ArticlePDF Available

Molecular modelling analysis of the metabolism of thymoquinone

Authors:

Abstract

Thymoquinone (TQ) is the major biologically active component (about 54%) of volatile oil of black seed that has been shown to exert anti-inflammatory, antioxidant and anti-neoplastic effects both in vitro and in vivo. TQ induces apoptosis, disrupts mitochondrial membrane potential and triggers the activation of caspases 8, 9 and 3 in myeloblastic leukaemia HL-60 cells. Although TQ acts as antioxidant and is found to inhibit inflammation in animal models and culture systems, it can also cause glutathione depletion thus acting as a prooxidant. It has been suggested that TQ may be metabolized to reactive species and increase oxidative stress that contributes to the depletion of antioxidant enzymes and damage to DNA in hepatocytes treated with high concentrations of the compound. Molecular modelling analyses based on molecular mechanics, semi-empirical (PM3) and DFT (at B3LYP/6-31G* level) calculations show that TQ and its metabolic products have LUMO-2 HOMO energy differences ranging from 3.8 to 5.4 eV indicating that the compounds would be moderately inert kinetically with THY being most inert. The molecular surfaces of TQ and DTQ are found to possess significant amounts of positively charged electron-deficient regions so that they may be subject to nucleophilic attacks by glutathione and nucleobases in DNA, thus causing cellular toxicity due to glutathione depletion and DNA damage due to oxidation of nucleobases.
1
Molecular modelling analysis of the metabolism of thymoquinone
Fazlul Huq and Ehsanul Hoque Mazumder
Discipline of Biomedical Science,
School of Medical Sciences,
Faculty of Medicine, Cumberland Campus, C42,
The University of Sydney,
Lidcombe, NSW, Australia.
Phone: +61 2 9351 9522; Fax: +61 2 9351 9520
Email: F.Huq@usyd.edu.au
Abstract
Thymoquinone (TQ) is the major biologically active component (about 54%) of volatile oil of black
seed that has been shown to exert anti-inflammatory, antioxidant and anti-neoplastic effects both in
vitro and in vivo. TQ induces apoptosis, disrupts mitochondrial membrane potential and triggers the
activation of caspases 8, 9 and 3 in myeloblastic leukaemia HL-60 cells. Although TQ acts as
antioxidant and is found to inhibit inflammation in animal models and culture systems, it can also
cause glutathione depletion thus acting as a prooxidant. It has been suggested that TQ may be
metabolized to reactive species and increase oxidative stress that contributes to the depletion of
antioxidant enzymes and damage to DNA in hepatocytes treated with high concentrations of the
compound. Molecular modelling analyses based on molecular mechanics, semi-empirical (PM3) and
DFT (at B3LYP/6-31G* level) calculations show that TQ and its metabolic products have LUMO-
2
HOMO energy differences ranging from 3.8 to 5.4 eV indicating that the compounds would be
moderately inert kinetically with THY being most inert.
The molecular surfaces of TQ and DTQ are found to possess significant amounts of positively charged
electron-deficient regions so that they may be subject to nucleophilic attacks by glutathione and
nucleobases in DNA, thus causing cellular toxicity due to glutathione depletion and DNA damage due
to oxidation of nucleobases.
Key words: Thymoquinone, black seed, antioxidant, apoptosis, molecular modelling
Introduction
Over the recent years, there has been a growing interest in tumour active phytochemicals because they
are relatively non-toxic, inexpensive and available in forms that can be easily ingested [1]. Nigella
sativa Linn, also known as black seed or black cumin, caraway seed, habbat-ul-baraka (meaning the
blessed seed) has been used to treat many different ailments including asthma, rheumatoid arthritis,
stomach aches, cancer and cough for thousands of years [2]. In the Middle East it is incorporated in
diets and everyday life style as it is regarded as a part of holistic approach to health. Traditionally,
there is a common Islamic belief that black seed is a universal healer that provides remedy for all
ailments except death and ageing [3]. Black seed was discovered in Tutankhamen’s tomb, implying
that it played an important role in ancient Egyptian practices as well.
Thymoquinone (TQ; 2-isopropyl-5-methyl-1,4-benzoquinone) is the major biologically active
component (about 54%) of volatile oil of black seed. TQ has been shown to exert anti-inflammatory,
antioxidant and anti-neoplastic effects both in vitro and in vivo [1]. Many investigations have shown
that the growth inhibitory effects of TQ are specific to cancer cells [4, 5]. It induces apoptosis, disrupts
3
mitochondrial membrane potential and triggers activation of caspases 8, 9 and 3 in myeloblastic
leukaemia HL-60 cells. Growth inhibition by TQ was found to be associated with inhibition of DNA
synthesis [6] and that of cell cycle arrest [5]. TQ significantly reduces proliferation of mouse
papilloma cells and mouse spindle cells [1]. Treatment of mouse papilloma cells with 30 M TQ was
found to cause a 22% increase in the number of cells in the G
1
phase within 24 h [1]. The TQ-induced
G
1
arrest was associated with a sharp increase in p16 protein levels as early as 2 h after treatment; the
increased level was sustained for up to 24 h [7]. The increase in expression of p16 protein is
considered to be of special significance as it increases the sensitivity of tumour to chemotherapeutic
drugs.
