1681
ISSN 0006-2979, Biochemistry (Moscow), 2024, Vol. 89, No. 10, pp. 1681-1691 © Pleiades Publishing, Ltd., 2024.
ISSN 0006-2979, Biochemistry (Moscow), 2024. © Pleiades Publishing, Ltd., 2024.
Computational Assessment of Carotenoids
as Keap1-Nrf2 Protein–Protein Interaction Inhibitors:
Implications for Antioxidant Strategies
Alessandro Medoro
1
, Tassadaq Hussain Jafar
1
, Fabio Sallustio
2
,
Giovanni Scapagnini
1
, Luciano Saso
3
, and Sergio Davinelli
1,a
*
1
Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
2
Department of Precision and Regenerative Medicine and Ionian Area (DIMEPRE-J), University of Bari, Bari, Italy
3
Department of Physiology and Pharmacology “Vittorio Erspamer”, La Sapienza University, Rome, 00185, Italy
a
e-mail: sergio.davinelli@unimol.it
Received May 8, 2024
Revised July 21, 2024
Accepted July 23, 2024
AbstractThe Keap1-Nrf2 pathway is an essential system that maintains redox homeostasis and modulates key
metabolic processes, including metabolism of amino acids to promote the synthesis of antioxidant enzymes.
Inhibitors of the protein-protein interaction (PPI) between Keap1 and Nrf2 have emerged as a promising strategy
for developing novel classes of antioxidant agents that selectively activate this pathway without off-target effects.
Carotenoids, a large family of lipophilic isoprenoids synthesized by all photosynthetic organisms, are well-known
for their antioxidant activities. However, the ability of carotenoids to inhibit the Keap1-Nrf2 PPI through the
involvement of specific amino acid residues has not yet been revealed. We utilized molecular docking, molecular
dynamic simulations, and pharmacokinetic prediction techniques to investigate the potential of eight oxygenat-
ed carotenoids, known as xanthophylls, to inhibit Keap1. Among the compounds investigated, fucoxanthin and
astaxanthin established multiple hydrogen-bonding and hydrophobic interactions within the Kelch domain of
Keap1, showing remarkable binding affinities. Furthermore, fucoxanthin and astaxanthin displayed the ability
toform astable complex with Keap1 and fit into the binding pocket of its Kelch domain. These analyses led to the
identification of critical amino acid residues in the binding pocket of Keap1 which are involved in the interac-
tion with carotenoid xanthophylls. Our analyses further revealed that fucoxanthin and astaxanthin demonstrate
a favorable safety profile and possess pharmacokinetic properties consistent with acceptable drug-like character-
istics. These findings lay the preliminary foundation for developing a novel class of Keap1-Nrf2 PPI inhibitors with
potential applications against oxidative stress-related diseases.
DOI: 10.1134/S0006297924100031
Keywords: Keap1, Nrf2, carotenoids, xanthophylls, antioxidants
* To whom correspondence should be addressed.
INTRODUCTION
As maintaining redox homeostasis is a continu-
ous challenge, multicellular organisms have evolved
various defense systems to mitigate the toxic effects
of numerous oxidants and electrophiles to which they
are exposed. A prominent aspect of oxidative stress
is the induction of a cellular stress response, mediat-
ed by molecular redox switches. These switches are
crucial in the activation of an extensive network of an-
tioxidant enzymes. Accordingly, this activation coun-
teracts the oxidative challenge, thereby preserving
redox homeostasis [1, 2]. The Nrf2 (nuclear factor
erythroid 2 [NF-E2]-related factor 2)-Keap1 (Kelch-like
ECH-associated protein 1) pathway is one of the ma-
jor regulators of oxidative and electrophilic stress re-
sponse, playing a pivotal role in the maintenance of
the cellular redox homeostasis. Nrf2, ubiquitously ex-
pressed across various organs, is a cap’n’collar (CNC)
basic region-leucine zipper (bZIP) transcription factor.
MEDORO et al.1682
BIOCHEMISTRY (Moscow)
It regulates theexpression levels of hundreds of genes
associated with homeostatic functions and oxidative
stress response. This regulation occurs through the
binding of Nrf2 to a DNA enhancer region known as
the antioxidant response element (ARE). Furthermore,
Nrf2 is involved in amino acid metabolism, promoting
the synthesis of antioxidant enzymes and increasing
the availability of amino acids such as cysteine, ser-
ine, glycine, and asparagine. In humans, Nrf2 consists
of 605 amino acids and includes seven distinct and
highly conserved Nrf2-ECH homology (Neh) domains
(Neh1–Neh7) [3-5].
Although the activity of Nrf2 is regulated by vari-
ous mechanisms, including at transcriptional and epi-
genetic levels, its proteasomal degradation is mainly
mediated by the repressor Keap1. Under basal condi-
tions, Nrf2 protein levels remain low due to its inter-
action with Keap1 in the cytoplasm. This interaction
involves the Neh2 domain of Nrf2, which binds to
Keap1 using the DLG and ETGE motifs. This binding is
essential for the ubiquitination and subsequent prote-
asomal degradation of Nrf2. Briefly, Keap1 recruits the
Cullin 3 (Cul3)-RING-box protein 1 (RBX1) E3 ubiquitin
ligase complex with subsequent ubiquitination of Nrf2
by the 26S proteasome [6]. Human Keap1 contains 627
amino acids and consists of five distinct domains: the
N-terminal region (NTR), the broad complex, tramtrack
and bric-a-brac (BTB) domain, the intervening region
(IVR), double glycine repeats (DGR) or Kelch domain,
and the C-terminal region (CTR). The BTB domain fa-
cilitates dimerization and interaction with Cul3, while
the Kelch domain, binds to Neh2 of Nrf2 [7]. Moreover,
Keap1 contains several highly reactive Cys residues
in these domains, particularly within the IVR domain,
that act as cellular redox status sensors. The most com-
mon electrophilic Nrf2 inducers trigger conformation-
al changes in Keap1 due to modifications of its cysteine
residues, disrupting the Keap1-Nrf2 protein–protein
interaction (PPI) and leading to the nuclear accumula-
tion of Nrf2. This, in turn, activates a cellular defense
response against oxidative stress [8,9]. However, since
the mechanism of action of these compounds involves
the covalent targeting of cysteine thiols, Nrf2 inducers
may lack selectivity for Keap1, potentially affecting
other cysteines within the cells and leading to unpre-
dictable off-target side effects [10].
