Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 128(11): 2013-2024, 2023 06.
Article in English | MEDLINE | ID: mdl-37012319

ABSTRACT

BACKGROUND: Cisplatin (CDDP) is a mainstay treatment for advanced head and neck squamous cell carcinomas (HNSCC) despite a high frequency of innate and acquired resistance. We hypothesised that tumours acquire CDDP resistance through an enhanced reductive state dependent on metabolic rewiring. METHODS: To validate this model and understand how an adaptive metabolic programme might be imprinted, we performed an integrated analysis of CDDP-resistant HNSCC clones from multiple genomic backgrounds by whole-exome sequencing, RNA-seq, mass spectrometry, steady state and flux metabolomics. RESULTS: Inactivating KEAP1 mutations or reductions in KEAP1 RNA correlated with Nrf2 activation in CDDP-resistant cells, which functionally contributed to resistance. Proteomics identified elevation of downstream Nrf2 targets and the enrichment of enzymes involved in generation of biomass and reducing equivalents, metabolism of glucose, glutathione, NAD(P), and oxoacids. This was accompanied by biochemical and metabolic evidence of an enhanced reductive state dependent on coordinated glucose and glutamine catabolism, associated with reduced energy production and proliferation, despite normal mitochondrial structure and function. CONCLUSIONS: Our analysis identified coordinated metabolic changes associated with CDDP resistance that may provide new therapeutic avenues through targeting of these convergent pathways.


Subject(s)
Antineoplastic Agents , Head and Neck Neoplasms , Humans , Cisplatin/metabolism , Squamous Cell Carcinoma of Head and Neck , Kelch-Like ECH-Associated Protein 1/genetics , NF-E2-Related Factor 2/genetics , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Glucose , Antineoplastic Agents/pharmacology
2.
J Immunother Cancer ; 10(8)2022 08.
Article in English | MEDLINE | ID: mdl-36002187

ABSTRACT

BACKGROUND: The existence of immunologically 'cold tumors' frequently found across a wide spectrum of tumor types represents a significant challenge for cancer immunotherapy. Cold tumors have poor baseline pan-leukocyte infiltration, including a low prevalence of cytotoxic lymphocytes, and not surprisingly respond unfavorably to immune checkpoint (IC) inhibitors. We hypothesized that cold tumors harbor a mechanism of immune escape upstream and independent of ICs that may be driven by tumor biology rather than differences in mutational neoantigen burden. METHODS: Using a bioinformatic approach to analyze TCGA (The Cancer Genome Atlas) RNA sequencing data we identified genes upregulated in cold versus hot tumors across four different smoking-related cancers, including squamous carcinomas from the oral cavity (OCSCC) and lung (LUSC), and adenocarcinomas of the bladder (BLCA) and lung (LUAD). Biological significance of the gene most robustly associated with a cold tumor phenotype across all four tumor types, glutathione peroxidase 2 (GPX2), was further evaluated using a combination of in silico analyses and functional genomic experiments performed both in vitro and in in vivo with preclinical models of oral cancer. RESULTS: Elevated RNA expression of five metabolic enzymes including GPX2, aldo-keto reductase family 1 members AKR1C1, AKR1C3, and cytochrome monoxygenases (CP4F11 and CYP4F3) co-occurred in cold tumors across all four smoking-related cancers. These genes have all been linked to negative regulation of arachidonic acid metabolism-a well-established inflammatory pathway-and are also known downstream targets of the redox sensitive Nrf2 transcription factor pathway. In OCSCC, LUSC, and LUAD, GPX2 expression was highly correlated with Nrf2 activation signatures, also elevated in cold tumors. In BLCA, however, GPX2 correlated more strongly than Nrf2 signatures with decreased infiltration of multiple leukocyte subtypes. GPX2 inversely correlated with expression of multiple pro- inflammatory cytokines/chemokines and NF-kB activation in cell lines and knockdown of GPX2 led to increased secretion of prostaglandin E2 (PGE2) and interleukin-6. Conversely, GPX2 overexpression led to reduced PGE2 production in a murine OCSCC model (MOC1). GPX2 overexpressing MOC1 tumors had a more suppressive tumor immune microenvironment and responded less favorably to anti-cytotoxic T-lymphocytes-associated protein 4 IC therapy in mice. CONCLUSION: GPX2 overexpression represents a novel potentially targetable effector of immune escape in cold tumors.


