Citation: | RamaRao Malla, Rakshmitha Marni, Anindita Chakraborty, Mohammad Amjad Kamal. Diallyl disulfide and diallyl trisulfide in garlic as novel therapeutic agents to overcome drug resistance in breast cancer[J]. Journal of Pharmaceutical Analysis, 2022, 12(2): 221-231. doi: 10.1016/j.jpha.2021.11.004 |
R. R. Malla, S. Kumari, M. M. Gavara, etal., A perspective on the diagnostics, prognostics, and therapeutics of microRNAs of triple-negative breast cancer, Biophys. Rev. 11 (2019) 227-234
|
M. Nedeljković, A. Damjanović, Mechanisms of chemotherapy resistance in triple-negative breast cancer-how we can rise to the challenge, Cells 8 (2019) E957
|
L. Luo, W. Gao, J. Wang, etal., Study on the mechanism of cell cycle checkpoint Kinase 2 (CHEK2) gene dysfunction in chemotherapeutic drug resistance of triple negative breast cancer cells, Med. Sci. Monit. 24 (2018) 3176-3183
|
S. Shu, C. Y. Lin, H. H. He, etal., Response and resistance to BET bromodomain inhibitors in triple-negative breast cancer, Nature 529 (2016) 413-417
|
C. Mani, S. Jonnalagadda, J. Lingareddy, etal., Prexasertib treatment induces homologous recombination deficiency and synergizes with olaparib in triple-negative breast cancer cells, Breast Cancer Res. 21 (2019) 104
|
X. Chen, K. H. Low, A. Alexander, etal., Cyclin E overexpression sensitizes triple-negative breast cancer to Wee1 Kinase inhibition, Clin .Cancer Res. 24 (2018) 6594-6610
|
K. L. Thu, J. Silvester, M. J. Elliott, etal., Disruption of the anaphase-promoting complex confers resistance to TTK inhibitors in triple-negative breast cancer, Proc. Natl. Acad. Sci. USA 115 (2018) E1570-E1577
|
T. M. MacDonald, L. N. Thomas, E. Daze, etal., Prolactin-inducible EDD E3 ubiquitin ligase promotes TORC1 signalling, anti-apoptotic protein expression, and drug resistance in breast cancer cells, Am.J .Cancer Res. 9 (2019) 1484-1503
|
P. M. M. Ozawa, F. Alkhilaiwi, I. J. Cavalli, etal., Extracellular vesicles from triple-negative breast cancer cells promote proliferation and drug resistance in non-tumorigenic breast cells, Breast Cancer Res.Treat. 172 (2018) 713-723
|
C. K. Das, B. Linder, F. Bonn, etal., BAG3 overexpression and cytoprotective autophagy mediate apoptosis resistance in chemoresistant breast cancer cells, Neoplasia 20 (2018) 263-279
|
C. C. Cheng, L. H. Shi, X. J. Wang, etal., Stat3/Oct-4/c-Myc signal circuit for regulating stemness-mediated doxorubicin resistance of triple-negative breast cancer cells and inhibitory effects of WP1066, Int. J. Oncol. 53 (2018) 339-348
|
Y. Liu, M. L. Burness, R. Martin-Trevino, etal., RAD51 mediates resistance of cancer stem cells to PARP inhibition in triple-negative breast cancer, Clin .Cancer Res. 23 (2017) 514-522
|
D. R. Chen, D. Y. Lu, H. Y. Lin, etal., Mesenchymal stem cell-induced doxorubicin resistance in triple negative breast cancer, Biomed. Res. Int. 2014 (2014) 532161
|
W. L. Yeh, C. F. Tsai, D. R. Chen, Peri-foci adipose-derived stem cells promote chemoresistance in breast cancer, Stem Cell Res. Ther. 8 (2017) 177
|
Z. Wang, N. Wang, W. Li, etal., Caveolin-1 mediates chemoresistance in breast cancer stem cells via β-catenin/ABCG2 signaling pathway, Carcinogenesis 35 (2014) 2346-2356
