inert reactive groups exhibit minimal interference with the chemical properties
of natural bioactive components, allowing them to bind to target proteins in live cells or cell lysates. Subsequently, through click
chemistry reactions, uorescent dyes or biotin can be appended to the probetarget complex, facilitating the enrichment and
Fig. 2. The structure of ABPs and the workow of competitive ABPP. The upper panel depicts the structure of ABPs, which include a reactive group,
a reporter group, and a linker facilitating their connection. The middle and lower panels denote the workow of a competitive ABPP. Initially, cell
lysates or live cells are incubated with either the native bioactive natural product or solvent and subsequently labeled with an ABPP probe.
Following this, SDS-PAGE or LCMS/MS are employed to either visualize or identify the targets labeled by the ABPP probe. Targets of the bioactive
natural product exhibit diminished signals in the active compound-treated samples compared to those treated with solvent controls.
X. Jiang et al.
Heliyon 10 (2024) e33917
6
identication of the target proteins [15].
The typical workow of ABPP entails incubating probes with live cells or cell lysates, facilitating the binding of the small-molecule
probe to the active site of its target enzyme. The probe forms a complex with the enzymes through enzymatic reactions or covalent
binding with the target protein, induced by ultraviolet (UV) irradiation using photoafnity groups. Subsequently, the labeled protein is
enriched and puried using the probes reporter group, such as uorescence or biotin. Finally, target identication is achieved using
techniques such as MS.
In ABPP experiments, competitive ABPP groups may be congured by pre-incubating the natural bioactive component with either
cell lysates or live cells prior to introducing the natural bioactive component probe (Fig. 2). Should the natural bioactive component
effectively compete for binding to the enzymatic active site of the target protein, it would competitively impede the labeling process of
the target protein by the probe. Compared to the control group treated solely with the probe, the competitive group exhibits
Table 2
Recent studies that conducted ABPP-based target identication of natural products.
No. Natural
products
Structure Category Bioactivity Target protein Reference
1 Nimbolide
Terpenoids Anticancer
activity
E3 ubiquitin ligase RNF114 [21]
2 Parthenolide Terpenoids Anticancer
activity
Ubiquitin carboxyl-terminal hydrolase
10 (USP10)
[22]
3 Dankastatin B Terpenoids Anticancer
activity
Mitochondrial VDAC3 [23]
4 Salinipostin A Cyclopeptide Antimalarial
activity
Essential
α
/β serine hydrolases [25]
5 Illudin
Terpenoids Anticancer
activity
DNA modication and unselective
protein binding
[26]
6 Withangulatin A Alkaloids Antitumor
activity
phosphoglycerate dehydrogenase
(PHGDH)
[27]
7 Piperlongumine Alkaloids Anticancer
activity
GSTO1 [28]
Note: The red-marked portion in the structure refers to the labeling binding site.
X. Jiang et al.
Heliyon 10 (2024) e33917
7
Table 3
Recent studies that performed PAL probe-based target identication of natural products.
No. Natural products Structure Category Bioactivity Target protein Reference
1 Matrine
Alkaloids Anticancer activity Annexin A2 [30]
2 Coibamide A Cyclopeptide Anticancer activity Sec61 [31]
3 Betulinic acid Terpenoids Anticancer activity Tropomyosin [32]
4 Harringtonolide Cyclopeptide Suppression of
cancer cell
migration
Receptor for activated C kinase 1 [33]
5 Pseudolaric acid
B
Terpenoids Anticancer activity CD147 [34]
6 Epothilone Macrolides Alleviation of CNS
injuries
The binding site on β-tubulin [35]
7 Quercetin Flavonoids Antitumor activity HSP70, HSP90, RuvB-like 2 ATPases,
and eukaryotic translation initiation
factor 3
[36]
8 Pladienolide Macrolides Antitumor activity Splicing factor SF3b [37]
X. Jiang et al.
Heliyon 10 (2024) e33917
8
diminished uorescence signal intensity or reduced peak intensity in MS, facilitating the identication of potential target proteins
[20].
Spradlin et al. [21] utilized ABPP platforms to reveal that nimbolide, a terpenoid natural product derived from the Neem tree, binds
to a novel functional cysteine crucial for substrate recognition in the E3 ubiquitin ligase RNF114. This interaction inhibits the
ubiquitination and degradation of tumor suppressors, thereby impeding cancer pathogenicity. Similarly, Zhao et al. [22] identied the
covalently modied target of parthenolide, another terpenoid natural product with potent antitumor activity, using ABPP platforms.
Moreover, ABPP has been employed to explore the mechanisms of action of dankastatin B, which exhibits anticancer activity [23],
spongiolactones, marine natural products with potential anticancer properties [24], the antimalarial natural product salinipostin A
[25], illudin, an anticancer natural product family [26], withangulatin A, a natural small molecule that inhibits the serine synthesis
pathway and proliferation of colon cancer cells [27], and piperlongumine, a cancer-selective killing natural product [28], all of which
possess electrophilic functional groups (Table 2).
This method offers the advantage of robust specicity. ABPs eliminate the need for pre-xation to a substrate material and can be
Note: The red-marked portion in the structure refers to the labeling binding site.
Fig. 3. The typical process of competitive ABPPSILAC. The heavy amino acid-labeled group is treated with the natural active compound, serving as
the competitive group, while the light amino acid-labeled group remains untreated. Subsequently, the two groups are combined in accordance with
either cell number or protein quantity, followed by the addition of the bioactive natural product probe for labeling the target proteins. LCMS/MS is
subsequently employed to identify the proteins captured by the probe. The SILAC ratio (light vs. heavy) quantied for each protein serves as an
indicator of the potential target of the active natural compound, with a higher ratio suggesting a stronger likelihood of interaction.
X. Jiang et al.
Heliyon 10 (2024) e33917
9
directly applied to live cells, elucidating drugtarget interactions under physiological conditions. However, its effectiveness is con-
strained to capturing target proteins that covalently bind with small molecules, thus limiting its applicability.
2.3. Photoafnity labeling probes (PAL probes)
When designing probes for natural active compounds that bind to targets through non-covalent interactions, it is necessary to
introduce photoreactive groups in addition to linker groups and reporter groups. These photoreactive groups, upon UV irradiation,
generate highly reactive intermediates known as carbenes, which can form stable covalent bonds between the probe and the target
protein. Such probes are categorized as PAL probes. Commonly employed photoafnity groups encompass diazirine, benzophenone,
and aryl azide [29]. Matrine is a plant alkaloid with potent anticancer activities. However, its molecular target(s) and mechanism
remain unknown. Wang et al. [30], employing the PAL approach, for the rst time, identied annexin A2 as a direct-binding target of
matrine in cancer cells. Moreover, photoafnity probes have been utilized to elucidate the targets of various natural compounds, such
as coibamide A, a marine natural product with potent antiproliferative activity against human cancer cells [31], betulinic acid [32],
harringtonolide, a bioactive diterpenoid tropone isolated from Cephalotaxus harringtonia with antiproliferation activity [33], pseu-
dolaric acid B possessing anticancer activity [34], the epothilone binding site on β-tubulin [35], quercetin, a avonoid natural product
found in various foods with a wide range of medicinal effects [36], and pladienolide, a naturally occurring antitumor macrolide [37]
(Table 3).
This method enables the analysis of proteins that cannot be targeted by ABPs. Nevertheless, photoafnity groups may still display
non-specic binding to abundant cellular proteins.
2.4. ABPP coupled with the stable isotope labeling by amino acids in cell culture (SILAC) technique
To enhance quantitative accuracy, Cravatt et al. integrated the SILAC technique and ABPP, yielding ABPPSILAC. SILAC operates
by substituting specic amino acids in cell culture media with either naturally occurring isotope (light) or stable isotope (heavy)-
labeled amino acids. Following ve to six cell cycles, stable isotope-labeled amino acids are fully incorporated into newly synthesized
proteins, replacing endogenous amino acids. Lysates from light- and heavy-isotope-labeled cells are equitably combined, separated via
gel electrophoresis, and subsequently identied via MS. Each peptide manifests as a pair in MS data, with the light-labeled peptide
containing the light amino acid and the heavy-labeled peptide containing the heavy amino acid. A 1:1 ratio in SILAC peptide pairs
signies unaltered protein abundance in the proteome. If the SILAC peptide pair ratio (light vs. heavy) is large, it indicates a potential
target of the natural product [38].
By integrating competitive ABPP with SILAC, the heavy amino acid-labeled group is subjected to treatment with the natural active
component within the cell lysate (or within live cells), serving as the competitive group, while the light amino acid-labeled group
remains untreated. Subsequently, the two groups are combined in accordance with cell number or protein quantity, followed by the
addition of the natural active component probe to label the target proteins. MS is then employed for target identication purposes
(Fig. 3). A diminished intensity of the heavy-labeled peptide peak implies potential binding of the natural product to the target protein.
Wang and colleagues employed the ABPPSILAC technique to explore the targets involved in the lipid-lowering effects of baicalin, the
principal active component of Scutellaria baicalensis. They synthesized a baicalin-based photoafnity probe and employed competitive
ABPPSILAC to capture its targets. LCMS/MS analysis, complemented by bioinformatics analysis and reverse validation, revealed
carnitine palmitoyltransferase 1A as a target of baicalin in the context of obesity amelioration [39]. In addition, Chen et al. [40]
identied peroxiredoxin 6 as a direct target of withangulatin A in the alleviation of non-small cell lung cancer (NSCLC) via
ABPPSILAC (Table 4).
Table 4
Recent studies that performed ABPPSILAC-based target identication of natural products.
No. Natural products Structure Category Bioactivity Target protein Reference
1 Baicalin
Flavonoids Alleviation of obesity Carnitine palmitoyltransferase 1A [39]
2 Withangulatin A Alkaloids Anti-NSCLC effects Peroxiredoxin 6 [40]
Note: The red-marked portion in the structure refers to the labeling binding site.
X. Jiang et al.
Heliyon 10 (2024) e33917
10
2.5. Proteome microarray assays
Firstly, biotin or uorescent-labeling natural active components were co-incubated with a protein chip. After washing away un-
bound components, the uorescence intensity of each spot on the chip is determined via uorescence scanning or laser confocal
scanning. When biotin-labeling natural active components are employed, they are co-incubated with uorescent-labeling streptavidin
and subjected to uorescence scanning to screen potential targets of the natural active components. Chen and colleagues utilized this
technique to explore the targets of arsenic [41]. Arsenic demonstrates excellent efcacy in acute promyelocytic leukemia treatment
and possesses signicant therapeutic potential for various tumors. However, its broad anticancer mechanisms remain unclear. Re-
searchers co-incubated biotinylated arsenic with a protein chip containing 16,368 proteins, utilizing Cy3-labeling streptavidin to
identify arsenic-binding proteins. Fluorescence scanning revealed enhanced uorescence signals in the probe group compared to the
negative control group. They identied 360 proteins directly interacting with arsenic, including the well-known arsenic target PML.
Previously, fewer than 20 proteins directly interacting with arsenic had been identied worldwide, underscoring the protein chip
platforms efcacy in rapidly and comprehensively identifying drug targets. The most signicantly enriched proteins are implicated in
the glycolysis pathway. Detailed biochemical and metabolomics analyses have identied hexokinase-2 (HK2) as a key target of arsenic,
functioning as a rate-limiting enzyme in glycolysis. This research was published in Proceedings of the National Academy of Sciences of
the United States of America. Liu et al. [42] identied bufalin, a natural product-derived molecular glue, which targets E2F2
degradation, leading to the transcriptional suppression of multiple oncogenes and the growth of hepatocellular carcinoma in vitro and
in vivo. In addition, proteome microarray assays have been employed to identify the targets of coelonin, an active anti-inammatory
component of Bletilla striata [43] (Table 5).
This method offers the advantages of necessitating minimal sample volumes and boasting high throughput, enabling the simul-
taneous analysis of thousands to millions of proteins. Additionally, protein chips are relatively user-friendly. However, they entail high
costs due to the need for a series of expensive, high-end instruments. Furthermore, standardization issues are observed with protein
chips.
