Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 292(42): 17461-17472, 2017 10 20.
Article in English | MEDLINE | ID: mdl-28900036

ABSTRACT

Polo-like kinase 1 (Plk1), a serine/threonine protein kinase normally expressed in mitosis, is frequently up-regulated in multiple types of human tumors regardless of the cell cycle stage. However, the causal relationship between Plk1 up-regulation and tumorigenesis is incompletely investigated. To this end, using a conditional expression system, here we generated Plk1 transgenic mouse lines to examine the role of Plk1 in tumorigenesis. Plk1 overexpression in mouse embryonic fibroblasts prepared from the transgenic mice led to aberrant mitosis followed by aneuploidy and apoptosis. Surprisingly, Plk1 overexpression had no apparent phenotypes in the mice. Given that no malignant tumor formation was observed even after a long period of Plk1 overexpression, we reasoned that additional factors are required for tumorigenesis in Plk1-overexpressing mice. Because Plk1 can directly participate in the regulation of the DNA damage response (DDR) pathway, we challenged Plk1-overexpressing mice with ionizing radiation (IR) and found that Plk1-overexpressing mice are much more sensitive to IR than their wild-type littermates. Analysis of tumor development in the Plk1-overexpressing mice indicated a marked decrease in the time required for tumor emergence after IR. At the molecular level, Plk1 overexpression led to reduced phosphorylation of the serine/threonine kinases ATM and Chk2 and of histone H2AX after IR treatment both in vivo and in vitro Furthermore, RNA-Seq analysis suggested that Plk1 elevation decreases the expression of several DDR genes. We conclude that Plk1 overexpression may contribute to tumor formation by both inducing chromosomal instability and suppressing the DDR pathway.


Subject(s)
Cell Cycle Proteins/biosynthesis , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Enzymologic/radiation effects , Gene Expression Regulation, Neoplastic/radiation effects , Neoplasms, Radiation-Induced/enzymology , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Radiation, Ionizing , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Transformation, Neoplastic/genetics , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , DNA Damage , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Neoplastic/genetics , Mice , Mice, Transgenic , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/pathology , Phosphorylation/genetics , Phosphorylation/radiation effects , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Polo-Like Kinase 1
2.
Toxicol Appl Pharmacol ; 352: 87-96, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29802912

ABSTRACT

Previously, we proved that caffeic acid (CA), a major dietary phenolic acid, prevents skin carcinogenesis by modulating inflammatory signaling in mouse skin. However, the actual mechanisms of CA against UVB (280-320 nm) induced photocarcinogenesis remains unclear. The present results confirms that CA significantly inhibits single UVB-induced CPDs formation, oxidative DNA damage, ROS generation and frequency of apoptotic cell death in human dermal fibroblasts (HDFa). Furthermore, CA prevents UVB-induced expression of PI3K and AKT kinases through activation of PTEN which subsequently promotes XPC dependant NER proteins such as XPC, XPE, TFIIH (p44) and ERCC1 in HDFa cells and mouse skin tissue. Further, CA directly activates PTEN through hydrogen bond and hydrophobic interactions. Taken together, these findings suggest that CA prevents UVB-induced photodamage through the activation of PTEN expression in human dermal fibroblasts and mouse skin.


Subject(s)
Anticarcinogenic Agents/pharmacology , Caffeic Acids/pharmacology , Cell Transformation, Neoplastic/drug effects , Fibroblasts/drug effects , Neoplasms, Experimental/prevention & control , Neoplasms, Radiation-Induced/prevention & control , PTEN Phosphohydrolase/metabolism , Signal Transduction/drug effects , Skin Neoplasms/prevention & control , Skin/drug effects , Ultraviolet Rays/adverse effects , Animals , Apoptosis/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cell Transformation, Neoplastic/radiation effects , DNA Damage/drug effects , DNA Repair/drug effects , DNA Repair Enzymes/metabolism , Fibroblasts/enzymology , Fibroblasts/pathology , Fibroblasts/radiation effects , Humans , Mice , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/pathology , Neoplasms, Radiation-Induced/enzymology , Neoplasms, Radiation-Induced/pathology , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Signal Transduction/radiation effects , Skin/enzymology , Skin/pathology , Skin/radiation effects , Skin Neoplasms/enzymology , Skin Neoplasms/pathology
3.
Toxicol Appl Pharmacol ; 284(1): 92-99, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25680589

