ArticlePDF Available

Anticancer effect of trehalose by modulating oxidative stress, apoptosis and autophagy

Authors:

Abstract

Trehalose is a novel disaccharide sugar that contains two glucose motifs. Nowadays, the major applications of trehalose have applied in the medical field. It has been investigated whether trehalose has inhibitory or protective effects on different types of tumor and non-tumor cells. Trehalose effect depends mainly on type of the target cells. Some studies reported that trehalose could potentially be employed as a novel anticancer agent that could be used safely and effectively to treat tumors by decreasing oxidative stress, autophagy, and increasing apoptosis. Some other studies found that trehalose is a potent autophagy inducer for some other cancer cell types. In this short review, we shed a light on the potential role played by trehalose as an anticancer, antioxidant or oxidative stress inducer, apoptotic or anti-apoptotic, and autophagy inhibitor or inducer agent.
1
Nasr Eldeen et al., 2018, AJMS 2(1):1-3, DOI:10.5455/ajms.13
Anticancer eect of trehalose by modulating oxidative stress, apoptosis and
autophagy
Arabian Journal of Medical Sciences
http://www.ajms.tk
Abstract
Article Info
Keywords:
Trehalose,
Autophagy,
Apoptosis,
Oxidative stress
Received Nov 7,
Revised Dec 2,
Published Dec 7, 2018
*Corresponding author:
sciencesamah@gmail.com
Trehalose is a novel disaccharide sugar that contains two glucose motifs. Nowadays, the major applications
of trehalose have applied in the medical eld. It has been investigated whether trehalose has inhibitory or
protective eects on dierent types of tumor and non-tumor cells. Trehalose eect depends mainly on type
of the target cells. Some studies reported that trehalose could potentially be employed as a novel anticancer
agent that could be used safely and eectively to treat tumors by decreasing oxidative stress, autophagy, and
increasing apoptosis. Some other studies found that trehalose is a potent autophagy inducer for some other
cancer cell types. In this short review, we shed a light on the potential role played by trehalose as an anticancer,
antioxidant or oxidative stress inducer, apoptotic or anti-apoptotic, and autophagy inhibitor or inducer agent.
Samah K Nasr Eldeen a*, Mohammed A. El-Magd b, Abeer Khamis a, Wafaa M. Ibrahim c, Afrah F. Salama a
a Department of Chemistry, Biochemistry Division, Faculty of Science, Tanta University, Egypt
b Department of Anatomy, Faculty Veterinary Medicine, Kafrelsheikh University, Egypt
c Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Egypt
Trehalose structure and properties
Trehalose(α-glucopyranosyl-(1→1)-α-D-glucopyranoside;
C12H22O11; molecular weight 342.31 g/mol) is a disaccharide
found in many microorganisms and plants. It has 45% sweetness
more than sucrose sugar so it is commonly used in the food industry
as a sweetener. There are many isomers of trehalose such as α,β (neo-
trehalose) and β, β (isotrehalose) which have been rst synthesized
in 1994. The α form is the isomer commonly dened as trehalose (α,
α -trehalose, α -D-glucopyranosyl α-D-glucopyranoside, mushroom
sugar, mycose).
Trehalose is very stable disaccharide sugar due to its unique
physicochemical properties, including high degree of optical rota-
tion, thermal stability (unique primary melting point at 97οC, crystal-
lization at 130 οC, and melting of its anhydrous form at 203 ο C) and
wide PH range (Elbein et al., 2003).
Trehalose extraction and production
Trehalose has been rst extracted from yeast in 1994 but this
method was too expensive and produced very low concentration. An-
other method for trehalose extraction was performed in 1995 using
two bacterial enzymes, malto-oligosyltrehalose synthase (or MTSase)
and malto-oligosyltrehalosetrehalohydorolase (or MTHase), isolated
from genus Arthrobacter sp. Q36 in the soil (Maruta et al., 1995).
MTSase catalyzes the transfer of one glucose molecule at the
reducing end of oligosaccharide from α-1, 4 bonds to α-1, 1bond,
to produce oligosyltrehalose, which contains trehalose residue at the
end of the saccharide chain. Subsequently, MTHase broke oligos-
yltrehalose for trehalose liberation. These enzymes can act on α-1,
4 glucose bond repeatedly and can produce up to 80 % trehalose.