|
I. G. Ryoo, B. H. Choi, S. K. Ku, etal., High CD44 expression mediates p62-associated NFE2L2/NRF2 activation in breast cancer stem cell-like cells: Implications for cancer stem cell resistance, Redox Biol. 17 (2018) 246-258
|
R. Chakravarthy, K. Mnich, A. M. Gorman, Nerve growth factor (NGF)-mediated regulation of p75NTR expression contributes to chemotherapeutic resistance in triple negative breast cancer cells, Biochem .Biophys. Res. Commun. 478 (2016) 1541-1547
|
I. N. Sari, L. T. H. Phi, N. Jun, etal., Hedgehog Signaling in Cancer: A Prospective Therapeutic Target for Eradicating Cancer Stem Cells, Cells 7 (2018)
|
P. Bhateja, M. Cherian, S. Majumder, etal., The hedgehog signaling pathway: a viable target in breast cancer?, Cancers (Basel) 11 (2019) E1126
|
D. J. Azzam, D. Zhao, J. Sun, etal., Triple negative breast cancer initiating cell subsets differ in functional and molecular characteristics and in γ-secretase inhibitor drug responses, EMBO Mol. Med. 5 (2013) 1502-1522
|
B. Darvishi, L. Farahmand, S. Z. Eslami, etal., NF-κB as the main node of resistance to receptor tyrosine kinase inhibitors in triple-negative breast cancer, Tumour Biol. 39 (2017) 1010428317706919
|
H. Song, D. Li, T. Wu, etal., MicroRNA-301b promotes cell proliferation and apoptosis resistance in triple-negative breast cancer by targeting CYLD, BMB Rep. 51 (2018) 602-607
|
J. Niu, A. Xue, Y. Chi, etal., Induction of miRNA-181a by genotoxic treatments promotes chemotherapeutic resistance and metastasis in breast cancer, Oncogene 35 (2016) 1302-1313
|
L. Chen, L. Y. Bourguignon, Hyaluronan-CD44 interaction promotes c-Jun signaling and miRNA21 expression leading to Bcl-2 expression and chemoresistance in breast cancer cells, Mol. Cancer 13 (2014) 52
|
L. De Mattos-Arruda, G. Bottai, P. G. Nuciforo, etal., MicroRNA-21 links epithelial-to-mesenchymal transition and inflammatory signals to confer resistance to neoadjuvant trastuzumab and chemotherapy in HER2-positive breast cancer patients, Oncotarget 6 (2015) 37269-37280
|
H. Korkaya, G. I. Kim, A. Davis, etal., Activation of an IL6 inflammatory loop mediates trastuzumab resistance in HER2+ breast cancer by expanding the cancer stem cell population, Mol. Cell 47 (2012) 570-584
|
L. H. Zhang, A. J. Yang, M. Wang, etal., Enhanced autophagy reveals vulnerability of P-gp mediated epirubicin resistance in triple negative breast cancer cells, Apoptosis 21 (2016) 473-488
|
S. Yin, L. Xu, R. D. Bonfil, etal., Tumor-initiating cells and FZD8 play a major role in drug resistance in triple-negative breast cancer, Mol. Cancer Ther. 12 (2013) 491-498
|
K. Wang, X. Zhu, K. Zhang, etal., Interleukin-6 contributes to chemoresistance in MDA-MB-231 cells via targeting HIF-1α, J. Biochem. Mol. Toxicol. 32 (2018) e22039
|
M. Kalimutho, D. Sinha, D. Mittal, etal., Blockade of PDGFRβ circumvents resistance to MEK-JAK inhibition via intratumoral CD8(+) T-cells infiltration in triple-negative breast cancer, J. Exp. Clin. Cancer Res. 38 (2019) 85
|