Currently, alternative strategies to target Nrf2
with higher selectivity have attracted increasing at-
tention. These alternative approaches focus on devel-
oping Keap1–Nrf2 PPI inhibitors to block their interac-
tion, thereby increasing Nrf2 activity and potentially
improving the safety and efficacy of treatments for
oxidative stress-related diseases. To date, several non-
covalent small-molecule Keap1-Nrf2 inhibitors have
been described [11]. These Keap1-Nrf2 PPI inhibitors
typically bind to the Kelch domain of Keap1, target-
ing the sites where the ETGE and DLG motifs of Nrf2
would normally bind. The Keap1‐Kelch domain, a
six‐bladed β‐propeller structure, is divided into six
subpockets (P1-P6). These subpockets accommodate
different amino acid residues that interact with DLG
and ETGE motifs located in the Neh2 domain of Nrf2.
Therefore, the higher selectivity of Keap1-Nrf2 PPI
inhibitors is determined by interactions between the
DLG and ETGE motifs in the Neh2 domain of Nrf2 and
the corresponding subpockets within the Keap1-Kelch
domain [12,13]. This mechanism of action selectively
inhibits the binding between Keap1 and Nrf2, thereby
enhancing Nrf2 activity and ensuring a more targeted
modulation of the pathway.
Carotenoids, which are terpenoid-based com-
pounds, are widely distributed in algae, fungi, bacteria,
and plants. They constitute a large family of fat-solu-
ble plant pigments known for their health-promoting
effects against major chronic diseases, including dia-
betes, cancer, and dementia [14-16]. Carotenoids are
typically classified into two groups, carotenes, and
xanthophylls, based on chemical and biochemical cri-
teria. Xanthophylls, such as fucoxanthin, astaxanthin,
lutein, zeaxanthin, and canthaxanthin, which are rich
in double bonds, exhibit higher antioxidant properties
than carotenes [17, 18]. Several experimental studies
have shown that marine-derived xanthophylls, such
as astaxanthin, zeaxanthin, and lutein, can activate
the Nrf2 pathway and induce the expression of anti-
oxidant enzymes, including glutathione (GSH), heme
oxygenase-1 (HO-1), and superoxide dismutase (SOD)
[19-21]. However, there is limited evidence suggesting
that these xanthophylls may disrupt the Keap1-Nrf2
PPI by interacting with Keap1, leading to the release
and activation of Nrf2 [22]. We selected from the
CHEMnetBASE – Dictionary of Marine Natural Prod-
ucts eight microalgal xanthophylls extensively studied
for their antioxidant characteristics (astaxanthin, can-
thaxanthin, echinenone, fucoxanthin, lutein, neoxan-
thin, violaxanthin, and zeaxanthin) [23-32]. We used
computational methods, including molecular docking
and molecular dynamic simulations (MDS), to inves-
tigate whether these xanthophyll carotenoids might
serve as potential candidates for developing selective
Keap1-Nrf2 PPI inhibitors.
MATERIALS AND METHODS
3D structural retrieval and preparation of the
Keap1 protein. Keap1 protein (2FLU) was retrieved
from the RCSB Protein Data Bank (https://www.rcsb.org/
structure/2FLU). In detail, it is the crystal structure of
the Kelch domain of Keap1 bound to a 16-mer pep-
tide from Nrf2 containing a highly conserved DxETGE
motif [33]. After conducting a 3D structural retrieval,
CAROTENOIDS AS KEAP1-NRF2 PPI INHIBITORS 1683
BIOCHEMISTRY (Moscow)
the protein underwent preparation using UCSF Chimera
software (v1.12) [34]. The retrieved structure was
checked for missing residues and refined removing
water molecules, additional protein chains, and li-
gands. To ensure protein stability, energy minimi-
zation, and geometry optimization were carried out
using UCSF Chimera with 1000 steps (step size 0.02 Å)
and the conjugate gradient method. The protonation
of wild-type histidine was done following the AMBER
ff98 method [35, 36]. Energy minimization was also
performed to maintain the stability of Keap1 using the
Chiron energy minimization online tool [37].
Xanthophyll carotenoids selection and struc-
ture preparation. A total of eight marine-derived
xanthophyll carotenoids were selected from the
CHEMnetBASE– Dictionary of Marine Natural Products
(https://dmnp.chemnetbase.com/chemical/Chemical
Search.xhtml?dswid=-7750) for the study: astaxanthin,
canthaxanthin, echinenone, fucoxanthin, lutein, neox-
anthin, violaxanthin, and zeaxanthin. The two-dimen-
sional (2D) chemical structures of the compounds were
searched in the PubChem database [38]. In addition,
2D chemical structures were converted into a .pdb for-
mat by Chem3D Pro, and the structural optimization
was performed by UCSF Chimera software (v1.12).
Molecular docking studies. AutoDock Vina soft-
ware was used for molecular docking [39]. The grid
size was set at 75×75×75 Å in the X, Y, and Z axes, with a
grid spacing of 0.650 Å covering Keap1 active domains.
Polar hydrogen atoms were added to the Keap1 pro-
tein structure, and each docking experiment consist-
ed of 100 runs. Default parameters for van der Waals
forces, electrostatic forces, AMBER force field, and in-
termolecular forces were used for the ligand-protein
complex. The genetic algorithm was employed as the
primary search protocol. All docked complexes were
analyzed, and the best-docked complex based on in-
termolecular interactions was visualized and analyzed
using Discovery Studio.
Molecular dynamics simulations (MDS). The
NAMD2 simulation software (version 2.14) was used
to conduct Molecular Dynamics Simulations (MDS) of
docked complexes for 120 ns in a water environment
[40]. The simulations utilized the Amber ff14SB force
field for proteins and a General Amber Force Field
(GAFF) for ligands [41, 42]. The antechamber package
generated topology and coordinate files for the li-
gands, while Xleap prepared the simulation system
[43]. The system was solvated in a cubic box with
TIP3P water molecules, and the protein was positioned
within 1 nm of the box edge to adhere to the mini-
mum image convention. Neutralization was achieved
by adding 10 Na
+
ions to the system. The complexes of
astaxanthin and fucoxanthin contained a total of 41363
and 41385 atoms, respectively, and were protonated
at a pH of 7.4. Energy minimization was performed
with the conjugate gradient method to eliminate ste-
ric clashes. The equilibration process involved settling
water molecules, gradual heating, and equilibration
in an NPT ensemble. The production run maintained
a constant temperature of 310.15 K and pressure of
1 atm with weak coupling using a Langevin thermostat
and barostat. The time step used is 0.2 fs. Bond lengths
involving hydrogen atoms were constrained using the
SHAKE algorithm, and Particle Mesh Ewald (PME) was
employed for electrostatic interactions beyond a cutoff
distance.