ABSTRACT

Extensive exposure of solar ultraviolet-B (UVB) radiation to skin induces oxidative stress and inflammation that play a crucial role in the induction of skin cancer. Photochemoprevention with natural products represents a simple but very effective strategy for the management of cutaneous neoplasia. In this study, we investigated whether blackberry extract (BBE) reduces chronic inflammatory responses induced by UVB irradiation in SKH-1 hairless mice skin. Mice were exposed to UVB radiation (100 mJ/cm(2)) on alternate days for 10 weeks, and BBE (10% and 20%) was applied topically a day before UVB exposure. Our results show that BBE suppressed UVB-induced hyperplasia and reduced infiltration of inflammatory cells in the SKH-1 hairless mice skin. BBE treatment reduced glutathione (GSH) depletion, lipid peroxidation (LPO), and myeloperoxidase (MPO) in mouse skin by chronic UVB exposure. BBE significantly decreased the level of pro-inflammatory cytokines IL-6 and TNF-α in UVB-exposed skin. Likewise, UVB-induced inflammatory responses were diminished by BBE as observed by a remarkable reduction in the levels of phosphorylated MAP Kinases, Erk1/2, p38, JNK1/2 and MKK4. Furthermore, BBE also reduced inflammatory mediators such as cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), and inducible nitric oxide synthase (iNOS) levels in UVB-exposed skin. Treatment with BBE inhibited UVB-induced nuclear translocation of NF-κB and degradation of IκBα in mouse skin. Immunohistochemistry analysis revealed that topical application of BBE inhibited the expression of 8-oxo-7, 8-dihydro-2'-deoxyguanosine (8-oxodG), cyclobutane pyrimidine dimers (CPD), proliferating cell nuclear antigen (PCNA), and cyclin D1 in UVB-exposed skin. Collectively, these data indicate that BBE protects from UVB-induced oxidative damage and inflammation by modulating MAP kinase and NF-κB signaling pathways.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Rubus , Signal Transduction/drug effects , Skin/drug effects , Sunburn/prevention & control , Sunscreening Agents/pharmacology , Ultraviolet Rays , Active Transport, Cell Nucleus , Animals , Anti-Inflammatory Agents/isolation & purification , Antioxidants/isolation & purification , Biomarkers/metabolism , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , DNA Damage , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Fruit , Inflammation Mediators/metabolism , Lipid Peroxidation/drug effects , Mice, Hairless , Neoplasms, Radiation-Induced/enzymology , Neoplasms, Radiation-Induced/immunology , Neoplasms, Radiation-Induced/prevention & control , Phosphorylation , Phytotherapy , Plant Extracts/isolation & purification , Plants, Medicinal , Rubus/chemistry , Skin/enzymology , Skin/immunology , Skin/pathology , Skin Neoplasms/enzymology , Skin Neoplasms/immunology , Skin Neoplasms/prevention & control , Sunburn/enzymology , Sunburn/immunology , Sunburn/pathology , Sunscreening Agents/isolation & purification , Time Factors
4.
Acta Derm Venereol ; 95(1): 40-4, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24535144

ABSTRACT

The detoxifying enzyme glutathione-s-transferase pi (GST-π) is present in keratinocytes and melanocytes and exerts a protective role against tumour progression. Melanomas close to melanocytic naevus remnants occur less frequently on sun-exposed areas, whereas solar dermal elastosis, hallmark of chronic sun-damage, characterise melanomas on sun-exposed skin. We evaluated the expression of GST-π in 113 melanomas associated to melanocytic naevus remnants or to solar dermal elastosis, classified according to clinical characteristics, history of sun exposure, histological subtypes and AJCC staging. Chronically sun-damaged melanomas, identified by moderate-severe solar dermal elastosis, showed a lower nuclear GST-π expression and a higher thickness than those related to melanocytic naevus remnants (p < 0.03). Multivariate logistic regression analysis demonstrated that male gender and chronic sun-exposure are independent risk factors significantly associated to melanomas localised on the trunk (OR = 3.36, 95% CI: 1.31-8.65; OR = 5.97, 95% CI: 1.71-20.87). If confirmed on a larger series, lower expression of nuclear GST-π in melanoma cells could represent a possible marker of chronically sun-damaged melanoma pathogenesis.


Subject(s)
Biomarkers, Tumor/analysis , Glutathione S-Transferase pi/analysis , Melanoma/enzymology , Melanoma/epidemiology , Neoplasms, Radiation-Induced/enzymology , Neoplasms, Radiation-Induced/epidemiology , Nevus, Pigmented/enzymology , Nevus, Pigmented/epidemiology , Sunlight/adverse effects , Adult , Aged , Biopsy , Chi-Square Distribution , Down-Regulation , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Italy/epidemiology , Logistic Models , Male , Melanoma/pathology , Middle Aged , Multivariate Analysis , Neoplasm Staging , Neoplasms, Radiation-Induced/pathology , Nevus, Pigmented/pathology , Odds Ratio , Risk Factors , Sex Factors , Time Factors
5.
Radiats Biol Radioecol ; 54(4): 350-9, 2014.
Article in Russian | MEDLINE | ID: mdl-25775823

ABSTRACT

An association between polymorphous (allelic) gene variants of phase II of enzymatic xenobiotic biotransformation (EXB) of multigene families of glutathione-S-transferase (GSTs) GSTM1*0, GSTT1*0, GSTP1*B Ile105Val, and N-acetyltransferase (NAT) NAT2*6 590G>A, NAT2*5 481C>T, as well as lung cancer in Mayak workers exposed occupationally to prolonged external γ-rays and internal α-radiation from incorporated 239Pu was studied. Analysis of the population frequency of genotypes and alleles of the studied genes in the cohort of Mayak workers revealed their compliance with the Hardy-Weinberg principle and with the corresponding frequency in the European population. The study was based on the case-control method. A case-group consisted of 49 Mayal workers with a verified diagnosis of lung cancer. The mean total absorbed dose from external γ-rays at the moment of diagnostics was 1.03 Gy; the mean total absorbed dose from internal α-radiation from incorporated 239Pu to lung was 0.35 Gy. Control consisted of 172 Mayak workers matched by the year of birth, gender, and age at the moment of employment at one of the main facilities with no lung cancer registered within the study period. No increase in the relative risk of lung cancer (odds ratio, OR) was revealed among the individuals with deletion variants of genes GSTM1*0 and GSTT1*0 (pp genotype, complete absence of gene products) as compared to the individuals with ww or wp genotype, which was determined in total for these genes (normal or partly decreased gene activity). An increase in OR of lung cancer in 1.849 times (p = 0.239) and in 2.439 times (p = 0.075) was found in the carriers with a complete absence of the product of genes GSTP1*B and NAT2*6 590G>A, correspondingly (pp genotype). A statistically significant decrease in OR of lung cancer was found in the wp genotype carriers of gene GSTP1*B (OR = 0.50, p = 0.041). Three variants of paired combinations of gene alleles were established in the carriers with a statistically significant increase in OR of lung cancer (ww GSTP1*B + pp GSTM1*0; ww GSTP1*B + pp NAT2*6 590G>A; pp GSTP1*B + pp NAT2*5 481C>T), and one combination in the carriers with a statistically significant decrease in OR of lung cancer (wp GSTP1*B and ww +wp GSTT1*0).


