ArticlePDF Available

Divergent Anticancer Activity of Free and Formulated Camel Milk α-Lactalbumin

Authors:

Abstract and Figures

Alpha-lactalbumin (α-LA), a small milk calcium-binding globular protein, is known to possess noticeable anticancer activity, which is determined by the ability of this protein to form complexes with oleic acid (OA). To date, in addition to human and bovine α-LA, the ability to form such anti-tumor complexes with OA was described for goat and camel α-LA. Although the mechanisms of the anticancer activity of human and bovine α-LA are already well-studied, little is currently known about the anticancer action of this camel protein. The goal of this study was to fill this gap and to analyze the anticancer and pro-apoptotic activities of camel α-LA in its free form (α-cLA) and as an OA-containing complex (OA-α-cLA) using four human cancer cell lines, including Caco-2 colon cancer cells, PC-3 prostate cancer cells, HepG-2 hepatoma cells, and MCF-7 breast cancer cells as targets. The anti-tumor activities of OA-α-cLA and α-cLA were analyzed using MTT test, annexin/PI staining, cell cycle analysis, nuclear staining, and tyrosine kinase (TK) inhibition methods. We show here that the OA-α-cLA complex does not affect normal cells but has noticeable anti-cancer activity, especially against MCF-7 cells, thus boosting the anticancer activity of α-cLA and improving the selectivity of OA. The OA-α-cLA complex mediated cancer cell death via selective induction of apoptosis and cell-cycle arrest at lower IC50 than that of free α-cLA by more than two folds. However, OA induced apoptosis at higher extent than OA-α-cLA and α-cLA. OA also caused unselective apoptosis-dependent cell death in both normal and cancer cells to a similar degree. The apoptosis and cell-cycle arresting effect of OA-α-cLA may be attributed to the TK inhibition activity of OA. Therefore, OA-α-cLA serves as efficient anticancer complex with two functional components, α-cLA and OA, possessing different activities. This study declared the effectiveness of OA-α-cLA complex as a promising entity with anticancer activity, and these formulated OA-camel protein complexes constitute an auspicious approach for cancer remedy, particularly for breast cancer.
Content may be subject to copyright.
Full Terms & Conditions of access and use can be found at
http://www.tandfonline.com/action/journalInformation?journalCode=icnv20
Download by: [Professor Vladimir N. Uversky] Date: 26 September 2017, At: 16:39
Cancer Investigation
ISSN: 0735-7907 (Print) 1532-4192 (Online) Journal homepage: http://www.tandfonline.com/loi/icnv20
Divergent Anticancer Activity of Free and
Formulated Camel Milk α-Lactalbumin
Vladimir N. Uversky, Esmail M. El-Fakharany, Marwa M. Abu-Serie, Hussein
A. Almehdar & Elrashdy M. Redwan
To cite this article: Vladimir N. Uversky, Esmail M. El-Fakharany, Marwa M. Abu-Serie, Hussein
A. Almehdar & Elrashdy M. Redwan (2017): Divergent Anticancer Activity of Free and Formulated
Camel Milk α-Lactalbumin, Cancer Investigation, DOI: 10.1080/07357907.2017.1373783
To link to this article: http://dx.doi.org/10.1080/07357907.2017.1373783
Published online: 26 Sep 2017.
Submit your article to this journal
View related articles
View Crossmark data
CANCER INVESTIGATION
, VOL. , NO. , –
https://doi.org/./..
Divergent Anticancer Activity of Free and Formulated Camel Milk α-Lactalbumin
Vladimir N. Uversky a,b,c, Esmail M. El-Fakharany d, Marwa M. Abu-Seriee, Hussein A. Almehdara,
and Elrashdy M. Redwana,d
aDepartment of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia; bInstitute for Biological
Instrumentation of the Russian Academy of Sciences, Pushchino, Moscow region, Russia; cDepartment of Molecular Medicine and USF Health
Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; dTherapeutic and Protective
Proteins Laboratory, Protein Research Department, Medical Biotechnology Department, Genetic Engineering and Biotechnology Research
Institute, City for Scientific Research and Technology Applications (SRTA-City), Alexandria, Egypt; eMedical Biotechnology Department, Genetic
Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications (SRTA-City), Alexandria, Egypt
ARTICLE HISTORY
Received  January 
Revised  August 
Accepted  August 
KEYWORDS
Camel milk; Milk protein;
α-Lactalbumin; Anticancer
activity
ABSTRACT
Alpha-lactalbumin (α-LA), a small milk calcium-binding globular protein, is known to possess notice-
able anticancer activity, which is determined by the ability of this protein to form complexes with
oleic acid (OA). To date, in addition to human and bovine α-LA, the ability to form such anti-tumor
complexes with OA was described for goat and camel α-LA. Although the mechanisms of the anti-
cancer activity of human and bovine α-LA are already well-studied, little is currently known about the
anticancer action of this camel protein. The goal of this study was to ll this gap and to analyze the
anticancer and pro-apoptotic activities of camel α-LA in its free form (α-cLA) and as an OA-containing
complex (OA-α-cLA) using four human cancer cell lines, including Caco-2 colon cancer cells, PC-3
prostate cancer cells, HepG-2 hepatoma cells, and MCF-7 breast cancer cells as targets. The anti-tumor
activities of OA-α-cLA and α-cLA were analyzed using MTT test, annexin/PI staining, cell cycle anal-
ysis, nuclear staining, and tyrosine kinase (TK) inhibition methods. We show here that the OA-α-cLA
complex does not aect normal cells but has noticeable anti-cancer activity, especially against MCF-7
cells, thus boosting the anticancer activity of α-cLA and improving the selectivity of OA. The OA-α-cLA
complex mediated cancer cell death via selective induction of apoptosis and cell-cycle arrest at lower
IC50 than that of free α-cLA by more than two folds. However, OA induced apoptosis at higher extent
than OA-α-cLA and α-cLA. OA also caused unselective apoptosis-dependent cell death in both nor-
mal and cancer cells to a similar degree. The apoptosis and cell-cycle arresting eect of OA-α-cLA may
be attributed to the TK inhibition activity of OA. Therefore, OA-α-cLA serves as ecient anticancer
complex with two functional components, α-cLA and OA, possessing dierent activities. This study
declared the eectiveness of OA-α-cLA complex as a promising entity with anticancer activity, and
these formulated OA-camel protein complexes constitute an auspicious approach for cancer remedy,
particularly for breast cancer.
Introduction
Recent research revealed that, in addition to their tra-
ditional biological roles (e.g., catalysis of various reac-
tions), various milk proteins and peptides can possess
multiple unrelated functions, exerting various moon-
lighting activities, such as anti-microbial, antiviral, and
antifungal activities, immuno-modulatory functions,
as well as playing roles in cancer cell apoptosis, anti-
metastatic activities, and antioxidant eects (1).Sev-
eral milk-derived biologics, such as HAMLET (human
CONTACT Elrashdy M. Redwan redwan@yahoo.com Therapeutic and Protective Proteins Laboratory, Protein Research Department, Medical Biotech-
nology Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications (SRTA-City), New Borg
EL-Arab , Alexandria, Egypt; Vladimir N. Uversky vuversky@health.usf.edu Department of Biological Sciences, Faculty of Sciences, King Abdulaziz
University, P.O. Box , Jeddah, Saudi Arabia.
Color versions of one or more of the figures in the article can be found online at www.tandfonline.com/icnv.
α-lactalbumin [α-LA] made lethal to tumor cells),
demonstrated some promising results in clinical trials
(2). The attractiveness of using milk-derived biologics
as potential drugs relies on the important fact that pro-
teins and peptides are well-tolerated by the organism
and exhibit oral bioavailability. This also suggests that
milk proteins and peptides may complement standard
therapies to boost overall success in cancer treatments
(1). Historically, the journey toward the understanding
of the molecular basis of the anticancer activity of milk
©  Taylor & FrancisGroup, LLC
Downloaded by [Professor Vladimir N. Uversky] at 16:39 26 September 2017
2V. N. UVERSKY ET AL.
components has started from the identication of a
complex of α-LAandoleicacid(OA)inhumanmilk
(2–5). HAMLET was characterized by a highly selec-
tive apoptotic activity against tumor cells both in vivo
(6–10) and in clinical trials (11–13).Sinceα-LA is
a highly conserved milk protein, it was interesting
to investigate anticancer activities that were ascribed
to the oleic acid-bound forms of α-LAs from other
species,suchasbovine,camel,andgoat,givingraiseto
BAMLET, CAMLET, and GAMLET, respectively (14–
18).Fragmentsofbovineα-LAobtainedbylimited
proteolysis can also bind OA and exhibit anti-tumor
activity (19).
For human and bovine α-LAs as well as for
some other proteins forming HAMLET-like com-
plexes with OA, the molecular mechanisms of HAM-
LET/BAMLET formation are well-understood (18).In
fact, the formation of the molten globule-like inter-
mediate (which can be achieved by the removal of
Ca2+from calcium-binding proteins, moderate heat-
ing, acidic or alkaline pH, etc.) seems to be an impor-
tant prerequisite for the eective interaction of a target
protein with OA, and for the formation of HAMLET-
like complexes (18). Therefore, the formation of the
HAMLET-like complexes by milk and some other pro-
teins involves the OA-induced formation of alterna-
tively folded conformations with unexpected benecial
functions (20).
In a recent study, we conducted a systematic com-
parative analysis of structural properties and con-
formational stabilities of bovine and camel α-LAs
usingasetofspectroscopicandcomputationaltech-
niques (21). These analyses revealed the existence
of several noticeable dierences between these two
proteins, with camel α-LA being more stable toward
thermal and pH-mediated denaturation but less stable
toward guanidine hydrochloride-mediated unfolding
(21). Furthermore, the camel α-LAwasshowntobe
more disordered and possessed stronger aggregation
propensities (21). Although noticeable anticancer and
pro-apoptotic activity was reported for some camel
milk proteins such as α-LA (15) and casein (22),the
mechanisms of the anti-cancer potential of camel
α-LA are poorly understood. The goals of the current
study were to analyze the anticancer activity of camel
α-LA and to validate our hypotheses that, similar to
other milk proteins from dierent species, camel α-LA
is able to form complexes with OA with cytotoxic
potential, and that the anticancer and pro-apoptotic
activities of camel milk α-LA are OA-dependent.