Subject(s)
Glutathione Peroxidase/metabolism , Immune Checkpoint Inhibitors , NF-E2-Related Factor 2 , Animals , Dinoprostone , Glutathione Peroxidase/genetics , Mice , NF-E2-Related Factor 2/metabolism , Tumor Microenvironment
3.
Mol Cell Biol ; 38(9)2018 05 01.
Article in English | MEDLINE | ID: mdl-29463647

ABSTRACT

The molecular mechanisms underlying resistance to radiotherapy in breast cancer cells remain elusive. Previously, we reported that elevated ß1-integrin is associated with enhanced breast cancer cell survival postirradiation, but how ß1-integrin conferred radioresistance was unclear. Ionizing radiation (IR) induced cell killing correlates with the efficiency of DNA double-strand break (DSB) repair, and we found that nonmalignant breast epithelial (S1) cells with low ß1-integrin expression have a higher frequency of S-phase-specific IR-induced chromosomal aberrations than the derivative malignant breast (T4-2) cells with high ß1-integrin expression. In addition, there was an increased frequency of IR-induced homologous recombination (HR) repairosome focus formation in T4-2 cells compared with that of S1 cells. Cellular levels of Rad51 in T4-2 cells, a critical factor in HR-mediated DSB repair, were significantly higher. Blocking or depleting ß1-integrin activity in T4-2 cells reduced Rad51 levels, while ectopic expression of ß1-integrin in S1 cells correspondingly increased Rad51 levels, suggesting that Rad51 is regulated by ß1-integrin. The low level of Rad51 protein in S1 cells was found to be due to rapid degradation by the ubiquitin proteasome pathway (UPP). Furthermore, the E3 ubiquitin ligase RING1 was highly upregulated in S1 cells compared to T4-2 cells. Ectopic ß1-integrin expression in S1 cells reduced RING1 levels and increased Rad51 accumulation. In contrast, ß1-integrin depletion in T4-2 cells significantly increased RING1 protein levels and potentiated Rad51 ubiquitination. These data suggest for the first time that elevated levels of the extracellular matrix receptor ß1-integrin can increase tumor cell radioresistance by decreasing Rad51 degradation through a RING1-mediated proteasomal pathway.


Subject(s)
Integrin beta1/physiology , Integrin beta1/radiation effects , Rad51 Recombinase/metabolism , Breast Neoplasms , Cell Line, Tumor , Cell Survival , DNA , DNA Breaks, Double-Stranded , DNA Damage , DNA Repair/physiology , Female , Homologous Recombination/physiology , Humans , Integrin beta1/metabolism , Rad51 Recombinase/physiology , Radiation, Ionizing , Recombinational DNA Repair/physiology
4.
Mol Cell Biol ; 38(6)2018 03 15.
Article in English | MEDLINE | ID: mdl-29298824

ABSTRACT

The human MOF (hMOF) protein belongs to the MYST family of histone acetyltransferases and plays a critical role in transcription and the DNA damage response. MOF is essential for cell proliferation; however, its role during replication and replicative stress is unknown. Here we demonstrate that cells depleted of MOF and under replicative stress induced by cisplatin, hydroxyurea, or camptothecin have reduced survival, a higher frequency of S-phase-specific chromosome damage, and increased R-loop formation. MOF depletion decreased replication fork speed and, when combined with replicative stress, also increased stalled replication forks as well as new origin firing. MOF interacted with PCNA, a key coordinator of replication and repair machinery at replication forks, and affected its ubiquitination and recruitment to the DNA damage site. Depletion of MOF, therefore, compromised the DNA damage repair response as evidenced by decreased Mre11, RPA70, Rad51, and PCNA focus formation, reduced DNA end resection, and decreased CHK1 phosphorylation in cells after exposure to hydroxyurea or cisplatin. These results support the argument that MOF plays an important role in suppressing replication stress induced by genotoxic agents at several stages during the DNA damage response.