There is another method for trehalose production from starch by the
eect of α-amylase followed by isoamylase, which can produce am-
ylose. The latter can then be converted eciently into trehalose by
MTHase and MTSase.
Applications of trehalose
Trehalose has unique functions that are dierent from sucrose,
lactose, and maltose, including the protective action against stress-
ors such as nutrient deprivation, oxidative stress, and cold (Jain and
Roy, 2009). It could also prevent inammation induced by endoge-
nous and exogenous toxins (Minutoli et al., 2008). Trehalose report-
edly inhibits protein denaturation of bacterial plasma membranes
(Leslie et al., 1995) and human cells (Guo et al., 2000).
In addition, the low cost of trehalose permits a widespread ap-
plication in the medical eld such as preservation of mammalian
embryos for a long time, preservation of cells under freeze-dried
conditions, cryopreservation of platelets, and treatment of Hunting-
ton’s disease and dry eye syndrome (Matsuo et al., 2002; Mori et
al., 2010; Nakamura et al., 2008; Takeuchi et al., 2011; Tanaka et
al., 2004).
It can increase the viability of preserved bovine embryos in liq-
uid nitrogen (Saha et al., 1996). It can also increase the viability of
mammalian cells producing insulin during freezing and so it can aid
in treatment of diabetes disease (Beattie et al., 1997). Trehalose has
a therapeutic and protective eect in various neurodegenerative dis-
eases, such as Alzheimer ’s disease, frontotemporal dementia, pro-
gressive supranuclear palsy and corticobasal degeneration (Kruger
et al., 2012). A recent study emphasized that trehalose also has a
therapeutic eect on cardiac remodeling after myocardial infarction
(Sciarretta et al., 2018).
Trehalose eect on oxidative stress, apoptosis and auto-
phagy
Trehalose suppresses the oxidative stress by reducing the harm-
ful free radicals in yeast cells by the binding with the membrane
and so it can suppress the oxidation of the unsaturated fatty acids
in the cell membranes (Herdeiro et al., 2006; Oku et al., 2003). On
the other hand, mycolic acid-containing glycolipids, such as cord
factor (CF) (trehalose 6, 6-dimycolate [TDM]), trehalose trimycola,
2
Nasr Eldeen et al., 2018, AJMS 2(1):1-3, DOI:10.5455/ajms.13
and sulfatide (SL) (2,3,6,6-tetraacyl trehalose 2-sulfate), are virulent
factors characteristic of acid-fast bacteria which exert various bio-
logical eects, including adjuvant activity and granuloma formation,
by the activation of macrophages, natural killer (NK) cells and T cells
(Natsuhara et al., 1990;Tabata, et al., 1996 ). In vivo administration of
mycobacterial CF induces marked thymic atrophy via the apoptotic
process (Hernandez et al., 1993; Yuriko et al., 1997).
Autophagy is induced by trehalose in a mTOR-independent man-
ner to protect various cells from dierent harmful conditions such as
dehydration, extreme heating and cooling and oxidation (Sarkar et
al., 2007). Trehalose is considered as an autophagy inducer in mam-
malian cells (as evidenced by its ability to induce autophagic genes
Atg5 and LC3-II expression) and so it can be used as an adjuvant for
various diseases treatment (Kang et al., 2010).
Anticancer eect of trehalose
Although trehalose has signicantly preservative eects on cel-
lular viabilities as far as there is few investigations that examined the
eects of trehalose on tumor cell growth. Mycobacterial CF (as a nat-
ural product containing trehalose) has the potential to kill tumor cells
in guinea pig and mice (Watanabe et al., 1999), and trehalose surfac-
tant can also trigger in vitro death of human hepatocellular carcinoma
(Hep-G2 and HuH-7) cells (Matsumoto, et al., 2013a), human colon
carcinoma and gastric carcinoma (Matsumoto et al., 2013b).
Trehalose could potentially be employed as a novel anticancer
agent that could be used safely and eectively to topically treat ma-
lignant tumors, however the exact mode of this anticancer eect
has not been elucidated yet (Takashi et al., 2012). A previous study
showed that trehalose can cause malignant melanoma cell death via
induction of apoptosis (Kudo et al., 2012).