3. Target identication and target validation of natural bioactive components using label-free techniques
Despite signicant advancements in afnity-based methods for screening natural product targets, limitations persist [44]. Some
natural products lack suitable modication sites or synthetic methods to introduce such sites. Additionally, exogenous modication
groups may affect the activity of natural active compounds or hinder their binding to true targets, thereby compromising target
identication accuracy. To overcome these challenges, researchers have devised various target screening methods that obviate the
need for structural modications of natural active molecules. Instead, they employ biophysical approaches to assess the impact of the
binding of natural active compounds to target proteins on protein stability, thus facilitating the discovery of targets for natural active
molecules. Moreover, label-free techniques can be categorized into target identication and target validation methods. For example,
drug afnity responsive target stability (DARTS), thermal proteome proling (TPP), and stability of proteins from rates of oxidation
(SPROX) methods are primarily used for target validation, while cellular thermal shift assays (CETSA) and isothermal titration
calorimetry (ITC) are often used for target identication.
Table 5
Recent studies that conducted proteome microarray-based target identication of natural products.
No. Natural
products
Structure Category Bioactivity Target protein Reference
1 Arsenic
Elemental Broad anticancer activity 360 proteins, among which HK2 is a
key target
[41]
2 Bufalin Steroids Suppression of hepatocellular
carcinoma growth
E2F2 [42]
3 Coelonin Alkaloids Anti-inammatory activity PTEN [43]
X. Jiang et al.
Heliyon 10 (2024) e33917
11
3.1. Target identication methods
3.1.1. DARTS
In 2009, Lomenick et al. introduced DARTS [45]. The methods principle posits that upon the binding of a small molecule to a target
protein, the small molecule occupies the recognition site of proteolytic enzymes, impeding their interaction with the cleavage site on
the target protein. Consequently, the target protein exhibits enhanced resistance to proteolytic cleavage and increased stability.
Changes in the resistance to proteolysis pre- and post-small molecule binding can be discerned via electrophoresis. Subsequent
identication of distinct bands on the protein gel can be achieved through MS to ascertain the target proteins of the small molecule. As
a proof-of-principle, researchers selected the FKBP12 protein, a known target of rapamycin, and FK506. Proteolysis was conducted
using subtilisin protease, and the resistance of FKBP12 to proteolysis subsequent to binding with the compound was observed via
SDS-PAGE, thus afrming the scientic validity and feasibility of this method [45].
The typical procedure of this method involves cell lysis, followed by dividing the lysate into two groups (Fig. 4): the active natural
compound group and the solvent group. The lysates of the active natural compound group are incubated with the bioactive compound,
while the lysates of the solvent group are treated with the solvent alone. This allows the bioactive compound to bind to the target
proteins. Subsequently, both the solvent and the bioactive compound groups are further divided into ve to six subgroups, and each
subgroup is treated with a gradient concentration of proteolytic enzyme for protein digestion. Commonly used proteolytic enzymes
include subtilisin protease, thermolysin, and protease from Streptomyces griseus. Finally, various methods, such as SDS-PAGE, 2D-
PAGE, gel staining techniques, and gel or non-gel MS, are employed to detect or identify the bound proteins.
The DARTS technique, which does not necessitate chemical modication of small molecules, offers a convenient means to identify
direct targets of natural products [46]. It has seen widespread application in recent years. Huang et al. [47] employed the DARTS
method and discerned that the antitumor natural active compound batzelladine A targets HSP90. The target of the primary active
compound, andrographolide, in the traditional Chinese medicinal Andrographis paniculata, known for its heat-clearing and detoxifying
properties, was also identied using the DARTS technique. As the concentration of proteolytic enzymes increased, proteins in the
solvent group gradually degraded, while andrographolide inhibited the hydrolysis of the target protein, dynamin-related protein 1
(DRP1), compared to the control group [48]. Moreover, DARTS conrmed the targets of polyphyllin D, a natural product effective
against NSCLC [49], ergolide, a sesquiterpene lactone natural product with anti-inammatory and anticancer activities used to treat
inammatory diseases [50], aconitine, an active compound exhibiting cardiotoxic effects found in Aconitum species [51], crellastatin
A, a cytotoxic sulfated bis-steroid isolated from the Vanuatu Island marine sponge Crella sp. [52], cryptotanshinone, isolated from the
roots of Salvia miltiorrhiza inhibiting the terminal differentiation of human keratinocytes [53], and grape seed extract in anti-colorectal
cancer applications [54] (Table 6).
The limitations of DARTS encompass the following: certain small molecules may bind to the target protein without inducing
signicant conformational changes of the target; certain proteins could exhibit reduced sensitivity to proteolytic enzymes, thereby
resulting in imperceptible alterations in DARTS outcomes; and the capacity to identify targets characterized by low protein abundance
remains limited.
3.1.2. TPP
TPP, a combination of CETSA and multiplexed quantitative MS, enables the monitoring of global protein thermal stability changes
during interactions with small molecules, thereby facilitating high-throughput screening of target proteins [55]. TPP operates on two
principles: the principles of CETSA and multiplexed quantitative MS. Unlike CETSA, TPP characterizes protein thermal stability based
on the melting temperature (Tm), denoting the temperature at which 50 % of the protein unfolds during thermal denaturation.
Typically, proteins bound to small molecules exhibit increased stability and higher Tm values compared to unbound proteins. By
comparing the Tm values and melting curves of proteins with and without small-molecule binding, target proteins can be identied.
Multiplexed quantitative MS primarily refers to tandem mass tag (TMT), a peptide labeling technology developed by Thermo Fisher in
the United States. TMT facilitates the labeling of peptides with up to 10 isotopic tags, enabling the simultaneous comparison of relative
protein levels across 10 different samples. Therefore, TMT serves as a high-throughput analysis method that is particularly suitable for
differential protein analysis of samples subjected to various treatments or treatment durations. Furthermore, through stable isotope
Fig. 4. The typical process of DARTS. Initially, cell lysates are incubated either with a solvent or with a natural active compound. Following this,
both groups are subdivided into ve to six subgroups. Subsequently, gradient proteolysis is performed for each subgroup. Finally, the samples are
subjected to SDS-PAGE for the detection of the target molecule.
X. Jiang et al.
Heliyon 10 (2024) e33917
12
labeling of different samples, all labeled samples can be pooled for subsequent processing, signicantly reducing experimental errors
and considerably improving quantication accuracy.
The typical protocol of this method involves treating cells or cell lysates with or without an active natural compound, followed by
dividing the samples into 10 equal portions and heating them at increasing temperatures to induce protein denaturation and aggre-
gation. The supernatant is then collected and digested using trypsin to generate peptides, which are subsequently labeled using
TMT10. These labeled samples are mixed and analyzed through LCMS/MS to calculate the Tm values of each protein, thereby
screening for target proteins (Fig. 5). Building upon the potent efcacy of the natural product vioprolide A against human acute
lymphoblastic leukemia cells, Kirsch et al. [56] conducted TPP, conrming nucleolar protein 14, essential in ribosome biogenesis, as a
specic target of vioprolide A in Jurkat cells. Furthermore, in a study published in the journal Science Translational Medicine, TPP
was utilized to identify the targets of antimalarial drugs, revealing that the antimalarial drug quinine directly binds to the parasites
intracellular purine nucleotide phosphorylases [57]. Chen et al. [58] demonstrated that rhodojaponin VI, a grayanotoxin from
Rhododendron molle, alleviates neuropathic pain by targeting N-ethylmaleimide-sensitive fusion protein through the TPP approach.
Moreover, TPP has been employed to elucidate the targets of tutin-induced epilepsy [59], the natural sesquiterpene lactone alanto-
lactone, which inhibits NSCLC [60], kurarinone, which attenuates MPTP-mediated neuroinammation [61], and arone, which pro-
tects against neuroinammation [62] (Table 7).
This method offers the advantage of monitoring global thermal stability changes in proteins during drug action, enabling high-
throughput detection of target proteins. However, it is unsuitable for heat-insensitive proteins, some of which require extreme tem-
perature conditions to exhibit changes in thermal stability. Additionally, certain proteins may not display signicant alterations in
thermal stability upon ligand binding, rendering their identication unfeasible through TPP. Furthermore, extracting additional
thermodynamic parameters from TPP data, aside from the Tm value, poses challenges. Moreover, the magnitude of the Tm shift
following proteinligand binding does not consistently correlate with ligand afnity.
Table 6
Recent studies that conducted DARTS-based target identication of natural products.
No. Natural products Category Bioactivity Target protein Reference
1 Batzelladine A Alkaloids Suppression of lung cancer
tumorigenesis
HSP90 [47]
2 Andrographolide Terpenoids Alleviation of parkinsonism DRP1 [48]
3 Polyphyllin D Saponins NSCLC therapy Distruption of HSC70LAMP2A interaction [49]
4 Ergolide Terpenoids Alleviation of inammatory diseases NLRP3 [50]
5 Aconitine Alkaloids Cardiotoxicity Cytosolic phospholipase A2 [51]
6 Crellastatin A Macrocyclic
depsipeptides
Anticancer activity PARP-1 [52]
7 Cryptotanshinone Diterpenoid quinone Alleviation of keratinopathic ichthyosis FKBP1A [53]
8 Grape seed extract Polyphenolic compounds Anticancer activity Endoplasmic reticulum stress response
proteins
[54]
Fig. 5. Schematic workow of TPP. Initially, cell lysates are incubated either with or without the active natural compound. Subsequently, aliquots
are incubated at 10 distinct temperatures and then centrifuged. Following this, the proteins are digested with trypsin to generate peptides. After
TMT10 labeling, the samples are mixed, fractionated, and subjected to LCMS/MS analysis to ascertain the Tm values of individual proteins, thereby
facilitating the screening of target proteins.
X. Jiang et al.
Heliyon 10 (2024) e33917
13
3.1.3. SPROX
SPROX, introduced by Fitzgerald and colleagues from Duke Universitys Department of Chemistry in 2008 [63], represents another
method for detecting natural product-induced target stability. This method capitalizes on the enhanced antioxidant capacity observed
in target proteins upon binding with natural products. Protein oxidation capacity is determined by assessing the oxidation rate of
methionine residues (most proteins typically contain at least one methionine residue, which can serve as a crucial marker for assessing
the global dynamics of protein folding and unfolding equilibrium). The underlying principle is that as the concentration of chemical
denaturants (such as guanidine hydrochloride or urea) increases, the protein unfolds, and increased exposure of its methionine res-
idues to solvent renders them susceptible to oxidation in the unfolded state. Target proteins bound to natural products exhibit
heightened structural stability. They exhibit slower unfolding in the presence of chemical denaturants. The addition of hydrogen
peroxide, aimed at oxidizing methionine residues, results in a decelerated rate of methionine residue oxidation. The oxidation level of
methionine residues is subsequently assessed via MS, and a concentration-dependent curve is generated, depicting the variation in
oxidation and non-oxidized product. The intersection of the two curves denotes the transition midpoint. Binding of the natural product
to the target protein enhances stability, necessitating a relatively higher denaturant concentration to achieve the equivalent oxidation
product levels. Consequently, the transition midpoint shifts towards higher denaturant concentrations. Therefore, the potential target
protein can be inferred by comparing the transition midpoints between the drug and control groups.
The typical procedure of SPROX is as follows (Fig. 6): Initially, a protein lysate is treated with a buffer containing varying denaturant
concentrations in the presence or absence of the active natural product to establish the unfolding/refolding equilibrium of the protein.
Subsequently, hydrogen peroxide is introduced to initiate the oxidation of methionine residues. Upon the completion of oxidation, an
excess of either methionine or hydrogen peroxide scavenger is added to quench the oxidation reaction. Subsequently, the sample is digested
using trypsin, following which the levels of oxidation and non-oxidation in methionine-containing peptide segments are quantied using
MS. Finally, a curve is generated with the quantity of oxidized and non-oxidized methionine-containing peptide segments plotted on the y-
axis and the denaturant concentration plotted on the x-axis. The shift in the transition midpoint is utilized to identify potential target
proteins. The inventors of this technique, the Fitzgerald team, employed SPROX in 2011 to investigate the target proteins of the active
compound resveratrol. They identied six potential target proteins in addition to the known target protein, cytoplasmic aldehyde dehy-
drogenase [64]. In addition, Fitzgerald employed the SPROX technique to analyze the thermodynamics of the geldanamycinHSP90
interaction [65], as well as elucidate the targets of manassantin A, which exhibits anticancer activity [66] (Table 8).
Table 7
Recent studies that conducted TPP-based target identication of natural products.