Subject(s)
Arylamine N-Acetyltransferase/genetics , Glutathione S-Transferase pi/genetics , Glutathione Transferase/genetics , Lung Neoplasms/genetics , Neoplasms, Radiation-Induced/genetics , Occupational Diseases/genetics , Polymorphism, Single Nucleotide , Radiation, Ionizing , Xenobiotics/pharmacokinetics , Aged , Alpha Particles/adverse effects , Case-Control Studies , Female , Gamma Rays/adverse effects , Genetic Predisposition to Disease , Genotype , Humans , Industry , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Male , Metabolic Detoxication, Phase II , Neoplasms, Radiation-Induced/enzymology , Neoplasms, Radiation-Induced/metabolism , Occupational Diseases/enzymology , Occupational Diseases/metabolism , Occupational Exposure/adverse effects , Polymorphism, Restriction Fragment Length , Russia
6.
J Nanosci Nanotechnol ; 13(1): 52-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23646697

ABSTRACT

We investigated the anti-melanogenetic efficacy of hydrogen-occluding silica microcluster (H2-Silica), which is a silsesquioxane-based compound with hydrogen interstitially embedded in a matrix of caged silica, against melanogenesis in HMV-II human melanoma cells and L-DOPA-tyrosinase reaction [EC1.14.18.1]. HMV-II cells were subjected to oxidative stress by ultraviolet ray-A (UVA) exposure of 3-times of 0.65 J/cm2 summed up to 1.95 J/cm2. After UVA irradiation, HMV-II cells were stimulated to produce melanin by 2.72-fold more abundantly than unirradiated control. When HMV-II cells were treated with H2-Silica of 20 ppm or kojic acid of 28.4 ppm before and after UVA-irradiation, the amount of melanin was repressed to 12.2% or 14.5% as compared to that of UVA-irradiated control, respectively. That is, H2-Silica exhibited a comparable efficacy to the whitening agent kojic acid. The H2-Silica could prevent melanogenesis in HMV-II cells by low-level doses at 1-10 ppm, and cell viability and apoptosis event did not change even by high-level doses at 100-1000 ppm. On the contrary, kojic acid was cytotoxic at the concentration of 14-28 ppm or more. By microscopic observation, H2-Silica suppressed such properties indicative of melanin-rich cells as cellular hypertrophy, cell process formation, and melanogenesis around the outside of nuclei. The enzymatic assay using L-DOPA and mushroom tyrosinase demonstrated that H2-Silica restrained UVA-mediated melanin formation owing to down-regulation of tyrosinase activity, which could be attributed to scavenging of free radicals and inhibition of L-DOPA-to-dopachrome oxidation by hydrogen released from H2-Silica. Thus H2-Silica has a potential to prevent melanin production against UVA and serves as a skin-lightening ingredient for supplements or cosmetics.


Subject(s)
Hydrogen/administration & dosage , Melanoma/etiology , Melanoma/prevention & control , Monophenol Monooxygenase/metabolism , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/prevention & control , Silicon Dioxide/administration & dosage , Capsules/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Humans , Melanoma/enzymology , Neoplasms, Radiation-Induced/enzymology , Treatment Outcome , Ultraviolet Rays/adverse effects
7.
Am J Dermatopathol ; 35(4): 412-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23051629

ABSTRACT

BACKGROUND: BRAF mutations have been implicated in initiating promutagenic cellular melanocytic proliferation mostly based on homogeneous Western-based cohorts. Data addressing the possible interaction between exposure to different solar ultraviolet radiation (UVR) magnitudes and BRAF mutation rate (BMR) in melanocytic nevi are limited. DESIGN: Extended BRAF testing for 9 mutations in 225 melanocytic nevus (MN) cases derived from 211 patients from 4 different UVR regions: Lebanon (n = 95; 110 kJ · m(-2) · yr), Syria (n = 23; 93.5 kJ · m(-2) · yr), Kingdom of Saudi Arabia (n = 70; 139 kJ · m(-2) · yr), and Pakistan (n = 37; 118 kJ · m(-2) · yr) was performed. Data collected included age, gender, anatomic location, and lesion size. Histological parameters recorded were MN type (junctional, compound, intradermal, classical blue, cellular blue, compound and intradermal spitz, and congenital) solar elastosis grade, and nevus pigmentation degree. Cumulative 21-year erythemally effective UV averages were derived from The National Center for Atmospheric Research. RESULTS: BRAF mutation status was obtained in 210 cases (6.7% failed polymerase chain reaction). Overall, BMR was 62.4% (131/210) with V600E mutation accounting for 98.5% of cases. Discordant mutation status was found in 2 of 10 patients with multiple nevi. BMR differed significantly, yet nonsystematically, among UVR regions; the highest was detected in nevi coming from Syria (18/23 cases, 78%), followed by Pakistan (21/30 cases, 70%), Kingdom of Saudi Arabia (47/70 cases, 67%), and Lebanon (45/87 cases, 52%). Mutation rates varied significantly across MN type (P < 0.001); the highest rate was recorded in the intradermal nevus type (33/39 cases, 84.6%), followed by the compound (26/32 cases, 81.2%) and congenital (60/74 cases 81.0%) nevi. Stratified by anatomic location, nevi occurring on the face (61/82, 74%) and trunk (58/78, 74%) had more frequent BMRs compared with those occurring on the upper (7/26, 27%) and lower extremities (5/24, 21%, P < 0.001). Severe pigmentation was less frequent in BRAF mutation-positive nevi [5/131 (4%) vs. 34/79 (43%); P < 0.001]. Multivariate independent predictors of BRAF mutation in MN were age [odds ratio (95% confidence interval ) = 1.43 (1.13-1.74) per 10 years; P = 0.004], anatomic location [P = 0.043 overall], and nevus type [P < 0.001 overall]. UVR region was not an independent predictor of BRAF mutation. CONCLUSIONS: Increased BRAF mutation with age along with the lack of a UVR magnitude-BRAF mutation association suggests that duration of exposure rather than UVR exposure dose is the more likely link to acquiring the mutation.