Trehalose can trigger autophagy in many tumor cell lines (Sarkar
et al., 2007). In contrast, a recent study by our group proved that tre-
halose can induce the antitumor potential of chemotherapeutic drug
methotrexate against mice bearing Ehrlich ascites carcinoma (EAC)
through induction of apoptosis and oxidative stress, and inhibition of
autophagy in EAC cells (El-Magd et al., 2017). These conicted nd-
ings may be due to variation in cancer types and stage as reported by
Shen et al., (2015) who found both protumorigenic and antitumori-
genic eect for autophagy, depending on the cell type, developmental
stage of cancer.
Summary
Trehalose can be used as a safe drug for treatment of many diseas-
es including cancers. However, its mode of action is quietly dierent
dependant on the tissue/cell type. In non-cancer cells, trehalose ex-
erts its eect through inhibition of apoptosis, reduction of oxidative
stress, and induction of autophagy. So in disease other than cancer,
trehalose acts as a potent anti-apoptotic, antioxidant, and autophagy
inducer agent. In contrast, in cancer cells, trehalose can act either
as in non-cancer cells or in a reverse direction (a potent apoptotic,
ROS inducer, and autophagy inhibitor agent) depending on type and
stage of tumor. Therefore, further investigation are required to denote
the actual mechanism of trehalose action when used for treatment of
dierent type of tumors. It is also worth to combine trehalose with
other chemotherapeutic agent and check whether it can increase their
anticancer potential and decrease their side eects.
References
Beattie, G.M., Crowe, J.H., Lopez, A.O., Ciralli, V., Ricordi, C., and
Hayek, A., 1997. Trehalose: a cryoprotectant that enhances re-
covery and preserves function of human pancreatic islets after
long-term storage. Diabetes, 46: 519–523.
Elbein, A.D., Pan, Y.T., Pastuszak, I., and Carroll, D. 2003. New
insights on trehalose: a multifunctional molecule. Glycobiol.,
13:17–27.
El-Magd, M.A., Khamis, A., Nasr Eldeen, S.K., Ibrahim, W.M.,
Salama, A.F.2017. Trehalose enhances the antitumor potential
of methotrexate against mice bearing Ehrlich ascites carcinoma.
Biomed Pharmacother.;92: 870-878.
Guo, N., Puhlev, I, Brown, D. R., Mansbridge, J., and Levine, F.
2000. Trehalose expression confers desiccation tolerance on hu-
man cells,” Nat. Biotech., 18 (2)168–171.
Herdeiro, R.S., Pereira, M.D., Panek, A.D., and Eleutherio, E.C.
2006. Trehalose protects Saccharomyces cerevisiae from lipid
peroxidation during oxidative stress. Biochim Biophys Acta.,
1760:340–346.
Hernandez-Caselles, T., and Stutuman, O. 1993. Immune functions
of tumor necrosis factor. I. Tumor necrosis factor induces ap-
optosis of mouse thymocytes and can also stimulate or inhibit
IL-6-induced proliferation depending on the concentration of
mitogenic costimulation. J. Immunol. 151:3999– 4012.
Jain, N.K., and Roy I. 2009. Eect of trehalose on protein structure.
Protein Sci., 18:24–36.
Kang, R., Tang, D., Schapiro, N.E., Livesey, K.M., Farkas, A.,
Loughran, P. et al. 2010. The receptor for advanced glycation
end products (RAGE) sustains autophagy and limits apoptosis,
promoting pancreatic tumor cell survival. Cell Death Dier; 17:
666–676.
Kruger, U., Wang, Y., Kumar, S. and Mandelkow, E.M. 2012. Auto-
phagic degradation of tau in primary neurons and its enhance-
ment by trehalose. Neurobiol., aging 33: 2291–2305.
Kudo, T. K. Takeuchi, Y. Ebina, M. Nakazawa, 2012. Inhibitory ef-
fects of trehalose on malignant melanoma cell growth: implica-
tions for a novel topical anticancer agent on the ocular surface,
ISRN Ophthalmol. 2012, 968493.
Leslie, S. B., Israeli E., Lighthart B., Crowe J. H., and Crowe L. M.,
1995. Trehalose and sucrose protect both membranes and pro-
teins in intact bacteria during drying, J. Appl. and Envir. Micro.,
61 (10): 3592–3597.
Maruta, K., Nakada, T., Kubota, M., H. Chaen, T., Sugimoto, M.,
Kurimoto, and Tsjisaka,Y. 1995. Formation of trehalose from
maltooligosaccharides by the novel enzymatic system. Biosci.