Prediction of drug-likeness, pharmacokinetics,
and toxicity. To estimate the physicochemical proper-
ties, absorption, distribution, metabolism, excretion,
toxicity (ADMET) profile, pharmacodynamic and drug-
likeness parameters of all the eight xanthophyll ca-
rotenoids, the mCule server, SwissADME, AdmetSAR,
Molsoft, and the ProTox-II server were used as previ-
ously described [44].
RESULTS AND DISCUSSION
Molecular docking and interaction analyses.
The molecular docking studies showed the best-fitted
conformational binding pose of the tested xanthophyll
carotenoids (astaxanthin, canthaxanthin, echinenone,
fucoxanthin, lutein, neoxanthin, violaxanthin, and zea-
xanthin) within Keap1. The identified binding pocket
is located in the Kelch domain of Keap1, in the shared
binding region with Nrf2. This domain is a β-propeller
structure composed of six blades, each consisting of a
four-stranded antiparallel β-sheet connected by loops.
This structure serves as the binding pocket for the
Nrf2 ETGE and DLG motifs and is divided into six sub-
pockets (P1-P6) based on interactions observed in co-
crystal structures of Keap1 with these motifs. Subpock-
ets P1 and P2 are polar, containing residues such as
Ser363, Arg380, Asn382, Asn414, Arg415, Ile461, Gly462,
Phe478, Arg483, and Ser508. In contrast, P4 and P5 are
hydrophobic, with residues Tyr525, Gln530 in P4, and
Tyr334, Tyr572, Phe577 in P5. P3, located at the center
of the channel formed by small polar residues, consists
of Gly509, Ser555, Ala556, Gly571, Ser602, and Gly603.
Finally, P6 includes residues Asp389, Ser431, His432,
Gly433, Cys434, Ile435, and His436, which interact with
the DLGex peptide but not with the ETGE motif [11].
Moreover, Nrf2-ETGE-motif-containing peptides, when
co-crystallized with the Keap1-Kelch domain, adopt a
β-hairpin structure within the binding site. This site
includes residues Asp77, Glu78, Glu79, Thr80, Gly81,
and Glu82. This β-hairpin conformation is stabilized
by a network of intramolecular hydrogen bonds in-
volving the backbone atoms of residues Gln75, Asp77,
Asp79, Thr80, Glu82, Leu89, and the sidechain atoms
of residues Asp77 and Thr80 [33, 45].
MEDORO et al.1684
BIOCHEMISTRY (Moscow)
Table 1. Binding affinities calculation of astaxanthin,
canthaxanthin, echinenone, fucoxanthin, lutein, neox-
anthin, violaxanthin, and zeaxanthin
Compounds Binding Affinities (Kcal/mol)
Astaxanthin –9.0
Canthaxanthin –8.1
Echinenone –8.8
Fucoxanthin –9.5
Lutein –8.6
Neoxanthin –8.9
Violaxanthin –8.5
Zeaxanthin –7.1
The molecular docking assessments of the eight
xanthophyll carotenoids revealed variations in the re-
corded binding energies, with the docked complex ex-
hibiting the lowest binding energy being chosen from
all the docking complexes created for each compound,
as detailed in Table 1. The generated docked complex-
es were evaluated based on docking energy scores and
the number of hydrogen bonds and hydrophobic inter-
actions. Fucoxanthin and astaxanthin (Fig. 1) showed
the lowest binding affinity scores (–9.5 to –9.0 kcal/mol,
respectively), while those of canthaxanthin, echinen-
one, lutein, neoxanthin, violaxanthin, and zeaxanthin,
were lower than –7.0 kcal/mol (ranging from –7.1 to
–8.9 kcal/mol), although good values that predicted a
stable binding.
All intermolecular interactions, including hydro-
gen bonds, and hydrophobic and electrostatic inter-
actions, contribute to the stability of the docked com-
plex and lower binding affinity score. In particular,
hydrogen bonds and hydrophobic interactions play a
significant role in ligand-receptor stability. Notably, fu-
coxanthin and astaxanthin, the two compounds with
the lowest binding scores, were firmly bound, predom-
inantly in a planar manner, in the Kelch domains with
potential hydrogen bonding interactions, as shown
in Figs. 2-3 and Table 2. In detail, in the docking com-
plex between fucoxanthin-Keap1, Arg326, Arg415, and
Arg416 were the three residues involved in hydrogen
bonding. Cys368, Val370, Val418, Val420, and Val467
are involved in hydrophobic interactions, contributing
to the fucoxanthin stability in the binding pocket of
Keap1 (Fig. 3, Table 2). Several potential residues iden-
tified in this docking analysis are already reported as
crucial for Keap1–Nrf2 interaction and the PPI inhib-
itors, including Arg415, Gly509, Ala556, and Gly603,
suggesting, due to the peculiar structure of this mole-
cule, interaction with polar and hydrophobic subpock-
ets [11]. These identified key residues for the fucoxan-
thin–Keap1 complex also showed good overlap with
8 previously reported potential residues, including
Arg415, Val465, Val512, and Ala556 [22]. In this study,
Wu et al. reported that fucoxanthin, extracted from
seaweeds, exhibits a neuroprotective effect by the po-
tential inhibition of the Keap1–Nrf2 complex. Biolayer
interferometry (BLI) assays showed that fucoxanthin
binds to Keap1 with a potential dissociation constant
(Kd) of 51.6 µM, confirming its potential as Keap1-Nrf2
PPI and inducer of Nrf2 nuclear translocation and acti-
vation of the antioxidant response element (ARE). Sim-
ilar to fucoxanthin, it was observed that astaxanthin
also showed a strong binding interaction with residues
in the Kelch domains of Keap1, including key residues
such as Arg415, Gly509, Ala556, and Gly603 (Table 2),
with one hydrogen bond observed at Arg415 and three
hydrophobic interactions with Val467, Val512, and
Cys513 (Fig. 3). For the neoxanthin–Keap1 complex,
Fig. 1. 2D chemical structures of (a) fucoxanthin, and (b) astaxanthin.