When more aggressive mouse spindle cancer cells were treated with TQ, it caused G
2
/M arrest (38%
increase) that was accompanied with a decrease in the expression of cyclin B1 protein. TQ has also
been found to suppress the proliferation of several human colon cancer cell lines including Caco-2,
LoVo, HT-29, DLD-1 and HCT-116 cells. It modulates the Bax/Bcl2 ratio by upregulation of
proapoptotic Bax and down-regulation of antiapoptotic Bcl2 proteins in p53-null myeloblastic-
leukemia HL-60 cells during apoptosis [8].
TQ has been found to inhibit inflammation in animal models and culture systems [4]. Administration
of TQ to rats has been found to protect against transient forebrain ischemia-induced damage in the
hippocampus [10]. It acts as a powerful antioxidant with greater ROS scavenging capacity than some
other natural flavavnoids including those isolated from olive oil and silibinin [9].
TQ is found to be equally sensitive to both the multi-drug resistant forms of human pancreatic
adenocarcinoma, uterine sarcoma and leukemic cancer cell lines and their parental controls whereas
commonly used anti-neoplastic agents such as doxorubicin and etopside are 10 times less active to the
resistant forms [6]. Orally administered TQ has been found to potentiate antitumour activity of
4
cisplatin and prevent cisplatin-induced nephrotoxicity in mice and rats [11]. In primary rat hepatocyte
cultures TQ has been found to exert cyto- and genotoxic effects in a concentration dependent manner;
it induced significant antiproliferative effects at 20 M and acute cytotoxicity at higher concentrations
[12]. It has been suggested that TQ may be metabolized to reactive oxygen species (ROS) and increase
oxidative stress that contributes to the depletion of antioxidant enzymes and damage to DNA in
hepatocytes treated with high concentrations of the compound [13, 14]. When TQ was orally
administered to mice, LD
50
was found to be 2.4 g/kg [15]. At high doses (2-3 g/kg) hypoactivity and
difficulty in respiration, and a significant reduction in tissue (liver, kidney and heart) glutathione
content were observed 24 h after administration. It is thought that TQ like other quinones can undergo
redox cycling so that it is metabolized in vivo to hydroquinones or semiquinone radicals by cellular
oxidoreductases leading to the formation of ROS.
Besides TQ, black seed is also found to contain dithymoquinone (DTQ), thymohydroquinone (THY)
and thymol [16]. These compounds are likely to be metabolites of TQ. Indeed under physiological
conditions, TQ is slowly reduced to thymol (THY) and dihydrothymoquinone (THQ). THQ reacts
rapidly with glutathione (GSH) to form glutathionyl-dihydrothymoquinone (THQSG) [17]. TQ may
also exist as dimymoquinone (DTQ). Several authors have reported that in vivo protection against
oxidative damage provided by TQ may be due to the combined action of TQ and THQ. In this regard
THQSG may also be playing an important role, since it has antioxidant activity similar to that of THQ.
The mechanism of metabolic activation of TQ remains unclear. However, it has been suggested that
the reduction of TQ may be catalysed by different cellular reductants possibly according to
mechanisms similar to those reported for the activation of other quinones such as menadione and
CoenzymeQ that have reduction poptential close to that of TQ [17].
5
O
O
CH
3
CH(CH
3
)
2
OH
OH
CH
3
CH(CH
3
)
2
OH
OH
CH
3
CH(CH
3
)
2
SG
CH
3
CH(CH
3
)
2
OH
TQ
O
O
CH
3
O
O
CH
3
CH
3
CH
3
CH
3
DTQ
THQSG
THY
THQ
Figure 1 Products formed from TQ under physiological conditions
In this study, molecular modelling analyses have been carried out using the program Spartan ’08 [19]
of TQ and its likely metabolic products, with the aim of providing a better understanding of their
relative toxicity. Previous studies have shown that xenobiotics and their metabolites which are
kinetically labile and abound in electron-deficient regions on the molecular surface tend to induce
cellular toxicity due to glutathione depletion and cause DNA damage due to oxidation of nucleobases
in DNA [20, 21]. The work was carried out in the Discipline of Biomedical Science, School of
Medical Sciences, The University of Sydney during February to May 2009.