Subject(s)
Mutation , Neoplasms, Radiation-Induced/genetics , Nevus, Pigmented/genetics , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/genetics , Sunlight/adverse effects , Ultraviolet Rays/adverse effects , Adolescent , Adult , Age Factors , Aged , Biopsy , Chi-Square Distribution , Child , Child, Preschool , DNA Mutational Analysis/methods , Female , Humans , Logistic Models , Male , Middle Aged , Middle East , Multivariate Analysis , Neoplasms, Radiation-Induced/enzymology , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/pathology , Nevus, Pigmented/enzymology , Nevus, Pigmented/etiology , Nevus, Pigmented/pathology , Odds Ratio , Pakistan , Polymerase Chain Reaction , Residence Characteristics , Risk Factors , Skin Neoplasms/enzymology , Skin Neoplasms/etiology , Skin Neoplasms/pathology , Time Factors , Young Adult
8.
J Neurooncol ; 109(3): 477-84, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22821382

ABSTRACT

Somatic mutations of the isocitrate dehydrogenase-1 gene (IDH1), most commonly resulting in replacement of arginine at position 132 by histidine (p.R132H), have been reported for WHO grade II and III diffuse gliomas and secondary glioblastomas. We investigated IDH1/2 mutations in a retrospective series of 165 pediatric brain tumors, including atypical teratoid/rhabdoid tumors (AT/RT) and choroid plexus tumors, which had not previously been investigated. Mutation analysis was performed by use of pyrosequencing and, additionally, data were validated for a cohort of 70 gliomas from among the series by use of the arrayed primer extension technique. We identified one tumor which harbored mutation of IDH1 at codon 132 and no alteration was identified in the matched-germline DNA. No IDH2 mutations were detected. Most noteworthy, the IDH1 mutant tumor was an anaplastic astrocytoma involving the cortex in the left frontal lobe which appeared seven years after radiation treatment for an extensive sellar/suprasellar craniopharyngioma. This anaplastic astrocytoma was regarded as secondary to radiation treatment because it seemed to originate within the irradiation field that received a dose varying from a maximum of 30.6 Gy of 4 MV X-rays down to very few Gy of lower-energy scattered radiation. In this work our observations agree with those in previous reports showing the rarity of IDH1/2 mutations in childhood tumors. The interesting identification of an IDH1 mutation in a radiation-induced secondary malignant glioma raises the likelihood that these types of tumor may develop IDH1/2 mutations. Thus, caution is needed when dealing with these tumors, and further genetic analysis is warranted.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Isocitrate Dehydrogenase/genetics , Mutation , Neoplasms, Radiation-Induced/genetics , Adolescent , Astrocytoma/enzymology , Astrocytoma/secondary , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Child , Child, Preschool , DNA Mutational Analysis , Humans , Infant , Male , Neoplasms, Radiation-Induced/enzymology , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction
9.
J Neurooncol ; 109(2): 399-404, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22744755

ABSTRACT

Meningiomas are the most frequent intracranial tumors. Although most are benign WHO grade I tumors, grade II and III tumors are aggressive and survival is poor. Treatment options for grade II and III meningiomas are limited, and molecular targets are few. The re-programming of metabolic pathways including glycolysis, lipogenesis, and nucleotide synthesis is a hallmark of the physiological changes in cancer cells. Because fatty acid synthase (FAS), the enzyme responsible for the de-novo synthesis of fatty acids, has emerged as a potential therapeutic target for several cancers, we investigated its involvement in meningiomas. We subjected 92 paraffin-embedded samples from 57 patients with grade I, 18 with grade II and III, and six with radiation-induced tumors to immunohistochemical study of FAS. Whereas its expression was increased in grade II and III meningiomas (62.9 %) compared with grade I tumors (29.8 %) (chi-squared test: p < 0.001), FAS was expressed in grade I tumors with a high MIB-1 index and infiltration into surrounded tissues. All radiation-induced meningiomas expressed FAS and its expression was positively correlated with the MIB-1 index (p < 0.005). Our findings suggest that increased FAS expression reflects the aggressiveness of meningiomas and that it may be a novel therapeutic target for treatment of unresectable or malignant tumors.