CAROTENOIDS AS KEAP1-NRF2 PPI INHIBITORS 1685
BIOCHEMISTRY (Moscow)
Table 2. Molecular interactions of astaxanthin and fucoxanthin with Keap1
Docked complexes Interactive residues of docking studies
Astaxanthin–Keap1
Gly364, Leu365, Ala366, Gly367, Cy368, Arg415, Ile416, Gly417, Val418, Gly419, Val420,
Asp422, Gly423, Gly462, Val463, Gly464, Val465, Ala466, Val467, Gly509, Ala510, Gly511,
Val512, Cys513, Leu557, Ile559, Gly603, Val604
Fucoxanthin–Keap1
Gly364, Leu365, Ala366, Gly367, Cys368, Val369, Val370, Gly371, Gly372, Arg415, Ile416,
Gly417, Val418, Gly419, Val420, Ile421, Asp422, Gly423, Val463, Gly464, Val467, Gly509,
Ala510, Gly511, Ala556, Leu557, Gly558, Ile559, Gly603, Val604, Gly605
Note. Bold residues indicate residues that are involved in potential hydrogen bonding.
a unique hydrogen bond with the residue Ile416 was
identified. No hydrogen bonding interactions were
reported in the docking complexes of canthaxanthin–
Keap1, echinenone–Keap1, lutein–Keap1, violaxanthin–
Keap1, and zeaxanthin–Keap1, although in the dock-
ing complex of echinenone–Keap1 some hydrophobic
interactions were detected (Fig. S1 in the Online Re-
source 1).
Moreover, studied carotenoids may bind many
Keap1 residues shared with Nrf2, including Arg415,
Gly509, Ala556, and Gly603, which are all residues lo-
cated in P1-P3 subpockets. Overall, these data may of-
fer further interpretation of the antioxidant activity
of xanthophyll carotenoids. Carotenoids are known to
be efficient antioxidants that directly scavenge singlet
molecular oxygen and peroxyl radicals, contributing
to the antioxidant defense system [46]. However, there
is multiple experimental evidence that xanthophyll
carotenoids, particularly fucoxanthin, and astaxan-
thin, may inhibit Nrf2 degradation and increase Nrf2-
dependent activity [20, 47-49]. Overall, our findings
provide evidence for a new potential mechanism by
which carotenoids may exert their antioxidant activi-
ty through the Nrf2 pathway. Among the carotenoids
analyzed in this study, astaxanthin and fucoxanthin
demonstrated potentially higher binding stability
with Keap1, as indicated by their binding energies
and the greater number of identified hydrogen bonds
and hydrophobic interactions.
MDS studies. To gain further insights into the
study of the interaction of the two most potential
xanthophyll carotenoids, fucoxanthin and astaxan-
thin, with Keap1, MDS studies were performed. MDS
is a method for analyzing the physical movements of
atoms and molecules, providing information on con-
formational changes in docking complexes. As ex-
pected, some differences in docking interactions of
protein- ligand complexes between AutoDock Vina and
MDS analyses were detected, considering that MDS
offers a dynamic view of the protein-ligand complex,
Fig. 2. 3D molecular representation of fucoxanthin (green) and astaxanthin (cyan) docking complexes with Keap1.
MEDORO et al.1686
BIOCHEMISTRY (Moscow)
Fig. 3. 2D molecular interaction analyses of (a)fucoxanthin, and (b)astaxanthin with Keap1.
aswell as revealed comprehensive and more realistic
molecular interactions during the simulation system
(Fig. S2 in the Online Resource 1). Overall, fucoxan-
thin and astaxanthin showed significant potential in
the binding of the Kelch domains of Keap1, generat-
ing a very stable and tight-docked complex, as shown
by the data below.
Root Mean Square Deviation (RMSD) is a measure
used to quantify the structural analysis of the docking
complex. In Fig. 4 RMSD values indicated the average
deviation for both ligands (astaxanthin and fucoxan-
thin) and protein. Both fucoxanthin and astaxanthin
simulation complexes with Keap1 showed that the
structure remained stable throughout the simulation
time with some fluctuation within the range of ~1 Å,
which is an expected behavior. Therefore, the RMSD
values (1.54 ± 0.08 Å in complex with astaxanthin and
1.67 ± 0.14 Å with fucoxanthin) indicate that the Keap1
underwent small local conformational changes with
apparently no change in its folding and stability
(Fig. 4a). Astaxanthin and fucoxanthin showed high
RMSD values (2.44 ± 0.47 Å and 3.39 ± 0.35 Å, respec-
tively), suggesting conformational changes. Fucoxan-
thin exhibited higher variation in ligand conformation,
as indicated by its higher RMSD value, and suggests at
least two different clusters for ligand conformations.
The ligand’s RMSD plot immediately reached a pla-
teau, indicating stability in conformation. Astaxanthin
at least exhibited two ensembles of ligands during sim-
ulation, which transformed into each other back and
forth (Fig.4b).
In addition, Root Mean Square Fluctuation (RMSF)
was used to estimate the binding orientations of the
ligands and structural alternations of the residues
that linked to targeted protein at a specific tempera-
ture and pressure. Fluctuations in the protein residues
CAROTENOIDS AS KEAP1-NRF2 PPI INHIBITORS 1687
BIOCHEMISTRY (Moscow)
Fig. 4. a, b) RMSD detailed calculation analyses of astaxanthin (black line), and fucoxanthin (red line) complexes with Keap1
during 120ns MDS.
ofKeap1 and both changes in the ligand atom position
of astaxanthin and fucoxanthin were calculated from
the 120 ns trajectory. These RMSF studies endorsed
that both complexes in our simulations exhibited
≤2.0 Å protein RMSF values indicating firmly bound li-
gands with great protein stability and rigidity in the
structures (Fig.S3A in the Online Resource1).
Both complexes showed a very stable radius of
gyration with a fluctuation of 1 Å, which indicates the
stability and the compactness of the 3D protein struc-
ture. The slight fluctuation within the 1 Å radius-of-
gyration value during the MDS time indicated a slight
opening and closing of the N- and C-terminal domains
(Fig. S3B in the Online Resource1). Notably, these were
very small fluctuations in the normal range of radi-
us-of-gyration, which indicates that the protein–ligand
system remains compact during molecular dynamics
simulation and that no unfolding event is observed.