M. G. Mendoza-Rodríguez, J. T. Ayala-Sumuano, L. García-Morales, etal., IL-1β inflammatory cytokine-induced TP63 isoform ΔNP63α signaling cascade contributes to cisplatin resistance in human breast cancer cells, Int. J. Mol. Sci. 20 (2019)
|
R. B. Penney, D. Roy, Thioredoxin-mediated redox regulation of resistance to endocrine therapy in breast cancer, Biochim. Biophys. Acta 1836 (2013) 60-79
|
L. Wang, C. Yang, X. B. Liu, etal., B7-H4 overexpression contributes to poor prognosis and drug-resistance in triple-negative breast cancer, Cancer Cell Int. 18 (2018) 100
|
D. J. Newman, G. M. Cragg, Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019, J. Nat. Prod. 83 (2020) 770-803
|
L. L. Koteswari, S. Kumari, A. B. Kumar, etal., A comparative anticancer study on procyanidin C1 against receptor positive and receptor negative breast cancer, Nat. Prod. Res. (2019) 1-8
|
N. Mamadalieva, N. Mamedov. Taxus brevifolia a High-Value Medicinal Plant, as a Source of Taxol. Medicinal and Aromatic Plants of North America: Springer; 2020. p. 201-218
|
M. A. Taher, M. A. B. Nyeem, M. M. Billah, etal., Vinca alkaloid-the second most used alkaloid for cancer treatment-A review, Inter. J. Physiol. Nutr. Phys. Educ. 2 (2017) 723-727
|
M. A. Kalam, A. H. Malik, A. H. Ganie, etal., Medicinal importance of Papra (Podophyllum hexandrum Royle) in Unani System of Medicine, J.Complement.Integ.Med. (2021)
|
M. S. Subramanian, G. Nandagopal MS, S. Amin Nordin, etal., Prevailing Knowledge on the Bioavailability and Biological Activities of Sulphur Compounds from Alliums: A Potential Drug Candidate, Molecules 25 (2020) 4111
|
S. H. Omar, N. A. Al-Wabel, Organosulfur compounds and possible mechanism of garlic in cancer, Saudi. Pharm. J. 18 (2010) 51-58
|
L. D. Lawson, S. M. Hunsaker, Allicin Bioavailability and Bioequivalence from Garlic Supplements and Garlic Foods, Nutrients 10 (2018)
|
S. A. Almatroodi, M. A. Alsahli, A. Almatroudi, etal., Garlic and its Active Compounds: A Potential Candidate in The Prevention of Cancer by Modulating Various Cell Signalling Pathways, Anticancer Agents Med. Chem. 19 (2019) 1314-1324
|
Y. Liu, A. Li, X. Feng, etal., Pharmacological Investigation of the anti-Inflammation and anti-oxidation activities of diallyl disulfide in a rat emphysema model induced by cigarette smoke extract, Nutrients 10 (2018) E79
|
H. He, Y. Ma, H. Huang, etal., A comprehensive understanding about the pharmacological effect of diallyl disulfide other than its anti-carcinogenic activities, Eur. J. Pharmacol. 893 (2021) 173803
|
A. Bordia, S. K. Verma, K. C. Srivastava, Effect of garlic (Allium sativum) on blood lipids, blood sugar, fibrinogen and fibrinolytic activity in patients with coronary artery disease, Prostaglandins Leukot. Essent. Fatty Acids 58 (1998) 257-263
|
I. C. Lee, S. H. Kim, H. S. Baek, etal., Protective effects of diallyl disulfide on carbon tetrachloride-induced hepatotoxicity through activation of Nrf2, Environ. Toxicol. 30 (2015) 538-548
|
A. Hosseinzadeh, D. Jafari, T. Kamarul, etal., Evaluating the protective effects and mechanisms of diallyl disulfide on interlukin-1β-induced oxidative stress and mitochondrial apoptotic signaling pathways in cultured chondrocytes, J. Cell. Biochem. 118 (2017) 1879-1888