Subject(s)
Biomarkers/metabolism , Fatty Acid Synthases/metabolism , Meningeal Neoplasms/enzymology , Meningioma/enzymology , Adult , Age Factors , Aged , Aged, 80 and over , Female , Humans , Male , Meningeal Neoplasms/mortality , Meningioma/mortality , Middle Aged , Neoplasms, Radiation-Induced/enzymology , Neoplasms, Radiation-Induced/pathology , Prognosis , Sex Factors , Statistics, Nonparametric , Survival Analysis , Time Factors , Young Adult
10.
Cancer Lett ; 512: 51-59, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33965452

ABSTRACT

As a treatment option for PDAC, radiation therapy induces good local control. However, radiation also reportedly enhances the malignant potential (e.g., invasion and migration ability) in various cancers, thus increasing the risk of distant metastasis. It remains unclear how radiation induces malignant potential, and how such enhanced malignant potential can be suppressed. In the current study, we evaluated the sequential change of c-Met expression in pancreatic cancer cells following irradiation. We found that irradiation transiently induced c-Met expression in vitro. In an in vivo subcutaneous tumor mouse model, irradiation also enhanced downstream phosphorylated Met (p-Met). Furthermore, this enhancement of p-Met protein expression was suppressed by oral administration of the c-Met inhibitor INC280. Irradiated pancreatic cancer cells with enhanced c-Met expression exhibited higher malignant potential, including invasion and migration ability, compared with cells showing low c-Met expression. Pancreatic cancer cells that overexpressed c-met also showed enhanced malignant potential, which was reversed by c-Met inhibition. Additionally, c-Met inhibitor suppressed the metastatic potential in a liver metastasis mouse model using c-met-overexpressing cells. Overall, our present results revealed that irradiation could induce c-met expression in pancreatic cancer cells, leading to enhanced malignant potential (e.g., invasion and migration ability) and thus promoting distant metastasis. Moreover, a c-Met inhibitor could reverse this enhanced malignant potential.


Subject(s)
Neoplasms, Radiation-Induced/drug therapy , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Animals , Benzamides/pharmacology , Cell Movement/radiation effects , Humans , Imidazoles/pharmacology , Mice , Mice, Inbred NOD , Neoplasms, Radiation-Induced/enzymology , Neoplasms, Radiation-Induced/pathology , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Triazines/pharmacology , Pancreatic Neoplasms
11.
Int J Cancer ; 126(1): 11-8, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19609953

ABSTRACT

Solid organ transplant recipients have a greatly increased risk for the development of non-melanoma skin cancers. We have previously shown in our mouse model that sirolimus given in combination with cyclosporine A resulted in fewer and smaller tumors than cyclosporine A alone. In the current study, we tested the hypothesis that an anti-inflammatory agent celecoxib applied topically after UVB exposure would further reduce UVB induced skin cancer in mice treated with cyclosporine A and sirolimus. The effect of celecoxib treatment on acute inflammation, initiation/promotion and tumor development was examined through a set of four experiments. Delayed tumor onset was observed in both tumor development experiments. Reduced tumor size and number compared to vehicle was observed when CX was administered concurrently with UVB and when CX was administered after cessation of UVB treatments, respectively. Prostaglandin E2 was confirmed to be significantly reduced in the dorsal skin of mice concurrently treated with immunosuppressants, CX and UVB for 13 weeks, suggesting a reduction in the inflammatory response could be the mechanism by which CX reduced tumorigenesis. Furthermore, topical celecoxib treatment following acute UVB exposure reduced dermal neutrophil number and activity compared to vehicle. In all of these experiments, unirradiated and vehicle treated mice were utilized as controls. In conclusion, these data suggest that even in the presence of cyclosporine A and sirolimus, topical celecoxib treatment can result in reduced inflammation, tumor number and size; properties which may be beneficial in the therapeutic reduction of skin cancer development in solid organ transplant recipients.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Immunosuppressive Agents/therapeutic use , Neoplasms, Radiation-Induced/prevention & control , Pyrazoles/pharmacology , Skin Neoplasms/prevention & control , Sulfonamides/pharmacology , Ultraviolet Rays , Animals , Blotting, Western , Caspase 3/metabolism , Celecoxib , Cyclosporine/administration & dosage , Dinoprostone/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Immunohistochemistry , Mice , Neoplasms, Radiation-Induced/enzymology , Neoplasms, Radiation-Induced/metabolism , Sirolimus/administration & dosage , Skin Neoplasms/enzymology , Skin Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism
12.
Clin Cancer Res ; 15(5): 1534-42, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19223498

ABSTRACT

PURPOSE: NRH:quinone oxidoreductase 2 (NQO2) is known to protect against myelogenous hyperplasia. However, the role of NQO2 in prevention of hematologic malignancies remains unknown. Present studies investigated in vivo role of NQO2 in prevention of myeloproliferative disease and lymphomas. EXPERIMENTAL DESIGN: Wild-type and NQO2-null mice were exposed to 0, 1, and 3 Gy gamma-radiation. One year later, the mice were analyzed for the development of myeloproliferative disease and lymphomas. Immunohistochemistry analysis determined the B- and T-cell origin of lymphomas. The mice were also sacrificed at 6 and 48 h after radiation exposure and bone marrow was collected and analyzed for p53, Bax, and B-cell apoptosis. Bone marrow cells were cultured and the rate of degradation of p53 was analyzed. RESULTS: Seventy-two percent NQO2-null mice showed development of B-cell lymphomas in multiple tissues compared with 11% in wild-type mice exposed to 3 Gy gamma-radiation. In contrast, only 22% NQO2-null mice showed myeloproliferation compared with none in wild-type mice. Further analysis revealed that bone marrow from NQO2-null mice contained lower levels of p53 compared with wild-type mice due to rapid degradation of p53. In addition, the exposure to radiation resulted in lower induction of p53 and Bax and decreased B-cell apoptosis in NQO2-null mice. CONCLUSION: NQO2-null mice are highly susceptible to develop radiation-induced B-cell lymphomas. The lack of significant induction of p53 and Bax and decrease in B-cell apoptosis presumably contributed to the development of lymphomas. NQO2 functions as endogenous factor in prevention against radiation-induced B-cell lymphomas.