6
Computational methods
The geometries of TQ, and its likely metabolites THY, THQ, THQSG and DTQ have been optimised
based on molecular mechanics, semi-empirical and DFT calculations, using the molecular modelling
program Spartan ’08. Molecular mechanics calculations were carried out using MMFF force field.
Semi-empirical calculations were carried out using the routine PM3. DFT calculations were carried at
B3LYP/6-31G* level. In optimization calculations, a RMS gradient of 0.001 was set as the
terminating condition. For the optimised structures, single point calculations were carried out to give
heat of formation, enthalpy, entropy, free energy, dipole moment, solvation energy, energies for
HOMO and LUMO. The order of calculations: molecular mechanics followed by semi-empirical
followed by DFT ensured that the structure was not embedded in a local minimum. To further check
whether the global minimum was reached, some calculations were carried out with improvable
structures. It was found that when the stated order was followed, structure corresponding to the global
minimum or close to that could ultimately be reached in all cases. Although RMS gradient of 0.001
may not be sufficiently low for vibrational analysis, it is believed to be sufficient for calculations
associated with electronic energy levels [20].
Results and discussion
Table 1 gives the total energy, heat of formation as per PM3 calculation, enthalpy, entropy, free
energy, surface area, volume, dipole moment, and energies of HOMO and LUMO as per both PM3
and DFT calculations for TQ, THY, THQ, THQSG and DTQ. Figures 2-6 give the regions of negative
electrostatic potential (greyish-white envelopes) in (a), HOMOs (where red indicates HOMOs with
high electron density) in (b), LUMOs in (c), and density of electrostatic potential on the molecular
surface (where red indicates negative, blue indicates positive and green indicates neutral) in (d) as
applied to optimised structures of TQ, THY, THQ, THQSG and DTQ.
7
The LUMO-HOMO energy differences for TQ, THY, THQ, THQSG and DTQ from DFT calculations
are found to range from 370.3 kJ/mol (3.8 eV) to 569.6 kJ/mol (5.9 eV), indicating that TQ and its
metabolites would be neither extremely inert nor extremely labile kinetically. The most labile
compound would be TQ itself and the most inert metabolite would be THY.
The solvation energies of TQ, THY, THQ, THQSG and DTQ obtained from PM3 calculations are
found to range from -10.5 to -97.3 kJ mol
-1
, indicating that the compounds would vary greatly in their
solubility in water. Whereas the conjugation product THQSG would be highly soluble in water, the
parent compound TQ is expected to be insoluble in water but soluble in fat.
In the case of TQ and DTQ, the electrostatic potential is found to be more negative around the
carbonyl oxygen atoms, indicating that the positions may be subject to electrophilic attack. In the case
of THY and THQ, the electrostatic potential is found to be more negative around the hydroxyl oxygen
atoms, indicating that the positions may be subject to electrophilic attack. In the case of THQSG, the
electrostatic potential is found to be more negative around various oxygen atoms, indicating that the
positions may be subject to electrophilic attack.
In the case of TQ, THY, THQ, THQSG and DTQ, both the HOMOs with high electron density and
LUMOs are found to be located close to the non-hydrogen atoms of the phenyl ring, indicating the
possibility of occurrence of electronic excitation from the HOMOs with high electron density to the
LUMOs.
5
Table 1 Calculated thermodynamic and other parameters of TQ and its metabolites
Molecule
Calculation
type
Total energy
(kJ mol
-1
)
Enthalpy
(kJ mol
-1
K
-1
)
Entropy
(J mol
-1
K
-1
)
Free energy
(kJ mol
-1
)
Solvation
energy (kJ
mol
-1
)
Area
2
)
Volume
3
)
Dipole
moment
(debye)
HOMO
(kJ/mol)
LUMO
(kJ/mol)
LUMO-HOMO
(kJ/mol)*
TQ
PM3
-244.4
554.9
450.7
420.7
-10.5
201.5
181.1
1.1
-1034.4
-146.9
887.5 (9.2)
DFT
-1414418.4
555.7
452.1
420.9
-7.4
202.9
182.0
0.2
-680.6
-310.3
370.3 (3.8)
THY
PM3
-206.6
603.5
429.7
475.4
-13.7
196.8
177.7
1.2
-867.0
26.8
893.8 (9.3)
DFT
-1220137.5
604.1
437.6
473.6
-13.0
198.3
178.8
1.4
-551.8
17.8
569.6 (5.9)
THQ
PM3
-389.2
621.2
449.2
487.3
-31.2
204.0
184.6
0.2
-830.0
15.2
845.2 (8.8)
DFT
-1417339.7
617.4
457.1
481.2
-31.2
205.5
185.6
0.1
-500.9
16.0
516.9 (5.4)
THQSG
PM3
-1236.2
1234.2
847.9
981.4
-97.3
426.7
395.4
3.5
-878.0
-74.2
803.8 (8.4)
DFT
-4557498.0
1231.6
851.3
977.9
-96.2
425.9
395.9
3.9
-555.0
-74.8
480.2 (5.0)
DTQ
PM3
-421.6
1116.1
665.1
917.5
-16.5
351.7
343.7
0.5
-1028.4
-98.5
929.9 (9.6)
DFT
-2828702.0
11155.2
667.5
913.6
-14.2
356.4
346.7
0.9
-650.9
-256.5
394.4 (4.1)
* The numbers in parentheses are the equivalent values in electron volt
6
O
O
CH
3
CH(CH
3
)
2
(a) (b)
(c) (d)
Figure 2 Structure of TQ giving in: (a) the electrostatic potential (greyish envelope denotes negative
electrostatic potential), (b) the HOMOs, (where red indicates HOMOs with high electron density) (c)
the LUMOs (where blue indicates LUMOs) and in (d) density of electrostatic potential on the
molecular surface (where red indicates negative, blue indicates positive and green indicates neutral).
