Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cancer Lett ; 450: 42-52, 2019 05 28.
Article in English | MEDLINE | ID: mdl-30790679

ABSTRACT

Piperlongumine (PL), naturally synthesized in long pepper, is known to selectively kill tumor cells via perturbation of reactive oxygen species (ROS) homeostasis. ROS are the primary effector molecules of radiation, and increase of ROS production by pharmacological modulation is known to enhance radioresponse. We therefore investigated the radiosensitizing effect of PL in colorectal cancer cells (CT26 and DLD-1) and CT26 tumor-bearing mice. Firstly, we found that PL induced excessive production of ROS due to depletion of glutathione and inhibition of thioredoxin reductase. Secondly, PL enhanced both the intrinsic and hypoxic radiosensitivity of tumor cells, linked to ROS-mediated increase of DNA damage, G2/M cell cycle arrest, and inhibition of cellular respiration. Finally, the radiosensitizing effect of PL was verified in vivo. PL improved the tumor response to both single and fractionated radiation, resulting in a significant increase of survival rate of tumor-bearing mice, while it was ineffective on its own. In line with in vitro findings, enhanced radioresponse is associated with inhibition of antioxidant systems. In conclusion, our results suggest that PL could be a potential radiosensitizer in colorectal cancer.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/radiotherapy , Dioxolanes/pharmacology , Glutathione/antagonists & inhibitors , Radiation-Sensitizing Agents/pharmacology , Reactive Oxygen Species/metabolism , Thioredoxins/antagonists & inhibitors , Animals , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Cell Line, Tumor , Colorectal Neoplasms/metabolism , DNA Damage , Glutathione/metabolism , Humans , Mice , Oxygen Consumption/drug effects , Oxygen Consumption/radiation effects , Signal Transduction/drug effects , Signal Transduction/radiation effects , Thioredoxins/metabolism
2.
Front Pharmacol ; 9: 1073, 2018.
Article in English | MEDLINE | ID: mdl-30337872

ABSTRACT

Background and Purpose: The anti-diabetic biguanide drugs metformin and phenformin exhibit antitumor activity in various models. However, their radiomodulatory effect under hypoxic conditions, particularly for phenformin, is largely unknown. This study therefore examines whether metformin and phenformin as mitochondrial complex I blockades could overcome hypoxic radioresistance through inhibition of oxygen consumption. Materials and Methods: A panel of colorectal cancer cells (HCT116, DLD-1, HT29, SW480, and CT26) was exposed to metformin or phenformin for 16 h at indicated concentrations. Afterward, cell viability was measured by MTT and colony formation assays. Apoptosis and reactive oxygen species (ROS) were detected by flow cytometry. Phosphorylation of AMP-activated protein kinase (AMPK) was examined by western blot. Mitochondria complexes activity and oxygen consumption rate (OCR) were measured by seahorse analyzer. The radiosensitivity of tumor cells was assessed by colony formation assay under aerobic and hypoxic conditions. The in vitro findings were further validated in colorectal CT26 tumor model. Results: Metformin and phenformin inhibited mitochondrial complex I activity and subsequently reduced OCR in a dose-dependent manner starting at 3 mM and 30 µM, respectively. As a result, the hypoxic radioresistance of tumor cells was counteracted by metformin and phenformin with an enhancement ratio about 2 at 9 mM and 100 µM, respectively. Regarding intrinsic radioresistance, both of them did not exhibit any effect although there was an increase of phosphorylation of AMPK and ROS production. In tumor-bearing mice, metformin or phenformin alone did not show any anti-tumor effect. While in combination with radiation, both of them substantially delayed tumor growth and enhanced radioresponse, respectively, by 1.3 and 1.5-fold. Conclusion: Our results demonstrate that metformin and phenformin overcome hypoxic radioresistance through inhibition of mitochondrial respiration, and provide a rationale to explore metformin and phenformin as hypoxic radiosensitizers.