Subject(s)
B-Lymphocytes/radiation effects , Gamma Rays , Lymphoma, B-Cell/etiology , Neoplasms, Radiation-Induced/etiology , Quinone Reductases/physiology , Animals , Apoptosis/radiation effects , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Blotting, Western , Bone Marrow/metabolism , Bone Marrow/pathology , Cells, Cultured , Female , Flow Cytometry , Lymphoma, B-Cell/enzymology , Lymphoma, B-Cell/pathology , Male , Mice , Mice, Knockout , Neoplasms, Radiation-Induced/enzymology , Neoplasms, Radiation-Induced/pathology , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
13.
Cancer Res ; 67(10): 4751-8, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17510403

ABSTRACT

Cutaneous squamous cell carcinomas (CSCC) are a common malignancy of keratinocytes that arise in sites of the skin exposed to excessive UV radiation. In the present study, we show that human SCC cell lines, preneoplastic solar keratoses (SK), and CSCC are associated with perturbations in glutathione peroxidase (GPX) activity and peroxide levels. Specifically, we found that two of three SKs and four of five CSCCs, in vivo, were associated with decreased GPX activity and all SKs and CSCCs were associated with an elevated peroxide burden. Given the association of decreased GPX activity with CSCC, we examined the basis for the GPX deficiency in the CSCCs. Our data indicated that GPX was inactivated by a post-translational mechanism and that GPX could be inactivated by increases in intracellular peroxide levels. We next tested whether the decreased peroxidase activity coupled with an elevated peroxidative burden might contribute to CSCC formation in vivo. This was tested in Gpx1(-/-) and Gpx2(-/-) mice exposed to solar-simulated UV radiation. These studies showed that Gpx2 deficiency predisposed mice to UV-induced CSCC formation. These results suggest that inactivation of GPX2 in human skin may be an early event in UV-induced SCC formation.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/etiology , Glutathione Peroxidase/metabolism , Neoplasms, Radiation-Induced/enzymology , Skin Neoplasms/enzymology , Skin Neoplasms/etiology , Enzyme Activation , Glutathione Peroxidase/biosynthesis , Glutathione Peroxidase/deficiency , Glutathione Peroxidase/genetics , Humans , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/metabolism , Keratinocytes/enzymology , Keratinocytes/pathology , Keratinocytes/radiation effects , Neoplasms, Radiation-Induced/etiology , Peroxides/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Ultraviolet Rays
14.
Sci Rep ; 9(1): 5301, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30923327

ABSTRACT

Stochastic long-term damages at relatively low doses have the potential for cancer induction. For the first time we investigated the occurrence of breast cancer in female patients after radiotherapy of non-malignant disorders of the shoulder and made a comparison with the estimated spontaneous incidence of mammary carcinoma for this cohort. In a geographically defined district with a population of approximately 100.000 inhabitants, comprehensive data of radiological diagnostics and radiotherapy were registered nearly completely for 41 years; data included mammography and radiotherapy of breast cancer patients as well as of non-malignant disorders. Within this population a collective of 158 women with radiotherapy of the shoulder was investigated. Radiotherapy was performed with cobalt-60 photons (Gammatron) with an average cumulative-dose of 6 Gy. The average follow-up time was 21.3 years. Patients were 55 years old (median) when radiotherapy of the shoulder was performed. Seven patients (4.4%) developed breast cancer after a median of 21 years. According to the incidence statistics, 9.4 +/- 1.8 (95%CI) cases (5.9%) would be expected. In regard to the irradiated shoulder neither the ipsilateral nor the contralateral breasts showed increased rates of breast cancer. An induction of additional breast cancer caused by radiation of non-malignant disorders of the shoulder wasn't detected in the investigated cohort.


Subject(s)
Breast Neoplasms/epidemiology , Breast/radiation effects , Neoplasms, Radiation-Induced/enzymology , Organs at Risk/radiation effects , Shoulder Pain/radiotherapy , Adult , Aged , Aged, 80 and over , Breast/pathology , Breast Neoplasms/etiology , Dose-Response Relationship, Radiation , Female , Follow-Up Studies , Humans , Incidence , Middle Aged , Neoplasms, Radiation-Induced/etiology , Organs at Risk/pathology , Phantoms, Imaging , Radiotherapy Dosage , Retrospective Studies
15.
Photochem Photobiol ; 84(2): 322-9, 2008.
Article in English | MEDLINE | ID: mdl-18194346

ABSTRACT

Besides induction of DNA damage and p53 mutations, chronic exposure to UV irradiation leads to the constitutive up-regulation of cyclo-oxygenase-2 (COX-2) expression and to increased production of its primary product in skin, prostaglandin E2 (PGE2). COX-2 has also been shown to be constitutively overexpressed in mouse, as well as human, UV-induced skin cancers and premalignant lesions. UV exposure results in ligand-independent activation of the epidermal growth factor receptor and subsequent activation of mitogen-activated protein kinase and phosphatidylinositol 3-kinase/Akt pathways leading to transcriptional activation of the COX-2 gene. Use of COX-2-specific inhibitors and genetic manipulation of COX-2 expression have demonstrated that UV induction of COX-2 in the skin contributes to the induction of epidermal hyperplasia, edema, inflammation, and counters the induction of apoptosis after UV exposure. Likewise, inhibition of COX-2 activity or reduced expression in COX-2 knockout mice resulted in significantly reduced UV-induced tumorigenesis, while overexpression of COX-2 in transgenic mice enhanced UV-induced tumor development. A combination of signaling from the PGE2 EP1, EP2 and/or EP4 receptors mediates the effects of COX-2 overexpression. These studies demonstrate the crucial role of COX-2 in the development of UV-related nonmelanoma skin cancers.