Subject(s)
Antineoplastic Agents/pharmacology , Camptothecin/pharmacology , Cisplatin/pharmacology , DNA Damage/drug effects , DNA Replication/drug effects , Histone Acetyltransferases/metabolism , Hydroxyurea/pharmacology , Cell Death/drug effects , DNA Repair/drug effects , HeLa Cells , Histone Acetyltransferases/genetics , Homologous Recombination/drug effects , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , S Phase/drug effects
5.
Nucleic Acids Res ; 45(5): 2585-2599, 2017 03 17.
Article in English | MEDLINE | ID: mdl-27994036

ABSTRACT

Microhomology-mediated end joining (MMEJ), an error-prone pathway for DNA double-strand break (DSB) repair, is implicated in genomic rearrangement and oncogenic transformation; however, its contribution to repair of radiation-induced DSBs has not been characterized. We used recircularization of a linearized plasmid with 3΄-P-blocked termini, mimicking those at X-ray-induced strand breaks, to recapitulate DSB repair via MMEJ or nonhomologous end-joining (NHEJ). Sequence analysis of the circularized plasmids allowed measurement of relative activity of MMEJ versus NHEJ. While we predictably observed NHEJ to be the predominant pathway for DSB repair in our assay, MMEJ was significantly enhanced in preirradiated cells, independent of their radiation-induced arrest in the G2/M phase. MMEJ activation was dependent on XRCC1 phosphorylation by casein kinase 2 (CK2), enhancing XRCC1's interaction with the end resection enzymes MRE11 and CtIP. Both endonuclease and exonuclease activities of MRE11 were required for MMEJ, as has been observed for homology-directed DSB repair (HDR). Furthermore, the XRCC1 co-immunoprecipitate complex (IP) displayed MMEJ activity in vitro, which was significantly elevated after irradiation. Our studies thus suggest that radiation-mediated enhancement of MMEJ in cells surviving radiation therapy may contribute to their radioresistance and could be therapeutically targeted.


Subject(s)
Casein Kinase II/metabolism , DNA End-Joining Repair , DNA-Binding Proteins/metabolism , Cell Line, Tumor , DNA Breaks, Double-Stranded , Humans , Phosphorylation , X-Rays , X-ray Repair Cross Complementing Protein 1
6.
Cancer Res ; 73(12): 3737-48, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23576567

ABSTRACT

ß1-integrin induction enhances breast cancer cell survival after exposure to ionizing radiation (IR), but the mechanisms of this effect remain unclear. Although NF-κB initiates prosurvival signaling pathways post-IR, the molecular function of NF-κB with other key elements in radioresistance, particularly with respect to extracellular matrix-induced signaling, is not known. We discovered a typical NF-κB-binding site in the ß1-integrin promoter region, indicating a possible regulatory role for NF-κB. Using three-dimensional laminin-rich extracellular matrix (3D lrECM) culture, we show that NF-κB is required for ß1-integrin transactivation in T4-2 breast cancer cells post-IR. Inhibition of NF-κB reduced clonogenic survival and induced apoptosis and cytostasis in formed tumor colonies. In addition, T4-2 tumors with inhibition of NF-κB activity exhibit decreased growth in athymic mice, which was further reduced by IR with downregulated ß1-integrin expression. Direct interactions between ß1-integrin and NF-κB p65 were induced in nonmalignant breast epithelial cells, but not in malignant cells, indicating context-specific regulation. As ß1-integrin also activates NF-κB, our findings reveal a novel forward feedback pathway that could be targeted to enhance therapy.