In detail, the radius of gyration of astaxanthin and fu-
coxanthin indicated no change in the protein compact-
ness, remaining highly compact with consistent values
and smaller standard deviations (18.09 ± 0.07 Å and
18.06 ± 0.06 Å, respectively).
To deepen the molecular interactions between
Keap1 active site residues and astaxanthin and fucox-
anthin, the free energy of the docked complexes was
calculated using the MM/GBSA method. Molecular in-
teractions between Keap1 binding sites residues and
astaxanthin, and fucoxanthin after every 1 ns (120
frames in total) portrayed highly favorable electrostatic
energies, and polar solvation-free energies, and showed
that van der Waals forces were highly favorable inter-
actions contributing to the stability of the molecular
docking systems (Fig. 5). The overall least binding en-
ergies obtained for astaxanthin (–46.84 ± 7.39 kcal/mol)
and fucoxanthin (–52.66 ± 4.77 kcal/mol) docking com-
Fig. 5. MM/GBSA protein–ligand binding energy calculated for astaxanthin (black line), and fucoxanthin (red line) after every
1ns (120 frames in total).
MEDORO et al.1688
BIOCHEMISTRY (Moscow)
Table 3. Drug-likeness analyses of astaxanthin, canthaxanthin, echinenone, fucoxanthin, lutein, neoxanthin,
violaxanthin, and zeaxanthin
Compounds RO5 Veber Rule Drug-likeness Bioavailability
Astaxanthin no yes –0.13 0.17
Canthaxanthin no yes 0.08 0.17
Echinenone no yes 0.19 0.17
Fucoxanthin no no –0.35 0.17
Lutein no yes –0.33 0.17
Neoxanthin no yes –0.78 0.17
Violaxanthin no yes –1.49 0.17
Zeaxanthin no yes –0.18 0.17
plexes represented binding flexibility. Although these
findings showed that both ligands remained in bound
form throughout the simulation time with very good
binding energy, fucoxanthin may bind with slightly
more favorable energy to Keap1 with respect to astax-
anthin. Moreover, ligand-protein hydrogen bonds play
a significant role in determining the binding effica-
cy of a ligand on the binding pockets of the protein.
Thetotal number of hydrogen bonds formed between
ligand and protein is shown in Fig.S4 in the Online Re-
source 1. Astaxanthin and fucoxanthin, during 120 ns
simulation time, maintained almost two hydrogen
bonds formed at the interface of protein at most points
of the simulation time.
Drug-likeness, pharmacokinetics, and toxicity
analyses. All compounds investigated in this study
were subjected to drug-likeness, pharmacodynamic,
and toxicity profiling to assess their potential safety,
with overall very good results for all the xanthophylls
tested. Table S1 in the Online Resource 1 shows the
cheminformatics analysis of the compounds. The mo-
lecular weight (g/mol) of all compounds was also com-
parable with the standard value (<5000 g/mol). The
SwissADME analyses presented in Table 3 show that
all the analyzed carotenoids have more than one viola-
tion in RO5, the five rule-based filters allow prediction
of whether a molecule is defined as an orally active
drug. However, as shown in Table 3 and Table S1 in
the Online Resource 1, these carotenoids, except fu-
coxanthin, met the requirements of drug-likeness of
Veber filters. Moreover, also drug-likeness scores of
all the compounds calculated using MolSoft and syn-
thetic bioavailability scores calculated by SwissADME
were in contrast to the RO5 results. Fucoxanthin and
astaxanthin displayed good drug-likeness scores (–0.35
and –0.13) and bioavailability scores within the range
of >10%. All the drug-likeness scores of the other
carotenoids were comprised between 0.08 and –1.49
(Table3 and TableS1 in the Online Resource1).
All compounds were predicted to be absorbed by
the human intestine and displayed promising results
for the blood brain barrier (BBB) permeability. More-
over, non-toxic and non-carcinogenic behaviors were
observed for all the xanthophyll carotenoids. Accord-
ing to the predicted results, overall, these compounds
showed lower toxicity (hepatotoxicity, immunotoxic-
ity, mutagenicity, and cytotoxicity), and carcinogenic-
ity. The overall predicted results justified their good
ADMET behavior (Table S2 in the Online Resource 1).
According to the predicted ADMET analysis, some of
the compounds had a very high lethal dose50(LD
50
),
such as astaxanthin (4600 mg/kg), canthaxanthin
(10,000 mg/kg), and echinenone (10,000 mg/kg), which
were classified as non-toxic for acute oral toxicity.
On the other side, because of their lower LD
50
, fu-
coxanthin (130 mg/kg), lutein (10 mg/kg), neoxanthin
(50 mg/kg), violaxanthin (55 mg/kg), and zeaxanthin
(not available) were classified as harmful if swallowed
(50 < LD
50
≤ 300) or fatal if swallowed (5 < LD
50
≤ 50).
In the toxicity analyses, astaxanthin confirmed its
non-toxic potential, showing no hepatotoxicity, car-
cinogenicity, mutagenicity, or cytotoxicity. On the oth-
er hand, fucoxanthin showed carcinogenicity and im-
munotoxicity potential with more probability.
In addition, all compounds were screened with
ProTox-II to predict whether they could interfere with
various biological pathways. The results revealed that
fucoxanthin and astaxanthin were inactive against all
the targeted biological pathways. In detail, astaxan-
thin, echinenone, fucoxanthin, neoxanthin, and vio-
laxanthin showed no potential activity against nucle-
ar receptor pathways and stress response pathways,
including aryl hydrocarbon receptor (Ahr), andro-
gen receptor (AR), androgen receptor ligand binding
CAROTENOIDS AS KEAP1-NRF2 PPI INHIBITORS 1689
BIOCHEMISTRY (Moscow)
domain (AR-LBD), estrogen receptor alpha (ER), Estro-
gen receptor ligand binding domain (ER-LBD), peroxi-
some proliferator-activated receptor gamma (PPAR-γ),
and p53. Canthaxanthin could interfere with ER and
ER-LBD, and lutein and zeaxanthin with p53 (TableS3
in the Online Resource1).