Subject(s)
Cyclooxygenase 2/metabolism , Neoplasms, Radiation-Induced/enzymology , Skin Neoplasms/enzymology , Ultraviolet Rays , Animals , Cyclooxygenase 2/biosynthesis , Enzyme Induction , Humans
16.
Oncogene ; 25(47): 6229-38, 2006 Oct 12.
Article in English | MEDLINE | ID: mdl-16682947

ABSTRACT

Ercc1 has an essential role in the nucleotide excision repair (NER) pathway that protects against ultraviolet (UV)-induced DNA damage and is also involved in additional repair pathways. The premature death of simple Ercc1 mouse knockouts meant that we were unable to study the role of Ercc1 in the skin. To do this, we have used the Cre-lox system to generate a skin-specific Ercc1 knockout. With a Cre transgene under control of the bovine keratin 5 promoter we achieved 100% recombination of the Ercc1 gene in the epidermis. Hairless mice with Ercc1-deficient skin were hypersensitive to the short-term effects of UV irradiation, showing a very low minimal erythemal dose and a dramatic hyperproliferative response. Ultraviolet-irradiated mice with Ercc1-deficient skin developed epidermal skin tumours much more rapidly than controls. These tumours appeared to arise earlier in actinic progression and grew more rapidly than tumours on control mice. These responses are more pronounced than have been reported for other NER-deficient mice, demonstrating that Ercc1 has a key role in protecting against UV-induced skin cancer.


Subject(s)
DNA Repair/genetics , DNA-Binding Proteins/physiology , Endonucleases/physiology , Epidermis/enzymology , Neoplasms, Radiation-Induced/enzymology , Skin Neoplasms/enzymology , Ultraviolet Rays/adverse effects , Animals , DNA/radiation effects , DNA Damage , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Disease Progression , Endonucleases/deficiency , Endonucleases/genetics , Epidermis/pathology , Epidermis/radiation effects , Female , Gene Targeting , Genes, Lethal , Integrases , Male , Mice , Mice, Hairless , Mice, Knockout , Neoplasms, Radiation-Induced/genetics , Organ Specificity , Skin Neoplasms/genetics , Transgenes
17.
Cancer Res ; 65(9): 3958-65, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15867397

ABSTRACT

The epidermal growth factor receptor (EGFR) is activated in skin cells following UV irradiation, the primary cause of nonmelanoma skin cancer. The EGFR inhibitor AG1478 prevented the UV-induced activation of EGFR and of downstream signaling pathways through c-Jun NH2-terminal kinases, extracellular signal-regulated kinases, p38 kinase, and phosphatidylinositol 3-kinase in the skin. The extent to which the UV-induced activation of EGFR influences skin tumorigenesis was determined in genetically initiated v-ras(Ha) transgenic Tg.AC mice, which have enhanced susceptibility to skin carcinogenesis. Topical treatment or i.p. injection of AG1478 before UV exposure blocked the UV-induced activation of EGFR in the skin and decreased skin tumorigenesis in Tg.AC mice. AG1478 treatment before each of several UV exposures decreased the number of papillomas arising and the growth of these tumors by approximately 50% and 80%, respectively. Inhibition of EGFR suppressed proliferation, increased apoptotic cell death, and delayed the onset of epidermal hyperplasia following UV irradiation. Genetic ablation of Egfr similarly delayed epidermal hyperplasia in response to UV exposure. Thus, the UV-induced activation of EGFR promotes skin tumorigenesis by suppressing cell death, augmenting cell proliferation, and accelerating epidermal hyperplasia in response to UV. These results suggest that EGFR may be an appropriate target for the chemoprevention of UV-induced skin cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Neoplasms, Radiation-Induced/prevention & control , Skin Neoplasms/prevention & control , Tyrphostins/pharmacology , Animals , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Enzyme Activation/radiation effects , Enzyme Inhibitors/pharmacology , ErbB Receptors/deficiency , ErbB Receptors/physiology , Hyperplasia , MAP Kinase Signaling System/radiation effects , Mice , Mice, Nude , Neoplasms, Radiation-Induced/enzymology , Neoplasms, Radiation-Induced/etiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Quinazolines , Skin/pathology , Skin/radiation effects , Skin Neoplasms/enzymology , Skin Neoplasms/etiology , Ultraviolet Rays/adverse effects
18.
Cancer Res ; 65(10): 4202-10, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15899811

ABSTRACT

We have assessed the potential of [18F]fluorothymidine positron emission tomography ([18F]FLT-PET) to measure early cytostasis and cytotoxicity induced by cisplatin treatment of radiation-induced fibrosarcoma 1 (RIF-1) tumor-bearing mice. Cisplatin-mediated arrest of tumor cell growth and induction of tumor shrinkage at 24 and 48 hours, respectively, were detectable by [18F]FLT-PET. At 24 and 48 hours, the normalized uptake at 60 minutes (tumor/liver radioactivity ratio at 60 minutes after radiotracer injection; NUV60) for [18F]FLT was 0.76 +/- 0.08 (P = 0.03) and 0.51 +/- 0.08 (P = 0.03), respectively, compared with controls (1.02 +/- 0.12). The decrease in [18F]FLT uptake at 24 hours was associated with a decrease in cell proliferation assessed immunohistochemically (a decrease in proliferating cell nuclear antigen labeling index, LI(PCNA), from 14.0 +/- 2.0% to 6.2 +/- 1.0%; P = 0.001), despite the lack of a change in tumor size. There were G1-S and G2-M phase arrests after cisplatin treatment, as determined by cell cycle analysis. For the quantitative measurement of tumor cell proliferation, [18F]FLT-PET was found to be superior to [18F]fluorodeoxyglucose-PET (NUV60 versus LIPCNA: r = 0.89, P = 0.001 and r = 0.55, P = 0.06, respectively). At the biochemical level, we found that the changes in [18F]FLT and [18F]fluorodeoxyglucose uptake were due to changes in levels of thymidine kinase 1 protein, hexokinase, and ATP. This work supports the further development of [18F]FLT-PET as a generic pharmacodynamic readout for early quantitative imaging of drug-induced changes in cell proliferation in vivo.