Subject(s)
Breast Neoplasms/metabolism , Cell Culture Techniques/methods , Integrin beta1/metabolism , NF-kappa B/metabolism , Animals , Binding Sites/genetics , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/radiation effects , Extracellular Matrix/metabolism , Female , Humans , Integrin beta1/genetics , Mice , Mice, Nude , Microscopy, Confocal , NF-kappa B/antagonists & inhibitors , Phenylenediamines/pharmacology , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Binding/radiation effects , Radiation Tolerance/genetics , Radiation, Ionizing , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/metabolism , Tumor Burden/drug effects , Tumor Burden/radiation effects , Xenograft Model Antitumor Assays
7.
J Biol Chem ; 285(7): 4464-71, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20007691

ABSTRACT

The high mobility group AT-hook 2 (HMGA2), a DNA architectural protein, is highly regulated during development and plays an important role in tumorigenesis. Indeed, HMGA2 was overexpressed in many different kinds of tumors. However, the mechanisms regulating HMGA2 expression remain elusive. Using microarray analysis, we found that HMGA2, along with a dozen of other genes, was co-repressed by ZBRK1, BRCA1, and CtIP. BRCA1 exerts its transcriptional repression activity through interaction with the transcriptional repressor ZBRK1 in the central domain, and with CtIP in the C-terminal BRCT domain. Here, we show that ZBRK1, BRCA1, and CtIP form a repression complex that coordinately regulates HMGA2 expression via a ZBRK1 recognition site in the HMGA2 promoter. Depletion of any of the proteins in this complex via adenoviral RNA interference in MCF10A mammary epithelial cells activates HMGA2 expression, resulting in increased colony formation in soft agar. Similarly, depletion of ZBRK1, or ectopic overexpression of HMGA2, in MCF10A cells induces abnormal acinar size with increased cell number and inhibits normal acinar formation. Consistently, many BRCA1-deficient mouse breast tumors express higher levels of HMGA2 than BRCA1-proficient tumors. These results suggest that activation of HMGA2 gene expression through derepression of the ZBRK1/BRCA1/CtIP complex is a significant step in accelerating breast tumorigenesis.


Subject(s)
BRCA1 Protein/metabolism , Breast Neoplasms/metabolism , Carrier Proteins/metabolism , HMGA2 Protein/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Animals , BRCA1 Protein/genetics , Blotting, Western , Breast Neoplasms/genetics , Carrier Proteins/genetics , Cell Line , Cell Line, Tumor , Chromatin Immunoprecipitation , Electrophoresis, Polyacrylamide Gel , Endodeoxyribonucleases , HMGA2 Protein/genetics , Humans , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/metabolism , Mice , Mice, Knockout , Nuclear Proteins/genetics , RNA, Small Interfering , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
8.
Free Radic Biol Med ; 46(11): 1543-50, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19324081