CONCLUSIONS
Keap1, a highly redox-sensitive member of the
BTB-Kelch substrate adaptor protein family, is known
to mediate the ubiquitination of Nrf2, a master regu-
lator of the antioxidant response. This study employs a
computational approach to explore the potential of ox-
ygenated carotenoids, known as xanthophylls, as can-
didates for inhibiting the Keap1-Nrf2 PPI. Recently, in-
hibitors of the Keap1-Nrf2 PPI have gained increasing
attention as a means to selectively target Nrf2 without
off-target effects, leading to the development of novel
classes of potential preventive and therapeutic agents
for a variety of diseases characterized by oxidative
stress. Although the reliability of in silico predictions
greatly depends on the accuracy of molecular dock-
ing simulations and scoring functions, computational
techniques remain crucial for accelerating the screen-
ing of Keap1-Nrf2 PPI inhibitors and diversifying their
chemical scaffolds. In this study, three-dimensional
structural information, molecular docking screening,
and interaction analysis revealed the structural deter-
minants underpinning the binding process between
carotenoid xanthophylls, especially fucoxanthin and
astaxanthin, and Keap1. These analyses led to the iden-
tification of critical amino acid residues in the binding
pocket of Keap1 which are involved in the interaction
with carotenoid xanthophylls. However, fucoxanthin
and astaxanthin exhibited a stronger binding interac-
tion with residues in the Kelch domains of Keap1 than
other carotenoid xanthophylls. We then performed
MDS studies to elucidate the dynamic behavior and
stability of the interactions between fucoxanthin, as-
taxanthin, and Keap1. Through a 120 ns simulation
trajectory, the results offered insights into the tempo-
ral evolution of protein–ligand complexes, deciphering
the forces underlying their associations. Notably, fu-
coxanthin and astaxanthin formed a highly stable and
tightly docked complex throughout the simulation pe-
riod, establishing multiple hydrogen bonds with Keap1
and characterized by very good binding energy. More-
over, favorable pharmacokinetic and safety profiles
have demonstrated the drug-like properties of these
compounds, enhancing their potential as Keap1-Nrf2
PPI inhibitors. Overall, this study provides theoretical
support that could stimulate further research into an-
tioxidant strategies involving carotenoid xanthophylls,
particularly focusing on their potential to inhibit the
Keap1-Nrf2 PPI in experimental models of oxidative
stress.
Supplementary information. The online version
contains supplementary material available at https://
doi.org/10.1134/S0006297924100031.
Contributions. Conceptualization, A.M., S.D., and
G.S.; methodology, A.M. and S.D.; formal analysis, A.M.
and T.H.J.; writing – original draft preparation, A.M.,
T.H.J., and F.S.; writing– review and editing, G.S., L.S.,
and S.D.
Funding. This work was supported by ongoing in-
stitutional funding. No additional grants to carry out
or direct this particular research were obtained.
Ethics declarations. This work does not contain
any studies involving human and animal subjects.
Theauthors of this work declare that they have nocon-
flicts of interest.
REFERENCES
1. Go, Y.M., and Jones, D.P. (2013) The redox proteome,
J. Biol. Chem., 288, 26512, https://doi.org/10.1074/
jbc.r113.464131.
2. Brigelius-Flohé, R., and Flohé, L. (2011) Basic prin-
ciples and emerging concepts in the redox control
of transcription factors, Antioxid. Redox Signal., 15,
2335-2381, https://doi.org/10.1089/ars.2010.3534.
3. Hybertson, B.M., Gao,B., Bose, S.K., and McCord, J.M.
(2011) Oxidative stress in health and disease: the ther-
apeutic potential of Nrf2 activation, Mol. Aspects Med.,
32, 234-246, https://doi.org/10.1016/j.mam.2011.10.006.
4. Medoro,A., Saso,L., Scapagnini,G., and Davinelli,S.
(2023) NRF2 signaling pathway and telomere length
in aging and age-related diseases, Mol. Cell. Biochem.,
https://doi.org/10.1007/s11010-023-04878-x.
5. Malhotra, D., Portales-Casamar, E., Singh, A., Sri-
vastava, S., Arenillas, D., Happel, C., Shyr, C., Wak-
abayashi, N., Kensler, T. W., Wasserman, W. W., and
Biswal, S. (2010) Global mapping of binding sites
for Nrf2 identifies novel targets in cell survival re-
sponse through chip-seq profiling and network analy-
sis, Nucleic Acids Res., 38, 5718-5734, https://doi.org/
10.1093/nar/gkq212.
6. Baird, L., and Yamamoto, M. (2020) The molecular
mechanisms regulating the KEAP1-NRF2 pathway,
Mol. Cell. Biol., 40, e00099-20, https://doi.org/10.1128/
mcb.00099-20.
7. Canning, P., Sorrell, F. J., and Bullock, A. N. (2015)
Structural basis of Keap1 interactions with Nrf2, Free
Radic. Biol. Med., 88, 101-107, https://doi.org/10.1016/
j.freeradbiomed.2015.05.034.
8. Davinelli,S., Medoro,A., Intrieri,M., Saso,L., Scapag-
nini,G., and Kang, J.X. (2022) Targeting NRF2-KEAP1
axis by Omega-3 fatty acids and their derivatives:
MEDORO et al.1690
BIOCHEMISTRY (Moscow)
emerging opportunities against aging and diseases,
Free Radic. Biol. Med., 193, 736-750, https://doi.org/
10.1016/j.freeradbiomed.2022.11.017.
9. Dayalan Naidu,S., and Dinkova-Kostova, A.T. (2020)
KEAP1, a cysteine-based sensor and a drug target for
the prevention and treatment of chronic disease, Open
Biol., 10, 200105, https://doi.org/10.1098/rsob.200105.
10. Lee,S., and Hu,L. (2020) Nrf2 activation through the
inhibition of Keap1-Nrf2 protein-protein interaction,
Med. Chem. Res., 29, 846-867, https://doi.org/10.1007/
s00044-020-02539-y.
11. Crisman, E., Duarte, P., Dauden, E., Cuadrado, A.,
Rodríguez-Franco, M. I., López, M. G., and León, R.
(2023) KEAP1-NRF2 protein-protein interaction inhib-
itors: design, pharmacological properties and ther-
apeutic potential, Med. Res. Rev., 43, 237-287, https://
doi.org/10.1002/med.21925.
12. Pallesen, J.S., Tran, K.T., and Bach,A. (2018) Non-co-
valent small-molecule Kelch-like ECH-associated
protein 1-nuclear factor erythroid 2-related Factor2
(Keap1-Nrf2) Inhibitors and their potential for target-
ing central nervous system diseases, J. Med. Chem.,
61, 8088-8103, https://doi.org/10.1021/acs.jmedchem.