No. Natural
products
Category Bioactivity Target protein Reference
1 Vioprolide A Cyclopeptide Anti-human acute lymphoblastic leukemia Nucleolar protein 14 [56]
2 Quinine Alkaloids Antimalarial activity Malarial parasites intracellular purine nucleotide
phosphorylase
[57]
3 Rhodojaponin
VI
Saponins Alleviation of neuropathic pain N-ethylmaleimide-sensitive fusion protein [58]
4 Tutin Ketones Induction of epilepsy Calcineurin [59]
5 Alantolactone Terpenoids Inhibition of NSCLC AKR1C1 [60]
6 Kurarinone Flavonoids Attenuation of MPTP-mediated
neuroinammation
Soluble epoxide hydrolase enzyme [61]
7 Arone Terpenoids Protection against neuroinammation Histone-remodeling chaperone ASF1a [62]
Fig. 6. The workow of SPROX. Initially, in both the presence and absence of the active natural product, cell lysates are combined with various
denaturant-containing buffers. Following equilibration of protein unfolding/refolding, hydrogen peroxide is added to induce the oxidation of
methionine residues. Subsequently, the oxidation reaction is quenched using methionine or a hydrogen peroxide scavenger. Finally, proteins in each
denaturant-containing buffer are digested into peptides for subsequent quantitative proteomics analysis.
X. Jiang et al.
Heliyon 10 (2024) e33917
14
This method offers the advantage of facilitating comprehensive assessment of protein folding states and potentially precise mea-
surement of the binding domains and peptide segments engaged in the interaction between natural products and target proteins.
Nonetheless, it is constrained by its ability to solely detect the binding of natural products to proteins containing methionine residues.
In addition, generating the standard curve necessitates a large sample volume, thereby limiting the widespread utilization of this
method.
3.2. Target validation methods
3.2.1. CETSA
In 2013, Martinez Molina et al. introduced CETSA [67], a ligand-induced target stability detection method similar to DARTS. This
technique capitalizes on ligand-induced alterations in target protein, such as alterations in protein conformation, increased hydro-
phobicity within the protein, and chemical cross-linking within or between subunits of the target protein, resulting in enhanced
thermal stability. As the temperature rises, proteins that are not bound to ligands unfold and undergo rapid denaturation, leading to
precipitation. In contrast, ligand-bound proteins maintain their folded state and remain relatively stable, thereby bolstering the
thermal stability of the target protein. Molina and colleagues applied this method to evaluate the thermal melting proles of four
distinct clinical drug targets in cell lysates, each displaying characteristic melting curves. The introduction of drugs capable of binding
to these proteins to the cell lysates resulted in signicant alterations in the melting proles, thereby validating the scientic integrity
and practical utility of CETSA.
The standard procedure of CETSA entails incubating equivalent quantities of cellular lysates or tissue homogenates with small
molecules, followed by heating at various temperatures to induce protein denaturation and aggregation. Proteins bound to drugs
remain relatively stable, whereas unbound proteins undergo rapid denaturation and precipitation with rising temperatures. Relative
quantication of target proteins in the supernatant is performed via western blotting, thereby conrming the target proteins of the
small molecules. Usenamine A, a novel product, was extracted from the lichen Usnea longissima. Yang et al. [68] identied myosin-9 as
the direct target of usenamine A through MS, SPR assays, and CETSA. In addition, CETSA has been employed in combination with other
Table 8
Recent studies that conducted SPROX-based target identication of natural products.
No. Natural products Category Bioactivity Target protein Reference
1 Resveratrol Flavonoids Antioxidant activity Six potential target proteins [64]
2 Geldanamycin Benzoquinone Anticancer activity HSP90 [65]
3 Manassantin A Terpenoids Anticancer activity 28 protein hits [66]
Table 9
Recent studies that conducted CETSA-based target identication of natural products.
No. Natural products Category Bioactivity Target protein Reference
1 Usenamine A Benzofuran
derivatives
Alleviation of parkinsonism Myosin-9 [68]
2 Paederosidic acid Terpenoids Suppression of osteoclast formation
and neuropathic pain
P2Y
14
receptor [69]
3 Echinatin Flavonoids Inhibition of the hemolytic activity
of methicillin-resistant S. aureus
Indirect binding to
α
-hemolysin
[70]
4
α
-Asarone Aromatic
hydrocarbons
Alleviation of ischemic stroke PI3K [71]
5 Nitidine chloride Alkaloids Alleviation of multiple myeloma ABCB6 [72]
6 Pentoxifylline Methylxanthine
derivatives
Alleviation of non-alcoholic fatty
liver
Toll-like receptor 4 (TLR4) [73]
7 Primary active constituents of the Xiang-lian
pill (evodiamine, rutaecarpine, and
stigmasterol)
Alkaloids Anti-pancreatic cancer PTGS2 and PTGS1 [74]
Steroids
8 Toosendanin Terpenoids Anti-colorectal cancer Shh [75]
9 Glytabastan B Flavonoids Prevention and alleviation of
rheumatoid arthritis
ERK2, JNK1 and class I
PI3K catalytic subunit p110
[76]
10 Tubocapsenolide A Terpenoids Therapeutic potential for
osteosarcoma
Src homology 2
phosphatase 2
[77]
11 Primary constituents of Zuojin capsule Alkaloids Anti-colorectal cancer activity CDKN1A, Bcl2, E2F1,
PRKCB, MYC, CDK2, and
MMP9
[78]
Flavonoids
Steroids
Indole
12 Proanthocyanidin A1 Flavonoids Amelioration of chemotherapy-
induced thrombocytopenia
JAK2 [79]
13 Cucurbitacin B Terpenoids Anti-NSCLC activity TLR4 [80]
14 Celastrol and gambogic acid Terpenoids Anti-breast cancer activity ER
α
Y537S mutant [81]
15 Shikonin Terpenoids Anti-triple-negative breast cancer
activity
IMPDH2 [82]
X. Jiang et al.
Heliyon 10 (2024) e33917
15
target identication methods to validate the targets of paederosidic acid, which suppresses osteoclast formation and neuropathic pain
[69], echinatin, a natural compound isolated from licorice, which effectively inhibits the hemolytic activity of methicillin-resistant
Staphylococcus aureus [70],
α
-asarone, an important component of the Huangxiong formula for treating ischemic stroke [71], niti-
dine chloride, an extract from Zanthoxylum nitidum, used in multiple myeloma treatment [72], pentoxifylline, which attenuates
non-alcoholic fatty liver [73], the primary active constituents of the Xiang-lian pill, which suppresses pancreatic tumor [74], too-
sendanin, a natural triterpenoid saponin with anti-colorectal cancer activity [75], glytabastan B, a coumestan isolated from Glycine
tabacina, which is used for the prevention and treatment of rheumatoid arthritis [76], tubocapsenolide A, a withanolide-type steroid
with therapeutic potential for osteosarcoma [77], the primary constituents of the Zuojin capsule, which is used to combat colorectal
cancer [78], proanthocyanidin A1, which ameliorates chemotherapy-induced thrombocytopenia [79], cucurbitacin B, which inhibits
NSCLC [80], celastrol and gambogic acid, which exhibit anti-breast cancer activity [81], shikonin, a natural bioactive component of
Lithospermum erythrorhizon with anti-triple-negative breast cancer activity [82] (Table 9).
One limitation of this method is its dependency on a relatively high concentration of the natural active compound for detection.
Additionally, quantication primarily hinges on western blotting, which exhibits low throughput. Consequently, this method is
typically utilized as a validation tool. Following the capture of small molecule-targeted proteins through alternative techniques, CETSA
is employed for validation purposes.
3.2.2. ITC
ITC enables the direct quantication of heat released or absorbed during biomolecular binding processes. When a compound binds
to a protein, it elicits a heat exchange detectable via ITC. In an ITC experiment, one reactant is placed in a temperature-controlled
sample cell, linked to a reference cell through a heat ow circuit, with both cells in the same external environment. A specic
titrant (such as a natural active compound) is incrementally injected into the sample cell. Upon reaction between the sample and
titrant, heat is either absorbed or released. After each injection of the sample, heat is absorbed or released, causing temperature
changes between the sample and reference cells. A heat-sensitive device detects temperature differences between the two cells upon
binding, providing feedback to a heater to rectify temperature disparities and equilibrate the sample and reference cells. Subsequent
titrant injections gradually increase the molar ratio between the titrant and protein, as the protein reaches saturation. The frequency of
titrant binding events decreases, and the heat change decreases until the number of titrations in the sample cell is in excess relative to
the protein, indicating saturation. Utilizing software integrated with the isothermal titration calorimeter, parameters such as binding
constants, stoichiometry (n), and enthalpy change can be obtained through data tting, thus yielding comprehensive thermodynamic
insights into molecular interactions [83].
Based on the aforementioned principle, the typical procedure entails conguring the reference and sample cells to the desired
experimental temperature. The natural active compound is loaded into a highly precise injection device, such as a syringe, which is
then inserted into the sample cell containing the target protein. The natural active compound is injected into the sample cell in multiple
Fig. 7. The principle and general workow of ITC. Initially, the reference and sample cells are equilibrated to the desired temperature. Subse-
quently, a highly accurate injection device loaded with the active compound is inserted into the sample cell containing the target protein. The
natural active compound is titrated into the sample cell through sequential injections. During each injection, the microcalorimeter measures the
released heat until the binding reaction reaches equilibrium. Thermodynamic parameters are derived by tting the titration curve.
X. Jiang et al.
Heliyon 10 (2024) e33917
16
increments. The slightest temperature variation, on the order of millionths of a degree Celsius, can be detected and quantied if
binding occurs between the natural active compound and the protein. During each injection, the microcalorimeter records all released
heat until equilibrium is reached in the binding reaction, with the measured heat being proportional to the degree of binding. Utilizing
the software provided with the isothermal titration calorimeter, the acquired data can be tted and processed to determine ther-
modynamic parameters (Fig. 7). For instance, polyphenols play an essential role in modulating the gut microbiota and enhancing the
integrity of the mucus barrier. Feng et al. [84] demonstrated that the pyrogallol-containing polyphenols epigallocatechin gallate and
tannic acid strongly bind to the nucleophilic thiol groups of mucins. This binding was shown to serve as cross-linkers within mucin
networks, thereby reinforcing the mucus barrier, via ITC. Xue et al. [85] revealed that thymoquinone, a signicant phytoconstituent of
Nigella sativa used in Alzheimers disease therapy, binds spontaneously to human transferrin. The binding constant was determined to
be 0.22 ×10
6
M
1
, forming a stable complex, using both ITC and spectroscopic methods. In addition, ITC has been employed to
elucidate the targets of glycyrrhizin derivatives, which suppress cancer chemoresistance [86], valonea tannin, which exhibits tyros-
inase inhibition activity [87], bioactive coffee compounds that protect against serotonin degradation for treating depression and
potentially type 2 diabetes [88], neoandrographolide, a novel inhibitor of Rab5 [89], piperine, a bioactive constituent of black pepper,
shown to alleviate experimental allergic encephalomyelitis [90], luteolin, which exhibits antibacterial activity [91], red clover iso-
avones, which inhibit cancer cell metastasis [92], and the molecular interactions of acetylcholinesterase with potential acetylcho-
linesterase inhibitors isolated from the root of Rhodiola crenulata [93] (Table 10).
This technique boasts several advantages, including its rapidity, accuracy, and minimal sample demand. It does not enforce
stringent requirements on the reaction system, such as transparency, turbidity, or viscosity. ITC furnishes detailed and comprehensive
thermodynamic insights into molecular interactions, facilitating precise quantication of molecular interaction levels. However, a
limitation of this method is its low throughput. Consequently, it is commonly employed as a supplementary screening tool in high-
throughput screening protocols.
4. Identication of novel drugs and drug targets based on the innate function of natural products
Notably, there is a novel avenue for rapidly exploring the targets of natural products based on their innate functions [94]. In the
course of evolution, organisms produce specialized secondary metabolites to exhibit important functions, such as defence, survival,
and reproduction, via interacting with specic target proteins in inter-kingdom entities. The structural complementarity between
ligands and their targets provide more opportunities for nding promising ligand analogues. For instance, tabtoxin, a dipeptide
monobactam synthesized by Pseudomonas syringae, inhibits glutamine synthetase (GS) in plants [95]. GS, which has a conserved active
site in both plants and mammals, is a signicant target for cancer therapy [96]. Consequently, structural analogues of tabtoxin hold
promise as potent human GS antagonist for cancer treatment [97,98].
Conversely, ligands may actively interact with several structural analogues of the receptors present in different branches of life.