7
CH
3
CH(CH
3
)
2
OH
(a) (b)
(c) (d)
Figure 3 Structure of THY giving in: (a) the electrostatic potential (greyish envelope denotes negative
electrostatic potential), (b) the HOMOs, (where red indicates HOMOs with high electron density) (c)
the LUMOs (where blue indicates LUMOs) and in (d) density of electrostatic potential on the
molecular surface (where red indicates negative, blue indicates positive and green indicates neutral).
8
OH
OH
CH
3
CH(CH
3
)
2
(a) (b)
(c) (d)
Figure 4 Structure of THQ giving in: (a) the electrostatic potential (greyish envelope denotes negative
electrostatic potential), (b) the HOMOs, (where red indicates HOMOs with high electron density) (c)
the LUMOs (where blue indicates LUMOs) and in (d) density of electrostatic potential on the
molecular surface (where red indicates negative, blue indicates positive and green indicates neutral).
9
OH
OH
CH
3
CH(CH
3
)
2
SG
(a) (b)
(c) (d)
Figure 5 Structure of THQSG giving in: (a) the electrostatic potential (greyish envelope denotes
negative electrostatic potential), (b) the HOMOs, (where red indicates HOMOs with high electron
density) (c) the LUMOs (where blue indicates LUMOs) and in (d) density of electrostatic potential on
the molecular surface (where red indicates negative, blue indicates positive and green indicates
neutral).
10
O
O
CH
3
O
O
CH
3
CH
3
CH
3
CH
3
(a) (b)
(c) (d)
Figure 6 Structure of DTQ giving in: (a) the electrostatic potential (greyish envelope denotes negative
electrostatic potential), (b) the HOMOs, (where red indicates HOMOs with high electron density) (c)
the LUMOs (where blue indicates LUMOs) and in (d) density of electrostatic potential on the
molecular surface (where red indicates negative, blue indicates positive and green indicates neutral).
The overlap of HOMO with high electron density and region of negative electrostatic potential at
some positions, gives further support to the idea that the positions may be subject to electrophilic
attack. It was stated earlier that Orally administered TQ potentiates antitumour activity of cisplatin and
prevents cisplatin-induced nephrotoxicity in mice and rats. The effects may be mediated by
antioxidant role played by TQ.
11
The molecular surfaces of TQ and DTQ are all found to possess significant amounts of electron-
deficient (blue) regions so that they may be subject to nucleophilic attacks such as those by glutathione
and nucleobases in DNA. Reaction with glutathione can induce cellular toxicity by compromising the
antioxidant status of the cell whereas that with nucleobases in DNA can cause DNA damage. The
reaction with glutathione and the resulting oxidative stress may explain why TQ in large doses can
cause acute toxicity.
Conclusion
Thymoquinone (TQ) is the major biologically active component (about 54%) of volatile oil of black
seed that has been shown to exert anti-inflammatory, antioxidant and anti-neoplastic effects both in
vitro and in vivo. It induces apoptosis, disrupts mitochondrial membrane potential and triggers
activation of caspases 8, 9 and 3 in myeloblastic leukaemia HL-60 cells. Molecular modelling analyses
based on molecular mechanics, semi-empirical (PM3) and DFT (at B3LYP/6-31G* level) calculations
show that TQ and its products have LUMO-HOMO energy differences ranging from 3.8 to 5.4 eV
indicating that the compounds would be moderately inert kinetically with THY being most inert. The
molecular surfaces of TQ and DTQ are found to possess significant amounts of electron-deficient
(blue) regions so that they may be subject to nucleophilic attacks such as those by glutathione and
nucleobases in DNA. Reaction with glutathione can induce cellular toxicity by compromising the
antioxidant status of the cell whereas that with nucleobases in DNA can cause DNA damage. The
acute toxicity from large doses of TQ may be associated with its reaction with glutathione.