|
Y.-R. Wu, L. Li, X.-C. Sun, etal., Diallyl disulfide improves lipid metabolism by inhibiting PCSK9 expression and increasing LDL uptake via PI3K/Akt-SREBP2 pathway in HepG2 cells, Nutr. Metab. Cardiovasc. Dis. 31 (2021) 322-332
|
P. Goudappala, C. Y. Gowda, R. T. Kashinath, Diallyl disulfide regulates purine metabolism and their metabolites in diabetes mellitus, Indian J. Physiol. Pharmacol. 65 (2021) 28-34
|
P. Mikaili, S. Maadirad, M. Moloudizargari, etal., Therapeutic uses and pharmacological properties of garlic, shallot, and their biologically active compounds, Iran. J. Basic Med. Sci. 16 (2013) 1031
|
A. A. Shaaban, D. S. El-Agamy, Cytoprotective effects of diallyl trisulfide against valproate-induced hepatotoxicity: new anticonvulsant strategy, Naunyn Schmiedebergs Arch. Pharmacol. 390 (2017) 919-928
|
L.-Y. Chen, Q. Chen, X.-J. Zhu, etal., Diallyl trisulfide protects against ethanol-induced oxidative stress and apoptosis via a hydrogen sulfide-mediated mechanism, Int. Immunopharmacol. 36 (2016) 23-30
|
C.-T. Liu, H. Hse, C.-K. Lii, etal., Effects of garlic oil and diallyl trisulfide on glycemic control in diabetic rats, Eur. J. Pharmacol. 516 (2005) 165-173
|
X. Zhu, F. Zhang, L. Zhou, etal., Diallyl trisulfide attenuates carbon tetrachloride-caused liver injury and fibrogenesis and reduces hepatic oxidative stress in rats, Naunyn Schmiedebergs Arch. Pharmacol. 387 (2014) 445-455
|
B. L. Predmore, K. Kondo, S. Bhushan, etal., The polysulfide diallyl trisulfide protects the ischemic myocardium by preservation of endogenous hydrogen sulfide and increasing nitric oxide bioavailability, Am. J. Physiol. Heart Circ. Physiol. 302 (2012) H2410-2418
|
G. Desai, M. Schelske-Santos, C. M. Nazario, etal., Onion and garlic intake and breast cancer, a case-control study in Puerto Rico, Nutr. Cancer 72 (2020) 791-800
|
A. Pourzand, A. Tajaddini, S. Pirouzpanah, etal., Associations between dietary Allium vegetables and risk of breast cancer: a hospital-based matched case-control study, J. Breast Cancer 19 (2016) 292-300
|
Y. Li, S. Li, X. Meng, etal., Dietary natural products for prevention and treatment of breast cancer, Nutrients 9 (2017) E728
|
I. Henao Castañeda, J. A. Pereañez, L. M. Preciado, etal., Sulfur Compounds as Inhibitors of Enzymatic Activity of a Snake Venom Phospholipase A2: Benzyl 4-nitrobenzenecarbodithioate as a Case of Study, Molecules 25 (2020) 1373
|
X. Wu, F. Kassie, V. Mersch-Sundermann, Induction of apoptosis in tumor cells by naturally occurring sulfur-containing compounds, Mutat. Res. Rev. Mutat. Res. 589 (2005) 81-102
|
S. H. Omar, N. A. Al-Wabel, Organosulfur compounds and possible mechanism of garlic in cancer, Saudi Pharm. J. 18 (2010) 51-58
|
P. Thejass, G. Kuttan, Inhibition of angiogenic differentiation of human umbilical vein endothelial cells by diallyl disulfide (DADS), Life Sci. 80 (2007) 515-521
|
S. V. Singh, A. A. Powolny, S. D. Stan, etal., Garlic constituent diallyl trisulfide prevents development of poorly differentiated prostate cancer and pulmonary metastasis multiplicity in TRAMP mice, Cancer Res. 68 (2008) 9503-9511