Inspired by this concept, we can identify therapeutic targets based on the innate functions of natural metabolites. For example, the
steroid hormone ecdysterone and its homologues interact with ecdysone receptors in arthropods. These receptors are composed of EcR
proteins and ultraspiracle protein (USP) heterodimers, functioning as nuclear receptors for ecdysone [99]. In humans, there are several
structural analogues of ecdysone receptors, including liver X receptor
α
(LXR
α
), LXRβ, estrogen receptor
α
(ER
α
), ERβ, retinoid X
receptor
α
(RXR
α
), and bile acid receptors. LXRβ is a promising therapeutic target for inammation [100], cancer [101], dyslipidemia
[102], and diabetes [103], while ER is a target for the treatment of breast cancer [104]. Some ecdysterone analogues have been
experimentally validated in cancer, diabetes, and inammation [105,106]. Therefore, ecdysterone and its analogues may act as se-
lective LXR agonists and ER modulators against human diseases.
Similarly, rhizobitoxin, a plant toxin produced by the fungus Bradyrhizobium elkanii, restrains the activity of β-cystathionase and 1-
aminocyclopropane-1-carboxylate (ACC) synthase to exert its natural effects [107,108]. Structural analysis reveals that the plant
Table 10
Recent studies that performed ITC-based target identication of natural products.
No. Natural products Category Bioactivity Target protein Reference
1 Epigallocatechin gallate and tannic
acid
Polyphenols Reinforcement of the mucus barrier Nucleophilic thiol groups of
mucins
[84]
2 Thymoquinone Isothiocyanates Alleviation of Alzheimers disease Transferrin [85]
3 Glycyrrhizin derivatives Terpenoids Suppression of cancer
chemoresistance
Progesterone receptor membrane
component 1
[86]
4 Valonea tannin Polyphenols Tyrosinase inhibition activity Skin whitening properties [87]
5 Coffee compounds Alkaloids Alleviation of depression and
potentially type 2 diabetes
Monoamine oxidase A [88]
Polyphenols
6 Neoandrographolide Terpenoids Anticancer activity Rab5 [89]
7 Piperine Alkaloids Alleviation of experimental allergic
encephalomyelitis
Dihydroorotate dehydrogenase [90]
8 Luteolin Flavonoids Antibacterial activity HK853 [91]
9 Red clover isoavones Flavonoids Inhibition of cancer cell metastasis Actin [92]
10 Constituents isolated from the root
of R. crenulata
Flavonoids Alleviation of Alzheimers disease Acetylcholinesterase [93]
Terpenoids
Phenols
X. Jiang et al.
Heliyon 10 (2024) e33917
17
Table 11
Benets and drawbacks of each target identication approach.
Approach Benets Drawbacks
Afnity
chromatography
1. The synthesis of immobilized probes is simple.
2. It facilitates the analysis of all adsorbed proteins without discrimination.
3. It can be easily conducted in individual laboratories.
1. Discrimination between truly high-afnity target proteins and high-abundance proteins with
low afnity is necessary.
2. The elution process relies on empirical knowledge; excessively strong washing conditions can
signicantly reduce the number of identied targets, while weak elution conditions can yield
false-positive results.
3. It does not reveal interactions between drugs and proteins in live cells under physiological
conditions.
4. Biotin afnity tags introduce steric hindrance, potentially reducing or even abolishing the
activity of natural active compounds and impeding probe molecule entry into cells.
ABPP 1. It exhibits strong specicity.
2. ABPs do not require pre-immobilization on matrix material.
3. It can be directly applied to live cells, elucidating drugtarget interactions under physiological
conditions.
1. Only proteins capable of forming covalent interactions with bioactive natural products can be
captured.
2. Synthesizing ABPs relies on the structure of the bioactive natural product and necessitates
expertise in medicinal chemistry.
PAL 1. It can facilitate the analysis of proteins that cannot be targeted by ABPs. 1. Photoafnity groups still exhibit non-specic binding to abundant cellular proteins.
2. The synthesis of PAL probes necessitates expertise in medicinal chemistry.
Proteome
microarray
assay
1. It requires minimal sample volumes.
2. High throughput enables parallel analysis of thousands to millions of proteins.
3. Utilizing protein chips is relatively simple.
1. It is relatively costly, necessitating a range of expensive, high-end instruments.
2. Standardization issues with the chips.
DARTS 1. No structural modication of natural bioactive compounds is necessary.
2. It can be easily implemented by individual laboratories.
1. Certain natural products do not signicantly affect the conformation of the target protein upon
binding.
2. Some proteins are less susceptible to proteases, resulting in undetectable changes in DARTS.
3. The ability to identify targets that exhibit low protein abundance is limited.
TPP 1. It monitors changes in protein thermal stability throughout the proteome during the action of
natural bioactive compounds.
2. It enables high-throughput detection of target proteins.
1. Inapplicable to heat-insensitive proteins.
2. The extraction of thermodynamic parameters besides the Tm value from TPP data is
challenging.
3. The magnitude of the Tm shift after protein binding with natural active compounds does not
consistently correlate with binding afnity.
4. Time-consuming and costly.
SPROX 1. It enables the large-scale assessment of protein folding states and potentially precise
determination of binding domains and peptide segments involved in the interactions between
compounds and target proteins.
1. It can only detect binding between proteins containing methionine residues and small
molecules.
2. Generating the standard curve requires a large sample volume.
3. The process is complex, costly, and requires substantial resources.
CETSA 1. No structural modication of natural bioactive compounds is required.
2. It can be easily implemented by individual laboratories.
1. Detection necessitates relatively high concentrations of the natural active compound.
2. The process exhibits low throughput in discovering new targets and is primarily utilized for
target validation.
ITC 1. It provides fast, accurate, and low-sample-volume analysis with minimal requirements for
reaction system transparency, turbidity, and viscosity.
2. It enables precise determination of comprehensive thermodynamic information.
1. It is typically utilized as an auxiliary screening technique in high-throughput screening due to
its low-throughput nature.
2. It requires specialized ITC equipment.
X. Jiang et al.
Heliyon 10 (2024) e33917
18
enzymes β-cystathionase and ACC synthase have structural analogues in human proteins, namely γ-cystathionase and kynurenine
aminotransferase, respectively. Over-activity of γ-cystathionase is linked to various diseases [109], while kynurenine aminotransferase
activity is associated with schizophrenia and cognitive impairment [110]. Hence, these proteins are potential therapeutic targets.
Some known inhibitors share structural similarities with rhizobitoxin [111], suggesting their potential action on these proteins.
Although rhizobitoxin is known to bind ACC synthase, there is no evidence of its binding to kynurenine aminotransferase. However,
structural analogues of rhizobitoxin have been found to suppress kynurenine aminotransferase.
In addition to the functions of metabolites, enzymes that catalyze their synthesis can also be potential therapeutic targets in
humans. For instance, strictosidine synthase, a plant enzyme, has a human structural homolog, regucalcin, which may serve as a
therapeutic target [94]. Although agonists or antagonists for regucalcin have not been studied yet, this concept can be applied to
develop relevant drugs.
5. Conclusions and prospects
Nature has been continuing to be a major source of medicinal products, and especially providing revolutionized structural leads in
the treatment of serious diseases for millennia, such as avonoids, alkaloids, steroidglycosides, sesquiterpene, essential oil and ste-
roids, all of which play an important role in medicinal chemistry. The identication of natural productstargets is as important as the
knowledge of natural products themselves because clarifying the former provides several new avenues for nding better drugs. Target
identication is the rst step in the early stage of drug discovery, followed by lead optimization.
Based on the foregoing, labeling methods facilitate direct labeling of target proteins while excluding indirectly targeted ones,
thereby signicantly reducing false-positive results in target identication. Nevertheless, these methods entail a high application
threshold and are unsuitable for investigating post-modied compounds with abolished activity, compounds exhibiting chemically
challenging synthesis, or analyzing trace amounts of natural active compounds. Conversely, non-labeling methods can be used to
identify targets of any natural active compound, even in trace amounts or multicomponent mixtures, albeit with lower reliability
compared to labeling methods. Challenges persist in eliminating the inuence of other proteins within the protein complex where the
natural active compound binds. Each method presents distinct advantages and limitations (Table 11). In practical applications, the
selection of appropriate target discovery methods is contingent upon the characteristics of small molecules or target proteins. Typi-
cally, a combination of multiple methods is employed to identify the targets of natural products [112115]. Furthermore, a recent
review outlined target identication and validation approaches for natural products categorized according to whether the natural
products were labeled [116]. Building upon this, this review claries the principles and processes of each method comprehensively.
Moreover, this review presents chemical structures and modifying moieties relevant to the labeling techniques.
New techniques continuously emerge, leveraging the aforementioned technologies, to aid in the identication of target proteins of
active natural products, thus signicantly impacting novel drug development.
In addition, there is a novel avenue for rapidly exploring the targets of natural products based on their innate functions. If a natural
metabolite (a) has been biosynthesized to fulll a specic role through its interaction with protein (A), the natural target of a, it
presents an ideal strategy to explore therapeutically important human proteins that are structurally analogous to protein A. The
aforementioned target validation approaches can be employed to validate the binding afnity of each target with a. Conversely, the
structural analogues of natural functional metabolite a may interact with protein A with varying degrees of afnity, thereby exerting
similar effects against human diseases.
Funding statement
This work was supported by the Matching Grant of the National Nature Science Foundation of China from Nanjing University of
Chinese Medicine, China (grant number NZY81903857), the National Natural Science Foundation of China (grant number 81903857),
the Natural Science Foundation of the Jiangsu Higher Education Institutions of China (grant number 24KJA360005) and the National
Natural Science Foundation of China (grant number 81961128020).
Ethics declarations
Not applicable.
Data availability
No data was used for the research described in the article.
CRediT authorship contribution statement
Xuan Jiang: Writing original draft. Kinyu Shon: Visualization. Xiaofeng Li: Visualization. Guoliang Cui: Writing review &
editing. Yuanyuan Wu: Project administration. Zhonghong Wei: Project administration. Aiyun Wang: Resources, Project admin-
istration. Xiaoman Li: Writing review & editing, Funding acquisition, Conceptualization. Yin Lu: Resources, Project administration,
Conceptualization.
X. Jiang et al.
Heliyon 10 (2024) e33917
19
Declaration of competing interest
The authors declare that they have no known competing nancial interests or personal relationships that could have appeared to
inuence the work reported in this paper.
References
[1] B. Shen, A new Golden age of natural products drug discovery, Cell 163 (6) (2015) 12971300.
[2] A. Parthasarathy, P.K. Mantravadi, K. Kalesh, Detectives and helpers: natural products as resources for chemical probes and compound libraries, Pharmacol.
Therapeut. 216 (2020) 107688.
[3] D.J. Newman, G.M. Cragg, Natural products as sources of new drugs from 1981 to 2014, J. Nat. Prod. 79 (3) (2016) 629661.
[4] L. Shaashua, M. Shabat-Simon, R. Haldar, P. Matzner, O. Zmora, M. Shabtai, et al., Perioperative COX-2 and β-adrenergic blockade improves metastatic
biomarkers in breast cancer patients in a phase-II randomized trial, Clin. Cancer Res. 23 (16) (2017) 46514661.
[5] F. Del Gaudio, F. Pollastro, M. Mozzicafreddo, R. Riccio, A. Minassi, M.C. Monti, Chemoproteomic shing identies arzanol as a positive modulator of brain
glycogen phosphorylase, Chem. Commun. 54 (91) (2018) 1286312866.
[6] R. Wu, R. Murali, Y. Kabe, S.W. French, Y.M. Chiang, S. Liu, et al., Baicalein targets GTPase-mediated autophagy to eliminate liver tumor-initiating stem cell-
like cells resistant to mTORC1 inhibition, Hepatology 68 (5) (2018) 17261740.
[7] F. Chen, K. Zhu, L. Chen, L. Ouyang, C. Chen, L. Gu, et al., Protein target identication of ginsenosides in skeletal muscle tissues: discovery of natural small-
molecule activators of muscle-type creatine kinase, Journal of ginseng research 44 (3) (2020) 461474.
[8] L. Liu, Y. Hua, D. Wang, L. Shan, Y. Zhang, J. Zhu, et al., A sesquiterpene lactone from a medicinal herb inhibits proinammatory activity of TNF-
α
by
inhibiting ubiquitin-conjugating enzyme UbcH5, Chem. Biol. 21 (10) (2014) 13411350.