8b00358.
13. Leung, C.H., Zhang, J.T., Yang, G.J., Liu,H., Han, Q.B.,
and Ma, D.L. (2019) Emerging screening approaches
in the development of Nrf2-Keap1 protein-protein in-
teraction inhibitors, Int.J. Mol. Sci., 20, 4445, https://
doi.org/10.3390/ijms20184445.
14. Davinelli, S., Ali, S., Solfrizzi, V., Scapagnini, G., and
Corbi, G. (2021) Carotenoids and cognitive out-
comes: a meta-analysis of randomized intervention
trials, Antioxidants, 10, 223, https://doi.org/10.3390/
antiox10020223.
15. Saini, R. K., Keum, Y. S., Daglia, M., and Rengasamy,
K. R. (2020) Dietary carotenoids in cancer chemo-
prevention and chemotherapy: a review of emerg-
ing evidence, Pharmacol. Res., 157, 104830, https://
doi.org/10.1016/j.phrs.2020.104830.
16. Medoro,A., Intrieri,M., Passarella,D., Willcox, D.C.,
Davinelli, S., and Scapagnini, G. (2024) Astaxanthin
as a metabolic regulator of glucose and lipid homeo-
stasis, J. Funct. Foods, 112, 105937, https://doi.org/
10.1016/j.jff.2023.105937.
17. Thomas, S.E., and Johnson, E.J. (2018) Xanthophylls,
Adv. Nutr., 9, 160, https://doi.org/10.1093/advances/
nmx005.
18. Pereira, A. G., Otero, P., Echave, J., Carreira-Casa-
is, A., Chamorro, F., Collazo, N., Jaboui, A., Lou-
renço-Lopes, C., Simal-Gandara, J., and Prieto, M. A.
(2021) Xanthophylls from the sea: algae as source
of bioactive carotenoids, Mar. Drugs, 19, 188, https://
doi.org/10.3390/md19040188.
19. Davinelli, S., Saso, L., D’angeli, F., Calabrese, V., In-
trieri, M., and Scapagnini, G. (2022) Astaxanthin
as a modulator of Nrf2, NF-κB, and their crosstalk:
molecular mechanisms and possible clinical appli-
cations, Molecules, 27, 502, https://doi.org/10.3390/
molecules27020502.
20. Zou,X., Gao,J., Zheng,Y., Wang,X., Chen,C., Cao,K.,
Xu,J., Li,Y., Lu,W., Liu,J., and Feng,Z. (2014) Zeaxan-
thin induces Nrf2-mediated phase II enzymes in pro-
tection of cell death, Cell Death Dis., 55, e1218, https://
doi.org/10.1038/cddis.2014.190.
21. Chang, J., Zhang, Y., Li, Y., Lu, K., Shen, Y., Guo, Y.,
Qi,Q., Wang,M., and Zhang,S. (2018) NrF2/ARE and
NF-κB pathway regulation may be the mechanism
for lutein inhibition of human breast cancer cell,
Futur. Oncol., 14, 719-726, https://doi.org/10.2217/
fon-2017-0584.
22. Wu, W., Han, H., Liu, J., Tang, M., Wu, X., Cao, X.,
Zhao,T., Lu,Y., Niu, T., Chen, J., and Chen, H. (2021)
Fucoxanthin prevents 6-OHDA-induced neurotox-
icity by targeting Keap1, Oxid. Med. Cell. Longev.,
2021, 6688708, https://doi.org/10.1155/2021/6688708.
23. Ahmed, F., Fanning, K., Netzel, M., and Schenk,
P. M. (2015) Induced carotenoid accumulation in
Dunaliella salina and Tetraselmis suecica by plant
hormones and UV-C radiation, Appl. Microbiol. Bio-
technol., 99, 9407-9416, https://doi.org/10.1007/s00253-
015-6792-x.
24. Pasquet,V., Morisset,P., Ihammouine,S., Chepied,A.,
Aumailley, L., Berard, J. B., Serive, B., Kaas, R., Lan-
neluc, I., Thiery, V., Lafferriere, M., Piot, J. M.,
Patrice,T., Cadoret, J.P., and Picot,L. (2011) Antiprolif-
erative activity of violaxanthin isolated from bioguid-
ed fractionation of Dunaliella tertiolecta extracts, Mar.
Drugs, 9, 819-831, https://doi.org/10.3390/md9050819.
25. Abe, K., Hattori, H., and Hirano, M. (2007) Accumu-
lation and antioxidant activity of secondary carot-
enoids in the aerial microalga Coelastrella striolata
var. multistriata, Food Chem., 100, 656-661, https://
doi.org/10.1016/j.foodchem.2005.10.026.
26. Xia,S., Wang,K., Wan,L., Li,A., Hu,Q., and Zhang,C.
(2013) Production, characterization, and antioxi-
dant activity of fucoxanthin from the marine diatom
Odontella aurita, Mar. Drugs, 11, 2667-2681, https://
doi.org/10.3390/md11072667.
27. Přibyl,P., Pilný,J., Cepák, V., and Kaštánek,P. (2016)
The role of light and nitrogen in growth and carot-
enoid accumulation in Scenedesmus sp., Algal Res.,
16, 69-75, https://doi.org/10.1016/j.algal.2016.02.028.
28. Neumann, U., Derwenskus, F., Flister, V. F., Schmid-
Staiger,U., Hirth,T., and Bischoff, S.C. (2019) Fucox-
anthin, A carotenoid derived from phaeodactylum
tricornutum exerts antiproliferative and antioxidant
activities in vitro, Antioxidants, 8, 183, https://doi.
org/10.3390/antiox8060183.
29. Medoro, A., Davinelli, S., Milella, L., Willcox, B. J.,
Allsopp, R. C., Scapagnini, G., and Willcox, D. C.
(2023) Dietary astaxanthin: a promising antioxi-
dant and anti-inflammatory agent for brain aging
CAROTENOIDS AS KEAP1-NRF2 PPI INHIBITORS 1691
BIOCHEMISTRY (Moscow)
and adult neurogenesis, Mar. Drugs, 21, 643, https://
doi.org/10.3390/md21120643.