3.
Oncotarget ; 8(22): 35728-35742, 2017 May 30.
Article in English | MEDLINE | ID: mdl-28415723

ABSTRACT

Auranofin (AF) is an anti-arthritic drug considered for combined chemotherapy due to its ability to impair the redox homeostasis in tumor cells. In this study, we asked whether AF may in addition radiosensitize tumor cells by targeting thioredoxin reductase (TrxR), a critical enzyme in the antioxidant defense system operating through the reductive protein thioredoxin. Our principal findings in murine 4T1 and EMT6 tumor cells are that AF at 3-10 µM is a potent radiosensitizer in vitro, and that at least two mechanisms are involved in TrxR-mediated radiosensitization. The first one is linked to an oxidative stress, as scavenging of reactive oxygen species (ROS) by N-acetyl cysteine counteracted radiosensitization. We also observed a decrease in mitochondrial oxygen consumption with spared oxygen acting as a radiosensitizer under hypoxic conditions. Overall, radiosensitization was accompanied by ROS overproduction, mitochondrial dysfunction, DNA damage and apoptosis, a common mechanism underlying both cytotoxic and antitumor effects of AF. In tumor-bearing mice, a simultaneous disruption of the thioredoxin and glutathione systems by the combination of AF and buthionine sulfoximine was shown to significantly improve tumor radioresponse. In conclusion, our findings illuminate TrxR in cancer cells as an exploitable radiobiological target and warrant further validation of AF in combination with radiotherapy.


Subject(s)
Auranofin/pharmacology , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Reactive Oxygen Species/metabolism , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , DNA Damage/drug effects , Disease Models, Animal , Glutathione/metabolism , Hypoxia/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Oxygen Consumption/drug effects , Thioredoxin-Disulfide Reductase/metabolism
4.
Radiother Oncol ; 119(2): 291-9, 2016 05.
Article in English | MEDLINE | ID: mdl-26874542

ABSTRACT

BACKGROUND AND PURPOSE: High arginase-1 (Arg) expression by myeloid-derived suppressor cells (MDSC) is known to inhibit antitumor T-cell responses through depletion of l-arginine. We have previously shown that nitric oxide (NO), an immune mediator produced from l-arginine, is a potent radiosensitizer of hypoxic tumor cells. This study therefore examines whether Arg(+) overexpressing MDSC may confer radioresistance through depleting the substrate for NO synthesis. MATERIAL AND METHODS: MDSC and Arg expression were studied in preclinical mouse CT26 and 4T1 tumor models and further validated in rectal cancer patients in comparison with healthy donors. The radioprotective effect of MDSC was analyzed in hypoxic tumor cells with regard to l-arginine depletion. RESULTS: In both mouse tumors and cancer patients, MDSC expansion was associated with Arg activation causing accelerated l-arginine consumption. l-Arginine depletion in turn profoundly suppressed the capacity of classically activated macrophages to synthesize NO resulting in impaired tumor cell radiosensitivity. In advanced cT3-4 rectal cancer, circulating neutrophils revealed Arg overexpression approaching that in MDSC, therefore mounting a protumor compartment wherein Arg(+) neutrophils increased from 17% to over 90%. CONCLUSIONS: Protumor Arg(+) MDSC reveal a unique ability to radioprotect tumor cells through l-arginine depletion, a common mechanism behind both T-cell and macrophage inhibition.


Subject(s)
Arginase/physiology , Arginine/metabolism , Myeloid-Derived Suppressor Cells/physiology , Rectal Neoplasms/radiotherapy , Animals , HCT116 Cells , Humans , Macrophages/physiology , Mice , Neutrophils/physiology , Nitric Oxide/biosynthesis , Rectal Neoplasms/metabolism , T-Lymphocytes/immunology
5.
Int J Radiat Oncol Biol Phys ; 76(5): 1520-7, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20338478