|
A. Malki, M. El-Saadani, A. S. Sultan, Garlic constituent diallyl trisulfide induced apoptosis in MCF7 human breast cancer cells, Cancer Biol. Ther. 8 (2009) 2175-2185
|
X. Xie, X. Huang, H. Tang, etal., Diallyl Disulfide Inhibits Breast Cancer Stem Cell Progression and Glucose Metabolism by Targeting CD44/PKM2/AMPK Signaling, Curr. Cancer Drug Targets 18 (2018) 592-599
|
H. Nakagawa, K. Tsuta, K. Kiuchi, etal., Growth inhibitory effects of diallyl disulfide on human breast cancer cell lines, Carcinogenesis 22 (2001) 891-897
|
R. Jan, G. E. Chaudhry, Understanding Apoptosis and Apoptotic Pathways Targeted Cancer Therapeutics, Adv. Pharm. Bull. 9 (2019) 205-218
|
G. P. Feeney, R. J. Errington, M. Wiltshire, etal., Tracking the cell cycle origins for escape from topotecan action by breast cancer cells, Br. J. Cancer 88 (2003) 1310-1317
|
Y. Nishiguchi, N. Oue, R. Fujiwara-Tani, etal., Role of metastasis-related genes in cisplatin chemoresistance in gastric cancer, Int. J. Mol. Sci. 21 (2019) E254
|
M. T. Puccinelli, S.D Stan, Dietary bioactive diallyl trisulfide in cancer prevention and treatment, Int. J. Mol. Sci. 18 (2017) E1645
|
E. R. Hahm, S. V. Singh, Diallyl trisulfide inhibits estrogen receptor-α activity in human breast cancer cells, Breast Cancer Res.Treat. 144 (2014) 47-57
|
H. A. Abdel-Hafiz, Epigenetic Mechanisms of Tamoxifen Resistance in Luminal Breast Cancer, Diseases 5 (2017)
|
C. H.Lee, Reversing agents for ATP-binding cassette drug transporters, Methods Mol. Biol. 596 (2010) 325-340
|
Y. S. Hong, Y. A. Ham, J. H. Choi, etal., Effects of allyl sulfur compounds and garlic extract on the expression of Bcl-2, Bax, and p53 in non small cell lung cancer cell lines, Exp. Mol. Med. 32 (2000) 127-134
|
S. Ohkubo, L. Dalla Via, S. Grancara, etal., The antioxidant, aged garlic extract, exerts cytotoxic effects on wild-type and multidrug-resistant human cancer cells by altering mitochondrial permeability, Int. J. Oncol. 53 (2018) 1257-1268
|
J. Huang, B. Yang, T. Xiang, etal., Diallyl disulfide inhibits growth and metastatic potential of human triple-negative breast cancer cells through inactivation of the β-catenin signaling pathway, Mol. Nutr. Food Res. 59 (2015) 1063-1075
|
T. Xiong, X. W. Liu, X. L. Huang, etal., Tristetraprolin: a novel target of diallyl disulfide that inhibits the progression of breast cancer, Oncol. Lett. 15 (2018) 7817-7827
|
M. Ciocci, E. Iorio, F. Carotenuto, etal., H2S-releasing nanoemulsions: a new formulation to inhibit tumor cells proliferation and improve tissue repair, Oncotarget 7 (2016) 84338-84358
|
S. V. Talluri, G. Kuppusamy, V. V. Karri, etal., Application of quality-by-design approach to optimize diallyl disulfide-loaded solid lipid nanoparticles, Artif. Cells Nanomed. Biotechnol. 45 (2017) 474-488
|
V. T. Siddhartha, S. Pindiprolu, P. K. Chintamaneni, etal., RAGE receptor targeted bioconjuguate lipid nanoparticles of diallyl disulfide for improved apoptotic activity in triple negative breast cancer: in vitro studies, Artif. Cells Nanomed. Biotechnol. 46 (2018) 387-397
|