[9] T. Ito, H. Ando, T. Suzuki, T. Ogura, K. Hotta, Y. Imamura, et al., Identication of a primary target of thalidomide teratogenicity, Science (New York, N.Y.) 327
(5971) (2010) 13451350.
[10] L.X. Liao, X.M. Song, L.C. Wang, H.N. Lv, J.F. Chen, D. Liu, et al., Highly selective inhibition of IMPDH2 provides the basis of antineuroinammation therapy,
Proc. Natl. Acad. Sci. U. S. A. 114 (29) (2017) E5986e5994.
[11] S. Du, X. Song, Y. Li, Y. Cao, F. Chu, O.A. Durojaye, et al., Celastrol inhibits ezrin-mediated migration of hepatocellular carcinoma cells, Sci. Rep. 10 (1) (2020)
11273.
[12] T. Dong, C. Li, X. Wang, L. Dian, X. Zhang, L. Li, et al., Ainsliadimer A selectively inhibits IKK
α
/β by covalently binding a conserved cysteine, Nat. Commun. 6
(2015) 6522.
[13] W.K. Low, Y. Dang, T. Schneider-Poetsch, Z. Shi, N.S. Choi, R.M. Rzasa, et al., Isolation and identication of eukaryotic initiation factor 4A as a molecular
target for the marine natural product Pateamine A, Methods Enzymol. 431 (2007) 303324.
[14] C.X. Liu, Q.Q. Yin, H.C. Zhou, Y.L. Wu, J.X. Pu, L. Xia, et al., Adenanthin targets peroxiredoxin I and II to induce differentiation of leukemic cells, Nat. Chem.
Biol. 8 (5) (2012) 486493.
[15] T. Yao, X. Xu, R. Huang, Recent advances about the applications of click reaction in chemical proteomics, Molecules 26 (17) (2021).
[16] Y. Liu, M.P. Patricelli, B.F. Cravatt, Activity-based protein proling: the serine hydrolases, Proc. Natl. Acad. Sci. U. S. A. 96 (26) (1999) 1469414699.
[17] X. Chen, Y. Wang, N. Ma, J. Tian, Y. Shao, B. Zhu, et al., Target identication of natural medicine with chemical proteomics approach: probe synthesis, target
shing and protein identication, Signal Transduct. Targeted Ther. 5 (1) (2020) 72.
[18] M. Gersch, J. Kreuzer, S.A. Sieber, Electrophilic natural products and their biological targets, Nat. Prod. Rep. 29 (6) (2012) 659682.
[19] A.E. Speers, B.F. Cravatt, Proling enzyme activities in vivo using click chemistry methods, Chem. Biol. 11 (4) (2004) 535546.
[20] A. Galmozzi, E. Dominguez, B.F. Cravatt, E. Saez, Application of activity-based protein proling to study enzyme function in adipocytes, Methods Enzymol.
538 (2014) 151169.
[21] J.N. Spradlin, X. Hu, C.C. Ward, S.M. Brittain, M.D. Jones, L. Ou, et al., Harnessing the anti-cancer natural product nimbolide for targeted protein degradation,
Nat. Chem. Biol. 15 (7) (2019) 747755.
[22] W.S. Zhao, K.F. Chen, M. Liu, X.L. Jia, Y.Q. Huang, B.B. Hao, et al., Investigation of targets and anticancer mechanisms of covalently acting natural products by
functional proteomics, Acta Pharmacol. Sin. 48 (8) (2023) 17011711.
[23] B.P. Belcher, P.A. Machicao, B. Tong, E. Ho, J. Friedli, B. So, et al., Chemoproteomic proling reveals that anti-cancer natural product dankastatin B covalently
targets mitochondrial VDAC3, Chembiochem : a European journal of chemical biology (2023) e202300111.
[24] M.H. Wright, Y. Tao, J. Drechsel, J. Krysiak, S. Chamni, A. Weigert-Munoz, et al., Quantitative chemoproteomic proling reveals multiple target interactions of
spongiolactone derivatives in leukemia cells, Chem. Commun. 53 (95) (2017) 1281812821.
[25] E. Yoo, C.J. Schulze, B.H. Stokes, O. Onguka, T. Yeo, S. Mok, et al., The antimalarial natural product salinipostin A identies essential
α
/β serine hydrolases
involved in lipid metabolism in P. Falciparum parasites, Cell Chem. Biol. 27 (2) (2020) 143157.e145.
[26] P. Le, M.B. Nodwell, J. Eirich, S.A. Sieber, A chemical proteomic analysis of illudin-interacting proteins, Chemistry 25 (54) (2019) 1264412651.
[27] C. Chen, T. Zhu, X. Liu, D. Zhu, Y. Zhang, S. Wu, et al., Identication of a novel PHGDH covalent inhibitor by chemical proteomics and phenotypic proling,
Acta Pharm. Sin. B 12 (1) (2022) 246261.
[28] L. Li, Y. Zhao, R. Cao, L. Li, G. Cai, J. Li, et al., Activity-based protein proling reveals GSTO1 as the covalent target of piperlongumine and a promising target
for combination therapy for cancer, Chem. Commun. 55 (30) (2019) 44074410.
[29] J. Sumranjit, S.J. Chung, Recent advances in target characterization and identication by photoafnity probes, Molecules 18 (9) (2013) 1042510451.
[30] D. Wang, Y. Cao, L. Zheng, D. Lv, L. Chen, X. Xing, et al., Identication of Annexin A2 as a target protein for plant alkaloid matrine, Chem. Commun. 53 (36)
(2017) 50205023.
[31] D. Tranter, A.O. Paatero, S. Kawaguchi, S. Kazemi, J.D. Serrill, J. Kellosalo, et al., Coibamide A targets Sec61 to prevent biogenesis of secretory and membrane
proteins, ACS Chem. Biol. 15 (8) (2020) 21252136.
[32] P. Martín-Acosta, Q. Meng, J. Klimek, A.P. Reddy, L. David, S.K. Petrie, et al., A clickable photoafnity probe of betulinic acid identies tropomyosin as a
target, Acta Pharm. Sin. B 12 (5) (2022) 24062416.
[33] T.Y. Zhu, X.T. Wu, C. Chen, X.Q. Liu, L. Zhu, J.G. Luo, et al., Photoafnity probe reveals the potential target of harringtonolide for cancer cell migration
inhibition, ACS Med. Chem. Lett. 13 (3) (2022) 449456.
[34] Y. Zhou, Z. Di, X. Li, Y. Shan, W. Li, H. Zhang, et al., Chemical proteomics reveal CD147 as a functional target of pseudolaric acid B in human cancer cells,
Chem. Commun. 53 (62) (2017) 86718674.
[35] A.R. Ranade, L. Higgins, T.W. Markowski, N. Glaser, D. Kashin, R. Bai, et al., Characterizing the epothilone binding site on β-tubulin by photoafnity labeling:
identication of β-tubulin peptides TARGSQQY and TSRGSQQY as targets of an epothilone photoprobe for polymerized tubulin, J. Med. Chem. 59 (7) (2016)
34993514.
[36] R.E. Wang, C.R. Hunt, J. Chen, J.S. Taylor, Biotinylated quercetin as an intrinsic photoafnity proteomics probe for the identication of quercetin target
proteins, Bioorg. Med. Chem. 19 (16) (2011) 47104720.
[37] Y. Kotake, K. Sagane, T. Owa, Y. Mimori-Kiyosue, H. Shimizu, M. Uesugi, et al., Splicing factor SF3b as a target of the antitumor natural product pladienolide,
Nat. Chem. Biol. 3 (9) (2007) 570575.
[38] S.E. Ong, B. Blagoev, I. Kratchmarova, D.B. Kristensen, H. Steen, A. Pandey, et al., Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and
accurate approach to expression proteomics, Mol. Cell. Proteomics : MCP 1 (5) (2002) 376386.
X. Jiang et al.
Heliyon 10 (2024) e33917
20
[39] J. Dai, K. Liang, S. Zhao, W. Jia, Y. Liu, H. Wu, et al., Chemoproteomics reveals baicalin activates hepatic CPT1 to ameliorate diet-induced obesity and hepatic
steatosis, Proc. Natl. Acad. Sci. U. S. A. 115 (26) (2018) E5896e5905.
[40] C. Chen, L. Gong, X. Liu, T. Zhu, W. Zhou, L. Kong, et al., Identication of peroxiredoxin 6 as a direct target of withangulatin A by quantitative chemical
proteomics in non-small cell lung cancer, Redox Biol. 46 (2021) 102130.
[41] H.N. Zhang, L. Yang, J.Y. Ling, D.M. Czajkowsky, J.F. Wang, X.W. Zhang, et al., Systematic identication of arsenic-binding proteins reveals that hexokinase-2
is inhibited by arsenic, Proc. Natl. Acad. Sci. U. S. A. 112 (49) (2015) 1508415089.
[42] T.T. Liu, H. Yang, F.F. Zhuo, Z. Yang, M.M. Zhao, Q. Guo, et al., Atypical E3 ligase ZFP91 promotes small-molecule-induced E2F2 transcription factor
degradation for cancer therapy, EBioMedicine 86 (2022) 104353.
[43] F. Jiang, M. Li, H. Wang, B. Ding, C. Zhang, Z. Ding, et al., Coelonin, an anti-inammation active component of Bletilla striata and its potential mechanism, Int.
J. Mol. Sci. 20 (18) (2019).
[44] J. Chang, Y. Kim, H.J. Kwon, Advances in identication and validation of protein targets of natural products without chemical modication, Nat. Prod. Rep. 33
(5) (2016) 719730.
[45] B. Lomenick, R. Hao, N. Jonai, R.M. Chin, M. Aghajan, S. Warburton, et al., Target identication using drug afnity responsive target stability (DARTS), Proc.
Natl. Acad. Sci. U. S. A. 106 (51) (2009) 2198421989.
[46] Y.S. Ren, H.L. Li, X.H. Piao, Z.Y. Yang, S.M. Wang, Y.W. Ge, Drug afnity responsive target stability (DARTS) accelerated small molecules target discovery:
principles and application, Biochem. Pharmacol. 194 (2021) 114798.
[47] X.H. Huang, X. Yan, Q.H. Zhang, P. Hong, W.X. Zhang, Y.P. Liu, et al., Direct targeting of HSP90 with daurisoline destabilizes β-catenin to suppress lung cancer
tumorigenesis, Cancer Lett. 489 (2020) 6678.
[48] J. Geng, W. Liu, J. Gao, C. Jiang, T. Fan, Y. Sun, et al., Andrographolide alleviates Parkinsonism in MPTP-PD mice via targeting mitochondrial ssion mediated
by dynamin-related protein 1, Br. J. Pharmacol. 176 (23) (2019) 45744591.
[49] R.F. Dong, C.J. Qin, Y. Yin, L.L. Han, C.M. Xiao, K.D. Wang, et al., Discovery of a potent inhibitor of chaperone-mediated autophagy that targets the HSC70-
LAMP2A interaction in non-small cell lung cancer therapy, Br. J. Pharmacol. (2023), https://doi.org/10.1111/bph.16165. Online ahead of print.
[50] M. Ren, J. Chen, H. Xu, W. Li, T. Wang, Z. Chi, et al., Ergolide covalently binds NLRP3 and inhibits NLRP3 inammasome-mediated pyroptosis, Int.
Immunopharm. 120 (2023) 110292.
[51] J. Wei, S. Fan, H. Yu, L. Shu, Y. Li, A new strategy for the rapid identication and validation of the direct targets of aconitine-induced cardiotoxicity, Drug Des.
Dev. Ther. 15 (2021) 46494664.
[52] E. Morretta, A. Tosco, C. Festa, M. Mozzicafreddo, M.C. Monti, A. Casapullo, A. Crellastatin, A PARP-1 inhibitor discovered by complementary proteomic
approaches, ChemMedChem 15 (3) (2020) 317323.
[53] S. Esch, S. K ¨
onig, B. Bopp, J. Jose, S. Brandt, A. Hensel, Cryptotanshinone from Salvia miltiorrhiza roots reduces cytokeratin CK1/10 expression in
keratinocytes by activation of peptidyl-prolyl-cis-trans-isomerase FKBP1A, Planta Med. 85 (7) (2019) 552562.
[54] M.M. Derry, R.R. Somasagara, K. Raina, S. Kumar, J. Gomez, M. Patel, et al., Target identication of grape seed extract in colorectal cancer using drug afnity
responsive target stability (DARTS) technique: role of endoplasmic reticulum stress response proteins, Curr. Cancer Drug Targets 14 (4) (2014) 323336.