Biotechnol. Biochem. 59 (10):1829–1834.
Matsumoto, Y., E. Cao, R. Ueoka. 2013a. Growth inhibition by
novel liposomes including trehalose surfactant against hepato-
carcinoma cells along with apoptosis, Anticancer Res. 33 (11):
4727–4740.
Matsumoto, Y., E. Cao, R. Ueoka. 2013b. Novel liposomes com-
posed of dimyristoylphosphatidylcholine and trehalose sur-
factants inhibit the growth of tumor cells along with apoptosis,
Biol. Pharm. Bull. 36 (8):1258–1262.
Matsuo, T. 2004. Trehalose versus hyaluronan or cellulose in eye-
drops for the treatment of dry eye, Japan. J. Ophthal., 48 (4):
321–327.
Matsuo, T., Tsuchida, Y., and Morimoto, N. 2002. Trehalose eye
drops in the treatment of dry eye syndrome, Ophthalmology, 109
(11): 2024–2029.
3
Nasr Eldeen et al., 2018, AJMS 2(1):1-3, DOI:10.5455/ajms.13
Minutoli, L., Altavilla, D., Bitto A., Polito, F., Bellocco, E., Laga-
na, G., Fiumara, T., Magazu, S., Migliardo, F., Venuti, F.S., and
Squadrito, F. 2008. Trehalose: a biophysics approach to modu-
late the inammatory response during endotoxic shock. Eur. J.
Pharmacol., 589:272–280.
Mori, Y., Yano, F., Shimohata, N., Suzuki, S., Chung, U.I., and Taka-
to, T. 2010. Trehalose inhibits oral dryness by protecting the cell
membrane. Int. J. Oral Maxillofac. Surg., 39:916–921.
Nakamura, T., Sekiyama, E., and Takaoka M. 2008. The use of the
trehalose-treated freeze-dried amniotic membrane for ocular sur-
face reconstruction, Biomaterials, 29 ( 27): 3729–3737.
Natsuhara, Y., Oka S., Kaneda, K., Kato Y., and Yano, I. 1990. Par-
allel antitumor, granuloma-forming and tumor-necrosis-fac-
tor-priming activities of mycoloyl glycolipids from Nocardia
Rubra that dier in carbohydrate moiety: structure-activity rela-
tionships. Canc. Immunol. Immunother. 31:99–106.
Oku, K., Watanabe, H., Kubota, M., Fukuda, S., Kurimoto, M., Tsu-
jisaka, Y., Komori, M., Inoue, Y., and Sakurai, M. 2003. NMR
and quantum chemical study on the OH. . pi and CH.. O interac-
tions between trehalose and unsaturated fatty acids: implication
for the mechanism of antioxidant function of trehalose. J. Am.
Chem. Soc., 125:12739–12748.
Saha, S., Rajamahendran, R., Boediono, A., Sumantri, C., and Su-
zuki, T., 1996.Viability of bovine blastocysts obtained after 7, 8,
or 9 days of culture in vitro following vitrication and one-step
rehydration. Theriogenology, 46:331–343.
Sarkar, S., Davies, J.E., Huang, Z., Tunnaclie, A., and Rubinsztein,
D.C. 2007. Trehalose, a novel mTOR-independent autophagy
enhancer, accelerates the clearance of mutant huntingtin and al-
pha-synuclein. J. Biol. chem., 282: 5641–5652.
Sciarretta, S., Yee, D., Nagarajan, N., Bianchi, F., Saito, T., Valenti,
V., Tong, M., Del Re, D.P., Vecchione, C., Schirone, L., Forte,
M., Rubattu, S., Shirakabe, A., Boppana, V.S., Volpe, M., Frati,
G., Zhai, P., and Sadoshima, J. 2018. Trehalose-Induced Activa-
tion of Autophagy Improves Cardiac Remodeling After Myocar-
dial Infarction. J Am Coll Cardiol. 8;71(18):1999-2010.
Shen, M., W.M. Duan, M.Y. Wu, W.J. Wang, L. Liu, M.D. Xu, J.
Zhu, D.M. Li, Q. Gui, L. Lian, F.R. Gong, K. Chen, W. Li, M.
Tao, 2015. Participation of autophagy in the cytotoxicity against
breast cancer cells by cisplatin, Oncol. Rep. 34 (1):359–367.