L.M. Yu, S. Li, X.L.Tang, etal., Diallyl trisulfide ameliorates myocardial ischemia-reperfusion injury by reducing oxidative stress and endoplasmic Reticulum stress-mediated apoptosis in type 1 diabetic rats: role of SIRT1 activation, Apoptosis 22 (2017) 942-954
|
Y.G. Li, Z.Y. Wang, J.M. Li, etal., Diallyl disulfide suppresses FOXM1-mediated proliferation and invasion in osteosarcoma by upregulating miR-134, J. Cell. Biochem. (2019) 7286-7296
|
Y.P.Liu, P.T. Zhu, Y.Y. Wang, etal., Antimetastatic therapies of the polysulfide diallyl tisulfide against triple-negative breast bancer (TNBC) via suppressing MMP2/9 by blocking NF-κB and ERK/MAPK signaling pathways, PLoS One 10 (2015) e0123781
|
Y.P.Liu, Y. Zhao, Z.H. Wei, etal., Targeting thioredoxin system with an organosulfur compound, diallyl trisulfide (DATS), attenuates progression and metastasis of triple-negative breast cancer (TNBC), Cell Physiol. Biochem. 50 (2018) 1945-1963
|
S. Y. Cheng, Y. C. Yang, K. L. Ting, etal., Lactate dehydrogenase downregulation mediates the inhibitory effect of diallyl trisulfide on proliferation, metastasis, and invasion in triple-negative breast cancer, Environ. Toxicol. 32 (2017) 1390-1398
|
Y. Liu, P. Zhu, Y. Wang, etal., Antimetastatic therapies of the polysulfide diallyl trisulfide against triple-negative breast cancer (TNBC) via suppressing MMP2/9 by blocking NF-κB and ERK/MAPK signaling pathways, PLoS One 10 (2015) e0123781
|
S. H. Kim, E. R. Hahm, K.B.Singh, Diallyl trisulfide inhibits leptin-induced oncogenic signaling in human breast cancer cells but fails to prevent chemically-induced luminal-type cancer in rats, J. Cancer Prev. 25 (2020) 1-12
|
K. C. Lai, S. C. Hsu, J. S. Yang, Diallyl trisulfide inhibits migration, invasion and angiogenesis of human colon cancer HT-29 cells and umbilical vein endothelial cells, and suppresses murine xenograft tumour growth, J. Cell Mol. Med. 19 (2015) 474-484
|
Q. Tao, C. Wu, R. Xu, etal., Diallyl trisulfide inhibits proliferation, invasion and angiogenesis of glioma cells by inactivating Wnt/β-catenin signaling, Cell Tissue Res. 370 (2017) 379-390
|
P. Sujatha, P. G. Anantharaju, P. M. Veeresh, Diallyl disulfide (DADS) retards the growth of breast cancer cells in vitro and in vivo through apoptosis induction, Biomed. Pharmacol. J. 10 (2017) 1619-1630
|
W.H. Talib, Consumption of garlic and lemon aqueous extracts combination reduces tumor burden by angiogenesis inhibition, apoptosis induction, and immune system modulation, Nutrition 43-44 (2017) 89-97
|
Y. P. Liu, Y. Zhao, Y. Y. Wang, Suppressive role of diallyl trisulfide in the activated platelet-mediated hematogenous metastasis of MDA-MB-231 human breast cancer cells, Int. J. Mol. Med. 39 (2017) 1516-1524
|
D. Bauer, N. Redmon, E. Mazzio, Diallyl disulfide inhibits TNFα induced CCL2 release through MAPK/ERK and NF-Kappa-B signaling, Cytokine 75 (2015) 117-126
|
V. A. Kiesel, S. D. Stan, Diallyl trisulfide, a chemopreventive agent from Allium vegetables, inhibits alpha-secretases in breast cancer cells, Biochem .Biophys. Res. Commun. 484 (2017) 833-838
|
X. Li, Y. Meng, C. Xie, Diallyl Trisulfide inhibits breast cancer stem cells via suppression of Wnt/β-catenin pathway, J. Cell. Biochem. 119 (2018) 4134-4141