We also conducted a control study assaying the anti-
cancer and pro-apoptotic activities of camel α-LA
or OA alone. The anticancer activities of puried
protein and prepared OA-containing complexes were
assayed using dierent human cancer cell lines as
targets.
Material and methods
Purication of camel α-lactalbumin
Camel skimmed milk was prepared according to the
procedure described in Almahdy et al. (22).Camel
α-lactalbumin (α-cLA) was puried from skimmed
milk according to method of Pettersson et al. (14)
with some modications. In brief, the camel skimmed
milk was exposed to decreased pH to reach the
isoelectric point (pI) of casein to cause precipita-
tion of this protein by adding 1 M HCl to milk
until the pH became 4.6. The casein-free prepara-
tion was treated with ammonium sulfate at concen-
tration of 45%, and after incubation for 24 h at 4°C,
the mixture was centrifuged at 6000 ×gfor 20 min.
The supernatant was collected and dialysed against
20 mM phosphate buer and 35 mM EDTA, pH
7.6. Proteins precipitated from skimmed milk in 45%
ammonium sulfate showed 7 bands on 12% SDS-
PAG E ( Figure 1), and this proteins mixture contained
α-cLA.
Thesamplecontainingα-cLA was applied to the
DEAE–cellulose column equilibrated with 50 mM
phosphate buer, pH 7.6. Both camel albumin (a
protein with a molecular weight of 67.0 kDa) and
α-cLA (a protein with a molecular weight of 14.4 kDa)
were eluted by a DEAE-cellulose column using gra-
dient 0.0–0.5 M NaCl. After dialysis of the eluted
samples, the fractions containing α-cLA were applied
into Sephadex-G50 gel ltration column equilibrated
with the same buer to separate α-cLA from albumin.
Homogeneity and purity of α-cLA was estimated by
SDS-PAGE. On 12% SDS-PAGE, puried camel α-LA
wascharacterizedbyasinglebandwithanexpected
molecular weight of 14.4 kDa (as shown in Figure 1).
The fractions contained puried α-cLA were pooled,
lyophilized, and kept at 80°C until use. The protein
concentration of α-cLA was estimated spectrophoto-
metrically at 280 nm using an extension coecient of
ε280 nm =32,470 M1cm1(23).Camelα-cLA was
standardized spectroscopicaly as described previously
by Redington et al. (21).
Downloaded by [Professor Vladimir N. Uversky] at 16:39 26 September 2017
CANCER INVESTIGATION 3
Figure . % SDS-PAGE of purified camel α-LA during purifica-
tion steps. Lane , molecular mass marker; lane , skimmed camel
milk after precipitation by % ammonium sulfate; lane , eluted
fractions from DEAE–cellulose column; lane , purified α-LA from
Sephadex G column.
Formation of α-LA-oleic acid complex
Lyophilized α-cLA was used for the preparation of the
OA-α-cLA complex by gentle heating. Here, native
α-cLA was dissolved in PBS at a concentration of
120 μM,andoleicacidwasaddeddirectlytothepro-
tein solution at the 50 molar equivalents (OA:α-LA=
50:1). The reaction mixture was incubated for 20 min
at 45°C after vortexing for 30 s. To remove unbound
OA, the mixture was centrifuged at 10,000 rpm fol-
lowed by ultraltration using the Centricone (3000 Da
cut-o membrane) (24).TheamountofOAboundto
α-cLA was estimated to be 318 μM.
Oleic acid determination
ConcentrationofadsorbedOAinproteinsampleswas
determined according to the colorimetric method of
Duncombe (25). In brief, the protein samples were
shaken with chloroform and copper nitrate solution,
OAwasremovedfromitscomplexwithα-LA and
formed copper-fatty acid soap which is soluble in chlo-
roform. The amount of copper in chloroform is equiv-
alent to the amount of oleic acid in the sample that is
assayed by addition of sodium diethylthiocarbamate as
a color developer. Here, 2.5 ml of the copper reagent
(nine volumes of 1 M triethanolamine; one volume of
1 N acetic acid and ten volumes of 10 g% copper sulfate)
were added to 500 μl samples and/or 500 μlstandard
oleic acid. All tubes were then shaken vigorously with
a vortex mixer, after which 5 ml of chloroform were
addedtothesolutionandshakenvigorouslyfor1min.
Then, 3 ml of the lower layer were carefully transferred
to another test tube containing 500 μlof0.1%sodium
diethylthiocarbamate in butanol and absorbencies of a
samples (Asam) and the standard (Ast) were read at
440 nm.
Endotoxin determination
The endotoxin levels in the camel milk, puried α-
cLA, and prepared compounds were tested to avoid its
cytotoxic eects on the cell lines. To this end, protein
samples and solutions of other compounds were mixed
with an equal volume of sample buer containing
20% glycerol or 20% sucrose, β-mercatoethanol, 1.5%
sodium deoxycholate (DOC), and 4 M urea. The
samples were boiled for 5 min at 100°C, then loaded
into the gel and separated at 20 mA/gel. The gels were
then transferred to the clean polyethylene containers
and stained according to the established protocol (26).
First, to remove the ions of the running buer, the gels
were washed with water for 5 min. Then, the gels were
xed in 10 volumes of 45% ethanol or methanol (ana-
lytical grade, Sigma-Aldrich, St. Louis, MO, USA) and
in 0.5% periodic acid for 15 min. Second, the gels were
additionally washed ve times over 5 min with 200 mL
water/wash, since washing less than three times gave a
yellow-brown color after the silver solution addition.
Then, aqueous silver nitrate (0.1%) was added, and gels
were for incubated for 15 min. Next, the stained gels
were rinsed in deionized water for 1 min, and then in
1 volume of developer (100 mL of 37% formaldehyde,
3% sodium carbonate) for just 30–40 s to remove
the silver nitrate ions from the gels surfaces. Then,
thegelsweresoakedin5volumesofdeveloperuntil
the desired color of bands was obtained (5 min).
Finally, the developed gels were soaked in water for
5 min and then overnight in 40% ethanol. Application
of this method, which can accurately detect less than
1 ng of bacterial lipopolysaccharide (26),revealedthat
all used proteins and prepared compounds were found
to be free from endotoxin.
Downloaded by [Professor Vladimir N. Uversky] at 16:39 26 September 2017
4V. N. UVERSKY ET AL.
Cytotoxicity assay against normal mammalian
epithelial cell line
The hydrogen acceptor 3-(4,5-dimethylthiazol-2-yl)-
2,5 diphenyltetrazolium bromide (MTT) assay is a
rapid and highly accurate colorimetric approach that
is widely used to determine cell metabolic activity in
order to evaluate cytotoxicity of various compounds
(including new drugs). Here, mitochondrial dehydro-
genases of viable cells cleave the tetrazolium ring and
then yield purple MTT formazan crystals. Therefore,
the optical density values of dissolved crystals after
treatment are proportional to the number of viable
cells (27, 28). The eect of samples containing query
compounds on the capability of cells to replicate is
used as an index of compound toxicity. In this study,
the concentration of the compound (e.g., the OA-α-
LA complex) at which 50 percent of the cells do not
multiply is called IC50 (the median inhibitory dose).
Vero (African green monkey kidney normal) cell line
(Sigma-Aldrich, USA) was cultured and seeded in a
DMEM medium (Lonza, USA) supplemented with
10% fetal bovine serum (FBS) as 5 ×103cells per
well (96-well cell culture plate). After 24 h incuba-
tion for cell attachment in 5% CO2incubator, dierent
concentrations of α-LA and OA-α-LA ranging from
25 μM to 100 μM were added. In addition, OA and
5-uorouracil (5-FU, which is a medication used in
the treatment of cancer) were added at dierent con-
centrations as positive control (for investigation the
cytotoxic eect of the fatty acid alone, OA was dis-
solved in ethanol and diluted in PBS, pH7.2). After 72 h
incubation in 5% CO2incubator, 200 μl of 0.5 mg/ml
MTT (Sigma-Aldrich) was dissolved in PBS, added to
each well, and the plate was incubated at 37°Cfor3h.
Then MTT solution was removed, 200 μlDMSOwas
added, and the absorbance of each well was measured
with a microplate reader (BMG LabTech, Germany)
at 570 nm. The half maximal inhibitory concentration
(IC50)andsafedose(EC
100)valuesofOA-α-LA were
analyzed by the Graphpad Instat software. Untreated
cellswereincludedasnegativecontrolandeachtested
sample was performed in triplicate.
Anti-cancer eect of the α-cLA-OA complexes
Cytotoxicity assay against human cancer cell lines
using MTT assay
The anti-cancer eect of OA-α-cLA was assayed
using four human cancer cell lines. Colon cancer cell
line (Caco-2) and prostate cancer cell line (PC-3) were
maintained as adherent cell cultures in DMEM (Lonza,
USA) containing 10% FBS (Gibco, Life Technologies,
Grand Island, NY, USA) while hepatoma cell line
(HepG-2) and breast cancer cell line (MCF-7) were
cultured in RPMI-1640 (Lonza, USA) supplemented
with 10% FBS. All cancer cell suspensions (2.5 ×103
cells/well) were seeded into sterile 96-well plates and
allowed to attach for 24 h. Then serial concentrations
of α-LA, OA-α-LA, OA, and 5-FU were added to
four cancer cell lines, and the plates were incubated
at 37°Cina5%CO
2incubator. After incubation
for 72 h, the sensitivity of tumor cells to OA-α-LA
was evaluated using MTT assay, as described above.
Moreover the anti-cancer activity of OA-α-LA was
investigated by phase contrast microscope in com-
parison with untreated cells as well as α- LA-, OA-,
and 5-FU-treated cells. The IC50 values was calculated
using Graphpad Instat software and the selectivity
index (SI) that was dened as the ratio of the IC50 on
normal mammalian cells (Vero) versus four cancer cell
lines was also calculated.
Flow cytometry analysis of cell death
Human cancer cell lines (HepG2, Caco-2, MCF-7, and
PC3) and normal mammalian cells (Vero) were treated
with IC50 concentration of α-cLA and OA-α-cLA for
72 h in control with IC50 of OA and 5-FU. The cells were
harvested by trypsinization, washed three time with
cold PBS, and resuspended in 200 μlof1×PBS buer.