ABSTRACT

Elucidating the molecular mechanism of the low-dose radiation (LDR)-mediated radioadaptive response is crucial for inventing potential therapeutic approaches to improving normal tissue protection in radiation therapy. ATM, a DNA-damage sensor, is known to activate the stress-sensitive transcription factor NF-kappaB upon exposure to ionizing radiation. This study provides evidence of the cooperative functions of ATM, ERK, and NF-kappaB in inducing a survival advantage through a radioadaptive response as a result of LDR treatment (10 cGy X-rays). By using p53-inhibited human skin keratinocytes, we show that phosphorylation of ATM, MEK, and ERK (but not JNK or p38) is enhanced along with a twofold increase in NF-kappaB luciferase activity at 24 h post-LDR. However, NF-kappaB reporter gene transactivation without a significant enhancement of p65 or p50 protein level suggests that NF-kappaB is activated as a rapid protein response via ATM without involving the transcriptional activation of NF-kappaB subunit genes. A direct interaction between ATM and NF-kappaB p65 is detected in the resting cells and this interaction is significantly increased with LDR treatment. Inhibition of ATM with caffeine, KU-55933, or siRNA or inhibition of the MEK/ERK pathway can block the LDR-induced NF-kappaB activation and eliminate the LDR-induced survival advantage. Altogether, these results suggest a p53-independent prosurvival network involving the coactivation of the ATM, MEK/ERK, and NF-kappaB pathways in LDR-treated human skin keratinocytes, which is absent from mutant IkappaB cells (HK18/mIkappaB), which fail to express NF-kappaB activity.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Keratinocytes/radiation effects , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Skin/radiation effects , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Caffeine/pharmacology , Cell Cycle Proteins/genetics , Cell Line, Transformed , Cell Survival/drug effects , DNA-Binding Proteins/genetics , Humans , I-kappa B Proteins/genetics , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/pathology , Morpholines/pharmacology , Mutation , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Pyrones/pharmacology , RNA, Small Interfering/genetics , Radiation, Ionizing , Skin/drug effects , Skin/metabolism , Skin/pathology , Tumor Suppressor Proteins/genetics
9.
Radiat Res ; 171(1): 9-21, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19138055

ABSTRACT

The molecular mechanisms governing acquired tumor resistance during radiotherapy remain to be elucidated. In breast cancer patients, overexpression of HER2 (human epidermal growth factor receptor 2) is correlated with aggressive tumor growth and increased recurrence. In the present study, we demonstrate that HER2 expression can be induced by radiation in breast cancer cells with a low basal level of HER2. Furthermore, HER2-postive tumors occur at a much higher frequency in recurrent invasive breast cancer (59%) compared to the primary tumors (41%). Interestingly, NF-kappaB is required for radiation-induced HER2 transactivation. HER2 was found to be co-activated with basal and radiation-induced NF-kappaB activity in radioresistant but not radiosensitive breast cancer cell lines after long-term radiation exposure, indicating that NF-kappaB-mediated HER2 overexpression is involved in radiation-induced repopulation in heterogeneous tumors. Finally, we found that inhibition of HER2 resensitizes the resistant cell lines to radiation. Since HER2 is shown to activate NF-kappaB, our data suggest a loop-like HER2-NF-kappaB-HER2 pathway in radiation-induced adaptive resistance in breast cancer cells.


Subject(s)
Adaptation, Physiological/genetics , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic/radiation effects , Genes, erbB-2/genetics , Genes, erbB-2/radiation effects , NF-kappa B/metabolism , Radiation Tolerance/genetics , Animals , Benzamides/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/prevention & control , Breast Neoplasms/radiotherapy , Cell Line, Tumor , Gamma Rays/adverse effects , Humans , Mice , NF-kappa B/antagonists & inhibitors , Phenotype , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/radiation effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recurrence , Signal Transduction/genetics , Signal Transduction/radiation effects , Up-Regulation/radiation effects
10.
Free Radic Biol Med ; 44(1): 1-13, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17967430

ABSTRACT

Ionizing radiation (IR) began to be a powerful medical modality soon after Wilhelm Röntgen's discovery of X-rays in 1895. Today, more than 50% of cancer patients receive radiotherapy at some time during the course of their disease. Recent technical developments have significantly increased the precision of dose delivery to the target tumor, making radiotherapy more efficient in cancer treatment. However, tumor cells have been shown to acquire a radioresistance that has been linked to increased recurrence and failure in many patients. The exact mechanisms by which tumor cells develop an adaptive resistance to therapeutic fractional irradiation are unknown, although low-dose IR has been well defined for radioadaptive protection of normal cells. This review will address the radioadaptive response, emphasizing recent studies of molecular-level reactions. A prosurvival signaling network initiated by the transcription factor NF-kappa B, DNA-damage sensor ATM, oncoprotein HER-2, cell cyclin elements (cyclin B1), and mitochondrial functions in radioadaptive resistance is discussed. Further elucidation of the key elements in this prosurvival network may generate novel targets for resensitizing the radioresistant tumor cells.