Tabata, A., Kaneda K., Watanabe H., Abo T., and Yano I. 1996. Ki-
netics of organ-associated natural killer cells and intermediate
CD3 cells during pulmonary and hepatic granulomatous inam-
mation induced by mycobacterial cord factor. Microbiol. Immu-
nol. 40:651–658.
Takashi ,K., Kimio T., Yu-ichi E., and Mitsuru N., Zaifu-cho, Hirosa-
ki, S. Jonuscheit and Ljubimov A. V. 2012. Inhibitory Eects
of Trehalose on Malignant Melanoma Cell Growth, ISRN Oph-
thalm., 2012:1-9.
Takeuchi, K., Nakazawa, M., and Ebina, Y. 2011. Eects of trehalose
on VEGF-stimulated angiogenesis and myobroblast prolifera-
tion: implications for glaucoma ltration surgery. Invest. Oph-
thal. and Vis. Sci. 52 (9): 6987–6993.
Tanaka, M., Machida, Y. and Niu, S. 2004. Trehalose alleviates poly-
glutamine-mediated pathology in a mouse model of Huntington
disease, Nat. Med., 10 (2): 148–154.
Watanabe, R. , Yoo, Y.C., K. Hata, M. Mitobe, Y. Koike, M. Nishiza-
wa, D.M. Garcia,Y. Nobuchi, H. Imagawa, H. Yamada, Azuma,
I.. 1999. Inhibitory eect of trehalose dimycolate (TDM) and its
stereoisometric derivatives, trehalose dicorynomycolates (TD-
CMs), with low toxicity on lung metastasis of tumour cells in mice,
Vaccine 17 (11-12): 1484–1492.
Yuriko, O., Kenji, K., Nagatoshi, F., Misayo, M., Shiro, O., and Ikuya,
Y. 1997. In Vivo Induction of Apoptosis in the Thymus by Adminis-
tration of Mycobacterial Cord Factor (Trehalose 6,69-Dimycolate).
Infect. Immun. 65 (5): 1793–1799.
... Indeed, the inability of autophagydeficient cells to degrade p62 causes this protein to accumulate abnormally, which promotes tumorigenesis (Mathew et al., 2009). It has been shown in our recent studies that trehalose acts as anticancer novel treatment by modulating oxidative stress with decreasing the autophagic activities and increasing the apoptotic activities of cancer cells (Eldeen et al., 2018;El-Magd et al., 2018). ...
... Indeed, the inability of autophagydeficient cells to degrade p62 causes this protein to accumulate abnormally, which promotes tumorigenesis (Mathew et al., 2009). It has been shown in our recent studies that trehalose acts as anticancer novel treatment by modulating oxidative stress with decreasing the autophagic activities and increasing the apoptotic activities of cancer cells (Eldeen et al., 2018;El-Magd et al., 2018). ...
Article
Full-text available
Autophagy is a survival process in which a cell preserves its components in case of nutrient deprivation by recycling its digested contents and cannibalizing itself. It is characterized by formation of double membrane autophagosomes and it has a role in cancer treatment by many mechanisms. Interestingly, autophagy has a significant effect in the treatment of many diseases by different mechanisms. Autophagy has been included as a treatment mechanism of various diseases, such as degenerative diseases of the muscle and nervous system. Abundant autophagic vacuoles were formed inside the damaged cells in many of these degenerative disorders. Autophagy can help both living cells and cancer cells to survive so some antitumor agents targeted autophagy in cancer cells. In this review, we illustrated the definition of autophagy, autophagic pathways, therapeutic implications of autophagy in cancer, the relationship between autophagy and cell death, inflammation, and necrosis.
... According to Nasr Eldeen et al. [26], trehalose induces autophagy processes, the first step of which was assumed to be endosome formation. On the other hand, Mackeh et al. [27] reported a relationship between autophagy and microtubules. ...