Then, the cells were incubated with 5 μlofannexin
V-biotin (Molecular ProbesTM,USA)and5μlofPIfor
15 min in the dark. After staining, the cells were washed
with 1 ×PBS and xed with 4% paraformaldehyde in
PBS for 10 min. After washing twice with cold PBS,
5μg/ml of streptavidine-uorescein (Sigma-Aldrich)
was added to the cells for 15 min; the cells were then
centrifuged and resuspended in PBS. The cell death
ratesweredetectedbyowcytometry(Ex=488 nm;
Em =530 nm) using FITC a signal detector (FL1)
and PI staining by a phycoerythrin emission signal
detector (FL2).
Flow cytometry analysis of the cell cycle
The cell cycle of untreated and treated cancer cells was
analyzed by ow cytometry according to the method
of Jass et al. (29). After treatment of cancer cells with
α-cLA and OA-α-cLA at IC50 concentration as
described above, about 1.0 ×106cells were de-attached
Downloaded by [Professor Vladimir N. Uversky] at 16:39 26 September 2017
CANCER INVESTIGATION 5
and suspended in 300 μlofPBS.Then,thecellswere
centrifuged at 1200 rpm at 4°Cfor5min,followedby
xation using the drop-wise addition of 700 μlofcold
ethanol (70%) to each tube while vortexing gently. The
cells can be left at 4°C (up to a few days) after xation.
The cells were centrifuged and washed three time with
cold PBS, resuspended in 250 μl of PBS containing
5μg/ml RNase A (Sigma-Aldrich), and incubated at
37°Cfor1h.Then10μl of 1 mg/ml PI (Sigma-Aldrich)
in deionized water was added and kept in the dark at
4°C until analysis on FACS (Partec, Germany) by read-
ing at 488 nm using Cell Quist and Mod Fit software.
Nuclear staining analysis of anti-cancer effect
The anti-cancer eect of OA-α-cLA was assayed by
uorescent nuclear dye PI (30, 31).Caco-2cellswere
seeded in 12 well plates and treated with OA-α-cLA
for 1 day under suitable conditions as described above.
Thetreatedcellswerewashedthreetimeswithcold
1.0 ×PBS, and xed with cold 4% paraformalde-
hyde for 10 min. After the cells permeabilization with
3% paraformaldehyde and 0.5% Triton X-100, the
cells were stained with PI dye (10 μg/ml). The cells
were viewed and counted after 15 min with a uo-
rescence inverted microscope with an excitation lter
(480/30 nm) and all tested samples were done in tripli-
cate. Untreated cells were involved as negative control
cells,andthecellshavingcondensedandfragmented
nuclei were considered apoptotic.
Tyrosine kinase (TK)–inhibition assay
The antibody BeaconTM TK assay kit (Molecular
Probes, USA) provides a simple and robust assay for
measuring the activity of TK and their inhibitors
and modulators. According to the instructions from
the manufacturer of the corresponding analytical kit,
12.5 μlofserialdilutionsofα-cLA, OA-α-cLA,OA,
and 5-FU were incubated with 12.5 μlofTK(25U/ml
in reaction buer) for 20 min at 37°C. The Antibody
Beacon TK detection complex (62.5 nM Oregon Green
488 ligand and 125 nM antiphosphotyrosine antibody),
500 μg/ml kinase substrate (poly(Glu:Tyr), 4:1), and
5μlof5mMATPwerethenaddedtoeachwell,
andthereactionswereincubatedat37°C. After 1 h,
the quenched uorescence was measured in a uo-
rescence microplate reader (BMG LabTech, Germany)
using excitation at 485 nm and emission at 535 nm for
estimation of TKi (the concentration required to cause
half maximum inhibition of enzyme activity).
Statistical analysis
Data were expressed as mean ±standard error of
the mean (SEM) for three measurements by the mul-
tiple comparisons Fisher’s least signicant dierence
(LSD)posthoctestoftheonewayanalysisofvari-
ance (ANOVA) using the SPSS16 software program,
with probability (p)-values <0.05 considered statisti-
cally signicant.
Results
In vitro cytotoxicity eects of OA-α-cLA complex on
mammalian Vero cells
We evaluated the viability of Vero cells after treatment
with α-cLA, OA, OA-α-cLA, and uorouracil (5-FU).
The results of these analyses are given in Table 1 ,which
clearly shows that the safe dose (EC100)fortheα-cLA
protein and OA-α-cLA complex were signicantly
(p<0.001) higher than the corresponding values
determined for OA and 5-FU. Furthermore, for OA
and 5-FU, the viability of Vero cells was decreased to
50 percent (IC50)atconcentrationslessthan18μM,
respectively, whereas for α-cLA and OA-α-cLA, the
IC50 values were 288.9 ±1.9 μM and 170.2 ±9.6 μM,
respectively (shown in Tab l e 1). These ndings indi-
cated a highly signicant (p<0.001) increase (by more
than 19 folds) in the safety of using the OA-α-cLA com-
plex instead of OA alone toward normal Vero cells.
In vitro cytotoxicity of OA-α-cLA complex on dierent
cancer cell lines
In vitro cytotoxicity of OA-α-cLA complex was inves-
tigated using dierent human cancer cell lines after
treatment for 72 h and using treatment with α-cLA,
OA, and 5-FU as a control. The cell viability and levels
of the compound cytotoxicity were estimated by the
MTT method. All tested cancer cell lines were found
Tab le . IC (µM) and EC (µM) values for effect of camel α-LA
and OA- α-LA on normal epithelial cell line (Vero) in control with
OA and -FU.
Sample EC IC
α-LA  ±.a. ±.a
OA- α-LA . ±.b. ±.b
OA . ±.c. ±. c
-FU . ±.d. ±. d
All values were expressed as mean±SEM. Different letters are significantly dif-
ferent within the same column at p<..
Downloaded by [Professor Vladimir N. Uversky] at 16:39 26 September 2017
6V. N. UVERSKY ET AL.
Tab le . IC (µM) and SI values for cytotoxic effect of camel α-LA and OA-α-LA complex against Caco-, HepG-, PC and MCF- cell lines
in control with OA and -FU.
Caco- HepG- PC- MCF-
IC SI IC SI IC SI IC SI
α-LA . ±.a. ±.a. ±.a. ±.a. ±a. ±. a. ±.a. ±. a
OA- α-LA . ±.b. ±.a. ±.b. ±.b. ±.b. ±. b. ±. b. ±. b
OA . ±.c. ±.b. ±.c. ±.c. ±.c. ±.c. ±. c. ±. c
-FU . ±.d. ±.b. ±.d. ±.c. ±.d. ±. c. ±. d. ±. c
All values were expressed as mean ±SEM. Different letters are significantly different within the same column at p<..
to be sensitive to OA-α-cLA complex at low concen-
trations, whereas free α-cLA showed cytotoxicity at
high concentrations (p<0.01). Tabl e 2 represents
the concentrations of OA-α-cLA complex and α-cLA
alone that were required to kill or reach 50% inhibition
the cells were varied according to the type of the cancer
cells. The MCF-7 cells were the most sensitive to the
treatment with OA-α-cLA, with the IC50 of 45.9 μM
and with substrate inhibition (SI) of 3.7, followed by
the HepG-2 cells, with IC50 of 70.5 μMandSIof
2.41, whereas the PC-3 and Caco-2 cells were the least
sensitive to the OA-α-cLA treatment, showing the IC50
value >80 μM and SI of 2 and 1.45, respectively. Free
α-cLA had a cytotoxic eect on all tested cancer cells
at high concentrations, with the IC50 values ranging
from 118.4 to 254.8 μMandtheSIvaluesrangingfrom
1.18 ±0.22 to 2.5 ±0.1. Our results revealed that the
oleic acid and 5-FU showed the highest cytotoxicity
against all tested cells including normal and cancer
cells, being characterized by the lowest IC50 values
(>66 and 20 μM, respectively) and the lowest SI values
below 0.85.
Figure 2 represents the results of the comparative
morphological analysis of human cancer cells of four
dierent cell lines untreated and treated with the IC50
concentrations of α-cLA, OA-α-cLA complex, OA, and
Figure . Morphological changes of human cancer cell lines (І) Caco- cells, (ІІ) HepG- cells, (ІІІ)PC-and(ІV) MCF- cells. (A) Untreated
cells, (B) cells after treatment with α- LA, (C) OA-α-LA, (D) OA, and (E) -FU (magnification ×).
Downloaded by [Professor Vladimir N. Uversky] at 16:39 26 September 2017
CANCER INVESTIGATION 7
Figure . Annexin/PI staining of cell lines (І) Caco- cells, (ІІ) HepG- cells, (ІІІ) PC-, (ІV) MCF- cells and (V) Vero cells. (A) Untreated cells,
cells after treatment with (B) α-LA, (C) OA-α-LA, (D) OA, and (E) -FU.
5-FU. These photomicrographs show that the mor-
phologies of the cancer cells were extremely altered as
aresultofthetreatmentwiththeOA-α-cLA complex,
OA-, or 5-FU, in comparison with the untreated cells
or free α-cLA-treated cells. Treatment with the OA-α-
cLA complex caused cells to become smaller in size,
and their cytoplasm to become denser, perhaps due to
cell apoptosis.
Apoptotic eect of OA-α-cLA complex assayed by
ow cytometry
The apoptosis-dependent anticancer activity of OA-
α-cLA complex at IC50 concentration was evaluated
using a double staining annexin-V/PI method using α-
cLA, OA, and 5-FU at IC50 concentrations as controls.
Figure 3 and Table 3 show that the OA-α-cLA complex
has the ability to induce apoptosis in all tested cancer
cell lines. At IC50 concentration, MCF-7 cells were the
most sensitive to OA-α-cLA complex, which induced
early apoptosis in 65.9% and late apoptosis in 8.2%
of the cells, followed by HepG-2 cells, in which early
and late apoptosis was induced in 45.1% and 10.7% of
thecells,respectively.Thepercentageoflateapoptotic
PC-3 cells (28%) was two-fold higher than the percent
of early apoptotic PC-3 cells after treatment with OA-
α-cLA. The colon cancer cells (Caco-2) were the least
sensitive to the treatment with OA-α-cLA complex,
Downloaded by [Professor Vladimir N. Uversky] at 16:39 26 September 2017
8V. N. UVERSKY ET AL.