Abbreviations
12
TQ: Thymoquinone; 2-isopropyl-5-methyl-1,4-benzoquinone
DTQ: Dithymoquinone
THY: Thymol
THQ: Dihydrothymoquinone
GSH: Glutathione
THQSG: Glutathionyl-dihydrothymoquinone
LD
50
:
Dose that kills 50% of the tested group
DFT: Density functional theory
LUMO: Lowest unoccupied molecular orbital
HOMO: Highest occupied molecular orbital
Acknowledgments
Fazlul Huq is grateful to the Discipline of Biomedical Science, School of Medical Sciences, The
University of Sydney for the time release from teaching.
13
References
1. Gali-Muhtasib H, Roessner A and Schneider-Stock R, Thymoquinone: A promising anti-
cancer drug from natural sources. Int. J. Biochem. Cell Biol., 2006, 38, 1249-1253.
2. El-Mahdy MA, Zhu Q, Wang Q-E, Wani G and Wani AA, Thymoquinone induces apoptosis
through activation of caspase-8 and mitochondrial events in p53-null myeloblastic leukemia
HL-60 cells. Int. J. Cancer, 2005, 117, 409-417.
3. Ali BH and Blunden G, Pharmacological and toxicological properties of Nigella sativa,
Phytother. Res., 2003, 17, 299-305.
4. Gali-Muhtasib HU, Diab-Assaf M, Boltze C, Al-Hmaria J, Hartig R, Roessner A, Roessner A
and Schneider-Stock R, Thymoquinone extracted from black seed triggers apoptotic cell death in
human colorectal cancer cells via a p53-dependent mechanism. Int. J. Oncol., 2004, 25, 857-
866.
5. Shoeeb AM, Elgayyar M, Dudrick PS, Bell JL and Tothof PK, In vitro inhibition of growth and
induction of apoptosis in cancer cell lines by thymoquinone. Int. J. Oncol., 2003, 22, 107-113.
 Worthen DR, Ghoseh OA and Crooks PA, The in vitro anti-tumor activity of some crude and
purified components of blackseed Nigella sativa L. Anticancer Res., 1998, 18, 1527-1532.
7. Gali-Muhtasib HU, Aboikheir W, Kheir L, Darwiche N and Crooks P, Molecular pathway for
Thymoquinone-induced cell cycle arrest and apoptosis in neoplastic keratinocytes. Anticancer
Drugs, 2004, 15, 389-399.
8. El-Mahdy MA, Zhu Q, Wang QE, Wani G and Wani AA, Thymoquinone induces apoptosis
through activation of caspase-8 and mitochondrial events in p53-null myeloblastic leukemia HL-
60 cells. Int. J. Cancer, 2005, 117, 409-417.
14
9. Badary OA, Taha RA, Gamel el-Din AM and Abdel-Wahab MH, Thymoquinone is a potent
superoxide anion scavenger. Drug. Chem. Toxicol., 2003, 26, 87-98.
10. Badary OA, Nagi MN, Al-Shabanah OA, Al-Sawaf HA, Al-Sohaibani MO and Al-Bekairi AM,
Thymoquinone ameliorates the nephrotoxicity induced by cisplatin in rodents and potentiates its
antitumor activity. Can. J. Physiol. Pharmacol., 1997, 75, 1356-1361.
11. Al-Majed AA, Al-Omar FA and Nagi MN, Neuroprotective effects of thymoquinone against
transient forebrain ischemia in the rat hippocampus, Euro. J. Pharmacol., 2006, 543, 40-47.
12. Khader M, Bresgen N and Eckl PM, In vitro toxicological properties of thymoquinone. Food
Chem. Toxicol., 2009, 47, 129-133.
13. Badary OA, Taha RA, El-Din AMG and Abdel-Wahab MH, Thymoquinone is a potent
superoxide anion scavenger. Drug Chem. Toxicol., 2009, 26(2), 87-98.
14. Mansour MA, Nagi MN, El-Khatib AS and Al-Bekhari AM, Effects of thymoquinone on
antioxidant enzyme activities, lipid peroxidation and DT-diaphorase in different tissues of mice:
a possible mechanism of action. Cell Biochem. Funct., 2002, 20, 143-151.