Article
Full-text available
Isolation and characterization of new biologically active substances affecting cancer cells is an important issue of fundamental research in biomedicine. Trehalose lipid was isolated from Rhodococcus wratislaviensis strain and purified by liquid chromatography. The effect of trehalose lipid on cell viability and migration, together with colony forming assays, were performed on two breast cancer (MCF7—low metastatic; MDA-MB231—high metastatic) and one “normal” (MCF10A) cell lines. Molecular modeling that details the structure of the neutral and anionic form (more stable at physiological pH) of the tetraester was carried out. The tentative sizes of the hydrophilic (7.5 Å) and hydrophobic (12.5 Å) portions of the molecule were also determined. Thus, the used trehalose lipid is supposed to interact as a single molecule. The changes in morphology, adhesion, viability, migration, and the possibility of forming colonies in cancer cell lines induced after treatment with trehalose lipid were found to be dose and time dependent. Based on the theoretical calculations, a possible mechanism of action and membrane asymmetry between outer and inner monolayers of the bilayer resulting in endosome formation were suggested. Initial data suggest a mechanism of antitumor activity of the purified trehalose lipid and its potential for biomedical application.
Article
Full-text available
Background: Trehalose (TRE) is a natural, nonreducing disaccharide synthesized by lower organisms. TRE exhibits an extraordinary ability to protect cells against different kinds of stresses through activation of autophagy. However, the effect of TRE on the heart during stress has never been tested. Objectives: This study evaluated the effects of TRE administration in a mouse model of chronic ischemic remodeling. Methods: Wild-type (WT) or beclin1+/- mice were subjected to permanent ligation of the left anterior descending artery (LAD) and then treated with either placebo or trehalose (1 mg/g/day intraperitoneally for 48 h, then 2% in the drinking water). After 4 weeks, echocardiographic, hemodynamic, gravimetric, histological, and biochemical analyses were conducted. Results: TRE reduced left ventricular (LV) dilation and increased ventricular function in mice with LAD ligation compared with placebo. Sucrose, another nonreducing disaccharide, did not exert protective effects during post-infarction LV remodeling. Trehalose administration to mice overexpressing GFP-tagged LC3 significantly increased the number of GFP-LC3 dots, both in the presence and absence of chloroquine administration. TRE also increased cardiac LC3-II levels after 4 weeks following myocardial infarction (MI), indicating that it induced autophagy in the heart in vivo. To evaluate whether TRE exerted beneficial effects through activation of autophagy, trehalose was administered to beclin 1+/- mice. The improvement of LV function, lung congestion, cardiac remodeling, apoptosis, and fibrosis following TRE treatment observed in WT mice were all significantly blunted in beclin 1+/- mice. Conclusions: TRE reduced MI-induced cardiac remodeling and dysfunction through activation of autophagy.
Article
Full-text available
Breast cancer is one of the most common cancers affecting women worldwide. Conventional chemotherapy is still one of the major approaches to the treatment of breast cancer. Autophagy, also termed as type II programmed cell death (PCD), exhibits either a protumorigenic or antitumorigenic function. In the present study, we investigated whether autophagy could be involved in the effect of chemotherapy against breast cancer. Epirubicin, docetaxel, methotrexate, cyclophosphamide, fluorouracil (5-FU) and cisplatin were applied in the present investigation. All of these chemotherapeutics presented cytotoxicity against breast cancer cells. DsRed-LC3 reporter assay revealed that only docetaxel and cisplatin induced autophagy. Autophagy inhibitor 3-methyladenine (3-MA) strengthened the cytotoxicity of docetaxel, yet impaired the cytotoxicity of cisplatin, suggesting that docetaxel stimulates protumorigenic autophagy, while cisplatin-induced autophagy could be antitumorigenic. Real-time PCR revealed that cisplatin upregulated multiple autophagy-related genes, including AMBRA1, ATG3, ATG4C, ATG4D, ATG5, ATG7, ATG13, ATG14, ATG16L2, Beclin1, DRAM1, GABARAP, GABARAPL1, GABARAPL2, HDAC6, IRGM, MAP1LC3B and ULK1, indicating that cisplatin induced autophagy through a multiple mechanism involved manner.