Tab le . Annexin/PI analysis of samples-induced apoptosis in Caco-, HepG-, PC- and MCF- cells as well as normal cell line (Vero).
Caco- HepG- PC- MCF- Vero
Cells
Apoptosis (%) Early Late Early Late Early Late Early Late Early Late
Untreated control . ±.a. ±.a. ±.a. ±.a. ±. a. ±. a. ±. a. ±. a. ±.a. ±.a
α-LA . ±.b,e . ±.b. ±.b. ±. b. ±.b. ±. b. ±.b. ±.b. ±.a. ±.b
OA-α-LA . ±. c. ±.c. ±.b. ±.c. ±.c. ±.c. ±.c. ±.c. ±.a. ±.c
OA . ±.d. ±.d. ±.a. ±.d. ±.d. ±.d. ±.d. ±.d. ±.b. ±.d
-FU . ±.e. ±.e. ±.c. ±.e. ±.e. ±.e. ±.e. ±.e. ±. c. ±.e
All values were expressed as mean ±SEM. Different letters are significantly different within the same column at p<..
since early and late apoptosis was induced in 27.8 and
3% of the cells, respectively. Furthermore, free α-cLA,
being present at IC50 concentrations, was able to induce
apoptosis in all cancer types, although they were less
ecient than the OA-α-cLA complex (see Figure 3 and
Table 3). In fact, at IC50 concentration of α-cLA, the
percentages of early (late) apoptotic cells in MCF-7,
HepG-2, PC-3, and Caco-2 were 57.6% (15.1), 43.9%
(3.7%), 35.9% (7.7%), and 14.4% (10.3%), respectively.
Additionally, OA and 5-FU had the highest ecacy
to induce the early and late stages of apoptosis in all
dierentcancercellsaswellasinnormalVerocells.
Importantly, although OA-α-cLA possessed higher
apoptosis eciency toward all tested cancer cells than
free α-cLA, no signicant dierence was found in the
percentage of early apoptosis (less than 2.5%) between
the OA-α-cLA- and α-cLA-treated Vero cells (Figure 3
and Table 3).
Analysis of the cell-cycle phases by ow cytometry
Analysis of the scanning of cell cycle phases was
conducted to explore the apoptotic and anti-cancer
mechanisms of OA-α-cLA complex, α-cLA, OA,
and 5-FU at their IC50 concentrations. Both G0/G1
andG2/Mphaseshaveasignicantroleandactas
the main check points in cell cycle progression (32).
Figure 4 and Table 4 show the results of the cell cycle
analysis for four human cancer cell lines before and
after their treatment with dierent compounds. The
percentage of G0/G1 and G2/M phases was around
97% in untreated cancer cells, as shown in Figure 4 and
Table 4. The G0/G1 phase was decreased signicantly
after treatment with IC50 doses of OA-α-cLA complex,
α-cLA, OA, and 5-FU. The sub-G1 phase was detected
and became observed (p<0.01), indicating all tested
compounds were able to induce apoptosis. However,
theaccumulationoftheOA-α-cLA complex-treated
cancercellsintheSphaseandtheG2/Mphasewas
suppressed. Our results indicated that the S phase
was suppressed in the Caco-2 and PC-3 cells after
treatment with OA (Tab le 4 ). Figure 4 and Ta b l e 5
show that the MCF-7 cells were the most sensitive to
the apoptosis induction after the treatment with the
OA-α-cLA complex. In fact, for these cells, a signif-
icant increase in the percentage of the sub-G1 phase
(55.8%) was observed in comparison with the other
studied cancer cells. The MCF-7 cells were followed by
the HepG-2 and PC-3 cells, for which 48.4 ±1.5% and
48.2 ±0.3% of the cell population were in the sub-G1
phase, respectively. The least sensitive were Caco-2
cells, with 30.3 ±0.4% of the cell population being in
the sub-G1 phase. Free α-cLA at IC50 dose was also
able to induce apoptosis in all analyzed cells, albeit less
eciently (p<0.001) than the OA-α-cLA complex.
More importantly, this OA-α-cLA complex was able to
reinforce all studied human cancer cell lines to enter
sub-G1 phase by two folds in control with α-cLA.
Apoptotic eects of OA-α-cLA complex on colon
cancer cells
Further support to the strong apoptotic and anti-cancer
potential of the OA-α-cLA complex was provided by
the analysis of morphological changes in Caco-2 cells
using PI dye for nuclear staining. PI dye binds to DNA
fragments and is used for identifying the apoptotic
eectofdrugs.AlthoughPIdyebindstoplasmamem-
brane and enters inside the intact cells, the eciency of
its entrance to the nucleus dramatically increases after
the cell integrity becomes disrupted. In the nucleus, PI
forms complexes with DNA which make the nucleus
highly uorescent and easily detectable in uores-
cence microscopy. Figure 5 shows that the Caco-2
cells lose their normal spindle shape and became more
round after treatment with the OA-α-cLA complex
for 24 h, than the cells treated by free α-cLA at the
IC50 concentrations for both compounds. Figure 5 also
shows that the nuclei became more condensed and
the chromatin was fragmented after the cell treatment
Downloaded by [Professor Vladimir N. Uversky] at 16:39 26 September 2017
CANCER INVESTIGATION 9
Figure . Cell cycle analysis of cell lines (І) Caco- cells, (ІІ) HepG- cells, (ІІІ)PC-and(ІV) MCF- cells. (A) Untreated cells, (B) cells after
treatment with α-LA, (C) OA-α-LA, (D) OA, and (E) -FU.
Tab le . Cell cycle analysis of samples-treated human cancer cell lines.
Cell lines Cell cycle phase Untreated control α-LA OA-α-LA OA -FU
Caco- Sub G . ±. a. ±. b. ±. c. ±. d. ±. e
G/G . ±. a. ±. b. ±. c. ±. d. ±. e
S.±. a. ±. b. ±. b±c. ±. d
G/M . ±. a. ±. b. ±. b±c. ±. b
HepG- Sub G . ±. a. ±. b. ±. c. ±. d. ±. c
G/G . ±. a. ±. b. ±. c. ±. d. ±. e
S.±. a. ±. b. ±. b. ±. c. ±. d
G/M . ±. a. ±. b. ±. c. ±. d. ±.e
PC- Sub G . ±. a. ±. b. ±. c. ±. d. ±. e
G/G . ±. a. ±. b. ±. c. ±. d. ±. e
S.±. a. ±. b. ±. c±d. ±. e
G/M . ±. a. ±. b. ±. c±d. ±. e
MCF- Sub G . ±. a. ±. b. ±. c. ±. d. ±. e
G/G . ±. a. ±. b. ±. c. ±. d. ±. e
S.±. a. ±. b. ±. c. ±. d. ±. e
G/M . ±. a. ±. b. ±. c. ±. d. ±. e
All values were expressed as mean ±SEM. Different letters are significantly different within the same raw at p<..
Downloaded by [Professor Vladimir N. Uversky] at 16:39 26 September 2017
10 V. N. UVERSKY ET AL.
Figure . Photograph of Caco- cells stained with PI dye under a light microscope (A) control “untreated” cells, cells exposed to IC (μM)
of (B) α-LA, (C) OA- α-LA,(D)OA,and(E)-FU.
with OA-α-cLA complex, whereas the untreated cells
were characterized by a normal cell shape and intact
nuclei, with a very minor number of cells being PI pos-
itive. It was observed that the nuclear condensation,
chromatin fragmentation, and shedding of apoptotic
bodies were the major features of the apoptotic eect in
the case of the treatment with OA-α-cLA complex, and
that these eects more noticeable than those caused by
the treatment with α-cLA or 5-FU.
Inhibition eect of OA-α-cLA complex on tyrosine
kinase
TheeectiveanticanceractivityofOA-α-cLA complex
may be attributed to the inhibition of the TK, as shown
in Table 5. In fact, data presented in Tab l e 5 demon-
strate that all the compounds tested were potent TK
inhibitors, excluding free α-cLA, which had the low-
est in vitro inhibitory activity. According to the Ki val-
ues, OA-α-cLA was found to be nearly 2.5-fold more
potent than α-cLA as a TK inhibitor. In addition, no
Tab le . The estimated Ki (µM) of α-LA and OA- α-LA for tyrosine
kinase inhibition in control with OA and -FU.
Sample Ki
α-LA . ±.a
OA-α-LA . ±. b
OA . ±. b
-FU . ±. c
All values were expressed as mean ±SEM. Different letters are significantly
different within the same column at p<..
signicant dierence between OA-α-cLA and OA was
observed.
Discussion
Although historically best-known for its ability to bind
calcium and regulate the catalysis of the nal step of the
lactose biosynthesis in the lactating mammary gland,
α-LA, which might constitute up to 20% of total milk
protein, is now recognized to have a multitude of bio-
logical activities (33–35),includingfattyacidbinding
(15, 36–38). It was pointed out that complexes of this
protein with some fatty acids (such as oleic or linoleic
acids), which are known now as HAMLET-like anti-
cancer compounds (39) or liprotides; i.e., complexes
consisting of protein and fatty acids (39, 40),possess
selective anticancer activity (6–13, 35, 39).α-LA-based
liprotides are believed to be formed as a result of bind-
ing of fatty acid molecules to the hydrophobic core
of the partially unfolded and aggregated protein (39),
where a micellar OA core is surrounded by a shell com-
prised of the partially unfolded protein molecules (41).
The HAMLET-like anticancer compounds were shown
to possess profound cytotoxicity and anticancer activ-
ity against all of the 50 dierent cancer cell types, for
which the representatives of the HAMLET family have
been trialed (39).
The majority of research on the anticancer activity
of α-LA-basedliprotideswasconductedusinghuman
and, to a much lesser degree, bovine protein, whereas
Downloaded by [Professor Vladimir N. Uversky] at 16:39 26 September 2017
CANCER INVESTIGATION 11
anticancer and pro-apoptotic activities of the com-
plexes between OA and camel α-LA are studied to a
muchlesserdegree.Infact,asearchofPubMedforarti-
cles discussing human, bovine, or camel LA and can-
cer generated 248, 55, and 3 hits, respectively. This is
instarkcontrasttothebroaduseofcamelmilkand
related products as an important nutritional source in
several world regions. Previous in vivo and in vitro
studiesreportedthepotencyofmilkproteinsthatwere
obtained from camel in halting breast, liver, and colon
cancer progression (42–44). It is therefore likely that
camel α-LA may have anticancer activity, in contrast to
the anticancer eects of HAMLET and BAMLET that
attributed only to OA (44, 46). Therefore, this study was
conductedtotesttheapplicabilityofcamelα-LA com-
plexed with OA as anticancer compounds with pro-
apoptotic activities against various cancer cells. These
eects were compared with those imposed on nor-
mal Vero and cancer cells by α-LA and OA alone, as
well as by a well-known anti-cancer drug 5-uorouracil
(5-FU).