Subject(s)
NF-kappa B/metabolism , Neoplasms/radiotherapy , Neoplastic Stem Cells/radiation effects , Radiation Tolerance , Signal Transduction/radiation effects , Animals , Cytokines/metabolism , Dose-Response Relationship, Radiation , Genes, erbB-2/radiation effects , Humans , NF-kappa B/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Radiation Tolerance/genetics , Radiation Tolerance/immunology , Radiation, Ionizing , Reactive Oxygen Species
11.
Curr Cancer Drug Targets ; 7(4): 335-42, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17979628

ABSTRACT

Ionizing radiation (IR) plays a key role in both areas of carcinogenesis and anticancer radiotherapy. The ATM (ataxia-telangiectasia mutated) protein, a sensor to IR and other DNA-damaging agents, activates a wide variety of effectors involved in multiple signaling pathways, cell cycle checkpoints, DNA repair and apoptosis. Accumulated evidence also indicates that the transcription factor NF-kappaB (nuclear factor-kappaB) plays a critical role in cellular protection against a variety of genotoxic agents including IR, and inhibition of NF-kappaB leads to radiosensitization in radioresistant cancer cells. NF-kappaB was found to be defective in cells from patients with A-T (ataxia-telangiectasia) who are highly sensitive to DNA damage induced by IR and UV lights. Cells derived from A-T individuals are hypersensitive to killing by IR. Both ATM and NF-kappaB deficiencies result in increased sensitivity to DNA double strand breaks. Therefore, identification of the molecular linkage between the kinase ATM and NF-kappaB signaling in tumor response to therapeutic IR will lead to a better understanding of cellular response to IR, and will promise novel molecular targets for therapy-associated tumor resistance. This review article focuses on recent findings related to the relationship between ATM and NF-kappaB in response to IR. Also, the association of ATM with the NF-kappaB subunit p65 in adaptive radiation response, recently observed in our lab, is also discussed.


Subject(s)
Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , NF-kappa B/physiology , Neoplasms/radiotherapy , Protein Serine-Threonine Kinases/physiology , Radiation Tolerance , Tumor Suppressor Proteins/physiology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Humans , NF-kappa B/metabolism , Neoplasms/metabolism , Neoplasms/physiopathology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism
12.
Int J Oncol ; 30(6): 1447-52, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17487365

ABSTRACT

MDM2 (murine double minute 2) is well-documented to play a key role in radiation response and tumor radiosensitivity, thus offering an attractive clinic drug target to enhance tumor sensitivity to anti-cancer radiotherapy. In this study, we designed and tested two siRNA fragments against human MDM2 in non-small cell human lung cancer A549 cells. Transfection of mammalian expression vector pUR/U6 containing either MDM2 siRNA1 or siRNA2 fragment was shown to reduce MDM2 mRNA levels by 72% and 31%, respectively. Western blotting detected a similar inhibition of MDM2 protein levels in cells transfected with MDM2 siRNA1. A549 cells transfected with the expression vector for siRNA1 significantly decreased cell proliferation and rendered cells more sensitive to radiation. The basal apoptotic and necrotic cells, 1% and 2%, respectively, detected among A549 cells were increased to 2.6% and 14.4% after gamma-irradiation with 5 Gy. Transfection of MDM2 siRNA1 induced 30.1% apoptosis and 12.7% necrosis while combined treatment of siRNA1 and 5-Gy radiation increased apoptosis and necrosis to 45.9% and 15.2%, respectively. These data provide the first evidence that specific siRNA fragment (MDM2 siRNA1) targeting human MDM2 mRNA is able to enhance lung cancer radiosensitivity.