[55] Y. Tu, L. Tan, H. Tao, Y. Li, H. Liu, CETSA and thermal proteome proling strategies for target identication and drug discovery of natural products,
Phytomedicine : international journal of phytotherapy and phytopharmacology 116 (2023) 154862.
[56] V.C. Kirsch, C. Orgler, S. Braig, I. Jeremias, D. Auerbach, R. Müller, et al., The cytotoxic natural product vioprolide A targets nucleolar protein 14, which is
essential for ribosome biogenesis, Angew. Chem. 59 (4) (2020) 15951600.
[57] J.M. Dziekan, H. Yu, D. Chen, L. Dai, G. Wirjanata, A. Larsson, et al., Identifying purine nucleoside phosphorylase as the target of quinine using cellular
thermal shift assay, Sci. Transl. Med. 11 (473) (2019).
[58] K. Chen, T. Wang, Y. Li, J. Wu, C.X. Zhao, S. Liu, et al., Rhodojaponin VI indirectly targets Cav 2.2 channels via N-ethylmaleimide-sensitive fusion protein to
alleviate neuropathic pain, Acta Pharm. Sin. B 13 (3) (2023) 13261336.
[59] Q.T. Han, W.Q. Yang, C. Zang, L. Zhou, C.J. Zhang, X. Bao, et al., The toxic natural product tutin causes epileptic seizures in mice by activating calcineurin,
Signal Transduct. Targeted Ther. 8 (1) (2023) 101.
[60] Z. Fu, S. Li, J. Liu, C. Zhang, C. Jian, L. Wang, et al., Natural product alantolactone targeting AKR1C1 suppresses cell proliferation and metastasis in non-small-
cell lung cancer, Front. Pharmacol. 13 (2022) 847906.
[61] C.P. Sun, J.J. Zhou, Z.L. Yu, X.K. Huo, J. Zhang, C. Morisseau, et al., Kurarinone alleviated Parkinsons disease via stabilization of epoxyeicosatrienoic acids in
animal model, Proc. Natl. Acad. Sci. U. S. A. 119 (9) (2022).
[62] X.W. Zhang, N. Feng, L.C. Wang, D. Liu, Y.M. Hua, C. Zhang, et al., Small-molecule arone protects from neuroinammation in LPS-activated microglia BV-2
cells by targeting histone-remodeling chaperone ASF1a, Biochem. Pharmacol. 177 (2020) 113932.
[63] G.M. West, L. Tang, M.C. Fitzgerald, Thermodynamic analysis of protein stability and ligand binding using a chemical modication- and mass spectrometry-
based strategy, Anal. Chem. 80 (11) (2008) 41754185.
[64] P.D. Dearmond, Y. Xu, E.C. Strickland, K.G. Daniels, M.C. Fitzgerald, Thermodynamic analysis of protein-ligand interactions in complex biological mixtures
using a shotgun proteomics approach, J. Proteome Res. 10 (11) (2011) 49484958.
[65] Y. Xu, M.A. Wallace, M.C. Fitzgerald, Thermodynamic analysis of the geldanamycin-hsp 90 interaction in a whole cell lysate using a mass spectrometry-based
proteomics approach, J. Am. Soc. Mass Spectrom. 27 (10) (2016) 16701676.
[66] M.A. Geer Wallace, D.Y. Kwon, D.H. Weitzel, C.T. Lee, T.N. Stephenson, J.T. Chi, et al., Discovery of manassantin A protein targets using large-scale protein
folding and stability measurements, J. Proteome Res. 15 (8) (2016) 26882696.
[67] D. Martinez Molina, R. Jafari, M. Ignatushchenko, T. Seki, E.A. Larsson, C. Dan, et al., Monitoring drug target engagement in cells and tissues using the cellular
thermal shift assay, Science (New York, N.Y.) 341 (6141) (2013) 8487.
[68] A. Yang, K. Zeng, H. Huang, D. Liu, X. Song, Y. Qian, et al., Usenamine A induces apoptosis and autophagic cell death of human hepatoma cells via interference
with the Myosin-9/actin-dependent cytoskeleton remodeling, Phytomedicine : international journal of phytotherapy and phytopharmacology 116 (2023)
154895.
[69] Y. Li, Y. Li, Y. Zhu, W. Ji, Y. Wang, X. Dong, et al., Structure-based virtual screening for discovery of paederosidic acid from Paederia scandens as novel P2Y
(14)R antagonist, Phytomedicine : international journal of phytotherapy and phytopharmacology 115 (2023) 154851.
[70] W. Zhang, Q. Gong, Z. Tang, X. Ma, Z. Wang, J. Guan, et al., The natural product, echinatin, protects mice from methicillin-resistant Staphylococcus aureus
pneumonia by inhibition of alpha-hemolysin expression, Front. Microbiol. 14 (2023) 1128144.
[71] S. Zhao, P. Zhang, Y. Yan, W. Xu, J. Li, L. Wang, et al., Network pharmacology-based prediction and validation of the active ingredients and potential
mechanisms of the Huangxiong formula for treating ischemic stroke, J. Ethnopharmacol. 312 (2023) 116507.
[72] Z. Yin, Y. Lv, L. Deng, G. Li, R. Ou, L. Chen, et al., Targeting ABCB6 with nitidine chloride inhibits PI3K/AKT signaling pathway to promote ferroptosis in
multiple myeloma, Free Radic. Biol. Med. 203 (2023) 86101.
[73] D. Xu, W. Zhao, Y. Feng, X. Wen, H. Liu, J. Ping, Pentoxifylline attenuates nonalcoholic fatty liver by inhibiting hepatic macrophage polarization to the M1
phenotype, Phytomedicine : international journal of phytotherapy and phytopharmacology 106 (2022) 154368.
[74] J. Jiao, C.S. Cheng, P. Xu, P. Yang, K. Zhang, Y. Jing, et al., Mechanisms of pancreatic tumor suppression mediated by Xiang-lian pill: an integrated in silico
exploration and experimental validation, J. Ethnopharmacol. 298 (2022) 115586.
[75] M. Zhang, Z. Tao, L. Gao, F. Chen, Y. Ye, S. Xu, et al., Toosendanin inhibits colorectal cancer cell growth through the Hedgehog pathway by targeting Shh, Drug
Dev. Res. 83 (5) (2022) 12011211.
[76] Y. Tu, L. Tan, T. Lu, K. Wang, H. Wang, B. Han, et al., Glytabastan B, a coumestan isolated from Glycine tabacina, alleviated synovial inammation,
osteoclastogenesis and collagen-induced arthritis through inhibiting MAPK and PI3K/AKT pathways, Biochem. Pharmacol. 197 (2022) 114912.
[77] D. Zhu, C. Chen, X. Liu, S. Wang, J. Zhu, H. Zhang, et al., Osteosarcoma cell proliferation suppression via SHP-2-mediated inactivation of the JAK/STAT3
pathway by tubocapsenolide A, J. Adv. Res. 34 (2021) 7991.
X. Jiang et al.
Heliyon 10 (2024) e33917
21
[78] J.H. Fan, M.M. Xu, L.M. Zhou, Z.W. Gui, L. Huang, X.G. Li, et al., Integrating network pharmacology deciphers the action mechanism of Zuojin capsule in
suppressing colorectal cancer, Phytomedicine : international journal of phytotherapy and phytopharmacology 96 (2022) 153881.
[79] R. Wang, X. Hu, J. Wang, L. Zhou, Y. Hong, Y. Zhang, et al., Proanthocyanidin A1 promotes the production of platelets to ameliorate chemotherapy-induced
thrombocytopenia through activating JAK2/STAT3 pathway, Phytomedicine : international journal of phytotherapy and phytopharmacology 95 (2022)
153880.
[80] R. Yuan, W. Zhao, Q.Q. Wang, J. He, S. Han, H. Gao, et al., Cucurbitacin B inhibits non-small cell lung cancer in vivo and in vitro by triggering TLR4/NLRP3/
GSDMD-dependent pyroptosis, Pharmacol. Res. 170 (2021) 105748.
[81] X. Liu, Q. Hu, W. Wang, H. Ma, J. Pu, J. Cui, et al., A protein-fragment complementation assay reveals that celastrol and gambogic acid suppress ER
α
mutants
in breast cancer, Biochem. Pharmacol. 188 (2021) 114583.
[82] W. Wang, Y. Wu, S. Chen, X. Liu, J. He, S. Wang, et al., Shikonin is a novel and selective IMPDH2 inhibitor that target triple-negative breast cancer, Phytother
Res. : PT 35 (1) (2021) 463476.
[83] O. Callies, A. Hern´
andez Daranas, Application of isothermal titration calorimetry as a tool to study natural product interactions, Nat. Prod. Rep. 33 (7) (2016)
881904.
[84] G. Feng, K. Han, Q. Yang, W. Feng, J. Guo, J. Wang, et al., Interaction of pyrogallol-containing polyphenols with mucin reinforces intestinal mucus barrier
properties, J. Agric. Food Chem. 70 (30) (2022) 95369546.
[85] B. Xue, D. DasGupta, M. Alam, M.S. Khan, S. Wang, A. Shamsi, et al., Investigating binding mechanism of thymoquinone to human transferrin, targeting
Alzheimers disease therapy, J. Cell. Biochem. 123 (8) (2022) 13811393.
[86] Y. Kabe, I. Koike, T. Yamamoto, M. Hirai, A. Kanai, R. Furuhata, et al., Glycyrrhizin derivatives suppress cancer chemoresistance by inhibiting progesterone
receptor membrane component 1, Cancers 13 (13) (2021).
[87] J. Liu, Y. Liu, X. He, B. Teng, J.M. McRae, Valonea tannin: tyrosinase inhibition activity, structural elucidation and insights into the inhibition mechanism,
Molecules 26 (9) (2021).
[88] J. Grzelczyk, G. Budryn, J. Pe˜
na-García, D. Szwajgier, I. Gałązka-Czarnecka, J. Oracz, et al., Evaluation of the inhibition of monoamine oxidase A by bioactive
coffee compounds protecting serotonin degradation, Food Chem. 348 (2021) 129108.
[89] J. Zhang, Y. Sun, L.Y. Zhong, N.N. Yu, L. Ouyang, R.D. Fang, et al., Structure-based discovery of neoandrographolide as a novel inhibitor of Rab5 to suppress
cancer growth, Comput. Struct. Biotechnol. J. 18 (2020) 39363946.
[90] Z. Liu, Q. Hu, W. Wang, S. Lu, D. Wu, S. Ze, et al., Natural product piperine alleviates experimental allergic encephalomyelitis in mice by targeting
dihydroorotate dehydrogenase, Biochem. Pharmacol. 177 (2020) 114000.
[91] Y. Zhou, L. Huang, S. Ji, S. Hou, L. Luo, C. Li, et al., Structural basis for the inhibition of the autophosphorylation activity of HK853 by luteolin, Molecules 24
(5) (2019).
[92] G. Budryn, J. Grzelczyk, H. P´
erez-S´
anchez, Binding of red clover isoavones to actin as A potential mechanism of anti-metastatic activity restricting the
migration of cancer cells, Molecules 23 (10) (2018).
[93] F.J. Li, Y. Liu, Y. Yuan, B. Yang, Z.M. Liu, L.Q. Huang, Molecular interaction studies of acetylcholinesterase with potential acetylcholinesterase inhibitors from
the root of Rhodiola crenulata using molecular docking and isothermal titration calorimetry methods, Int. J. Biol. Macromol. 104 (Pt A) (2017) 527532.
[94] P.S. Suresh, S. Kumari, D. Sahal, U. Sharma, Innate functions of natural products: a promising path for the identication of novel therapeutics, Eur. J. Med.
Chem. 260 (2023) 115748.
[95] M.D. Thomas, P.J. Langston-Unkefer, T.F. Uchytil, R.D. Durbin, Inhibition of glutamine synthetase from pea by tabtoxinine-beta-lactam, Plant Physiol. 71 (4)
(1983) 912915.
[96] M.M. Yamashita, R.J. Almassy, C.A. Janson, D. Cascio, D. Eisenberg, Rened atomic model of glutamine synthetase at 3.5 A resolution, J. Biol. Chem. 264 (30)
(1989) 1768117690.