Our analysis revealed that although OA, 5-FU,
and OA-α-cLA showed noticeable cytotoxic eects
on all studied cancer cells when compared to α-cLA
alone, OA-α-cLA was the only compound that had
the minimal eects on the viability of normal cells
with the highest SI. However, both OA and 5-FU
showed signicant cytotoxicity against Vero cells
with the lowest SI. These results illustrated that this
complex potentiated the cytotoxic activity of protein
portion (α-cLA) against cancer cells and improved
the selectivity of fatty acid portion (OA) to be more
specic to cancer cells rather than normal cells. Beside
amoderateanticancereectofcamelα-LA, previous
studies declared that protein portions of OA-α-LA
complex play an essential role in transporting OA
from membrane to cytoplasm then accumulated in
nucleus, subsequently causing nuclear shrinkage and
damage (47, 48).Aswasshowninthisstudy,PIpen-
etrated and stained the nucleus of treated cancer cells,
particularly those with an OA-α-cLA complex. We also
showed that the OA-α-cLA complex serves as an e-
cientinductorofcelldeathintheinvestigatedcancer
cell lines by causing severe damage in their morphol-
ogy, signicant increase of annexin-stained apoptotic
cell populations, and induction of cell cycle arrest
at lower IC50 than that of α-cLA. Our recent study
proved that OA-α-LA complex mediated cancer cell
death through induction of apoptosis (49).Also,other
previous studies demonstrated the anticancer activi-
tiesofHAMLETandBAMLETmediatedmainlyvia
apoptosis (45, 50).
Here, the observed cell cycle arrest and apoptosis
induction may be related to the capacity of OA-α-
cLAfortheTKinhibitionatlowconcentrationlike
OA. TK activation is required for several fundamen-
tal processes in cancer cells, such as oncogenic activa-
tion, angiogenesis, apoptosis inhibition, uncontrolled
cell cycle, and abnormal proliferation in several tumor
types. Therefore, TK inhibitors are considered as eec-
tive anticancer agents through targeting and disrupting
cancer cell signaling pathways and subsequent apopto-
sis induction (51–53). Khamaisie et al. (54) found that
oleic acid, being one of active component of mushroom
Daedalea gibbosa extract, inhibited TK autophospho-
rylation that mediated its anti-leukemic eect in an ani-
malmodel.OurresultsrevealedthattheOA-α-cLA
complex inhibited the activity of tyrosine kinase as e-
ciently as OA, while free α-cLA showed less inhibition
activity for tyrosine kinase.
Conclusion
We show here that camel α-cLA is able to bind OA
toformacomplex,whichcanbenamedCAMLET
(camel α-lactalbumin made lethal to tumor cells), with
a highly selective anticancer activity against four cancer
cells, associated with tumors in diverse tissues of the
humanbody,andcharacterizedbyavariablesensitivity.
The selectivity and potency of the anticancer activity
of CAMLET are related to α-cLA and OA, respectively.
The OA-α-cLA complex destroys cancerous cells, par-
ticularlybreastcancercells,byenhancingtheselective
apoptosic process and causing arrest of the cell cycle.
Unlike OA, this complex is able to specically induce
apoptosis only in cancer cells. All tested cancer cells in
G0/G1 and G2/M phase were intensely reduced, while
the cells in sub G1 phase were dramatically induced
when treated with IC50 of OA-α-cLA, compared to the
untreated control cells. Furthermore, the OA-α-cLA
complex has the ability to induce apoptosis and cell
cycle arrest through inhibition of tyrosine kinase activ-
ity.Thisstudymayprovidefoundationsforthedevel-
opment of a competent and cost-eective approach
fortheanalysisofproteinsthatformcomplexeswith
small molecules, such as oleic acid, characterized
by new functions and highly promising biological
activities.
Downloaded by [Professor Vladimir N. Uversky] at 16:39 26 September 2017
12 V. N. UVERSKY ET AL.
Declaration of interest
The authors report no declarations of interest.
Acknowledgments
This work was supported in part by a grant from the King Abdu-
laziz University (56-130-35- HiCi). We are thankful to Alexey
Uversky for careful reading and editing this manuscript.
ORCID
Vladimir N. Uversky http://orcid.org/0000-0002-4037-5857
Esmail M. El-Fakharany http://orcid.org/0000-0001-8246-
0075
References
1. Chen HY, Mollstedt O, Tsai MH, Kreider RB. Poten-
tial clinical applications of multi-functional milk proteins
and peptides in cancer management. Curr Med Chem.
2014;21:2424–2437.
2. Svensson M, Duringer C, Hallgren O, Mossberg AK,
Hakansson A, Linse S, et al. Hamlet–a complex from
human milk that induces apoptosis in tumor cells but
spares healthy cells. Adv Exp Med Biol. 2002;503:125–132.
3. Hakansson AP, Roche-Hakansson H, Mossberg AK, Svan-
borg C. Apoptosis-like death in bacteria induced by HAM-
LET, a human milk lipid-protein complex. PLoS One.
2011;6:e17717.
4. Yoo YC, Watanabe R, Koike Y, Mitobe M, Shimazaki K,
Watanabe S, et al. Apoptosis in human leukemic cells
induced by lactoferricin, a bovine milk protein-derived
peptide: involvement of reactive oxygen species. Biochem
Biophys Res Commun. 1997;237:624–628.
5. Hakansson A, Zhivotovsky B, Orrenius S, Sabharwal H,
Svanborg C. Apoptosis induced by a human milk protein.
Proc Natl Acad Sci U S A. 1995;92:8064–8068.
6. Casbarra A, Birolo L, Infusini G, Dal Piaz F, Svensson
M, Pucci P, et al. Conformational analysis of HAMLET,
the folding variant of human alpha-lactalbumin associated
with apoptosis. Protein Sci. 2004;13:1322–1330.
7. Fischer W, Gustafsson L, Mossberg AK, Gronli J, Mork
S, Bjerkvig R, et al. Human alpha-lactalbumin made
lethal to tumor cells (HAMLET) kills human glioblas-
toma cells in brain xenografts by an apoptosis-like
mechanism and prolongs survival. Cancer Res. 2004;64:
2105–2112.
8. Mossberg AK, Hun Mok K, Morozova-Roche LA, Svan-
borg C Structure and function of human alpha-lactalbumin
made lethal to tumor cells (HAMLET)-type complexes.
FEBS J. 2010;277:4614–4625.
9. Mossberg AK, Puchades M, Halskau O, Baumann A,
LanekoI,ChaoY,etal.HAMLETinteractswithlipid
membranes and perturbs their structure and integrity.
PLoS One. 2010;5:e9384.
10. Mossberg AK, Hou Y, Svensson M, Holmqvist
B, Svanborg C HAMLET treatment delays blad-
der cancer development. J Urol. 2010;183:1590–
1597.[https://doi.org/10.1016/j.juro.2009.12.008]
PMID:20172551
11. Gustafsson L, Leijonhufvud I, Aronsson A, Mossberg
AK, Svanborg C Treatment of skin papillomas with
topical alpha-lactalbumin-oleic acid. N Engl J Med.
2004;350:2663–2672.
12. MossbergAK,WulltB,GustafssonL,ManssonW,Ljung-
gren E, Svanborg C Bladder cancers respond to intravesical
instillation of HAMLET (human alphalactalbumin made
lethal to tumor cells). Int J Cancer. 2007;121:1352–1359.
13. Puthia M, Storm P, Nadeem A, Hsiung S, Svanborg C Pre-
vention and treatment of colon cancer by peroral admin-
istration of HAMLET (human alpha-lactalbumin made
lethal to tumor cells). Gut 2014;63:131–142.
14. Pettersson J, Mossberg AK, Svanborg C alpha-Lactalbumin
species variation, HAMLET formation, and tumor cell
death. Biochem Biophys Res Commun. 2006;345:
260–270.
15. Atri MS, Saboury AA, Moosavi-Movahedi AA, Goliaei B,
SedbakhtY,AlijanvandHH,etal.Structureandstability
analysis of cytotoxic complex of camel alpha-lactalbumin
and unsaturated fatty acids produced at high temperature.
J Biomol Struct Dyn. 2011;28:919–928.
16. Permyakov SE, Pershikova IV, Khokhlova TI, Uversky
VN, Permyakov EA No need to be HAMLET or BAM-
LET to interact with histones: binding of monomeric
alpha-lactalbumin to histones and basic poly-amino acids.
Biochemistry2004;43:5575–5582.
17. Svensson M, Fast J, Mossberg AK, Duringer C, Gustafsson
L, Hallgren O, et al. Alpha-lactalbumin unfolding is not
sucient to cause apoptosis, but is required for the conver-
sion to HAMLET (human alpha-lactalbumin made lethal
to tumor cells). Protein Sci. 2003;12:2794–2804.
18. Nakamura T, Aizawa T, Kariya R, Okada S, Demura M,
Kawano K, et al. Molecular mechanisms of the cytotoxic-
ity of human alphalactalbumin made lethal to tumor cells
(HAMLET) and other protein-oleic acid complexes. J Biol
Chem. 2013;288:14408–14416.
19. TolinS,DeFranceschiG,SpolaoreB,FrareE,CantonM,
Polverino de Laureto P, et al. The oleic acid complexes of
proteolytic fragments of alpha-lactalbumin displayy apop-
totic activity. FEBS J. 2010;277:163–173.
20. Min S, Meehan J, Sullivan LM, Harte NP, Xie Y, Davey GP,
et al. Alternatively folded proteins with unexpected bene-
cial functions. Biochem Soc Trans. 2012;40:746–751.
21. Redington JM, Breydo L, Almehdar HA, Redwan EM,
UverskyVNalpha-Lactalbumin:ofcamelsandcows.Pro-
tein Pept Lett. 2016;23:1072–1080.