15. Badary OA, Al-Shahib OA, Nagi MN, Al-Bekairi AM and Almazar MMA, Acute and
subchronic toxicity of thymoquinone in mice, Drug Dev. Res., 1998, 44, 56-61.
16. Ghosheh OA, Houdi AA and Crooks PA, High performance liquid chromatographic analysis of
the pharmacologically active quinones and related compounds in the oil of black seed [13]
(Nigella sativa L.). J. Pharm. Biomed. Anal., 1999, 19, 757-762.
17. Khaliffe KH, Lupidi G, Nonenzymatic reduction of thymoquinone in physiological conditions.
Free Rad. Res., 2009, 41(2), 153-161.
18. Spartan ’08, Wavefunction, Inc. Irvine, CA, USA, 2006.
15
19. Huq F, Molecular modelling analysis of the metabolism of tamoxifen. Int. J. Pure Appl. Chem.,
2006, 1(2), 155-163.
20. Huq F, Molecular modelling analysis of the metabolism of zaleplon. J Pharmacol. Toxicol.,
2006, 1(4), 328-336.
21. Huq F and Alshehri A, Molecular Modelling Analysis of the Metabolism of Diclofenac. Int. J.
Pure Appl. Chem., 2006, 1(3), 359-373.
... The binding of CMPF could be reduced leading to the decreased chances of acute renal failure. Assuming this, we hypothetically considered a compound which has been earlier proved to be beneficial for human health, such as cancer, tumor, and more [11][12][13][14][15][16][17]. This compound is dithymoquinone (DTQ), one of the active compounds found in black cumin seed (Nigella sativa), which is well known for its positive effects on human in various conditions such as tumor, cancer, clearance of hepato-renal toxicity, diabetes, [8][9][10][11][12][13][14][15][16][17][18].and ...
... Assuming this, we hypothetically considered a compound which has been earlier proved to be beneficial for human health, such as cancer, tumor, and more [11][12][13][14][15][16][17]. This compound is dithymoquinone (DTQ), one of the active compounds found in black cumin seed (Nigella sativa), which is well known for its positive effects on human in various conditions such as tumor, cancer, clearance of hepato-renal toxicity, diabetes, [8][9][10][11][12][13][14][15][16][17][18].and found to possess relatively higher binding affinity than CMPFWe predicted the inhibitory effect of DTQ against CMPF using in silico docking performed by AutoDock tools [19]. ...
Article
Full-text available
3-carboxy-4-methyl-5-propyl-2-furanpropanoic acid (CMPF) is a major endogenous ligand found in the human serum albumin (HSA) of renal failure patients. It gets accumulated in the HSA and its concentration in sera of patients may reflect the chronicity of renal failure [1-4]. It is considered uremic toxin due to its damaging effect on the renal cells. The high concentrations of CMPF inhibit the binding of other ligands to HSA. Removal of CMPF is difficult through conventional hemodialysis due to its strong binding affinity. We hypothesized that the competitive inhibition may be helpful in removal of CMPF binding to HSA. A compound with higher HSA binding affinity than CMPF could be useful to prevent CMPF from binding so that CMPF could be excreted by the body through the urine. We studied an active compound dihydrothymoquinone/ dithymoquinone (DTQ) found in black cumin seed (Nigella sativa), which has higher binding affinity for HSA. Molecular docking simulations were performed to find the binding affinity of CMPF and DTQ with HSA. DTQ was found to have higher binding affinity possessing more interactions with the binding residues than the CMPF. We studied the binding pocket flexibility of CMPF and DTQ to analyze the binding abilities of both the compounds. We have also predicted the ADME properties for DTQ which shows higher lipophilicity, higher gastrointestinal (GI) absorption, and blood-brain barrier (BBB) permeability. We discovered that DTQ has potential to act as an inhibitor of CMPF and can be considered as a candidate for the formation of the therapeutic drug against CMPF.
Article
Full-text available
Article
Full-text available
Nigella sativa has been traditionally used for the treatment of inflammations, liver disorders, and arthritis. Experimentally, it has been demonstrated that N. sativa extracts and the main constituent of their volatile oil, thymoquinone, possess antioxidant, anti-inflammatory and hepato-protective properties. To further evaluate the toxicological properties in a metabolically competent cellular system, thymoquinone was applied to primary rat hepatocyte cultures, and both cyto- and genotoxic effects were tested. Mitotic indices and the rates of apoptoses and necroses were determined as endpoints of cytotoxicity, while chromosomal aberrations and micronucleated cells served as endpoints of genotoxicity. In this approach thymoquinone demonstrated cyto- and genotoxic effects in a concentration dependent manner: it induced significant anti-proliferative effects at 20 microM and acute cytotoxicity at higher concentrations. Thymoquinone significantly increased the rates of necrotic cells at concentrations between 2.5 and 20 microM. Furthermore, it induced significant genotoxicity at concentrations > or =1.25 microM. These observations support the previous finding that thymoquinone causes glutathione depletion and liver damage, but contradict the reports indicating antioxidant and anti-clastogenic effects. Thymoquinone might be metabolised to reactive species and increase oxidative stress, which contributes to the depletion of antioxidant enzymes and damage to DNA in hepatocytes treated with high thymoquinone concentrations.
