143 research outputs found
Thymoquinone enhances the anticancer activity of doxorubicin against adult T-cell leukemia in vitro and in vivo through ROS-dependent mechanisms
Aims: Adult T-cell leukemia (ATL) is a mature T-cell neoplasm associated with human T-cell lymphotropic virus (HTLV-1) infection. Major limitations in Doxorubicin (Dox) chemotherapy are tumor resistance and severe drug complications. Here, we combined Thymoquinone (TQ) with low concentrations of Dox and determined anticancer effects against ATL in cell culture and animal model. Main methods: HTLV-1 positive (HuT-102) and HTLV-1 negative (Jurkat) CD4+ malignant T-cell lines were treated with TQ, Dox and combinations. Viability and cell cycle effects were determined by MTT assay and flow cytometry analysis, respectively. Combination effects on mitochondrial membrane potential and generation of reactive oxygen species (ROS) were assessed. Expression levels of key cell death proteins were investigated by western blotting. A mouse xenograft model of ATL in NOD/SCID was used for testing drug effects and tumor tissues were stained for Ki67 and TUNEL. Key findings: TQ and Dox caused greater inhibition of cell viability and increased sub-G1 cells in both cell lines compared to Dox or TQ alone. The combination induced apoptosis by increasing ROS and causing disruption of mitochondrial membrane potential. Pretreatment with N-acetyl cysteine (NAC) or pan caspase inhibitor significantly inhibited the apoptotic response suggesting that cell death is ROS- and caspase-dependent. TQ and Dox combination reduced tumor volume in NOD/SCID mice more significantly than single treatments through enhanced apoptosis without affecting the survival of mice. Significance: Our combination model offers the possibility to use up to twofold lower doses of Dox against ATL while exhibiting the same cancer inhibitory effects. © 201
Thymoquinone inhibits tumor growth and induces apoptosis in a breast cancer xenograft mouse model: The role of p38 MAPK and ROS
Due to narrow therapeutic window of cancer therapeutic agents and the development of resistance against these agents, there is a need to discover novel agents to treat breast cancer. The antitumor activities of thymoquinone (TQ), a compound isolated from Nigella sativa oil, were investigated in breast carcinoma in vitro and in vivo. Cell responses after TQ treatment were assessed by using different assays including MTT assay, annexin V-propidium iodide staining, Mitosox staining and Western blot. The antitumor effect was studied by breast tumor xenograft mouse model, and the tumor tissues were examined by histology and immunohistochemistry. The level of antioxidant enzymes/molecules in mouse liver tissues was measured by commercial kits. Here, we show that TQ induced p38 phosphorylation and ROS production in breast cancer cells. These inductions were found to be responsible for TQ’s anti-proliferative and pro-apoptotic effects. Moreover, TQ-induced ROS production regulated p38 phosphorylation but not vice versa. TQ treatment was found to suppress the tumor growth and this effect was further enhanced by combination with doxorubicin. TQ also inhibited the protein expression of anti-apoptotic genes, such as XIAP, survivin, Bcl-xL and Bcl-2, in breast cancer cells and breast tumor xenograft. Reduced Ki67 and increased TUNEL staining were observed in TQ-treated tumors. TQ was also found to increase the level of catalase, superoxide dismutase and glutathione in mouse liver tissues. Overall, our results demonstrated that the antiproliferative and pro-apoptotic effects of TQ in breast cancer are mediated through p38 phosphorylation via ROS generation