|
X. Jiang, X. Zhu, N. Liu, Diallyl trisulfide Inhibits growth of NCI-H460 in vitro and in vivo, and ameliorates cisplatin-induced oxidative injury in the treatment of lung carcinoma in xenograft mice, Int. J . Biol. Sci. 13 (2017) 167-178
|
H. C. Wang, Y. L. Chu, S. C. Hsieh, etal., Diallyl trisulfide inhibits cell migration and invasion of human melanoma a375 cells via inhibiting integrin/facal adhesion kinase pathway, Environ. Toxicol. 32 (2017) 2352-2359
|
A. K. Singh, A. Bishayee, A. K. Pandey, Targeting histone deacetylases with natural and synthetic agents: an emerging anticancer strategy, Nutrients 10 (2018)
|
Y. M. Pan, S. Y. Lin, R. Xing, Epigenetic upregulation of metallothionein 2A by diallyl trisulfide enhances chemosensitivity of human gastric cancer cells to docetaxel through attenuating NF-κB activation, Antioxid. Redox Signal. 24 (2016) 839-854
|
S. Lin, X. Wang, Y. Pan, Transcription factor myeloid zinc-finger 1 suppresses human gastric carcinogenesis by interacting with metallothionein 2A, Clin .Cancer Res. 25 (2019) 1050-1062
|
X. Xiao, B. Chen, X. Liu, Diallyl disulfide suppresses SRC/Ras/ERK signaling-mediated proliferation and metastasis in human breast cancer by up-regulating miR-34a, PLoS One 9 (2014) e112720
|
Y. Li, J. Zhang, L. Zhang, etal., Diallyl trisulfide inhibits proliferation, invasion and angiogenesis of osteosarcoma cells by switching on suppressor microRNAs and inactivating of Notch-1 signaling, Carcinogenesis 34 (2013) 1601-1610
|
E. P. Chiang, S. C. Chiu, M. H. Pai, Organosulfur garlic compounds induce neovasculogenesis in human endothelial progenitor cells through a modulation of MicroRNA 221 and the PI3-K/Akt signaling pathways, J. Agric. Food Chem. 61 (2013) 4839-4849
|
Z. H. Wei, Y. L. Shan, L. Tao, Diallyl trisulfides, a natural histone deacetylase inhibitor, attenuate HIF-1α synthesis, and decreases breast cancer metastasis, Mol. Carcinog. 56 (2017) 2317-2331
|
H. K. Na, E. H. Kim, M. A. Choi, Diallyl trisulfide induces apoptosis in human breast cancer cells through ROS-mediated activation of JNK and AP-1, Biochem. Pharmacol. 84 (2012) 1241-1250
|
X. Xie, X. Huang, H. Tang, et al., Diallyl disulfide inhibits breast cancer stem cell progression and glucose metabolism by targeting CD44/PKM2/AMPK signaling, Curr. Cancer Drug Targets 18 (2018) 592–599
|
S.H. Kim, C.H. Kaschula, N. Priedigkeit, et al., Forkhead box Q1 is a novel target of breast cancer stem cell inhibition by diallyl trisulfide, J. Biol. Chem. 291 (2016) 13495–13508
|
A. Liskova, P. Kubatka, M. Samec, et al., Dietary phytochemicals targeting cancer stem cells, Molecules 24 (2019), 899
|
Y. Qiao, X. Jiang, S. T. Lee, FOXQ1 regulates epithelial-mesenchymal transition in human cancers, Cancer Res. 71 (2011) 3076-3086
|
H. Zhang, F. Meng, G. Liu, Forkhead transcription factor foxq1 promotes epithelial-mesenchymal transition and breast cancer metastasis, Cancer Res. 71 (2011) 1292-13301
|
Y. Q. Chen, R. Xue, X. C. Jin, Antiarthritic activity of diallyl disulfide against freund's adjuvant-induced arthritic rat model, J. Environ. Pathol. Toxicol. Oncol. 37 (2018) 291-303