Article
Full-text available
Purpose . To investigate the inhibitory effects of trehalose on malignant melanoma cell growth. Methods . We cultured human malignant melanoma cells in a medium containing trehalose (control/2.5%/5.0%/7.5%/10.0%) and used the MTT assay to evaluate the growth activities. Subsequently, trehalose was topically instilled on subconjunctivally inoculated melanoma cells in F334/NJcl-rmu/rmu rats, followed by a histopathological evaluation of tumor growth. Using flow cytometry, we compared the distribution of the cell cycle, rate of apoptotic cells, and intracellular factors related to the cell cycle in cultured melanoma cells after trehalose treatment. Results . The MTT study showed that proliferation of melanoma cells was significantly inhibited by ≧ 5% of trehalose concentrations in the culture media. Subconjunctivally inoculated melanoma cell masses were significantly smaller in eyes administered trehalose as compared to controls. Flow cytometry analyses demonstrated that the trehalose groups had increased rates of G2/M phase cells and apoptotic cells in the cell culture. These cells also exhibited increased expressions of cell-cycle inhibitory factors. Conclusions . The current results show trehalose inhibits malignant melanoma cell growth by inducing G2/M cell cycle arrest and apoptosis, suggesting trehalose as a potential candidate for a topical agent to inhibit proliferation of malignant tumor cells of the ocular surface.
Article
Full-text available
To investigate whether trehalose inhibits VEGF-stimulated or inflammatory angiogenesis and the proliferation of myofibroblasts. Normal human dermal fibroblasts and human umbilical vein endothelial cells (HUVECs)were cocultured in trehalose-containing medium (2.5/5.0/7.5/10.0%) with or without VEGF (10 ng/mL). After 11 days, the area, length, joint, and path of neovascularization were evaluated. The effect of topical trehalose on corneal neovascularization was examined in vivo by treating Balb/c mice with alkali burn-induced corneal neovascularization. After 14 days of trehalose treatment, corneal vessels were visualized in flatmounts. The expressions of VEGFR2, phospho-VEGFR2, and vimentin were observed. Then a separate coculture model of the myofibroblasts and HUVECs was used to observe the morphologic changes of the myofibroblasts by trehalose. Furthermore, myofibroblasts were cultured with trehalose to examine the cytokeratin and E-cadherin expressions. In the in vitro models, there was a significant trehalose dose-dependent inhibition of neovascularization. In the in vivo alkali burn models, corneal neovascularization was significantly inhibited by treatments using ≥ 2.5% trehalose eyedrops. The expressions of VEGFR2, phospho-VEGFR2, and vimentin were downregulated by trehalose. When trehalose was added to the medium, the myofibroblasts were transformed into epithelial cell-like cells. The transformed myofibroblasts expressed cytokeratin but not E-cadherin. Trehalose prevents angiogenesis by partially downregulating VEGFR2 expression. In addition, trehalose inhibits the proliferation of myofibroblasts partially by inducing mesenchymal-epithelial transition. These findings suggest that trehalose has potential for use as a new agent that can control angiogenesis and fibrosis and potential for use in glaucoma surgery.
Article
Full-text available
Activation of the induced receptor for advanced glycation end products (RAGE) leads to initiation of NF-kappaB and MAP kinase signaling pathways, resulting in propagation and perpetuation of inflammation. RAGE-knockout animals are less susceptible to acute inflammation and carcinogen-induced tumor development. We have reported that most forms of tumor cell death result in release of the RAGE ligand, high-mobility group protein 1 (HMGB1). We now report a novel role for RAGE in the tumor cell response to stress. Targeted knockdown of RAGE in the tumor cell, leads to increased apoptosis, diminished autophagy and decreased tumor cell survival . In contrast, overexpression of RAGE is associated with enhanced autophagy, diminished apoptosis and greater tumor cell viability. RAGE limits apoptosis through a p53-dependent mitochondrial pathway. Moreover, RAGE-sustained autophagy is associated with decreased phosphorylation of mammalian target of rapamycin (mTOR) and increased Beclin-1/VPS34 autophagosome formation. These findings show that the inflammatory receptor, RAGE, has a heretofore unrecognized role in the tumor cell response to stress. Furthermore, these studies establish a direct link between inflammatory mediators in the tumor microenvironment and resistance to programmed cell death. Our data suggest that targeted inhibition of RAGE or its ligands may serve as novel targets to enhance current cancer therapies.