[97] J. Kreuzer, N.C. Bach, D. Forler, S.A. Sieber, Target discovery of acivicin in cancer cells elucidates its mechanism of growth inhibitionElectronic
supplementary information (ESI) available: synthesis, cloning, protein expression, purication and biochemical assays. See DOI: 10.1039/c4sc02339k, Chem.
Sci. 6 (1) (2014) 237245.
[98] H.L. Kindler, H.A. Burris 3rd, A.B. Sandler, I.A. Oliff, A phase II multicenter study of L-alanosine, a potent inhibitor of adenine biosynthesis, in patients with
MTAP-decient cancer, Invest. N. Drugs 27 (1) (2009) 7581.
[99] L.M. Riddiford, P. Cherbas, J.W. Truman, Ecdysone receptors and their biological actions, Vitam. Horm. 60 (2000) 173.
[100] M.B. Fessler, The challenges and promise of targeting the Liver X Receptors for treatment of inammatory disease, Pharmacol. Therapeut. 181 (2018) 112.
[101] C.Y. Lin, J. Gustafsson, Targeting liver X receptors in cancer therapeutics, Nat. Rev. Cancer 15 (4) (2015) 216224.
[102] J. Bełtowski, Liver X receptors (LXR) as therapeutic targets in dyslipidemia, Cardiovascular therapeutics 26 (4) (2008) 297316.
[103] S. Hazra, A. Rasheed, A. Bhatwadekar, X. Wang, L.C. Shaw, M. Patel, et al., Liver X receptor modulates diabetic retinopathy outcome in a mouse model of
streptozotocin-induced diabetes, Diabetes 61 (12) (2012) 32703279.
[104] S. Sommer, S.A. Fuqua, Estrogen receptor and breast cancer, Semin. Cancer Biol. 11 (5) (2001) 339352.
[105] J. Malíkov´
a, J. Swaczynov´
a, Z. Kol´
ar, M. Strnad, Anticancer and antiproliferative activity of natural brassinosteroids, Phytochemistry 69 (2) (2008) 418426.
[106] R. Sundaram, R. Naresh, P. Shanthi, P. Sachdanandam, Efcacy of 20-OH-ecdysone on hepatic key enzymes of carbohydrate metabolism in streptozotocin
induced diabetic rats, Phytomedicine : international journal of phytotherapy and phytopharmacology 19 (89) (2012) 725729.
[107] J. Giovanelli, L.D. Owens, S.H. Mudd, Mechanism of inhibition of spinach beta-cystathionase by rhizobitoxine, Biochim. Biophys. Acta 227 (3) (1971)
671684.
[108] T. Yasuta, S. Satoh, K. Minamisawa, New assay for rhizobitoxine based on inhibition of 1-aminocyclopropane-1-carboxylate synthase, Appl. Environ.
Microbiol. 65 (2) (1999) 849852.
[109] Y.Y. Mok, M.S. Atan, Ping C. Yoke, Jing W. Zhong, M. Bhatia, S. Moochhala, et al., Role of hydrogen sulphide in haemorrhagic shock in the rat: protective effect
of inhibitors of hydrogen sulphide biosynthesis, Br. J. Pharmacol. 143 (7) (2004) 881889.
[110] L.K. Nilsson, K.R. Linderholm, G. Engberg, L. Paulson, K. Blennow, L.H. Lindstr¨
om, et al., Elevated levels of kynurenic acid in the cerebrospinal uid of male
patients with schizophrenia, Schizophr. Res. 80 (23) (2005) 315322.
[111] A. Asimakopoulou, P. Panopoulos, C.T. Chasapis, C. Coletta, Z. Zhou, G. Cirino, et al., Selectivity of commonly used pharmacological inhibitors for
cystathionine β synthase (CBS) and cystathionine γ lyase (CSE), Br. J. Pharmacol. 169 (4) (2013) 922932.
[112] S. Yun, Y.J. Lee, J. Choi, N.D. Kim, D.C. Han, B.M. Kwon, Acacetin inhibits the growth of STAT3-activated DU145 prostate cancer cells by directly binding to
signal transducer and activator of transcription 3 (STAT3), Molecules 26 (20) (2021).
[113] Y.C. Liu, N. Feng, W.W. Li, P.F. Tu, J.P. Chen, J.Y. Han, et al., Costunolide plays an anti-neuroinammation role in lipopolysaccharide-induced BV2 microglial
activation by targeting cyclin-dependent kinase 2, Molecules 25 (12) (2020).
[114] C. Cassiano, R. Esposito, A. Tosco, A. Casapullo, M. Mozzicafreddo, C. Tringali, et al., Chemical proteomics-guided identication of a novel biological target of
the bioactive neolignan magnolol, Front. Chem. 7 (2019) 53.
[115] F. Lu, D. Wang, R.L. Li, L.Y. He, L. Ai, C.J. Wu, Current strategies and technologies for nding drug targets of active components from traditional Chinese
medicine, Front. Biosci. 26 (9) (2021) 572589.
[116] Y. Zhu, Z. Ouyang, H. Du, M. Wang, J. Wang, H. Sun, et al., New opportunities and challenges of natural products research: when target identication meets
single-cell multiomics, Acta Pharm. Sin. B 12 (11) (2022) 40114039.
X. Jiang et al.
Article
Full-text available
Many antiparasitic drugs have become obsolete and ineffective in treating parasitic diseases. This ineffectiveness arises from parasite drug resistance, high toxicity, and low drug efficacy. Thus, the discovery of novel agents is urgently needed to control parasitic diseases. Various strategies are employed in drug discovery, design, and development. This review highlights the paradigm of searching for bioactive molecules produced during inter- and intraspecific competition among organisms, particularly between microbes and parasites, as a strategy for de novo antiparasitic drug discovery. Competitive interactions occur when individuals of the same or different species coexist in overlapping niches and compete for space and resources. These interactions are well recognized. Therefore, bioactive molecules released during these interactions are promising targets for novel drug discovery. Compelling data indicate that microbes remain a potential source for the discovery of novel antiparasitic drugs because of their diversity. Many antimicrobial producers in nature have yet to be isolated and investigated. This body of evidence underscores the success of numerous therapeutic drugs, including penicillin, β-lactams, and tetracyclines, which have been successfully discovered and developed for treating infectious diseases. This review comprehensively covers these concepts, with a particular focus on inter- and intraspecific competition in the discovery of novel antiparasitic agents. This approach will pave the way for identifying alternative strategies to control and eradicate parasitic diseases that continue to threaten human health. Additionally, this review discusses current antiparasitic drugs and their mechanisms of action, limitations, and existing gaps. This discussion emphasizes the ongoing need to explore novel antiparasitic drugs.
Article
Full-text available
Background and Purpose Chaperone‐mediated autophagy (CMA) is a selective type of autophagy targeting protein degradation and maintains high activity in many malignancies. Inhibition of the combination of HSC70 and LAMP2A can potently block CMA. At present, knockdown of LAMP2A remains the most specific method for inhibiting CMA and chemical inhibitors against CMA have not yet been discovered. Experimental Approach Levels of CMA in non‐small cell lung cancer (NSCLC) tissue samples were confirmed by tyramide signal amplification dual immunofluorescence assay. High‐content screening was performed based on CMA activity, to identify potential inhibitors of CMA. Inhibitor targets were determined by drug affinity responsive target stability‐mass spectrum and confirmed by protein mass spectrometry. CMA was inhibited and activated to elucidate the molecular mechanism of the CMA inhibitor. Key Results Suppression of interactions between HSC70 and LAMP2A blocked CMA in NSCLC, restraining tumour growth. Polyphyllin D (PPD) was identified as a targeted CMA small‐molecule inhibitor through disrupting HSC70–LAMP2A interactions. The binding sites for PPD were E129 and T278 at the nucleotide‐binding domain of HSC70 and C‐terminal of LAMP2A, respectively. PPD accelerated unfolded protein generation to induce reactive oxygen species (ROS) accumulation by inhibiting HSC70–LAMP2A–eIF2α signalling axis. Also, PPD prevented regulatory compensation of macroautophagy induced by CMA inhibition via blocking the STX17–SNAP29–VAMP8 signalling axis. Conclusions and Implications PPD is a targeted CMA inhibitor that blocked both HSC70–LAMP2A interactions and LAMP2A homo‐multimerization. CMA suppression without increasing the regulatory compensation from macroautophagy is a good strategy for NSCLC therapy. LINKED ARTICLES This article is part of a themed issue Natural Products and Cancer: From Drug Discovery to Prevention and Therapy. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.10/issuetoc
Article
Full-text available
Chlorinated gymnastatin and dankastatin alkaloids derived from the fungal strain Gymnascella dankaliensis have been reported to possess significant anticancer activity but their mode of action is unknown. These members possess electrophilic functional groups that can might undergo covalent bond formation with specific proteins to exert their biological activity. To better understand the mechanism of action of this class of natural products, we mapped the proteome‐wide cysteine reactivity of the most potent of these alkaloids, dankastatin B, by using activity‐based protein profiling chemoproteomic approaches. We identified a primary target of dankastatin B in breast cancer cells as cysteine C65 of the voltage‐dependent anion‐selective channel on the outer mitochondrial membrane VDAC3. We demonstrated direct and covalent interaction of dankastatin B with VDAC3. VDAC3 knockdown conferred hypersensitivity to dankastatin B‐mediated antiproliferative effects in breast cancer cells, thus indicating that VDAC3 was at least partially involved in the anticancer effects of this natural product. Our study reveals a potential mode of action of dankastatin B through covalent targeting of VDAC3 and highlights the utility of chemoproteomic approaches in gaining mechanistic understanding of electrophilic natural products.
Article
Full-text available
Antimicrobial resistance (AMR) is a global, multifaceted crisis that poses significant challenges to the successful eradication of devastating pathogens, particularly methicillin-resistant Staphylococcus aureus (MRSA), a persistent superbug that causes devastating infections. The scarcity of new antibacterial drugs is obvious, and antivirulence strategies that reduce the pathogenicity of bacteria by weakening their virulence have become the subject of intense investigation. Alpha-hemolysin (Hla), a cytolytic pore-forming toxin, has a pivotal role in S. aureus pathogenesis. Here, we demonstrated that echinatin, a natural compound isolated from licorice, effectively inhibited the hemolytic activity of MRSA at 32 μg/mL. In addition, echinatin did not interfere with bacterial growth and had no significant cytotoxicity at the inhibitory concentration of S. aureus hemolysis. Heptamer formation tightly correlated with Hla-mediated cell invasion, whereas echinatin did not affect deoxycholic acid-induced oligomerization of Hla. Echinatin affected hemolytic activity through indirect binding to Hla as confirmed by the neutralization assay and cellular thermal shift assay (CETSA). Furthermore, qRT–PCR and western blot analyses revealed that echinatin suppressed Hla expression at both the mRNA and protein levels as well as the transcript levels of Agr quorum-sensing system-related genes. Additionally, when echinatin was added to a coculture system of A549 cells and S. aureus, it significantly reduced cell damage. Importantly, echinatin exhibited a significant therapeutic effect in an MRSA-induced mouse pneumonia model. In conclusion, the present findings demonstrated that echinatin significantly inhibits the hemolysin effect and may be a potential candidate compound for combating drug-resistant MRSA infections.
Article
In the course of evolution, living organisms have become well equipped with diverse natural products that serve important functions, including defence from biotic and abiotic stress, growth regulation, reproduction, metabolism, and epigenetic regulation. It seems to be the organism's ecological niche that influences the natural product's structural and functional diversity. Indeed, natural products constitute the nuts and bolts of molecular co-evolution and ecological relationships among different life forms. Since natural products in the form of specialized secondary metabolites exhibit biological functions via interactions with specific target proteins, they can provide a simultaneous glimpse of both new therapeutics and therapeutic targets in humans as well. In this review, we have discussed the innate role of natural products in the ecosystem and how this intrinsic role provides a futuristic opportunity to identify new drugs and therapeutic targets rapidly.