|
N. Zhang, Y. L. Wang, J. L. Zhang, Diallyl disulfide attenuates non-alcoholic steatohepatitis by suppressing key regulators of lipid metabolism, lipid peroxidation and inflammation in mice, Mol .Med. Rep. 20 (2019) 1363-1372
|
C. Feng, Y. Luo, Y. Nian, Diallyl disulfide suppresses the inflammation and apoptosis resistance induced by DCA through ROS and the NF-κB signaling pathway in human barrett's epithelial cells, Inflammation 40 (2017) 818-831
|
S. Miltonprabu, N. C. Sumedha, P. Senthilraja, Diallyl trisulfide, a garlic polysulfide protects against As-induced renal oxidative nephrotoxicity, apoptosis and inflammation in rats by activating the Nrf2/ARE signaling pathway, Int. Immunopharmacol. 50 (2017) 107-120
|
H. H. Lee, J. W. Jeong, S. H. Hong, Diallyl trisulfide suppresses the production of lipopolysaccharide-induced inflammatory mediators in BV2 microglia by decreasing the NF-κB pathway activity associated with toll-like receptor 4 and CXCL12/CXCR4 pathway blockade, J. Cancer Prev. 23 (2018) 134-140
|
J. Liu, W. Liu, H. Yang, Balancing apoptosis and autophagy for parkinson's disease therapy: targeting BCL-2, ACS Chem. Neurosci. 10 (2019) 792-802
|
S. Nakamura, M. Izumi, Chlorophagy is ATG gene-dependent microautophagy process, Plant Signal. Behav. 14 (2019) 1554469
|
L. Li, W. L. Liu, L. Su, The role of autophagy in cancer radiotherapy, Curr. Mol. Pharmacol. 13 (2020) 31-40
|
T. Suangtamai, D. I. Tanyong, Diallyl disulfide induces apoptosis and autophagy via mTOR pathway in myeloid leukemic cell line, Tumour Biol. 37 (2016) 10993-10999
|
Y. Y. Wu, Y. Q. Hu, H. Y. Zhou, Organosulfur compounds induce cytoprotective autophagy against apoptosis by inhibiting mTOR phosphorylation activity in macrophages, Acta Biochim. Biophys. Sin. 50 (2018) 1085-1093
|
Z. Q. Yue, X. Guan, R. Chao, Diallyl disulfide induces apoptosis and autophagy in human osteosarcoma MG-63 cells through the PI3K/Akt/mTOR pathway, Molecules 24 (2019) E2665
|
K. G. K. Deepak, R. Vempati, G. P. Nagaraju, Tumor microenvironment: Challenges and opportunities in targeting metastasis of triple negative breast cancer, Pharmacol . Res 153 (2020) 104683
|
R. R. Malla, K. Deepak, N. Merchant, Breast Tumor Microenvironment: Emerging target of therapeutic phytochemicals, Phytomedicine 70 (2020) 153227
|
N. Horn, G. Miller, K. M. Ajuwon, Garlic diallyl disulfide and diallyl trisulfide mitigates effects of pro-oxidant induced cellular stress and has immune modulatory function in LPS-stimulated porcine epithelial cells, J. Anim. Sci. 95 (2017) 4045-4051
|
I. A. Schepetkin, L. N. Kirpotina, A. I. Khlebnikov, Neutrophil immunomodulatory activity of natural organosulfur compounds, Molecules 24 (2019) 1809
|
Y. Hashizume, K. Shirato, I. Abe, Diallyl disulfide reduced dose-dependently the number of lymphocyte subsets and monocytes in rats, J. Nutr .Sci .Vitaminol. (Tokoyo) 58 (2012) 292-296
|
M. Ebrahimi, Z. Mohammad Hassan, A. Mostafaie, Purified protein fraction of garlic extract modulates cellular immune response against breast transplanted tumors in BALB/c mice model, Cell J. 15 (2013) 65-75
|