Article
Methotrexate (MTX) is commonly used as a standard chemotherapy for many cancers, however its usage required high doses thereby leading to severe adverse effects. In a trial to find a suitable neoadjuvant therapy to decrease MTX dosage without lowering its chemotherapeutic efficacy, we investigated the antitumor effect of trehalose (TRE) on mice bearing Ehrlich ascites carcinoma (EAC) and checked whether TRE can enhance the antitumor potential of MTX. Treatment with TRE induced anti-tumor effects against EAC as reveled by a remarkable decrease in body weight, tumor volume, count of viable tumor cells, expression of the anti-apoptotic gene Bcl2 as well as by a significant increase in mean survival time, life span and expression of the apoptotic gene caspase-3. TRE also caused a significant decrease in autophagic activity of EAC cells as evident by reduction in the expression of the autophagic gene Beclin 1 (Bec1) and the fluorescence intensity of autophagosome marker. Additionally, TRE restored the altered hematological and biochemical parameters and improved the disrupted hepatic tissues of EAC-bearing mice. Interestingly, co-administration of TRE and MTX showed highest anti-tumor effect against EAC. These data indicate that TRE enhances the antitumor potential of MTX and could be used as neoadjuvant drug to increase the efficacy of the antitumor drug, MTX.
Article
Novel liposomes composed of L-α-dimyristoylphosphatidylcholine (DMPC) and trehalose surfactant (DMTreCn) were produced by the method of sonication in buffer solution. The thickness of fixed aqueous layer of DMTreCn was larger than that of DMPC liposomes and increased in a dose-dependent manner. The remarkable inhibitory effects of DMTreCn on the growth of human hepatocellular carcinoma (HCC) (Hep-G2 and HuH-7) cells were obtained along with apoptosis, without affecting the growth of normal cells. DMTreCn induced apoptosis of Hep-G2 and HuH-7 cells through the activation of caspase-3, 8 and 9. Release of cytochrome c from mitochondria and activation of Bcl-2 family protein (BAX) were recorded, indicating that DMTreCn induced apoptosis of Hep-G2 and HuH-7 cells through mitochondrial pathway via BAX. It is noteworthy that the remarkable inhibitory effects of DMTreCn on the growth of human HCC cells were obtained along with apoptosis for the first time.
Article
Novel hybrid liposomes (DMTre) composed of L-α-dimyristoylphosphatidylcholine (DMPC) and trehalose surfactant were produced and inhibitory effects of DMTre on the growth of human colon carcinoma (HCT116) and gastric carcinoma (MKN45) in vitro were examined. The remarkably high inhibitory effects of DMTre on the growth of HCT116 and MKN45 cells were obtained without affecting the growth of normal cells. The thickness of fixed aqueous layer of DMTre was larger than that of DMPC liposomes and increased in a dose-dependent manner. The induction of apoptosis by DMTre was revealed on the basis of flow cytometric analysis. DMTre induced apoptosis through the activation of caspases and mitochondria via Bax. It is noteworthy that remarkable inhibitory effects of DMTre on the growth of human colon and gastric carcinoma cells leading to apoptosis were obtained for the first time.
Article
Modulating the tau level may represent a therapeutic target for Alzheimer's disease (AD), as accumulating evidence shows that Abeta-induced neurodegeneration is mediated by tau. It is therefore important to understand the expression and degradation of tau in neurons. Recently we showed that overexpressed mutant tau and tau aggregates are degraded via the autophagic pathway in an N2a cell model. Here we investigated whether autophagy is involved in the degradation of endogenous tau in cultured primary neurons. We activated this pathway in primary neurons with trehalose, an enhancer of autophagy. This resulted in the reduction of endogenous tau protein. Tau phosphorylation at several sites elevated in AD pathology had little influence on its degradation by autophagy. Furthermore, by using a neuronal cell model of tauopathy, we showed that activation of autophagy suppresses tau aggregation and eliminates cytotoxicity. Notably, apart from activating autophagy, trehalose also inhibits tau aggregation directly. Thus, trehalose may be a good candidate for developing therapeutic strategies for AD and other tauopathies.
Article
This study assessed the clinical efficacy and acceptability of trehalose solution for oral dryness caused by dental treatment. The efficacy of trehalose on oral dryness under drying conditions was assessed by measuring the surface area of the fungiform papillae and the moisture content of the tongue in seven healthy volunteers. Based on the data from this pilot study, a clinical study was performed, in which the efficacy of oral trehalose spray was evaluated on oral dryness in 10 patients undergoing root canal treatment. The effects of trehalose on cell viability were also assessed under drying conditions in vitro. Trehalose suppressed oral dryness and associated pain caused by dental treatment and protected cells from dryness-related damage. These results indicate that pretreatment application of trehalose solution on the oral mucosa is effective in preventing oral dryness caused by dental treatment.