Article
The effects of acute and subchronic administration of thymoquinone (TQ), the main constituent of the volatile oil of the black seeds Nigella sativa, with significant cytoprotective properties, were studied in male Swiss albino mice.After acute oral administration, the LD50 value (95% CL) was 2.4 g/kg (1.52–3.77). Signs of toxicity at high doses were hypoactivity and difficulty in respiration. Twenty-four hours after TQ (2 and 3 g/kg) administration, a significant reduction in tissue (liver, kidneys, and heart) reduced glutathione (GSH) content was observed. Plasma urea and creatinine concentrations and the enzyme activities of alanine amino transferase (ALT), lactate dehydrogenase (LDH), and creatine phosphokinase (CPK) were significantly increased.In the subchronic study, mice received TQ in drinking water at concentrations of 0.01, 0.02, and 0.03% for 90 days with no resulting mortality or signs of toxicity. The average daily intake of the compound was approximately 30, 60, or 90 mg/kg/day. There were no changes of toxicological significance in body and organ weights, food and water intake, or urine and feces output. Tissue GSH, plasma concentrations of TP, urea, creatinine and triglycerides, and enzyme activities of ALT, LDH, and CPK were also not affected. Histological examination revealed no gross or microscopic tissue damage. TQ, however, produced a significant decrease in fasting plasma glucose level.The results indicate that the acute oral toxicity of TQ in mice is of a low order and it is generally well tolerated when given subchronically at doses previously shown to have cytoprotective activity. Drug Dev. Res. 44:56–61, 1998. © 1998 Wiley-Liss, Inc.
Article
The effects of thymoquinone (TQ) on cisplatin-induced nephrotoxicity in mice and rats were studied. Oral administration of TQ (50 mg/L in drinking water) for 5 days before and 5 days after single injections of cisplatin (5 mg/kg, i.v., in rats and 7 or 14 mg/kg, i.p., in mice) greatly ameliorated cisplatin-induced nephrotoxicity in both species. In rats, i.v. cisplatin caused 4- and 5-fold elevations in serum urea and creatinine, a 235% increase in urine volume, a 41% increase in kidney weight, 8.5-fold decrease in creatinine clearance, and extensive histological damage 5 days after treatment. In mice, similar alterations in kidney function were observed. TQ-induced amelioration of cisplatin nephrotoxicity was evident by significant reductions in serum urea and creatinine and significant improvement in polyuria, kidney weight, and creatinine clearance. The protective effects of TQ against cisplatin-induced nephrotoxicity in the rat were further confirmed by histopathological examination. To evaluate the possible modification of the antitumor activity of cisplatin by TQ, we studied their interaction in Ehrlich ascites carcinoma (EAC) bearing mice. The results revealed that TQ potentiated the antitumor activity of cisplatin. The current study suggests that TQ may improve the therapeutic index of cisplatin.
Article
A crude gum, a fixed oil and two purified components of Nigella sativa seed, thymoquinone (TQ) and dithymoquinone (DIM), were assayed in vitro for their cytotoxicity for several parental and multi-drug resistant (MDR) human tumor cell lines. Although as much as 1% w/v of the gum or oil was devoid of cytotoxicity, both TQ and DIM were cytotoxic for all of the tested cell lines (IC50's 78 to 393 microM). Both the parental cell lines and their corresponding MDR variants, over 10-fold more resistant to the standard antineoplastic agents doxorubicin (DOX) and etoposide (ETP), as compared to their respective parental controls, were equally sensitive to TQ and DIM. The inclusion of the competitive MDR modulator quinine in the assay reversed MDR Dx-5 cell resistance to DOX and ETP by 6- to 16-fold, but had no effect on the cytotoxicity of TQ or DIM. Quinine also increased MDR Dx-5 cell accumulation of the P-glycoprotein substrate 3H-taxol in a dose-dependent manner. However, neither TQ nor DIM significantly altered cellular accumulation of 3H-taxol. The inclusion of 0.5% v/v of the radical scavenger DMSO in the assay reduced the cytotoxicity of DOX by as much as 39%, but did not affect that of TQ or DIM. These studies suggest that TQ and DIM, which are cytotoxic for several types of human tumor cells, may not be MDR substrates, and that radical generation may not be critical to their cytotoxic activity.