22. Almahdy O, El-Fakharany EM, El-Dabaa E, Ng TB, Red-
wan EM Examination of the activity of camel milk casein
against hepatitis C virus (genotype-4a) and its apoptotic
Downloaded by [Professor Vladimir N. Uversky] at 16:39 26 September 2017
CANCER INVESTIGATION 13
potential in hepatoma and hela cell lines. Hepat Mon.
2011;11:724–730.
23. Atri MS, Saboury AA, Moosavi-Movahedi AA, Kavousi K,
Ariaeenejad S Eects of zinc binding on the structure and
thermal stability of camel alpha-lactalbumin. J Therm Anal
Calorim. 2015;120:481–488.
24. Fang B, Zhang M, Tian M, Jiang L, Guo HY, Ren FZ
Bovine lactoferrin binds oleic acid to form an anti-tumor
complex similar to HAMLET. Biochim Biophys Acta.
2014;1841:535–543.
25. Duncombe WG The colorimetric micro-determination of
long-chain fatty acids. Biochem J. 1963;88:7–10.
26. Redwan E Simple, sensitive and quick protocol to detect
less than 1 ng of bacterial lipopolysaccharide. Prep
Biochem Biotechnol. 2012;42:171–182.
27. Vistica DT, Skehan P, Scudiero D, Monks A, Pittman A,
Boyd MR Tetrazolium-based assays for cellular viability:
a critical examination of selected parameters aecting for-
mazan production. Cancer Res. 1991;51:2515–2520.
28. Mosmann T Rapid colorimetric assay for cellular growth
and survival: application to proliferation and cytotoxicity
assays. J Immunol Methods. 1983;65:55–63.
29. Jass JR, Mukawa K, Goh HS, Love SB, Capellaro D Clinical
importance of DNA content in rectal cancer measured by
ow cytometry. J Clin Pathol. 1989;42:254–259.
30. Cao P, Cai X, Lu W, Zhou F, Huo J Growth inhibition and
induction of apoptosis in SHG-44 Glioma cells by chinese
medicine formula “Pingliu Keli”. Evid Based Complement
Alternat Med. 2011;2011:Article ID 958243, 9 pages.
31. Shanmugaprakash M, Jayashree C, Vinothkumar V,
Senthilkumar SNS, Siddiqui S, Rawat V, et al. Biochemical
characterization and antitumor activity of three phase
partitioned L-asparaginase from Capsicum annuum L. Sep
Purif Technol. 2015;142:258–267.
32. Chakravarti B, Maurya R, Siddiqui JA, Kumar Bid H,
Rajendran SM, Yadav PP, et al. In vitro anti-breast cancer
activity of ethanolic extract of Wri ght i a tom e nto s a:roleof
pro-apoptotic eects of oleanolic acid and urosolic acid. J
Ethnopharmacol. 2012;142:72–79.
33. Permyakov EA, Permyakov SE, Breydo L, Redwan EM,
Almehdar HA, Uversky VN Disorder in milk proteins:
alpha-lactalbumin. Part C. peculiarities of metal binding.
Curr Protein Pept Sci. 2016;17:735–745.
34. Permyakov EA, Permyakov SE, Breydo L, Redwan EM,
Almehdar HA, Uversky VN Disorder in milk proteins:
alpha -lactalbumin. Part A. Structural properties and con-
formational behavior. Curr Protein Pept Sci. 2016;17:352–
367.
35. Uversky VN, Permyakov SE, Breydo L, Redwan EM, Alme-
hdar HA, Permyakov EA Disorder in milk proteins: alpha-
lactalbumin. Part B. A multifunctional whey protein acting
as an oligomeric molten globular “Oil Container” in the
anti-tumorigenic drugs, liprotides. Curr Protein Pept Sci.
2016;17:612–628.
36. Wehbi Z, Perez MD, Dalgalarrondo M, Sanchez L, Calvo
M, Chobert JM, et al. Study of ethanol-induced confor-
mationalchangesofholoandapoalphalactalbuminby
spectroscopy and limited proteolysis. Mol Nutr Food Res.
2006;50:34–43.
37. Barbana C, Sanchez L, Perez MD Bioactivity of alpha-
lactalbumin related to its interaction with fatty acids: a
review. Crit Rev Food Sci Nutr. 2011;51:783–794.
38. Brinkmann CR, Thiel S, Otzen DE Protein-fatty acid com-
plexes: biochemistry, biophysics and function. FEBS J.
2013;280:1733–1749.
39. Rath EM, Du AP, Hakansson AP, Vacher CS, Liu GJ,
Knott RB, et al. Structure and potential cellular targets
of HAMLET-like anti-cancer compounds made from milk
components. J Pharm Pharm Sci. 2015;18:773–824.
40. PedersenJN,PedersenJS,OtzenDETheuseofliprotidesto
stabilize and transport hydrophobic molecules. Biochem-
istry. 2015;54:4815–4823.
41. Frislev HS, Jessen CM, Oliveira CL, Pedersen JS, Otzen DE
Liprotides made of alpha-lactalbumin and cis fatty acids
form core-shell and multi-layer structures with a common
membrane-targeting mechanism. Biochim Biophys Acta.
2016;1864:847–859.
42. El Miniawya HMF, Ahmed KA, Mansourc SA, Khattab
MMS Camel milk inhibits murine hepatic carcinogenesis,
initiated by diethylnitrosamine and promoted by pheno-
barbitone. Pharm Biol. 2017;55:1513–1520.
43. Korashy HM, Maayah ZH, Abd-Allah AR, El-Kadi AOS,
Alhaider AA J Biomed Biotechnol. 2012;2012:1–9.
44. Habib HM, Ibrahim WH, Schneider-Stock R, Hassan
HM Camel milk lactoferrin reduces the proliferation of
colorectal cancer cells and exerts antioxidant and DNA
damage inhibitory activities. Food Chem. 2013;141(1):
148–152.
45. TolinS,DeFranceschiG,SpolaoreB,FrareE,CantonM,de
Polverino LP, et al. The oleic acid complexes of proteolytic
fragments of alpha-lactalbumin display apoptotic activity.
FEBS J. 2010;277: 163–173.
46. Hoque M, Dave S, Gupta P, Saleemuddin M Oleic acid
may be the key contributor in the BAMLET-induced ery-
throcyte hemolysis and tumoricidal action. PLoS One.
2013;8:e68390–e68405.
47. FangB,ZhangM,WuH,FanX,RenFInternalizationprop-
erties of the anti-tumor α-lactalbumin-oleic acid complex.
Int J Biol Macromol. 2017;96:44–51.
48. Nakamura T, Aizawa T, Kariya R, Okada S, Demura M,
Kawano K, et al. Molecular mechanisms of the cytotoxicity
of human α-lactalbumin made lethal to tumor cells (HAM-
LET) and other protein-oleic acid complexes. J Biol Chem
2013;288(20):14408–14416.
49. Uversky VN, Permyakov SE, Breydo L, Redwan EM, Alme-
hdar HA, Permyakov EA Disorder in milk proteins: α-
lactalbumin. part b. a multifunctional whey protein act-
ing as an oligomeric molten globular “oil container” in the
anti-tumorigenic drugs, liprotides. Curr Protein Pept Sci.
2016;17(6):612–628.
50. Svanborg C, Agerstam H, Aronson A, Bjerkvig R, Duringer
C, Fischer W, et al. HAMLET kills tumor cells by
apoptosis-like mechanism-cellular, molecular, and thera-
peutic aspects. Adv Cancer Res. 2003;88:1–29.
Downloaded by [Professor Vladimir N. Uversky] at 16:39 26 September 2017
14 V. N. UVERSKY ET AL.
51. Paul MK, Anup K, Mukhopadhyay AK Tyrosine
kinase—role and signicance in cancer. Int J Med
Sci. 2004;1(2):101–115.
52. Vlahovic G, Crawfor J Activation of tyrosine kinases in
cancer. Oncologist. 2003;8:531–538.
53. El-Miligy MM, Abd El Razik HA, Abu-Serie MM. Synthe-
sis of piperazine-based thiazolidinones as VEGFR2 tyro-
sine kinase inhibitors inducing apoptosis. Fut Med Chem.
2017 [in press].
54. Khamaisie H, Sussan S, Tal M, Najajreh Y, Ruthardt
M, Mahajna J Oleic acid is the active component in
the mushroom Daedalea gibbosa inhibiting Bcr-Abl
kinase autophosphorylation activity. Anticancer Res.
2011;31(1):177–183.
Downloaded by [Professor Vladimir N. Uversky] at 16:39 26 September 2017
... Here, the prepared albumin-coated liposomes were very useful in demonstrating a reduced cytotoxic effect in healthy cells, which is very important in the search for new, less invasive, and more targeted mechanisms for the treatment of HCC. Albumin was initially purified from camel milk through column chromatography and indicated as one single protein band at 67 KDa on 12% SDS-PAGE, which is completely matched with the different albumins' molecular weights (15,38,39). The purified albumin encapsulation into a liposome was evaluated in several ratios, whereas 0.62: 2.2 mg of albumin: liposome revealed the maximum albumin loading. ...
... The results asserted the ability of both uncoated and albumin-coated liposomes to induce apoptosis (40.61 and 80.7%) in treated HepG-2 cells as compared to free OA (27.46%). The results are in accordance with other studies that reported the potency of albumin-OA for apoptosis induction by inhibiting tyrosine kinase (TK) activity in treated tumor cells (15,38). Moreover, albumin-coated liposomes significantly exhibited the maximum potential for down-regulation of NF-kB and Bcl2 genes compared with uncoated liposomes and free OA in the treated HepG-2 cells. ...