Copper chelation selectively kills colon cancer cells through redox cycling and generation of reactive oxygen species
Background: Metals including iron, copper and zinc are essential for physiological processes yet can be toxic at high concentrations. However the role of these metals in the progression of cancer is not well defined. Here we study the anti-tumor activity of the metal chelator, TPEN, and define its mechanism of action.Methods: Multiple approaches were employed, including cell viability, cell cycle analysis, multiple measurements of apoptosis, and mitochondrial function. In addition we measured cellular metal contents and employed EPR to record redox cycling of TPEN-metal complexes. Mouse xenografts were also performed to test the efficacy of TPEN in vivo.Results: We show that metal chelation using TPEN (5μM) selectively induces cell death in HCT116 colon cancer cells without affecting the viability of non-cancerous colon or intestinal cells. Cell death was associated with increased levels of reactive oxygen species (ROS) and was inhibited by antioxidants and by prior chelation of copper. Interestingly, HCT116 cells accumulate copper to 7-folds higher levels than normal colon cells, and the TPEN-copper complex engages in redox cycling to generate hydroxyl radicals. Consistently, TPEN exhibits robust anti-tumor activity in vivo in colon cancer mouse xenografts.Conclusion: Our data show that TPEN induces cell death by chelating copper to produce TPEN-copper complexes that engage in redox cycling to selectively eliminate colon cancer cells. © 2014 Fatfat et al.; licensee BioMed Central Ltd
Uptake, delivery, and anticancer activity of thymoquinone nanoparticles in breast cancer cells
Abstract: Thymoquinone (TQ) is a promising anticancer molecule but its development is hindered by its limited bioavailability. Drug encapsulation is commonly used to overcome low drug solubility, limited bioavailability, and nonspecific targeting. In this project, TQ nanoparticles (TQ-NP) were synthesized and characterized. The cytotoxicity of the NP was investigated in nontumorigenic MCF-10-A breast cells, while the uptake, distribution, as well as the anticancer potential were investigated in MCF-7 and MDA-MB-231 breast cancer cells. Flash Nanoprecipitation and dynamic light scattering coupled with scanning electron microscopy were used to prepare and characterize TQ-NP prior to measuring their anticancer potential by MTT assay. The uptake and subcellular intake of TQ-NP were evaluated by fluorometry and confocal microscopy. TQ-NP were stable with a hydrodynamic average diameter size around 100 nm. Entrapment efficiency and loading content of TQ-NP were high (around 80 and 50 %, respectively). In vitro, TQ-NP had equal or enhanced anticancer activity effects compared to TQ in MCF-7 and aggressive MDA-MB-231 breast cancer cells, respectively, with no significant cytotoxicity of the blank NP. In addition, TQ and TQ-NP were relatively nontoxic to MCF-10-A normal breast cells. TQ-NP uptake mechanism was both time and concentration dependent. Treatment with inhibitors of endocytosis suggested the involvement of caveolin in TQ-NP uptake. This was further confirmed by subcellular localization findings showing the colocalization of TQ-NP with caveolin and transferrin as well as with the early and late markers of endocytosis. Altogether, the results describe an approach for the enhancement of TQ anticancer activity and uncover the mechanisms behind cell-TQ-NP interaction. Graphical Abstract: [Figure not available: see fulltext.] © 2016, Springer Science+Business Media Dordrecht