Article
An HPLC method for quantifying the putative pharmacologically active constituents: thymoquinone (TQ), dithymoquinone (DTQ), thymohydroquinone (THQ), and thymol (THY), in the oil of Nigella sativa seed is described. Extraction of the constituents from the oil was carried out using C18 PrepSep mini columns followed by quantification of the recovered constituents by HPLC on a reversed-phase muBondapak C18 analytical column, using an isocratic mobile phase of water:methanol:2-propanol (50:45:5% v/v) at a flow rate of 2 ml min(-1). UV detection was at 254 nm for TQ, DTQ, and THY, and at 294 nm for THQ. The above four compounds were separated with good resolution, reproducibility, and sensitivity under these conditions. This analytical method was used to quantify the above four constituents in a commercial sample of N. sativa seed oil, and provides a good quality control methodology for the pharmacologically active components in this widely used natural remedy.
Article
The present investigation focused, firstly, on the effects of oral administration of thymoquinone (TQ) on antioxidant enzyme activities, lipid peroxidation and DT-diaphorase activity in hepatic, cardiac and kidney tissues of normal mice. Superoxide dismutase (SOD; E.C:1.15.1.1), catalase (CAT; E.C:1.11.1.6), glutathione peroxidase (GSH-Px; E.C:1.11.1.9), glutathione-S-transferase (GST; E.C:2.5.1.18), and DT-diaphorase (E.C:1.6.99.2) enzyme activities in each tissue type were determined. Treatment of mice with the different doses of TQ (25, 50 and 100 mg kg(-1) day(-1) orally) for 5 successive days, produced significant reductions in hepatic SOD, CAT and GSH-Px activities. In addition cardiac SOD activity was markedly inhibited with the higher doses of TQ, (namely 50 and 100 mg kg(-1)). Moreover, TQ (100 mg kg(-1)) significantly reduced hepatic and cardiac lipid peroxidation as compared with the respective control group. Conversely, TQ (50,100 mg kg(-1)) and TQ (100 mg kg(-1)) enhanced cardiac and renal DT-diaphorase activity respectively. However, the selected doses of TQ neither produced any change in GST activity nor influenced reduced glutathione content in all tissues studied. TQ was tested, secondly, as a substrate for hepatic, cardiac and renal DT-diaphorase of normal mice in the presence of NADPH. Kinetic parameters for the reduction of TQ to dihydrothymoquinone (DHTQ) indicated that DT-diaphorase of different tissues can efficiently reduce TQ to DHTQ. K(m) and V(max) values revealed that hepatic DT-diaphorase exhibited the higher values, while the lower values were associated with renal DT-diaphorase. TQ and DHTQ were tested, thirdly, as specific scavengers for superoxide anion (generated biochemically) or as general scavengers for free radicals (generated photochemically). The results revealed that TQ and DHTQ acted not only as superoxide anion scavengers but also as general free radical scavengers. The IC(50) for TQ and DHTQ in biochemical and photochemical assays were in the nanomolar and micromolar range respectively. Our data may explain at least partly the reported beneficial in vivo protective effects of TQ through the combined antioxidant properties of TQ and its metabolite DHTQ.
Article
The seeds of Nigella sativa Linn. (Ranunculaceae), commonly known as black seed or black cumin, are used in folk (herbal) medicine all over the world for the treatment and prevention of a number of diseases and conditions that include asthma, diarrhoea and dyslipidaemia. This article reviews the main reports of the pharmacological and toxicological properties of N. sativa and its constituents. The seeds contain both fixed and essential oils, proteins, alkaloids and saponin. Much of the biological activity of the seeds has been shown to be due to thymoquinone, the major component of the essential oil, but which is also present in the fixed oil. The pharmacological actions of the crude extracts of the seeds (and some of its active constituents, e.g. volatile oil and thymoquinone) that have been reported include protection against nephrotoxicity and hepatotoxicity induced by either disease or chemicals. The seeds/oil have antiinflammatory, analgesic, antipyretic, antimicrobial and antineoplastic activity. The oil decreases blood pressure and increases respiration. Treatment of rats with the seed extract for up to 12 weeks has been reported to induce changes in the haemogram that include an increase in both the packed cell volume (PCV) and haemoglobin (Hb), and a decrease in plasma concentrations of cholesterol, triglycerides and glucose. The seeds are characterized by a very low degree of toxicity. Two cases of contact dermatitis in two individuals have been reported following topical use. Administration of either the seed extract or its oil has been shown not to induce significant adverse effects on liver or kidney functions. It would appear that the beneficial effects of the use of the seeds and thymoquinone might be related to their cytoprotective and antioxidant actions, and to their effect on some mediators of inflammation.Copyright