Article
Full-text available
Hepatocellular carcinoma (HCC) is one of the most lethal and prevalent cancers, closely associated with cirrhosis and fibrosis. This study aimed to assess the antitumor activity of oleic acid-liposomes (uncoated liposomes) upon coating with albumin against HCC. The in vitro studies revealed the high safety of the prepared uncoated and albumin-coated liposomes to normal HFB-4 cells (EC100 of 35.57 ± 0.17 and 79.133 ± 2.92 µM, respectively) with significant anticancer activity against HepG-2 cells with IC50 of 56.29 ± 0.91 and 26.74 ± 0.64 µM, respectively. The albumin-coated liposomes revealed superior apoptosis induction potential (80.7%) with significant upregulation of p53 gene expression ( >7.0-folds), compared to OA. The in vivo study revealed that the administration of uncoated or albumin-coated liposomes (100 mg/kg) for six weeks markedly retarded the DENA-induced HCC in Wistar albino rates through regulating the liver enzymes, total bilirubin level, pro-inflammatory cytokines, and oxidative stress. Accordingly, the current study supports the in vitro and in vivo chemo-preventive feature of albumin-coated liposomes against HCC through modulation of apoptosis, improvement of the immune response, reduction of inflammation, and restoration of impaired oxidative stress, which is the first reported to the best of our knowledge.
... A nonnative globule state of α-LA, stabilized by its interaction with oleic acid in a complex named HAMLET (Human Alpha-lactalbumin Made LEthal to Tumor cells), has shown apoptotic activity against cancerous cells (Svensson et al., 1999(Svensson et al., , 2000. More recently, a biological activity similar to HAMLET has been shown also from bovine, equine, porcine, caprine, and camel α-LA complexed with oleic acid (Pettersson et al., 2006;Uversky et al., 2017). Many other biological functions have been demonstrated for α-LA. ...
... According to our study, the biomolecules present in camel milk that is associated with an inhibitory potential against cancer are likely to be lactoferrin, TRAIL (a soluble form of the apoptosis-inducing ligand related to TNF) and α-lactoglobulin, the latter exhibiting its potential upon the combination with oleic acid forming HAMLET in humans (Lal et al, 2020). It has been shown that the OA-α-cLA complex from camel milk does not affect normal cells but has noticeable anti-cancer activity especially, against MCF-7 cells (Uversky et al, 2017). Interestingly, the structure and stability analysis of cytotoxic complex of camel α-lactalbumin and unsaturated fatty acids produced at high temperature has also been studied (Maliheh et al, 2017). ...
Article
Full-text available
Camel milk and its products are gaining higher demand due to their attractive nutritional and therapeutic profile. However, manufacturing of camel milk cheese (CMC) is a challenging task. This study aimed to compare the quality of camel milk cheese made using different starter cultures. Four groups CM (camel milk + mesophilic cultures), CT (camel milk + thermophilic cultures), CBM (camel milk + 10% buffalo milk using mesophilic cultures), and CBT (camel milk + 10% buffalo milk using thermophilic cultures) were created. The cheeses were stored for 60 days at 4 °C to assess their physicochemical, sensory, and proteolytic properties. Results indicated highest moisture content in CM (70.19%) as opposed to the lowest moisture in CBT (55.19%). In addition, CM and CBM revealed lower acidity (0.63% and 0.66%) in contrast with CT and CBT (0.78% and 0.83%, respectively). On the other hand, CBM and CBT exhibited higher contents of protein (21.04, 21.57%) and fat (17.65, 17.70%) during storage. All quality indices of cheese samples significantly declined (P < 0.01) during storage excluding acidity. Sensory analyses revealed that panellists highly preferred CBT treatment followed by CBM and CT. Furthermore, urea‐PAGE and RP‐HPLC were performed for the proteolytic study of cheese samples. The study revealed that the presence of buffalo milk and thermophilic cultures in cheese resulted in a decrease concentration of intact caseins (αs1, β‐, and κ‐caseins) as storage days passed. The degree of proteolysis was found to be higher in CBT compared to CBM, CT, and CM. Based on the results, it was concluded that the use of pasteurisation at 65 °C for 30 min, pH of 5.5, 0.06% CaCl2, and the incorporation of 10% buffalo milk using thermophilic cultures (Lactobaccilus bulgaricus and Streptococcus thermophilus) resulted in desirable quality characteristics.
Article
α-Lactalbumin (α-LA) is an active protein with multiple biological functions, which can provide the body with abundant essential amino acids, such as Trp and Lys. It is the main whey protein in human milk, accounting for about 22% of its total protein, and provides energy and nutrition for infants’ body and brain development. However, the α-LA in bovine milk is relatively low, so bovine α-LA is often added as a supplement to improve α-LA content and the balance of amino acids in infant formula. In addition, more studies have focused on the health benefits of α-LA in humans, proposing novel physiological roles or supplementing more substantial research evidence, including improving metabolism-related chronic diseases, promoting intestinal health, strengthening bone and muscle health, reducing oxidative stress, delaying aging, and enhancing sleep and cognitive performance. Nonetheless, a considerable proportion of the research remains confined to cell and animal experiments, and it remains uncertain whether these findings will be replicated in humans, and at what dosage. This review describes the basic properties and the structural characteristics focusing on α-LA and specifically discusses its digestion and absorption in infants, adults, and the elderly. Further, the review summarizes the beneficial effects and mechanisms of action of α-LA supplementation in both infants and adults, as well as the future applications of α-LA are also discussed.
Article
Extensive research from large prospective cohort studies and meta-analytical investigations over recent decades have consistently indicated that dairy foods have protective effects, reducing the risk of colorectal cancer. Most of the literature has explored the potential role of milk minerals and vitamins in managing colorectal cancer. Yet, there is a paucity of a comprehensive summary of the anticancer attributes of milk protein components and their underlying mechanisms of action. Recent advancements have spotlighted the potential of whey proteins, including β-lactoglobulin, α-lactalbumin, serum albumin, and lactoferrin, as promising candidates for both the prevention and treatment of colorectal cancer. Notably, whey proteins have demonstrated a more pronounced capacity for suppressing carcinogen-induced tumors when compared to casein. Their strong binding affinity enables them to serve as effective carriers for small molecules or drugs targeting colon cancer therapy. Furthermore, numerous studies have underscored the anti-inflammatory and antioxidant prowess of whey proteins in cancer prevention. Additionally, whey proteins have been shown to trigger apoptosis, hinder tumor cell proliferation, and impede metastasis. This comprehensive review, therefore, not only substantiates the significance of incorporating whey protein components into a balanced daily diet but also underscores their potential in safeguarding against the onset and progression of colorectal cancer.
Article
Full-text available
As the most aggressive subtype, triple‐negative breast cancer (TNBC) without definitive targets represents a high probability of metastasis. Nevertheless, the presence of high‐level tumor‐infiltrating lymphocytes in the TNBC tumor microenvironment (TME) suggests patients may benefit from immunotherapy. In this study, bovine α‐lactalbumin coupled with oleic acid that forms tumoricidal lipid–protein complex (BAMLET) is electrostatically stabilized on the surface of amino‐functionalized mesoporous silica nanoparticles (MSN‐NH2). It is found that MSN‐NH2 may cause partial conformational changes of immobilized proteins due to electrostatic interactions. Therefore, BAMLET covered MSN‐NH2 (BMSN) as a drug cargo that can not only induce selectively oncolytic effect but kill tumor cells with rapid kinetics. Moreover, the hemolytic activity of BMSN conjugated with pre‐formed serum protein corona is largely reduced which may avoid the poor hemocompatibility caused by BAMLET. Furthermore, the in vivo study indicates that the combinational use of drug‐loaded BMSN and immune checkpoint small‐molecule inhibitor can efficiently inhibit the growth of solid TNBC tumors and activate immune response by sufficient infiltration of immune cells in the TME and prevent the seeding of circulating tumor cells. Therefore, the constructed BMSN provides an integrated nanotherapeutics and a nanoadjuvant with superior anticancer performance to combat TNBC.
Article
Full-text available
Colorectal cancer (CRC) is a malignant tumor recognized as a major cause of morbidity and mortality throughout the world. Therefore, novel liposomes of oleic acid coated with camel α-lactalbumin (α-LA coated liposomes) were developed for their potential antitumor activity against CRC, both in vitro and in DMH-induced CRC-modeled animal. In vitro results indicated the high safety of α-LA coated liposomes towards normal human cells with potent antitumor activity against Caco-2 cells at an IC50 value of 57.01 ± 3.55 µM with selectivity index of 6.92 ± 0.48. This antitumor activity has been attributed to induction of the apoptotic mechanism, as demonstrated by nuclear condensation and arrest of Caco-2 cells in sub-G1 populations. α-LA coated liposomes also revealed a significant up-regulation of the p53 gene combined with a down-regulation of the Bcl2 gene. Moreover, in vivo results revealed that treatment of induced-CRC modeled animals with α-LA coated liposomes for six weeks markedly improved the CRC-disorders; this was achieved from the significant reduction in the values of AFP, CEA, CA19.9, TNF-α, IL-1β, MDA, and NO coupled with remarkable rise in SOD, GPx, GSH, CAT, and CD4+ levels. The histopathological findings asserted the therapeutic potential of α-LA coated liposomes in the treatment of CRC. Therefore, the present results proved the antitumor activity of α-LA coated liposomes against CRC through the restoration of impaired oxidative stress, improved immune response, and reduced inflammation.Communicated by Ramaswamy H. Sarma.
Article
Full-text available
The present study aims to develop a novel nanocombination with high selectivity against several invasive cancer cells, sparing normal cells and tissues. Bovine lactoferrin (bLF) has recently captured the interest of numerous medical fields owing to its biological activities and well-known immunomodulatory effects. BLF is an ideal protein to be encapsulated or adsorbed into selenium nanocomposites (Se NPs) in order to produce stable nanocombinations with potent anticancer effects and improved immunological functions. The biosynthesis of the functionalized Se NPs was achieved using Rhodotorula sp. strain MZ312359 via a simultaneous bio-reduction approach to selenium sodium salts. The physicochemical properties of Se NPs using SEM, TEM, FTIR, UV Vis, XRD, and EDX confirmed the formation of uniform agglomerated spheres with a size of 18–40 nm. Se NPs were successfully embedded in apo-LF (ALF), forming a novel nanocombination of ALF-Se NPs with a spherical shape and an average nanosize of less than 200 nm. The developed ALF-Se NPs significantly displayed an effective anti-proliferation efficiency against many cancer cells, including MCF-7, HepG-2, and Caco-2 cell lines, as compared to Se NPs and ALF in free forms. ALF-Se NPs showed a significant selectivity impact (> 64) against all treated cancer cells at IC50 63.10 ≤ μg/mL, as well as the strongest upregulation of p53 and suppression of Bcl-2, MMP-9, and VEGF genes. Besides, ALF-Se NPs were able to show the maximum activation of transcrition of key redox mediator (Nrf2) with suppression in reactive oxygen species (ROS) levels inside all treated cancer cells. This study demonstrates that this novel nanocombination of ALF-Se NPs has superior selectivity and apoptosis-mediating anticancer activity over free ALF or individual form of Se NPs.