Article
Background: Monitoring target engagement at various stages of drug development is essential for natural product (NP)-based drug discovery and development. The cellular thermal shift assay (CETSA) developed in 2013 is a novel, broadly applicable, label-free biophysical assay based on the principle of ligand-induced thermal stabilization of target proteins, which enables direct assessment of drug-target engagement in physiologically relevant contexts, including intact cells, cell lysates and tissues. This review aims to provide an overview of the work principles of CETSA and its derivative strategies and their recent progress in protein target validation, target identification and drug lead discovery of NPs. Methods: A literature-based survey was conducted using the Web of Science and PubMed databases. The required information was reviewed and discussed to highlight the important role of CETSA-derived strategies in NP studies. Results: After nearly ten years of upgrading and evolution, CETSA has been mainly developed into three formats: classic Western blotting (WB)-CETSA for target validation, thermal proteome profiling (TPP, also known as MS-CETSA) for unbiased proteome-wide target identification, and high-throughput (HT)-CETSA for drug hit discovery and lead optimization. Importantly, the application possibilities of a variety of TPP approaches for the target discovery of bioactive NPs are highlighted and discussed, including TPP-temperature range (TPP-TR), TPP-compound concentration range (TPP-CCR), two-dimensional TPP (2D-TPP), cell surface-TPP (CS-TPP), simplified TPP (STPP), thermal stability shift-based fluorescence difference in 2D gel electrophoresis (TS-FITGE) and precipitate supported TPP (PSTPP). In addition, the key advantages, limitations and future outlook of CETSA strategies for NP studies are discussed. Conclusion: The accumulation of CETSA-based data can significantly accelerate the elucidation of the mechanism of action and drug lead discovery of NPs, and provide strong evidence for NP treatment against certain diseases. The CETSA strategy will certainly bring a great return far beyond the initial investment and open up more possibilities for future NP-based drug research and development.
Article
Background: Hepatocellular carcinoma (HCC) is a major cause of cancer-associated mortality worldwide. Myosin-9's role in HCC and the anti-HCC effect of the drugs targeting Myosin-9 remain poorly understood so far. Candidate antitumor agents obtained from natural products have attracted worldwide attention. Usenamine A is a novel product, which was first extracted in our laboratory from the lichen Usnea longissima. According to published reports, usenamine A exhibits good antitumor activity, while the mechanisms underlying its antitumor effects remain to be elucidated. Purpose: The present study investigated the anti-hepatoma effect of usenamine A and the underlying molecular mechanisms, along with evaluating the therapeutic potential of targeting Myosin-9 in HCC. Methods: The CCK-8, Hoechst staining, and FACS assays were conducted in the present study to investigate how usenamine A affected the growth and apoptosis of human hepatoma cells. Moreover, TEM, acridine orange staining, and immunofluorescence assay were performed to explore the induction of autophagy by usenamine A in human hepatoma cells. The usenamine A-mediated regulation of protein expression in human hepatoma cells was analyzed using immunoblotting. MS analysis, SPR assay, CETSA, and molecular modeling were performed to identify the direct target of usenamine A. Immunofluorescence assay and co-immunoprecipitation assay were conducted to determine whether usenamine A affected the interaction between Myosin-9 and the actin present in human hepatoma cells. In addition, the anti-hepatoma effect of usenamine A was investigated in vivo using a xenograft tumor model and the IHC analysis. Results: The present study initially revealed that usenamine A could suppress the proliferation of HepG2 and SK-HEP-1 cells (hepatoma cell lines). Furthermore, usenamine A induced cell apoptosis via the activation of caspase-3. In addition, usenamine A enhanced autophagy. Moreover, usenamine A administration could dramatically suppress the carcinogenic ability of HepG2 cells, as evidenced by the nude mouse xenograft tumor model. Importantly, it was initially revealed that Myosin-9 was a direct target of usenamine A. Usenamine A could block cytoskeleton remodeling through the disruption of the interaction between Myosin-9 and actin. Myosin-9 participated in suppressing proliferation while inducing apoptosis and autophagy in response to treatment with usenamine A. In addition, Myosin-9 was revealed as a potential oncogene in HCC. Conclusions: Usenamine A was initially revealed to suppress human hepatoma cells growth by interfering with the Myosin-9/actin-dependent cytoskeleton remodeling through the direct targeting of Myosin-9. Myosin-9 is, therefore, a promising candidate target for HCC treatment, while usenamine A may be utilized as a possible anti-HCC therapeutic, particularly in the treatment of HCC with aberrant Myosin-9.
Article
Background: NLR family pyrin domain-containing 3 (NLRP3)-mediated pyroptosis plays a key role in various acute and chronic inflammatory diseases. Targeted inhibition of NLRP3-mediated pyroptosis may be a potential therapeutic strategy for various inflammatory diseases. Ergolide (ERG) is a sesquiterpene lactone natural product derived from the traditional Chinese medicinal herb, Inula britannica. ERG has been shown to have anti-inflammatory and anti-cancer activities, but the target is remains unknown. Hypothesis/purpose: This study performed an in-depth investigation of the anti-inflammatory mechanism of ERG in NLRP3-mediated pyroptosis and NLPR3 inflammasome related sepsis and acute lung injury model. Methods: ELISA and Western blot were used to determine the IL-1β and P20 levels. Co-immunoprecipitation assays were used to detect the interaction between proteins. Drug affinity response target stability (DARTS) assays were used to explore the potential target of ERG. C57BL/6J mice were intraperitoneally injected with E. coli DH5α (2 × 109 CFU/mouse) to establish a sepsis model. Acute lung injury was induced by intratracheal administrationof lipopolysaccharide in wild-type mice and NLRP3 knockout mice with or without ERG treatment. Results: We showed that ERG is an efficient inhibitor of NLRP3-mediated pyroptosis in the first and second signals of NLRP3 inflammasome activation. Furthermore, we demonstrated that ERG irreversibly bound to the NACHT domain of NLRP3 to prevent the assembly and activation of the NLRP3 inflammasome. ERG remarkably improved the survival rate of wild-type septic mice. In lipopolysaccharide-induced acute lung injury model, ERG alleviated acute lung injury of wild-type mice but not NLRP3 knockout mice. Conclusion: Our results revealed that the anti-pyroptosis effect of ERG are dependent on NLRP3 and NLRP3 NACHT domain is ERG's direct target. Therefore, ERG can serve as a precursor drug for the development of novel NLRP3 inhibitors to treat NLRP3 inflammasome mediated inflammatory diseases.
Article
Background: The activation of P2Y14 receptor (P2Y14R) promotes osteoclast formation and causes neuropathic pain, exhibiting possible link to osteoarthritis (OA). Given lack of P2Y14R antagonist, the present study aims to search a novel P2Y14R antagonist with low toxicity and high activity from natural products as a possible drug candidate in treatment of OA. Methods: The role of P2Y14R on OA was verified using P2Y14R knockout (KO) rats. Molecular docking virtual screening strategy and activity test in P2Y14R stably-expressed HEK293 cells were used to screen target compound from natural product library. The MM/GBSA free energy calculation/decomposition technique was used to determine the principal interaction mechanism. Next, the binding of target compound to P2Y14R was examined using cellular thermal shift assay and drug affinity responsive target stability test. Finally, the therapeutic effect of target compound was performed in monosodium iodoacetate (MIA)-induced OA mouse model. To verify whether the effect of target compound was attributed to P2Y14R, we establish the osteoarthritis model in P2Y14R KO mice to perform pharmacodynamic evaluation. Importantly, to investigate the potential mechanism by which target compound attenuate OA, expressions of the major transcription factors involved in osteoclast differentiation were detected by western blot, while markers of nerve damage in dorsal root ganglion (DRG) were evaluated by RT-qPCR and immunofluorescence techniques. Results: Deficiency of P2Y14R alleviated pain behavior and cartilage destruction in MIA-induced OA rats. 14 natural compounds were screened by Glide docking-based virtual screening, among which paederosidic acid exhibited the highest antagonistic activity to P2Y14R with IC50 of 8.287 μM. As a bioactive component extracted from Paederia scandens, paederosidic acid directly interacted with P2Y14R to enhance the thermostability and decrease the protease sensitivity of target protein, which significantly inhibited receptor activator for nuclear factor-κB ligand (RANKL)-mediated osteoclastogenesis. More importantly, paederosidic acid suppressed osteoclast formation by downregulating expressions of NFAT2 and ATP6V0D2, as well as relieved neuropathic pain by decreasing expressions of CGRP, CSF1 and galanin in DRG. Conclusions: Paederosidic acid targeted P2Y14R to improve OA through alleviating osteoclast formation and neuropathic pain, which provided an available strategy for developing novel drug leads for treatment of OA.
Article
Ethnopharmacological relevance: Huangxiong Formula (HXF) is composed of four herbs: Rheum palmatum L., Ligusticum striatum DC., Curcuma aromatica Salisb., and Acorus gramineus Aiton. HXF is clinically used for the treatment of ischemic stroke (IS). However, its molecular mechanism remains unclear. Aim of the study: A network pharmacology-based strategy combined with experimental study in vivo and in vitro to were used to investigate the bioactive components, potential targets, and molecular mechanisms of HXF in the treatment of IS. Materials and methods: The components of HXF were detected by ultra-performance liquid chromatography (UPLC). The potential active ingredients of HXF were acquired from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) and literature, and corresponding targets were discerned through the Swiss TargetPrediction database. IS-related targets were obtained from Genecards, Online Mendelian Inheritance in Man (OMIM), Therapeutic Target Database (TTD), and DisGeNET. The intersection of ingredient and disease targets was screened, and a herbal-compound-target network was constructed. A protein-protein interaction (PPI) network was created, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed. Based on these analyses, we established a compound-target-pathway (C-T-P) network. A cerebral ischemia-reperfusion (I/R) animal model was established, and the cerebral protective effect of HXF was assessed. The accuracy of the predicted targets was verified by real-time quantitative polymerase chain reaction (RT-qPCR). Hippocampal neuronal injury cell model induced by oxygen-glucose deprivation and reperfusion (OGD/R) was used to evaluate the protective effect of α-Asarone. Furthermore, molecular docking, drug affinity responsive target stability (DARTS) assay, and cellular thermal shift assay (CETSA) were performed to verify whether α-Asarone can bind to PI3K. Results: A total of 44 active ingredients and 795 gene targets were identified through network pharmacology. Network analysis showed that naringenin, eupatin, kaempferol, and α-Asarone were possible drug candidates. SRC, AKT1, TP53, MAPK3, STAT3, HRAS, CTNNB1, EGFR, VEGFA, PIK3R1 could serve as potential drug targets. KEGG analysis implied that the PI3K/AKT signaling pathway might play an important role in treating IS by HXF. Moreover, HXF significantly reduced neurological impairment, cerebral infarct volume, brain index, and brain histopathological damage in I/R rats. The mRNA expression of the top 10 potential targets was verified in the brain tissue. The C-T-P network and UPLC analysis suggested that α-Asarone might be an important component of HXF and can inhibit oxidative stress and apoptosis in HT22 cells by activating the PI3K/AKT signaling pathway. Molecular docking, DARTS, and CETSA assay analysis confirmed that there were direct interactions between α-Asarone and PI3K. Conclusion: HXF had a therapeutic effect in IS with multi-component, multi-target, and multi-approach features. α-Asarone, identified as one of the major active components of HXF, could alleviate oxidative stress and apoptosis by targeting PI3K/AKT pathway.
Article
Since multiple myeloma (MM) remains a cureless malignancy of plasma cells to date, it becomes imperative to develop novel drugs and therapeutic targets for MM. We screened a small molecule library comprising 3633 natural product drugs, which demonstrated that Nitidine Chloride (NC), an extract from traditional Chinese medicine Zanthoxylum nitidum. We used Surface Plasmon Resonance-High Performance Liquid Chromatography-Protein Mass Spectrometry (SPR-HPLC-MS), Cellular Thermal Shift Assay (CETSA), molecular docking, and SPR assay to identify the potential targets of NC, in which ABCB6 was the unique target of NC. The effects of ABCB6 on cellular proliferation and drug resistance were determined by CCK8, western blot, flow cytometry, site-mutation cells, transmission electron microscopy, immunohistochemistry staining and xenograft model in vitro and in vivo. NC induced MM cell death by promoting ferroptosis. ABCB6 is the direct target of NC. ABCB6 expression was increased in MM samples compared to normal controls, which was significantly associated with MM relapse and poor outcomes. VGSK was the inferred binding epitope of NC on the ABCB6 protein. In the ABCB6-mutated MM cells, NC did not display cancer resistance, implying the vital role of ABCB6 in NC's bioactivity. Moreover, the silencing of ABCB6 significantly inhibited MM cell growth. Mechanistically, the direct binding of NC to ABCB6 suppressed PI3K/AKT signaling pathway to promote ferroptosis. In conclusion, ABCB6 can be a potential therapeutic target and prognostic biomarker in MM, while NC can be considered a novel drug for MM treatment.