30. Terao,J. (2023) Revisiting carotenoids as dietary an-
tioxidants for human health and disease prevention,
Food Funct., 14, 7799-7824, https://doi.org/10.1039/
d3fo02330c.
31. Srivastava, R. (2021) Physicochemical, antioxidant
properties of carotenoids and its optoelectronic and
interaction studies with chlorophyll pigments, Sci. Rep.,
11, 18365, https://doi.org/10.1038/s41598-021-97747-w.
32. Miller, N.J., Sampson,J., Candeias, L.P., Bramley, P.M.,
and Rice-Evans, C.A. (1996) Antioxidant activities of
carotenes and xanthophylls, FEBS Lett., 384, 240-242,
https://doi.org/10.1016/0014-5793(96)00323-7.
33. Lo, S. C., Li, X., Henzl, M. T., Beamer, L. J., and Han-
nink, M. (2006) Structure of the Keap1:Nrf2 inter-
face provides mechanistic insight into Nrf2 signal-
ing, EMBO J., 25, 3605-3617, https://doi.org/10.1038/
sj.emboj.7601243.
34. Pettersen, E. F., Goddard, T. D., Huang, C. C., Couch,
G.S., Greenblatt, D.M., Meng, E.C., and Ferrin, T. E.
(2004) UCSF Chimera – a visualization system for ex-
ploratory research and analysis, J.Comput. Chem., 25,
1605-1612, https://doi.org/10.1002/jcc.20084.
35. Huang, Z., Lin, Z., and Song, C. (2007) Protonation
processes and electronic spectra of histidine and re-
lated ions, J. Phys. Chem. A, 111, 4340-4352, https://
doi.org/10.1021/jp067280a.
36. Uranga, J., Mikulskis, P., Genheden, S., and Ryde, U.
(2012) Can the protonation state of histidine resi-
dues be determined from molecular dynamics sim-
ulations? Comput. Theor. Chem., 1000, 75-84, https://
doi.org/10.1016/j.comptc.2012.09.025.
37. Ramachandran,S., Kota,P., Ding,F., and Dokholyan,
N.V. (2011) Automated minimization of steric clash-
es in protein structures, Proteins, 79, 261-270, https://
doi.org/10.1002/prot.22879.
38. Kim, S., Thiessen, P. A., Bolton, E. E., Chen, J., Fu,G.,
Gindulyte,A., Han,L., He,J., He,S., Shoemaker, B.A.,
Wang, J., Yu, B., Zhang, J., and Bryant, S. H. (2016)
PubChem substance and compound databases, Nucle-
ic Acids Res., 44, D1202-D1213, https://doi.org/10.1093/
nar/gkv951.
39. Trott, O., and Olson, A. J. (2010) AutoDock Vina: im-
proving the speed and accuracy of docking with a
new scoring function, efficient optimization and
multithreading, J. Comput. Chem., 31, 455, https://
doi.org/10.1002/jcc.21334.
40. Phillips, J. C., Braun,R., Wang,W., Gumbart,J., Tajk-
horshid, E., Villa, E., Chipot, C., Skeel, R.D., Kalé,L.,
and Schulten,K. (2005) Scalable molecular dynamics
with NAMD, J. Comput. Chem., 26, 1781-1802, https://
doi.org/10.1002/jcc.20289.
41. Wang, J., Wolf, R. M., Caldwell, J. W., Kollman, P. A.,
and Case, D. A. (2004) Development and testing of
a general amber force field, J. Comput. Chem., 25,
1157-1174, https://doi.org/10.1002/jcc.20035.
42. Maier, J.A., Martinez,C., Kasavajhala,K., Wickstrom,L.,
Hauser, K. E., and Simmerling, C. (2015) ff14SB: im-
proving the accuracy of protein side chain and back-
bone parameters from ff99SB, J.Chem. Theory Comput.,
11, 3696-3713, https://doi.org/10.1021/acs.jctc.5b00255.
43. Yu,Y., Xu,S., He,R., and Liang,G. (2023) Application
of molecular simulation methods in food science: sta-
tus and prospects, J.Agric. Food Chem., 71, 2684-2703,
https://doi.org/10.1021/acs.jafc.2c06789.
44. Medoro, A., Jafar, T. H., Ali, S., Trung, T. T., Sorren-
ti, V., Intrieri, M., Scapagnini, G., and Davinelli, S.
(2023) In silico evaluation of geroprotective phyto-
chemicals as potential sirtuin 1 interactors, Biomed.
Pharmacother., 161, 114425, https://doi.org/10.1016/
j.biopha.2023.114425.
45. Padmanabhan,B., Tong, K.I., Ohta,T., Nakamura,Y.,
Scharlock,M., Ohtsuji,M., Kang, M.I., Kobayashi,A.,
Yokoyama, S., and Yamamoto, M. (2006) Structural
basis for defects of Keap1 activity provoked by its
point mutations in lung cancer, Mol. Cell, 21, 689-700,
https://doi.org/10.1016/j.molcel.2006.01.013.
46. Stahl, W., and Sies, H. (2023) Antioxidant activity of
carotenoids, Mol. Aspects Med., 24, 345-351, https://
doi.org/10.1016/s0098-2997(03)00030-x.
47. Roselli, S., Pundavela, J., Demont, Y., Faulkner, S.,
Keene, S., Attia, J., Jiang, C. C., Zhang, X. D., Walker,
M.M., and Hondermarck,H. (2015) Sortilin is associat-
ed with breast cancer aggressiveness and contributes
to tumor cell adhesion and invasion, Oncotarget, 6,
10473-10486, https://doi.org/10.18632/oncotarget.3401.
48. Frede, K., Ebert, F., Kipp, A. P., Schwerdtle, T., and
Baldermann,S. (2017) Lutein activates the transcrip-
tion factor Nrf2 in human retinal pigment epithe-
lial cells, J. Agric. Food Chem., 65, 5944-5952, https://
doi.org/10.1021/acs.jafc.7b01929.
49. Li, Z., Dong, X., Liu, H., Chen, X., Shi, H., Fan, Y.,
Hou, D., and Zhang, X. (2013) Astaxanthin protects
ARPE-19 cells from oxidative stress via upregulation
of Nrf2-regulated phaseII enzymes through activation
of PI3K/Akt, Mol. Vis., 19, 1656.
Publishers Note. Pleiades Publishing remains
neutral with regard to jurisdictional claims in published
maps and institutional affiliations. AI tools may have
been used in the translation or editing of this article.