Article
Full-text available
Aim: VEGFR2 tyrosine kinase is a main target in suppressing cancer growth and metastasis. Materials & methods: Piperazine-based thiazolidinones were synthesized and screened for their anticancer and VEGFR2 tyrosine kinase inhibitory activity. Results: Compounds 11, 13 and 16 displayed potent anticancer activity against HepG-2 with IC50 values 0.03-0.06 μM. They were safe on normal human fibroblasts with selectivity indices 8.09, 11.40 and 4.37, respectively. Also, these compounds showed VEGFR2 tyrosine kinase inhibitory activities more than the reference staurosporine with IC50 values <0.3 μM. Lineweaver-Burk plot revealed that these compounds behaved as uncompetitive VEGFR2 tyrosine kinase inhibitors. They also induced caspase-dependent apoptosis in HepG-2. In addition, these compounds revealed good binding within VEGFR2 tyrosine kinase enzyme in comparison with sorafenib reference. Conclusion: Compounds 11, 13 and 16 comprise a new promising scaffold of selective VEGFR2 tyrosine kinase inhibitors with caspase-dependent apoptotic activities.
Article
Full-text available
Context: Camel milk (CM) is recommended for liver disease patients in Egypt for a strong belief that it has a curative effect. Objective: The effect of consumption of CM with or without chemotherapeutic drug cisplatin was evaluated on induced hepatocarcinogenesis in rats. Materials and methods: Wistar male rats (56) were divided into eight groups (7 rats each). Group I was control. Hepatocarcinogenesis was initiated by a single dose of intraperitoneal injection of diethylnitrosamine (DENA) (200 mg/kg BW) and promoted by phenobarbitone (500 ppm) in drinking water in groups V, VI, VII and VIII. Treatment started from 28th till 38th week using CM (5 mL/day) and/or cisplatin (5 mg/kg/3 weeks) in groups II, III IV, VI, VII and VIII. Biochemical analysis, lipid peroxidation and superoxide dismutase (SOD) activity in liver tissue were performed. Histopathology of liver and kidney and immunohistochemistry of placental glutathione-S-transferase (P-GST) in liver were performed and analyzed using image analysis. Results: Albumin concentration and SOD activity were 3.13 ± 0.23 and 311.45 ± 41.71 in group VII (DENA & cisplatin), whereas they were 4.3 ± 0.15 and 540.5 ± 29.94 in group VII (DENA, CM and cisplatin). The mean area of altered hepatocellular foci and P-GST altered foci decreased in group VI (DENA and CM) (1049.6 ± 174.78 and 829.1 ± 261) and group VIII (cisplatin and CM) (1615.12 ± 436 and 543.9 ± 127) compared to group V (DENA only) (4173.74 ± 510.7 and 3169.49 ± 538.61). Cisplatin caused chronic interstitial nephritis, which was slightly alleviated in group VIII (CM and cisplatin). Conclusions: CM had an antioxidant effect and together with cisplatin managed to decrease hepatocarcinogenesis.
Article
Full-text available
This is a concluding part of the three-part article from a series of reviews on the abundance and roles of intrinsic disorder in milk proteins. In this paper, we describe the peculiarities of metal binding to a multifunctional milk protein, α -lactalbumin, which has two domains, a large α-helical domain and a small β-sheet domain connected by a calcium binding loop. It is known that in addition to four disulfide bonds, the native fold of this protein is stabilized by binding of a calcium ion. In fact, although in various mammals, α-lactalbumins are rather poorly conserved possessing the overall sequence identity of ~16%, the positions of all eight cysteines and a calcium binding site (residues DKFLDDDITDDI in human protein) are strongly conserved. Curiously, this conserved calcium binding loop is located within a region with increased structural flexibility. Besides canonical calcium binding, α-lactalbumin is known to interact with other metals, such as zinc (for which it has a specific binding site), and, in its apo-form, it can bind other divalent and monovalent cations. The binding of Mg2+, Na+, and K+ to the Ca2+ site increases α-lactalbumin stability against action of heat and various denaturing agents, with the higher stabilization effects being imposed by the stronger bound metal ions.
Article
Full-text available
Since camel milk has been attributed with various medicinal properties not found in bovine milk, we are systematically examining the differences between different proteins in bovine and camel milk. The purpose of this study is to investigate the structural differences between the bovine and camel α-lactalbumins. α-Lactalbumin is a highly abundant protein present in the milk of all mammalian species. Here we found several structural differences between bovine and camel α-lactalbumins: camel protein is more stable towards thermal and pH-mediated denaturation but less stable towards guanidine hydrochloride-mediated unfolding, aggregates faster and is predicted to be more disordered than bovine α-lactalbumin.
Article
Full-text available
This is a second part of the three-part article from a series of reviews on the abundance and roles of intrinsic disorder in milk proteins. We continue to describe α-lactalbumin, a small globular Ca2+-binding protein, which besides being one of the two components of lactose synthase that catalyzes the final step of the lactose biosynthesis in the lactating mammary gland, possesses a multitude of other functions. In fact, recent studies indicated that some partially folded forms of this protein possess noticeable bactericidal activity and other forms might be related to induction of the apoptosis of tumor cells. In its anti-tumorigenic function, oligomeric α-lactalbumin serves as a founding member of a new family of anticancer drugs termed liprotides (for lipids and partially denatured proteins), where an oligomeric molten globular protein acts as an "oil container" or cargo for the delivery of oleic acid to the cell membranes.
Article
Full-text available
p>The HAMLET family of compounds (Human Alpha-lactalbumin Made Lethal to Tumours) was discovered during studies on the properties of human milk, and is a class of protein-lipid complexes having broad spectrum anti-cancer, and some specific anti-bacterial properties. The structure of HAMLET-like compounds consists of an aggregation of partially unfolded protein making up the majority of the compound's mass, with fatty acid molecules bound in the hydrophobic core. This is a novel protein-lipid structure and has only recently been derived by small-angle X-ray scattering analysis. The structure is the basis of a novel cytotoxicity mechanism responsible for anti-cancer activity to all of the around 50 different cancer cell types for which the HAMLET family has been trialled. Multiple cytotoxic mechanisms have been hypothesised for the HAMLET-like compounds, but it is not yet clear which of those are the initiating cytotoxic mechanism(s) and which are subsequent activities triggered by the initiating mechanism(s). In addition to the studies into the structure of these compounds, this review presents the state of knowledge of the anti-cancer aspects of HAMLET-like compounds, the HAMLET-induced cytotoxic activities to cancer and non-cancer cells, and the several prospective cell membrane and intracellular targets of the HAMLET family. The emerging picture is that HAMLET-like compounds initiate their cytotoxic effects on what may be a cancer-specific target in the cell membrane that has yet to be identified. This article is open to POST-PUBLICATION REVIEW . Registered readers (see “For Readers”) may comment by clicking on ABSTRACT on the issue’s contents page.</p
Article
α-Lactalbumin (α-LA) can bind oleic acid (OA) to form the anti-tumor α-LA-OA complex. Previous studies suggested α-LA-OA induced apoptosis or autophagy in an independent way. Furthermore, as a large molecule, α-LA-OA could enter tumor cells and accumulated in the nucleus, which was speculated as the basis of its anti-tumor activity. In this study, we evaluated the internalization property of α-LA-OA with and without endocytosis inhibitors using flow cytometry and laser scanning confocal microscopy. It was shown α-LA-OA transported from the cell membrane to the cytoplasm, then accumulated around the nucleus, which consequently began to shrink and condense. The α-LA component only located in the membrane whereas the OA component entered cytoplasm. When pre-treated cells with these inhibitors, the internalization of OA would all decrease while the interaction of α-LA with membrane did not influence. As for the complex, the internalization of α-LA-OA was completely blocked at 4 °C and significantly decreased in the presence of cytochalasin D, an inhibitor of phagocytosis (p < 0.01). In conclusion, the anti-tumor activity of α-LA-OA was mainly dependent on the OA component whereas the internalization mechanism was related to the α-LA component to be temperature-dependent and have a close relationship with the phagocytosis pathway.
Article
α-lactalbumin (aLA) has been shown to form complexes with oleic acid (OA), which may target cancer cells. We recently showed that aLA and several other proteins all form protein-OA complexes called liprotides with a generic structure consisting of a micellar OA core surrounded by a shell of partially denatured protein. Here we report that a heat treatment and an alkaline treatment method both allow us to prepare liprotide complexes composed of aLA and a range of unsaturated fatty acids (FA), provided the FAs contain cis (but not trans) double bonds. All liprotides containing cis-FA form both small and large species, which all consist of partially denatured aLA, though the overall shape of the species differs. Small liprotides have a simple core-shell structure while the larger liprotides are multi-layered, i.e. they have an additional layer of both FA and aLA surrounding the outside of the core-shell structure. All liprotides can transfer their entire FA content to vesicles, releasing aLA as monomers and softening the lipid membrane. The more similar to OA, the more efficiently the different FAs induce hemolysis. We conclude that aLA can take up and transfer a wide variety of FA to membranes, provided they contain a cis-bond. This highlights liprotides as a general class of complexes where both protein and cis-FA component can be varied without departing from a generic (though sometimes multi-layered) core-shell structure.
Article
This is a first part of the two-part article that continues a series of reviews on the abundance and roles of intrinsic disorder in milk proteins. We introduce here α-lactalbumin, a small (Mr 14 200), simple, acidic (pI 4-5), Ca<sup>2+</sup>-binding protein that might constitute up to 20% of total milk protein. Although function (it is one of the two components of lactose synthase that catalyzes the final step of the lactose biosynthesis in the lactating mammary gland), structure (protein has two domains, a large α-helical domain and a small β-sheet domain connected by a calcium binding loop), and folding mechanisms (α-lactalbumin is well-known as a classic example of the molten globule state) of this model globular protein are relatively well understood, α-lactalbumin continues to surprise researchers and clearly continues to have high discovery potential. The goal of this review is to summarize some recent advances in the field of α-lactalbumin research and to analyze the peculiarities of the "intrinsic disorder code" of this protein.