ABSTRACT

PURPOSE: Nitric oxide (NO), synthesized by the inducible nitric oxide synthase (iNOS), is known to inhibit metabolic oxygen consumption because of interference with mitochondrial respiratory activity. This study examined whether activation of iNOS (a) directly in tumor cells or (b) in bystander macrophages may improve radioresponse through sparing of oxygen. METHODS AND MATERIALS: EMT-6 tumor cells and RAW 264.7 macrophages were exposed to bacterial lipopolysaccharide plus interferon-gamma, and examined for iNOS expression by reverse transcription polymerase chain reaction, Western blotting and enzymatic activity. Tumor cells alone, or combined with macrophages were subjected to metabolic hypoxia and analyzed for radiosensitivity by clonogenic assay, and for oxygen consumption by electron paramagnetic resonance and a Clark-type electrode. RESULTS: Both tumor cells and macrophages displayed a coherent picture of iNOS induction at transcriptional/translational levels and NO/nitrite production, whereas macrophages showed also co-induction of the inducible heme oxygenase-1, which is associated with carbon monoxide (CO) and bilirubin production. Activation of iNOS in tumor cells resulted in a profound oxygen sparing and a 2.3-fold radiosensitization. Bystander NO-producing, but not CO-producing, macrophages were able to block oxygen consumption by 1.9-fold and to radiosensitize tumor cells by 2.2-fold. Both effects could be neutralized by aminoguanidine, a metabolic iNOS inhibitor. An improved radioresponse was clearly observed at macrophages to tumor cells ratios ranging between 1:16 to 1:1. CONCLUSIONS: Our study is the first, as far as we are aware, to provide evidence that iNOS may induce radiosensitization through oxygen sparing, and illuminates NO-producing macrophages as a novel determinant of tumor cell radioresponse within the hypoxic tumor microenvironment.


Subject(s)
Macrophage Activation , Macrophages/enzymology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/biosynthesis , Oxygen Consumption/physiology , Radiation Tolerance/physiology , Animals , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Respiration , Electron Spin Resonance Spectroscopy , Enzyme Activation , Guanidines/pharmacology , Heme Oxygenase-1/biosynthesis , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Mice , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors
6.
Org Lett ; 11(15): 3302-5, 2009 Aug 06.
Article in English | MEDLINE | ID: mdl-19591453

ABSTRACT

A robust and reliable method has been developed for reductive amination of primary amines with various aldehydes and ketones using Zn(ClO(4))(2) x 6 H(2)O as a catalyst. [In-H] generated in situ via a combination of InCl(3) and Et(3)SiH is employed as an effective reducing system. A variety of secondary amines can be synthesized in a one-pot procedure in excellent yields.


Subject(s)
Aldehydes/chemistry , Amines/chemical synthesis , Indium/chemistry , Ketones/chemistry , Zinc Compounds/chemistry , Amination
7.
J Org Chem ; 73(22): 8829-37, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-18939879

ABSTRACT

A new strategy has been developed for reductive amination of aldehydes and ketones with the InCl3/Et3SiH/MeOH system, which is a nontoxic system with highly chemoselective and nonwater sensitive properties. The methodology can be applied to a variety of cyclic, acyclic, aromatic, and aliphatic amines. Functionalities including ester, hydroxyl, carboxylic acid, and olefin are found to be stable under our conditions. The reaction shows a first-order kinetics profile with respect to both InCl3 and Et3SiH. Spectroscopic techniques such as NMR and ESI-MS have been employed to probe the active and resulting species arising from InCl3 and Et3SiH in MeOH, which are important in deriving a mechanistic proposal. In the ESI-MS studies, we have first discovered the existence of stable methanol-coordinated indium(III) species which are presumably responsible for the gentle generation of indium hydride at room temperature. The solvent attribution was crucial in tuning the reactivity of [In-H] species, leading to the establishment of mild reaction conditions. The system is superior in flexible tuning of hydride reactivity, resulting in the system being highly chemoselective.

8.
Int J Radiat Oncol Biol Phys ; 71(3): 647-51, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18514774

ABSTRACT

Activated T lymphocytes are known to kill tumor cells by triggering cytolytic mechanisms; however, their ability to enhance radiation responses remains unclear. This study examined the radiosensitizing potential of mouse CD8+ T cells, obtained by T-cell-tailored expansion and immunomagnetic purification. Activated CD8+ T cells displayed an interferon (IFN)-gamma+ phenotype and enhanced by 1.8-fold the radiosensitivity of EMT-6 tumor cells in 1% oxygen, which modeled tumor-relevant hypoxia. Radiosensitization was counteracted by neutralizing IFN-gamma or by blocking the inducible isoform of nitric oxide synthase, thus delineating the immune-tumor cell interaction through the IFN-gamma secretion pathway. Reverse transcriptase-polymerase chain reaction, enzyme-linked immunosorbent assay, and fluorescence-activated cell sorter data in agreement detected downregulation of the IFN-gamma gene by hypoxia, which caused IFN-gamma deficiency next to radioresistance. Therefore, immune and radiation responses are likely to be allied in the hypoxic tumor microenvironment, and CD8+ T cells may bridge immunostimulatory and radiosensitizing strategies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Immunity, Innate/immunology , Interferon-gamma/immunology , Mammary Neoplasms, Animal/immunology , Animals , Cell Hypoxia/immunology , Cell Hypoxia/radiation effects , Cells, Cultured , Dose-Response Relationship, Radiation , Immunity, Innate/radiation effects , Mice , Mice, Inbred BALB C , Radiation Dosage
9.
Int J Radiat Oncol Biol Phys ; 66(5): 1473-80, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17056198

ABSTRACT

PURPOSE: To explore whether antitumor immunoadjuvant OM-174 can stimulate immune cells to produce interferon-gamma (IFN-gamma) and thereby radiosensitize tumor cells. METHODS AND MATERIALS: Splenocytes from BALB/c mice were stimulated by OM-174 at plasma-achievable concentrations (0.03-3 mug/mL), and afterward analyzed for the expression and secretion of IFN-gamma by reverse transcriptase-polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. Stimulated splenocytes were used as a source of IFN-gamma to radiosensitize hypoxic EMT-6 tumor cells through the cytokine-inducible isoform of nitric oxide synthase (iNOS). RESULTS: OM-174 activated the production of IFN-gamma at high levels that reached 70 ng/mL in normoxia (21% oxygen) and 27 ng/mL in tumor-relevant hypoxia (1% oxygen). This caused up to 2.1-fold radiosensitization of EMT-6 tumor cells, which was associated with the iNOS-mediated production of the radiosensitizing molecule nitric oxide, as confirmed by accumulation of its oxidative metabolite nitrite, Western blot analysis, and reverse transcriptase-polymerase chain reaction. Both iNOS activation and radiosensitization were counteracted by neutralizing antibodies against IFN-gamma. The same mechanism of radiosensitization through the IFN-gamma secretion pathway was identified for IL-12 + IL-18, which are known to mediate IFN-gamma responses. Hypoxia displayed a dual effect on the immune-tumor cell interaction, by downregulating the expression of the IFN-gamma gene while upregulating iNOS at transcriptional level. CONCLUSION: Immunoadjuvant OM-174 is an efficient radiosensitizer of tumor cells through activation of the IFN-gamma secretion pathway in immune cells. This finding indicates a rationale for combining immunostimulatory and radiosensitizing strategies and extends the potential therapeutic applications of OM-174.


Subject(s)
Adjuvants, Immunologic/pharmacology , Interferon-gamma/metabolism , Lipopolysaccharides/pharmacology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/biosynthesis , Radiation Tolerance , Animals , Cell Hypoxia/physiology , Cell Line, Tumor , Down-Regulation , Enzyme Activation , Immunity, Cellular , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/genetics , Interleukin-12/metabolism , Interleukin-18/metabolism , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/genetics , RNA, Messenger/metabolism , Radiation Tolerance/drug effects , Radiation Tolerance/immunology , Spleen/cytology , Up-Regulation
10.
J Am Chem Soc ; 128(10): 3130-1, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16522078

ABSTRACT

We have developed an efficient Pd(II)-catalyzed enantioselective oxidative tandem cyclization strategy using molecular oxygen as a green oxidant for the double 5-exo-trig cyclizations of N-(2-allylaryl) amides to afford a variety of indolines in good yields without the formation of undesired monocyclization products. By employing Pd(TFA)2/(-)-sparteine as the chiral catalyst, we obtained tandem cyclization products with high enantioselectivity (up to 91% ee).


Subject(s)
Indoles/chemical synthesis , Catalysis , Cyclization , Oxidation-Reduction , Oxygen/chemistry , Palladium/chemistry , Sparteine/chemistry , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...