Thymoquinone Induces Telomere Shortening, DNA Damage and Apoptosis in Human Glioblastoma Cells
Background: A major concern of cancer chemotherapy is the side effects caused by the non-specific targeting of both normal and cancerous cells by therapeutic drugs. Much emphasis has been placed on discovering new compounds that target tumour cells more efficiently and selectively with minimal toxic effects on normal cells. Methodology/Principal Findings: The cytotoxic effect of thymoquinone, a component derived from the plant Nigella sativa, was tested on human glioblastoma and normal cells. Our findings demonstrated that glioblastoma cells were more sensitive to thymoquinone-induced antiproliferative effects. Thymoquinone induced DNA damage, cell cycle arrest and apoptosis in the glioblastoma cells. It was also observed that thymoquinone facilitated telomere attrition by inhibiting the activity of telomerase. In addition to these, we investigated the role of DNA-PKcs on thymoquinone mediated changes in telomere length. Telomeres in glioblastoma cells with DNA-PKcs were more sensitive to thymoquinone mediated effects as compared to those cells deficient in DNA-PKcs. Conclusions/Significance: Our results indicate that thymoquinone induces DNA damage, telomere attrition by inhibiting telomerase and cell death in glioblastoma cells. Telomere shortening was found to be dependent on the status of DNA-PKcs. Collectively, these data suggest that thymoquinone could be useful as a potential chemotherapeutic agent in th
MR imaging of therapy-induced changes of bone marrow
MR imaging of bone marrow infiltration by hematologic malignancies provides non-invasive assays of bone marrow cellularity and vascularity to supplement the information provided by bone marrow biopsies. This article will review the MR imaging findings of bone marrow infiltration by hematologic malignancies with special focus on treatment effects. MR imaging findings of the bone marrow after radiation therapy and chemotherapy will be described. In addition, changes in bone marrow microcirculation and metabolism after anti-angiogenesis treatment will be reviewed. Finally, new specific imaging techniques for the depiction of regulatory events that control blood vessel growth and cell proliferation will be discussed. Future developments are directed to yield comprehensive information about bone marrow structure, function and microenvironment
Ursolic acid-mediated changes in glycolytic pathway promote cytotoxic autophagy and apoptosis in phenotypically different breast cancer cells
Slug grazing effects on seedling and adult life stages of North American Prairie plants used in designed urban landscapes
A journey under the sea: The quest for marine anti-cancer alkaloids
The alarming increase in the global cancer death toll has fueled the quest for new effective anti-tumor drugs thorough biological screening of both terrestrial and marine organisms. Several plant-derived alkaloids are leading drugs in the treatment of different types of cancer and many are now being tested in various phases of clinical trials. Recently, marine-derived alkaloids, isolated from aquatic fungi, cyanobacteria, sponges, algae, and tunicates, have been found to also exhibit various anti-cancer activities including anti-angiogenic, anti-proliferative, inhibition of topoisomerase activities and tubulin polymerization, and induction of apoptosis and cytotoxicity. Two tunicate-derived alkaloids, aplidin and trabectedin, offer promising drug profiles, and are currently in phase II clinical trials against several solid and hematologic tumors. This review sheds light on the rich array of anti-cancer alkaloids in the marine ecosystem and introduces the most investigated compounds and their mechanisms of action. © 2011 by the authors; licensee MDPI, Basel, Switzerland.Aiello A, 2000, J NAT PROD, V63, P517, DOI 10.1021-np990513n; Allavena P, 2005, CANCER RES, V65, P2964, DOI 10.1158-0008-5472.CAN-04-4037; Anderson HJ, 1997, CANCER CHEMOTH PHARM, V39, P223; Aoki S, 2006, BIOCHEM BIOPH RES CO, V342, P101, DOI 10.1016-j.bbrc.2006.01.119; Aoki S, 2006, ANTI-CANCER DRUG, V17, P269, DOI 10.1097-00001813-200603000-00005; Appleton DR, 2002, J ORG CHEM, V67, P5402, DOI 10.1021-jo0201427; Aune GJ, 2008, CLIN CANCER RES, V14, P6449, DOI 10.1158-1078-0432.CCR-08-0730; Baldwin JE, 2001, ORG LETT, V3, P1145, DOI 10.1021-ol15646q; BARROWS LR, 1993, ANTI-CANCER DRUG DES, V8, P333; Baunbk D, 2008, MAR DRUGS, V6, P514, DOI 10.3390-md20080026; Belarbi E, 2003, BIOTECHNOL ADV, V21, P585, DOI 10.1016-S0734-9750(03)00100-9; Bergamaschi D, 1999, BRIT J CANCER, V79, P267, DOI 10.1038-sj.bjc.6690044; Berlinck RGS, 1998, J ORG CHEM, V63, P9850, DOI 10.1021-jo981607p; Biscardi M, 2005, ANN ONCOL, V16, P1667, DOI 10.1093-annonc-mdi311; Brahic C, 2002, BIOORGAN MED CHEM, V10, P2845, DOI 10.1016-S0968-0896(02)00148-7; Broggini M, 2003, LEUKEMIA, V17, P52, DOI 10.1038-sj.leu.2402788; Buchanan MS, 2007, BIOORG MED CHEM LETT, V17, P6860, DOI 10.1016-j.bmcl.2007.10.021; CAFIERI F, 1995, TETRAHEDRON LETT, V36, P7893, DOI 10.1016-0040-4039(95)01626-S; Carletti I, 2000, J NAT PROD, V63, P981, DOI 10.1021-np990408d; CARMELY S, 1989, TETRAHEDRON, V45, P2193, DOI 10.1016-S0040-4020(01)80079-X; CARNEY JR, 1993, J NAT PROD, V56, P153, DOI 10.1021-np50091a025; Casapullo A, 2000, J NAT PROD, V63, P447, DOI 10.1021-np9903292; Champoux JJ, 2001, ANNU REV BIOCHEM, V70, P369, DOI 10.1146-annurev.biochem.70.1.369; Chanvorachote P, 2006, CANCER RES, V66, P6353, DOI 10.1158-0008-5472.CAN-05-4533; Chen JH, 2003, J AM CHEM SOC, V125, P8734, DOI 10.1021-ja036050w; Cigler T, 2010, EXPERT OPIN PHARMACO, V11, P1587, DOI 10.1517-14656566.2010.486790; Clement JA, 2008, BIOORGAN MED CHEM, V16, P10022, DOI 10.1016-j.bmc.2008.10.024; Cole KE, 2011, J AM CHEM SOC, V133, P12474, DOI 10.1021-ja205972n; COLEMAN JE, 1995, TETRAHEDRON, V51, P10653, DOI 10.1016-0040-4020(95)00646-P; Cuadrado A, 2003, J BIOL CHEM, V278, P241, DOI 10.1074-jbc.M201010200; Dassonneville L, 2000, BIOCHEM PHARMACOL, V60, P527, DOI 10.1016-S0006-2952(00)00351-8; DEGUZMAN FS, 1999, CHEMINFORM, V30, DOI DOI 10.1002-CHIN.199929203; Dembitsky VM, 2005, MINI-REV MED CHEM, V5, P319; DeVita VT, 2008, CANCER RES, V68, P8643, DOI 10.1158-0008-5472.CAN-07-6611; D'Incalci M, 2010, MOL CANCER THER, V9, P2157, DOI 10.1158-1535-7163.MCT-10-0263; Edwards DJ, 2004, CHEM BIOL, V11, P817, DOI 10.1016-j.chembiol.2004.03.030; ELDEIRY WS, 1993, CELL, V75, P817, DOI 10.1016-0092-8674(93)90500-P; Erba E, 2003, BRIT J CANCER, V89, P763, DOI 10.1038-sj.bjc.6601130; Fedorov SN, 2004, PHARM RES, V21, P2307, DOI 10.1007-s11095-004-7683-5; Friedman D, 2002, CANCER RES, V62, P3377; Gajate C, 2003, CLIN CANCER RES, V9, P1535; Gallego MA, 2008, ONCOGENE, V27, P1981, DOI 10.1038-sj.onc.1210833; Gamble WR, 1999, BIOORGAN MED CHEM, V7, P1611, DOI 10.1016-S0968-0896(99)00089-9; Garcia-Fernandez LF, 2002, ONCOGENE, V21, P7533, DOI 10.1038-sj.onc.1205972; Garrido L, 2003, J ORG CHEM, V68, P293, DOI 10.1021-jo020487p; Germano G, 2010, CANCER RES, V70, P2235, DOI 10.1158-0008-5472.CAN-09-2335; Goldring WPD, 1999, ORG LETT, V1, P1471, DOI 10.1021-ol991029e; Gonzalez-Santiago L, 2006, CELL DEATH DIFFER, V13, P1968, DOI 10.1038-sj.cdd.4401898; Gordaliza M, 2007, CLIN TRANSL ONCOL, V9, P767, DOI 10.1007-s12094-007-0138-9; Gross H, 2006, J NAT PROD, V69, P640, DOI 10.1021-np050519e; Guven KC, 2010, MAR DRUGS, V8, P269, DOI 10.3390-md8020269; Halim H, 2011, ANTICANCER RES, V31, P193; Han BN, 2005, TETRAHEDRON, V61, P11723, DOI 10.1016-j.tet.2005.09.036; Hanahan D, 2011, CELL, V144, P646, DOI 10.1016-j.cell.2011.02.013; HARPER JW, 1993, CELL, V75, P805; Herrero AB, 2006, CANCER RES, V66, P8155, DOI 10.1158-0008-5472.CAN-06-0179; Hirano K, 2000, TETRAHEDRON, V56, P8107, DOI 10.1016-S0040-4020(00)00732-8; Hirano K, 2000, CHEM PHARM BULL, V48, P974; HOLDEN JA, 1985, J BIOL CHEM, V260, P4491; Hu JF, 2002, J NAT PROD, V65, P476, DOI 10.1021-np010471e; Hurley LH, 2001, ADV EXP MED BIOL, V500, P289; Jin S, 2000, P NATL ACAD SCI USA, V97, P6775, DOI 10.1073-pnas.97.12.6775; JORDAN MA, 1992, J CELL SCI, V102, P401; Jordan MA, 2004, NAT REV CANCER, V4, P253, DOI 10.1038-nr1317; Kashman Y, 1999, TETRAHEDRON LETT, V40, P997, DOI 10.1016-S0040-4039(98)02467-8; Kijjoa A, 2007, MAR DRUGS, V5, P6, DOI 10.3390-md502006; Kluza J, 2006, CANCER RES, V66, P3177, DOI 10.1158-0008-5472.CAN-05-1929; Kuznetsov G, 2009, MOL CANCER THER, V8, P2852, DOI 10.1158-1535-7163.MCT-09-0301; LaBarbera DV, 2009, ANTI-CANCER DRUG, V20, P425, DOI 10.1097-CAD.0b013e32832ae55f; LIBERRA K, 1995, PHARMAZIE, V50, P583; Liu R, 2007, ARCH PHARM RES, V30, P270, DOI 10.1007-BF02977605; LOW RL, 1984, J BIOL CHEM, V259, P4576; Luesch H, 2002, TETRAHEDRON, V58, P7959, DOI 10.1016-S0040-4020(02)00895-5; Luesch H, 2000, J NAT PROD, V63, P611, DOI 10.1021-np990543q; Luesch H, 2001, J AM CHEM SOC, V123, P5418, DOI 10.1021-ja010453j; Luesch H, 2006, NAT CHEM BIOL, V2, P158, DOI 10.1038-nchembio769; Makarieva TN, 2001, J NAT PROD, V64, P1559, DOI 10.1021-np010161w; Marco E, 2005, J MED CHEM, V48, P3796, DOI 10.1021-jm049060w; Marquez BL, 2002, J NAT PROD, V65, P866, DOI 10.1021-np0106283; Marshall KM, 2003, BIOCHEM PHARMACOL, V66, P447, DOI 10.1016-S0006-2952(03)00209-0; Martinez JD, 2010, FUTURE ONCOL, V6, P1857, DOI [10.2217-fon.10.132, 10.2217-FON.10.132]; Meijer L, 2000, CHEM BIOL, V7, P51, DOI 10.1016-S1074-5521(00)00063-6; Milligan KE, 2000, J NAT PROD, V63, P1440, DOI 10.1021-np000133y; Minuzzo M, 2005, MOL PHARMACOL, V68, P1496, DOI 10.1124-mol.105.013615; Minuzzo M, 2000, P NATL ACAD SCI USA, V97, P6780, DOI 10.1073-pnas.97.12.6780; Molinski TF, 2009, NAT REV DRUG DISCOV, V8, P69, DOI 10.1038-nrd2487; Moneo V, 2007, MOL CANCER THER, V6, P1310, DOI 10.1158-1535-7163.MCT-06-0729; Morrison WB, 2010, J VET INTERN MED, V24, P1249, DOI 10.1111-j.1939-1676.2010.0590.x; MUNRO MHG, 1999, PROG IND MICROBIOL, V35, P15; Nobili S, 2009, PHARMACOL RES, V59, P365, DOI 10.1016-j.phrs.2009.01.017; Pawlik JR, 2002, J CHEM ECOL, V28, P1103, DOI 10.1023-A:1016221415028; Pietra F, 1997, NAT PROD REP, V14, P453, DOI 10.1039-np9971400453; Pla D, 2008, ANTI-CANCER AGENT ME, V8, P746; Pommier Y, 2006, NAT REV CANCER, V6, P789, DOI 10.1038-nrc1977; Prager G.W., 2011, HAMOSTASEOLOGIE, V32, P105, DOI DOI 10.5482-HA-1163; Ramaswamy AV, 2007, J NAT PROD, V70, P1977, DOI 10.1021-np0704250; Rashid MA, 2001, J NAT PROD, V64, P1454, DOI 10.1021-np010214+; Rhinehart K. L., 2004, DRUGS R D, Patent No. [2003-406997, 20040059112]; Richelle-Maurer E, 2003, J BIOTECHNOL, V100, P169, DOI 10.1016-S0168-1656(02)00251-1; Rinehart KL, 2000, MED RES REV, V20, P1; Rockwell S, 2010, INT J RADIAT BIOL, V86, P63, DOI 10.3109-09553000903264531; Roskelley CD, 2001, CANCER RES, V61, P6788; Saito N, 2004, TETRAHEDRON, V60, P3873, DOI 10.1016-j.tet.2004.02.071; SAKEMI S, 1991, J ORG CHEM, V56, P4304, DOI 10.1021-jo00013a044; Schmidt EW, 1995, J NAT PROD, V58, P1861, DOI 10.1021-np50126a008; Scotto KW, 2002, ANTI-CANCER DRUG, V13, pS3; Sessa C, 2011, ANN ONCOL, V22, P7, DOI 10.1093-annonc-mdq641; Sherr CJ, 1996, SCIENCE, V274, P1672, DOI 10.1126-science.274.5293.1672; Shinkre BA, 2007, BIOORG MED CHEM LETT, V17, P2890, DOI 10.1016-j.bmcl.2007.02.065; Simmons TL, 2005, MOL CANCER THER, V4, P333; Smetanina OF, 2007, J NAT PROD, V70, P906, DOI 10.1021-np060396d; Soares DG, 2007, P NATL ACAD SCI USA, V104, P13062, DOI 10.1073-pnas.0609877104; TALPIR R, 1994, TETRAHEDRON LETT, V35, P4453, DOI 10.1016-S0040-4039(00)73382-X; Tan LT, 2010, J APPL PHYCOL, V22, P659, DOI 10.1007-s10811-010-9506-x; Tan LT, 2007, PHYTOCHEMISTRY, V68, P954, DOI 10.1016-j.phytochem.2007.01.012; Taraboletti G, 2004, BRIT J CANCER, V90, P2418, DOI 10.1038-sj.bjc.6601864; Tasdemir D, 2002, J MED CHEM, V45, P529, DOI 10.1021-jm0102856; Tavecchio M, 2008, EUR J CANCER, V44, P609, DOI 10.1016-j.ejca.2008.01.003; Thomas T, 2010, ISME J, V4, P1557, DOI 10.1038-ismej.2010.74; Torres YR, 2002, J ORG CHEM, V67, P5429, DOI 10.1021-jo011174h; Torres YR, 2000, J NAT PROD, V63, P1098, DOI 10.1021-np9905618; Uddin MJ, 2001, J NAT PROD, V64, P1169, DOI 10.1021-np010066n; Umeyama A, 1998, J NAT PROD, V61, P1433, DOI 10.1021-np980207u; Urban S, 2002, J NAT PROD, V65, P1371, DOI 10.1021-np010594z; Reddy AV, 2006, BIOORGAN MED CHEM, V14, P4452, DOI 10.1016-j.bmc.2006.02.033; Verbitski SM, 2002, J ORG CHEM, V67, P7124, DOI 10.1021-jo026012f; Wade RH, 2009, MOL BIOTECHNOL, V43, P177, DOI 10.1007-s12033-009-9193-5; Wang B, 2011, J ORG CHEM, V76, P1140, DOI 10.1021-jo102478x; Wang JC, 2002, NAT REV MOL CELL BIO, V3, P430, DOI 10.1038-nrm831; Weissman K, 2004, CHEM BIOL, V11, P743, DOI 10.1016-j.chembiol.2004.06.003; Williams DE, 2002, J ORG CHEM, V67, P245, DOI 10.1021-jo016101c; Winter-Vann AM, 2005, NAT REV CANCER, V5, P405, DOI 10.1038-nrc1612; Xin ZH, 2007, J NAT PROD, V70, P853, DOI 10.1021-np060516h; Yamada HY, 2010, CURR CANCER DRUG TAR, V10, P623; Yokokawa F, 2001, TETRAHEDRON LETT, V42, P4171, DOI 10.1016-S0040-4039(01)00678-5; Zhang M, 2008, J NAT PROD, V71, P985, DOI 10.1021-np700737g; Zhang W, 2003, J BIOTECHNOL, V100, P161, DOI 10.1016-S0168-1656(02)00255-9; Zhou BN, 2000, TETRAHEDRON, V56, P5781, DOI 10.1016-S0040-4020(00)00534-2; Zubia E, 2005, MINI-REV ORG CHEM, V2, P389, DOI 10.2174-15701930577432270720242
- …