Subject(s)
Lung Neoplasms , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , RNA, Small Interfering , Radiation Tolerance/physiology , Radiation-Sensitizing Agents , Blotting, Western , Cell Death/radiation effects , Cell Line, Tumor , Flow Cytometry , Humans , Reverse Transcriptase Polymerase Chain Reaction , Transfection
13.
Int J Mol Med ; 19(6): 865-73, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17487418

ABSTRACT

Pulmonary fibrosis (PF) is a major side effect of radiotherapy and chemotherapy. Recent clinical trials, unfortunately, have failed to identify any therapeutic agent which has the potential to reduce the consequences of this devastating condition. Reactive oxygen species and tissue remodeling regulators, such as metalloproteinases (MMPs) and their inhibitors (TIMPs), are thought to be involved in the development of PF. We investigated these factors to determine the protective effects of antioxidant alpha-lipoic acid (LA) against antineoplastic agent bleomycin (BLM)-induced oxidant lung toxicity in Sprague-Dawley rats. At different time intervals after BLM administration, pathological changes of the lung were analyzed with the measurement of total protein in bronchoalveolar lavage fluid (BALF), hydroxyproline (HYP) content and the level of three oxidative stress markers, i.e. malondialdehyde (MDA), the GSH/GSSG ratio, and total antioxidative capability (T-AOC). Also, the expression changes of MMP-1 and TIMP-1 were measured. At day 14 or 28 after BLM administration, protein content in BALF, and HYP, MDA and T-AOC contents of the lung increased significantly with a decreased GSH/GSSG ratio, implicating an increased efflux of GSSG from the lung and consumption of GSH. In contrast, treatment with LA protected BLM-induced pulmonary injury by suppressing oxidative stress with the reduction of MDA, and the enhancement of the GSH/GSSG ratio and T-AOC. The BLM-stimulated symptoms of PF were relieved with significant reduction of HYP and total proteins in LA-treated rats. LA also ameliorated the MMP-1/TIMP-1 ratio. These results suggest that LA inhibits BLM-induced lung toxicity associated with oxidative damage. Therefore, antioxidant LA has a potential therapeutic effect in the prevention and alleviation of PF.


Subject(s)
Bleomycin , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Thioctic Acid/therapeutic use , Animals , Antioxidants/therapeutic use , Bronchoalveolar Lavage Fluid/chemistry , Gene Expression Profiling , Glutathione/analysis , Glutathione Disulfide/analysis , Hydroxyproline/analysis , Lung/chemistry , Lung/metabolism , Male , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Oxidative Stress , Pulmonary Fibrosis/pathology , Rats , Rats, Sprague-Dawley , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism
15.
Cancer Lett ; 180(2): 159-63, 2002 Jun 28.
Article in English | MEDLINE | ID: mdl-12175547

ABSTRACT

Midkine (MK) is a heparin-binding growth factor specified by a retinoic acid responsive gene. It plays important roles in development and carcinogenesis. The MK gene is located on chromosome 11q11.2 in humans. A heterozygous G to T transition at the 62nd base in intron 3 of this gene has been identified in sporadic colorectal and gastric cancers (Int. J. Mol. Med. 6 (2000) 281). To clarify whether this polymorphism is associated with a cancer risk, a case-control study was conducted. We examined 98 colorectal, 60 gastric, 59 esophagus, 32 lung and 37 breast cancer tissue specimens and their corresponding non-neoplastic tissues. Also, 86 unaffected control specimens were examined. The G/T genotype frequency in colorectal cancers was higher than that in normal samples (11.2 versus 2.3%; P=0.017). Therefore, this genotype could represent a risk factor for tumorigenesis in the colon and rectum of Japanese.


Subject(s)
Carrier Proteins/genetics , Colorectal Neoplasms/genetics , Cytokines , Introns , Colorectal Neoplasms/etiology , Female , Humans , Male , Midkine , Polymorphism, Genetic , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...