Subject(s)
Cisplatin/pharmacology , Dideoxynucleosides , Fibrosarcoma/diagnostic imaging , Fibrosarcoma/drug therapy , Fluorine Radioisotopes , Neoplasms, Radiation-Induced/diagnostic imaging , Neoplasms, Radiation-Induced/drug therapy , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Dideoxynucleosides/pharmacokinetics , Fibrosarcoma/enzymology , Fibrosarcoma/etiology , Hexokinase/metabolism , Male , Mice , Mice, Inbred C3H , Neoplasms, Radiation-Induced/enzymology , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Thymidine Kinase/metabolism
19.
J Natl Cancer Inst ; 76(1): 151-8, 1986 Jan.
Article in English | MEDLINE | ID: mdl-3455737

ABSTRACT

Skin tumors were induced in (C3H/HeN X C3H/HeN-PGK-1a)F1 female mice, heterozygous at the X-linked phosphoglycerate kinase-1 (PGK-1) locus, by exposure to the carcinogenic influence of ultraviolet radiation (UVR), 3-methylcholanthrene [(MCA) CAS: 56-49-5], or benz[a]pyrene [(BP) CAS: 50-32-8]. An assessment of the clonal origin of these tumors was accomplished through an analysis of the PGK-1 enzyme phenotype expressed by the transformed cells. In vitro culture was employed as a means of depleting nontransformed cells of host origin from the induced tumors. Cultured lines derived from tumors induced by each of the above agents were found to express only one of the two enzyme forms encoded by the host genotype, consistent with the probability that all the UVR-, MCA-, and BP-induced tumors examined in this study were monoclonal in origin. For further substantiation of the monoclonality of UVR-induced tumors, 2 UVR-induced tumors were enzymatically dissociated immediately following excision from the primary hosts, and the resulting cell suspensions were cloned in soft agar. Upon analysis, each set of clones selected in soft agar expressed only a single PGK-1 enzyme form. To rule out the possibility that the apparent monoclonality of culture-adapted tumor lines was due to in vitro selection, tumors that arose in UVR-treated PGK-1a/b female heterozygote mice were transplanted into PGK-1a and PGK-1b homozygous recipients. These transplanted tumors expressed a single PGK-1 allozyme following growth in recipients that were genetically homozygous for the major PGK-1 enzyme form expressed by the tumor prior to transplantation. These data strongly support the concept that most, if not all, UVR-induced tumors are derived from the progeny of a single transformed cell. This observation is important to the understanding of the nature of tumor-specific transplantation antigens expressed by individual UVR-induced tumors and indicates that such antigens also are clone specific. In addition, the results of this study indicate that polyclonality does not play a significant role in the generation of cellular and phenotypic heterogeneity known to be present within individual UVR-induced tumors.


Subject(s)
Neoplasms, Radiation-Induced/pathology , Animals , Benzo(a)pyrene , Female , Methylcholanthrene , Mice , Mice, Inbred C3H , Neoplasm Transplantation , Neoplasms, Radiation-Induced/enzymology , Phosphoglycerate Kinase/genetics , Ultraviolet Rays
20.
Cancer Res ; 57(3): 370-4, 1997 Feb 01.
Article in English | MEDLINE | ID: mdl-9012457

ABSTRACT

Telomerase activation plays a crucial role in the immortalization of human cells and carcinogenesis; however, the temporal and pathophysiological aspects of the activation in vivo are poorly understood. We found telomerase activity not only in malignant tumors (91%) but also in most benign (60%) and premalignant (89%) skin tumors. This suggests the involvement of telomerase activation in a crucial biological step of human skin carcinogenesis. Because UV light is a major factor in skin carcinogenesis, we further examined telomerase activity in normal skin samples and in normal skin samples adjacent to benign, premalignant, and malignant skin lesions. Data for chronically sun-exposed body sites were compared with those for covered sites. Among normal skin samples, 39% (26 of 67) had telomerase activity, and this activity was unrelated to neighboring lesions but strongly associated with the level of sun exposure. Fifty-four % (21 of 39) of normal skin samples from chronically sun-exposed sites were telomerase-positive, compared with only 12% (3 of 26) of samples from covered sites. When we examined telomerase activity and CC to TT mutations at codons 247/8 of the p53 gene (which are considered to be UV specific) in the same normal skin samples, only 43% (7 of 16) of telomerase-positive normal skin samples at sun-exposed sites contained the p53 mutations, whereas all (7 of 7) of the samples with UV-specific p53 mutations showed telomerase activity (P = 0.019). These data suggest that telomerase activation is involved at an early stage of human skin carcinogenesis and that activation may precede the acquisition of UV-associated p53 mutations in the skin. Telomerase activity was also found in plucked hair follicles and enzymatically separated epidermis, which may be associated with the presence of stem cells in the skin.


Subject(s)
Neoplasms, Radiation-Induced/etiology , Skin Neoplasms/etiology , Skin/radiation effects , Telomerase/metabolism , Enzyme Activation/radiation effects , Female , Genes, p53 , Hair Follicle/enzymology , Humans , Male , Mutation , Neoplasms, Radiation-Induced/enzymology , Precancerous Conditions/enzymology , Skin/enzymology , Skin Neoplasms/enzymology , Sunlight , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL