Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Int J Mol Sci ; 23(17)2022 Aug 31.
Article in English | MEDLINE | ID: mdl-36077308

ABSTRACT

Regardless of the significant improvements in treatment of melanoma, the majority of patients develop resistance whose mechanisms are still not completely understood. Hence, we generated and characterized two melanoma-derived cell lines, primary WM793B and metastatic A375M, with acquired resistance to the RAF inhibitor vemurafenib. The morphology of the resistant primary WM793B melanoma cells showed EMT-like features and exhibited a hybrid phenotype with both epithelial and mesenchymal characteristics. Surprisingly, the vemurafenib-resistant melanoma cells showed a decreased migration ability but also displayed a tendency to collective migration. Signaling pathway analysis revealed the reactivation of MAPK and the activation of the PI3K/AKT pathway depending on the vemurafenib-resistant cell line. The acquired resistance to vemurafenib caused resistance to chemotherapy in primary WM793B melanoma cells. Furthermore, the cell-cycle analysis and altered levels of cell-cycle regulators revealed that resistant cells likely transiently enter into cell cycle arrest at the G0/G1 phase and gain slow-cycling cell features. A decreased level of NME1 and NME2 metastasis suppressor proteins were found in WM793B-resistant primary melanoma, which is possibly the result of vemurafenib-acquired resistance and is one of the causes of increased PI3K/AKT signaling. Further studies are needed to reveal the vemurafenib-dependent negative regulators of NME proteins, their role in PI3K/AKT signaling, and their influence on vemurafenib-resistant melanoma cell characteristics.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Humans , Indoles/pharmacology , Indoles/therapeutic use , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Vemurafenib/pharmacology , Vemurafenib/therapeutic use
2.
Int J Cancer ; 150(2): 374-386, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34569060

ABSTRACT

Recurrent upper tract urothelial carcinomas (UTUCs) arise in the context of nephropathy linked to exposure to the herbal carcinogen aristolochic acid (AA). Here we delineated the molecular programs underlying UTUC tumorigenesis in patients from endemic aristolochic acid nephropathy (AAN) regions in Southern Europe. We applied an integrative multiomics analysis of UTUCs, corresponding unaffected tissues and of patient urines. Quantitative microRNA (miRNA) and messenger ribonucleic acid (mRNA) expression profiling, immunohistochemical analysis by tissue microarrays and exome and transcriptome sequencing were performed in UTUC and nontumor tissues. Urinary miRNAs of cases undergoing surgery were profiled before and after tumor resection. Ribonucleic acid (RNA) and protein levels were analyzed using appropriate statistical tests and trend assessment. Dedicated bioinformatic tools were used for analysis of pathways, mutational signatures and result visualization. The results delineate UTUC-specific miRNA:mRNA networks comprising 89 miRNAs associated with 1,862 target mRNAs, involving deregulation of cell cycle, deoxyribonucleic acid (DNA) damage response, DNA repair, bladder cancer, oncogenes, tumor suppressors, chromatin structure regulators and developmental signaling pathways. Key UTUC-specific transcripts were confirmed at the protein level. Exome and transcriptome sequencing of UTUCs revealed AA-specific mutational signature SBS22, with 68% to 76% AA-specific, deleterious mutations propagated at the transcript level, a possible basis for neoantigen formation and immunotherapy targeting. We next identified a signature of UTUC-specific miRNAs consistently more abundant in the patients' urine prior to tumor resection, thereby defining biomarkers of tumor presence. The complex gene regulation programs of AAN-associated UTUC tumors involve regulatory miRNAs prospectively applicable to noninvasive urine-based screening of AAN patients for cancer presence and recurrence.


Subject(s)
Aristolochic Acids/adverse effects , Biomarkers, Tumor/genetics , Carcinoma, Transitional Cell/pathology , Gene Expression Regulation, Neoplastic/drug effects , MicroRNAs/urine , Mutation , Urinary Bladder Neoplasms/pathology , Biomarkers, Tumor/urine , Carcinoma, Transitional Cell/chemically induced , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/urine , Exome , Follow-Up Studies , Humans , Prognosis , Proteome/analysis , Proteome/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Urinary Bladder Neoplasms/chemically induced , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/urine
3.
Int J Mol Sci ; 24(1)2022 Dec 21.
Article in English | MEDLINE | ID: mdl-36613518

ABSTRACT

Metastatic melanoma is one of the most aggressive tumors, with frequent mutations affecting components of the MAPK pathway, mainly protein kinase BRAF. Despite promising initial response to BRAF inhibitors, melanoma progresses due to development of resistance. In addition to frequent reactivation of MAPK or activation of PI3K/AKT signaling pathways, recently, the p53 pathway has been shown to contribute to acquired resistance to targeted MAPK inhibitor therapy. Canonical tumor suppressor p53 is inactivated in melanoma by diverse mechanisms. The TP53 gene and two other family members, TP63 and TP73, encode numerous protein isoforms that exhibit diverse functions during tumorigenesis. The p53 family isoforms can be produced by usage of alternative promoters and/or splicing on the C- and N-terminus. Various p53 family isoforms are expressed in melanoma cell lines and tumor samples, and several of them have already shown to have specific functions in melanoma, affecting proliferation, survival, metastatic potential, invasion, migration, and response to therapy. Of special interest are p53 family isoforms with increased expression and direct involvement in acquired resistance to MAPK inhibitors in melanoma cells, implying that modulating their expression or targeting their functional pathways could be a potential therapeutic strategy to overcome resistance to MAPK inhibitors in melanoma.


Subject(s)
Melanoma , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/genetics , Genes, p53 , Proto-Oncogene Proteins B-raf/genetics , Phosphatidylinositol 3-Kinases/metabolism , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Mutation , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor
4.
Cancers (Basel) ; 13(20)2021 Oct 18.
Article in English | MEDLINE | ID: mdl-34680379

ABSTRACT

Cutaneous melanoma is the most aggressive form of skin cancer. Despite the significant advances in the management of melanoma in recent decades, it still represents a challenge for clinicians. The TP53 gene, the guardian of the genome, which is altered in more than 50% of human cancers, is rarely mutated in melanoma. More recently, researchers started to appreciate the importance of shorter p53 isoforms as potential modifiers of the p53-dependent responses. We analyzed the expression of p53 and p73 isoforms both at the RNA and protein level in a panel of melanoma-derived cell lines with different TP53 and BRAF status, in normal conditions or upon treatment with common anti-cancer DNA damaging agents or targeted therapy. Using lentiviral vectors, we also generated stable clones of H1299 p53 null cells over-expressing the less characterized isoforms Δ160p53α, Δ160p53ß, and Δ160p53γ. Further, we obtained two melanoma-derived cell lines resistant to BRAF inhibitor vemurafenib. We observed that melanoma cell lines expressed a wide array of p53 and p73 isoforms, with Δ160p53α as the most variable one. We demonstrated for the first time that Δ160p53α, and to a lesser extent Δ160p53ß, can be recruited on chromatin, and that Δ160p53γ can localize in perinuclear foci; moreover, all Δ160p53 isoforms can stimulate proliferation and in vitro migration. Lastly, vemurafenib-resistant melanoma cells showed an altered expression of p53 and p73 isoforms, namely an increased expression of potentially pro-oncogenic Δ40p53ß and a decrease in tumor-suppressive TAp73ß. We therefore propose that p53 family isoforms can play a role in melanoma cells' aggressiveness.

5.
Cancers (Basel) ; 13(12)2021 Jun 09.
Article in English | MEDLINE | ID: mdl-34207603

ABSTRACT

The p53 tumor suppressor protein is crucial for cell growth control and the maintenance of genomic stability. Later discovered, p63 and p73 share structural and functional similarity with p53. To understand the p53 pathways more profoundly, all family members should be considered. Each family member possesses two promoters and alternative translation initiation sites, and they undergo alternative splicing, generating multiple isoforms. The resulting isoforms have important roles in carcinogenesis, while their expression is dysregulated in several human tumors including colorectal carcinoma, which makes them potential targets in cancer treatment. Their activities arise, at least in part, from the ability to form tetramers that bind to specific DNA sequences and activate the transcription of target genes. In this review, we summarize the current understanding of the biological activities and regulation of the p53/p73 isoforms, highlighting their role in colorectal tumorigenesis. The analysis of the expression patterns of the p53/p73 isoforms in human cancers provides an important step in the improvement of cancer therapy. Furthermore, the interactions among the p53 family members which could modulate normal functions of the canonical p53 in tumor tissue are described. Lastly, we emphasize the importance of clinical studies to assess the significance of combining the deregulation of different members of the p53 family to define the outcome of the disease.

6.
Int J Mol Sci ; 21(7)2020 Mar 29.
Article in English | MEDLINE | ID: mdl-32235358

ABSTRACT

Nucleoside diphosphate kinases (NDPK/NME/Nm23) are enzymes composed of subunits NME1/NDPK A and NME2/NDPK B, responsible for the maintenance of the cellular (d)NTP pool and involved in other cellular processes, such as metastasis suppression and DNA damage repair. Although eukaryotic NDPKs are active only as hexamers, it is unclear whether other NME functions require the hexameric form, and how the isoenzyme composition varies in different cellular compartments. To examine the effect of DNA damage on intracellular localization of NME1 and NME2 and the composition of NME oligomers in the nucleus and the cytoplasm, we used live-cell imaging and the FRET/FLIM technique. We showed that exogenous NME1 and NME2 proteins co-localize in the cytoplasm of non-irradiated cells, and move simultaneously to the nucleus after gamma irradiation. The FRET/FLIM experiments imply that, after DNA damage, there is a slight shift in the homomer/heteromer balance between the nucleus and the cytoplasm. Collectively, our results indicate that, after irradiation, NME1 and NME2 engage in mutual functions in the nucleus, possibly performing specific functions in their homomeric states. Finally, we demonstrated that fluorophores fused to the N-termini of NME polypeptides produce the largest FRET effect and thus recommend this orientation for use in similar studies.


Subject(s)
DNA Damage/genetics , DNA Damage/radiation effects , NM23 Nucleoside Diphosphate Kinases/genetics , Radiation, Ionizing , Animals , Biomarkers , Cell Line , Cell Nucleus/metabolism , Fluorescent Antibody Technique , Gamma Rays , Humans , NM23 Nucleoside Diphosphate Kinases/chemistry , NM23 Nucleoside Diphosphate Kinases/metabolism , Protein Binding , Protein Multimerization , Protein Transport
7.
Sci Rep ; 9(1): 12470, 2019 08 28.
Article in English | MEDLINE | ID: mdl-31462745

ABSTRACT

Unlike other tumours, TP53 is rarely mutated in melanoma; however, it fails to function as a tumour suppressor. We assume that its functions might be altered through interactions with several families of proteins, including p53/p73, NME and GLI. To elucidate the potential interplay among these families we analysed the expression profiles of aforementioned genes and proteins in a panel of melanoma cell lines, metastatic melanoma specimens and healthy corresponding tissue. Using qPCR a higher level of NME1 gene expression and lower levels of Δ40p53ß, ΔNp73, GLI1, GLI2 and PTCH1 were observed in tumour samples compared to healthy tissue. Protein expression of Δ133p53α, Δ160p53α and ΔNp73α isoforms, NME1 and NME2, and N'ΔGLI1, GLI1FL, GLI2ΔN isoforms was elevated in tumour tissue, whereas ∆Np73ß was downregulated. The results in melanoma cell lines, in general, support these findings. In addition, we correlated expression profiles with clinical features and outcome. Higher Δ133p53ß and p53α mRNA and both GLI1 mRNA and GLI3R protein expression had a negative impact on the overall survival. Shorter overall survival was also connected with lower p53ß and NME1 gene expression levels. In conclusion, all examined genes may have implications in melanoma development and functional inactivity of TP53.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Nucleoside-Diphosphate Kinase/biosynthesis , Tumor Protein p73/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , Cell Line, Tumor , Disease-Free Survival , Female , Humans , Male , Melanoma/genetics , Melanoma/mortality , Melanoma/pathology , Neoplasm Metastasis , Nucleoside-Diphosphate Kinase/genetics , Survival Rate , Tumor Protein p73/genetics , Tumor Suppressor Protein p53/genetics
8.
Urol Oncol ; 37(9): 578.e1-578.e10, 2019 09.
Article in English | MEDLINE | ID: mdl-30948335

ABSTRACT

OBJECTIVES: To analyze p53 mutations and gene expression of p53, ∆40p53, and ∆133p53 isoforms in renal cell cancer (RCC) tissues and normal adjacent tissue (NAT) and to associate them to clinical features and outcome. PATIENTS AND METHODS: Forty-one randomly selected patients, with primary, previously untreated RCC, with complete clinicopathohistological data were analyzed. NAT samples were available for 37 cases. Expression of p53, ∆40p53 and ∆133p53 was determined using RT-qPCR. A functional yeast-based assay was performed to analyze p53 mutations. RESULTS: More than half (56.1%) of patients harbored functional p53 mutations, and they were significantly younger than those with wild type (WT) p53 (P = 0.032). Expression of p53, ∆40p53, and ∆133p53 was upregulated in mutant (MT) p53 RCC compared to WT p53 RCC tissues. However, there was no difference in expression of these isoforms between MT p53 RCC tissues and NAT. Expression of ∆133p53 was significantly downregulated in WT p53 tissues compared to NAT (P = 0.006). Patients that harbored functional p53 mutation had better overall survival (hazard ratio 4.32, 95% confidence interval 1.46-18.82, P = 0.006). Multivariate analysis demonstrated that tumor stage and p53 mutation might be used as independent prognostic marker for overall survival in RCC patients. CONCLUSIONS: Our findings support the specific events in the carcinogenesis of RCC. p53 isoforms can be differentially expressed depending on p53 mutational status.


Subject(s)
Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Tumor Suppressor Protein p53/biosynthesis , Adult , Aged , Aged, 80 and over , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Female , Humans , Kidney Neoplasms/genetics , Male , Middle Aged , Mutation , Prognosis , Prospective Studies , Protein Isoforms , Tumor Suppressor Protein p53/genetics
9.
Mater Sci Eng C Mater Biol Appl ; 91: 486-495, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-30033280

ABSTRACT

Gold nanoparticles (AuNPs) were synthesized in the presence of citrate (Au-CIT), glutathione (Au-GSH) and aminodextran (Au-DEX) in order to modify AuNPs surfaces and to increase their cellular uptake in the breast cancer cells MDA-MB-231. AuNPs were characterized with respect to their particle size, shape and colloidal stability in an aqueous solution and cell media. The mass accumulation of each AuNP type inside cancer cells was determined quantitatively, using Inductive Coupled Plasma - mass spectroscopy. The sub-cellular accumulation was studied using Transmission Electron Microscopy (TEM). It was found that gold nanoparticles applied to cancer cells were localized in cytoplasmic vesicles and that the highest uptake was shown in the presence of Au-GSH nanoparticles. The effect of AuNPs on the cell cycle was investigated using flow cytometry and Western blot analysis. The gold nanoparticles alone did not affect the cell cycle, as shown by flow cytometry. Furthermore, the cancer cells were irradiated using conventional clinically relevant high-energy X-ray radiation of 6 MV in the dose of 4 Gy. The results on cells only irradiated showed an S phase arrest six and 8 h after irradiation, and a G2/M arrest 24 and 48 h after irradiation. The irradiation of breast cancer cells treated with AuNPs has shown no significant variation in cell cycle distribution as opposed to X-ray radiation alone.


Subject(s)
Breast Neoplasms/pathology , Cell Cycle , Gold/chemistry , Metal Nanoparticles/chemistry , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Survival/radiation effects , Endocytosis/drug effects , Female , Humans , Hydrodynamics , Metal Nanoparticles/ultrastructure , Spectrophotometry, Ultraviolet , Subcellular Fractions/metabolism , X-Rays
10.
Prog Neurobiol ; 168: 104-127, 2018 09.
Article in English | MEDLINE | ID: mdl-29733887

ABSTRACT

Alzheimer's disease (AD), the most common progressive neurodegenerative disorder, is characterized by severe cognitive decline and personality changes as a result of synaptic and neuronal loss. The defining clinicopathological hallmarks of the disease are deposits of amyloid precursor protein (APP)-derived amyloid-ß peptides (Aß) in the brain parenchyma, and intracellular aggregates of truncated and hyperphosphorylated tau protein in neurofibrillary tangles (NFT). At the cellular and molecular levels, many intertwined pathological mechanisms that relate Aß and tau pathology with a transcription factor p53 have been revealed. p53 is activated in response to various stressors that threaten genomic stability. Depending on damage severity, it promotes neuronal death or survival, predominantly via transcription-dependent mechanisms that affect expression of apoptosis-related target genes. Levels of p53 are enhanced in the AD brain and maintain sustained tau hyperphosphorylation, whereas intracellular Aß directly contributes to p53 pool and promotes downstream p53 effects. The review summarizes the role of p53 in neuronal function, discusses the interactions of p53, tau, and Aß in the normal brain and during the progression of AD pathology, and considers the impact of the most prominent hereditary risk factors of AD on p53/tau/Aß interactions. A better understanding of this intricate interplay would provide deeper insight into AD pathology and might offer some novel therapeutic targets for the improvement of treatment options. In this regard, drugs and natural compounds targeting the p53 pathway are of growing interest in neuroprotection as they may represent promising therapeutic approaches in the prevention of oxidative stress-dependent pathological processes underlying AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Tumor Suppressor Protein p53/metabolism , tau Proteins/metabolism , Animals , Humans
11.
Mech Ageing Dev ; 174: 55-62, 2018 09.
Article in English | MEDLINE | ID: mdl-28847485

ABSTRACT

Advances in epigenetics now enable us to better understand environmental influences on the genetic background of human diseases. This refers especially to fetal development where an adverse intrauterine environment impacts oxygen and nutrient supply to the fetus. Recently, differences in telomere length and telomere loss dynamics among individuals born with intrauterine growth restriction compared to normal controls have been described. In this paper we propose possible molecular mechanisms that (pre)program telomere epigenetics during pregnancy. This programming sets differences in telomere lengths and dynamics of telomere shortening in adulthood and therefore dictates the dynamics of aging and morbidity in later life.


Subject(s)
Epigenesis, Genetic/physiology , Fetal Development/physiology , Fetus/metabolism , Gene Expression Regulation, Developmental/physiology , Telomere Homeostasis/physiology , Animals , Female , Humans , Pregnancy
12.
J Hypertens ; 32(8): 1613-19; discussion 1619-20, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24805953

ABSTRACT

OBJECTIVE: The mechanisms that increase cardiovascular risk in individuals born small for gestational age (SGA) are not well understood. Telomere shortening has been suggested to be a predictor of disease onset. Our aim was to determine whether impaired intrauterine growth is associated with early signs of vascular aging and whether telomere length could be a biomarker of this pathway. METHODS: One hundred and fourteen healthy young men born SGA or after normal pregnancy [appropriate for gestational age (AGA)] were enrolled. Patient data were gathered from questionnaires and clinical exams, including blood pressure (BP) measurement routine laboratory analyses, and carotid intima-media thickness (cIMT). Leukocyte telomere length (LTL) was assessed by quantitative PCR. Birth data were obtained from medical records. RESULTS: The SGA group had significantly higher pulse pressure and cIMT, and a trend to increased SBP and heart rate in comparison to the AGA group. Interestingly, SGA men exhibited a 42% longer LTL than the AGA group. LTL was inversely associated with age, BMI, BP and birth parameters. In multiple regression analysis, BMI was the key determinant of SBP and cIMT. CONCLUSION: Young men born SGA show early signs of vascular aging. Unexpectedly, in our cohort, the SGA group had longer telomeres than the normal controls. Although longer telomeres are predictive of better health in the future, our findings could indicate a faster telomere attrition rate and probable early onset of cardiovascular risk in SGA participants. Follow-up of this cohort will clarify hypothesis and validate telomere dynamics as indicators of future health risks.


Subject(s)
Blood Pressure/physiology , Cellular Senescence , Fetal Growth Retardation/physiopathology , Telomere , Adult , Cardiovascular Diseases/etiology , Carotid Intima-Media Thickness , Cross-Sectional Studies , Humans , Infant, Small for Gestational Age , Linear Models , Male , Young Adult
13.
Carcinogenesis ; 34(3): 522-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23188674

ABSTRACT

The p53 activities are due, at least in part, to its ability to form oligomers that bind to specific DNA sequences and activate transcription. Since some mutant p53 proteins and ΔNp73 isoforms form heterocomplexes with TAp73, we asked whether p53 isoforms can do the same and potentially act as dominant-negative inhibitors of TAp73. Moreover, it has already been found that some isoforms form complex with wtp53 and some of them inhibit p53 tumor-suppressor functions. Therefore, we studied the complex formation and co-immunoprecipitation assays show that all six p53 isoforms examined can form complexes with TAp73ß, whereas only Δ133p53α/ß/γ isoforms form complex with TAp73α. All p53 isoforms counteract TAp73ß transactivation function but with different efficiency and in a promoter-dependent manner. Furthermore, apoptotic activity of TAp73ß was augmented by coexpression of p53ß, whereas Δ133p53α and ß inhibit its apoptotic activity most efficiently. We have determined the half-life of different p53 isoforms: p53γ isoform has the shortest half-life, whereas Δ133p53γ has the longest half-life. Inhibitory interactions of two proteins in complex often lead to their stabilization. However, only three isoforms (Δ133p53α, Δ133p53ß and Δ40p53α) stabilize TAp73ß. We are convinced that defining the interactions between p53/p73 would give a new insight into how the p53 isoforms modulate the p73 functions in tumorigenesis.


Subject(s)
Apoptosis , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Genes, Reporter , Half-Life , Humans , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Protein Binding , Protein Isoforms/metabolism , Protein Stability , Transcription, Genetic , Tumor Protein p73
14.
Kidney Int ; 81(6): 559-67, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22071594

ABSTRACT

Endemic (Balkan) nephropathy is a chronic tubulointerstitial disease frequently accompanied by urothelial cell carcinomas of the upper urinary tract. This disorder has recently been linked to exposure to aristolochic acid, a powerful nephrotoxin and human carcinogen. Following metabolic activation, aristolochic acid reacts with genomic DNA to form aristolactam-DNA adducts that generate a unique TP53 mutational spectrum in the urothelium. The aristolactam-DNA adducts are concentrated in the renal cortex, thus serving as biomarkers of internal exposure to aristolochic acid. Here, we present molecular epidemiologic evidence relating carcinomas of the upper urinary tract to dietary exposure to aristolochic acid. DNA was extracted from the renal cortex and urothelial tumor tissue of 67 patients that underwent nephroureterectomy for carcinomas of the upper urinary tract and resided in regions of known endemic nephropathy. Ten patients from nonendemic regions with carcinomas of the upper urinary tract served as controls. Aristolactam-DNA adducts were quantified by (32)P-postlabeling, the adduct was confirmed by mass spectrometry, and TP53 mutations in tumor tissues were identified by chip sequencing. Adducts were present in 70% of the endemic cohort and in 94% of patients with specific A:T to T:A mutations in TP53. In contrast, neither aristolactam-DNA adducts nor specific mutations were detected in tissues of patients residing in nonendemic regions. Thus, in genetically susceptible individuals, dietary exposure to aristolochic acid is causally related to endemic nephropathy and carcinomas of the upper urinary tract.


Subject(s)
Aristolochic Acids/adverse effects , Balkan Nephropathy/chemically induced , Carcinogens, Environmental/adverse effects , Carcinoma/chemically induced , DNA Adducts/analysis , Environmental Exposure , Kidney Cortex/drug effects , Urologic Neoplasms/chemically induced , Adult , Aged , Aged, 80 and over , Aristolochic Acids/metabolism , Balkan Nephropathy/diagnosis , Balkan Nephropathy/epidemiology , Balkan Nephropathy/genetics , Balkan Nephropathy/metabolism , Biomarkers/analysis , Biotransformation , Bosnia and Herzegovina/epidemiology , Carcinogens, Environmental/metabolism , Carcinoma/diagnosis , Carcinoma/epidemiology , Carcinoma/genetics , Carcinoma/metabolism , Case-Control Studies , Croatia/epidemiology , DNA Mutational Analysis , Diet , Environmental Monitoring/methods , Epidemiological Monitoring , Female , Genetic Predisposition to Disease , Humans , Kidney Cortex/chemistry , Kidney Cortex/pathology , Male , Mass Spectrometry , Middle Aged , Molecular Epidemiology , Mutation , Residence Characteristics , Risk Assessment , Risk Factors , Serbia/epidemiology , Tumor Suppressor Protein p53/genetics , Urologic Neoplasms/diagnosis , Urologic Neoplasms/epidemiology , Urologic Neoplasms/genetics , Urologic Neoplasms/metabolism
15.
Curr Pharm Des ; 17(6): 591-602, 2011.
Article in English | MEDLINE | ID: mdl-21391909

ABSTRACT

The p53 family consists of three members: p53, p63 and p73. All three of them have a role in cell cycle arrest and induction of apoptosis. However, despite structural and partly functional similarity, there are striking differences in their functions and each of them has its own and unique identity. All three genes encode multiple variants with opposing functions in cancer development - full length transactivation forms with proapoptotic and antiproliferative functions, and dominant-negative transactivation-deficient forms with anti-apoptotic (oncogenic) functions. The functional interactions between family members are crucial to gain insight and understand their role in cancer biology. The discovery of p53/p73 network could have a major clinical impact in prognostic use and targeted drug design. In the current review we present the recent achievements in p73 research including very complex and sophisticated p73 regulation and response to DNA damage, and functional interactions among family members. We discuss how p73 has affected drug discovery. According to the p73 tumor suppressor function, we outline current aspects of anticancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Antineoplastic Agents/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Drug Resistance, Neoplasm , Humans , Molecular Targeted Therapy , Nuclear Proteins/antagonists & inhibitors , Tumor Protein p73 , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors
16.
Int J Cancer ; 129(6): 1532-6, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21413016

ABSTRACT

This study was designed to establish the TP53 mutational spectrum of aristolochic acid (AA), examined in the context of endemic (Balkan) nephropathy, an environmental disease associated with transitional cell (urothelial) carcinomas of the upper urinary tract (UUC). Tumor tissue was obtained from residents of regions in Bosnia, Croatia and Serbia where endemic nephropathy has been prevalent for over 50 years. Fifty-nine TP53 mutations were detected in 42 of the 97 tumors analyzed. Mutational spectra were dominated by A:T to T:A transversions with the mutated adenines located almost exclusively on the nontranscribed strand. This marked strand bias is attributed to selective processing of aristolactam-dA adducts by transcription-coupled nucleotide excision repair. Hotspots for A:T to T:A mutations include codons 131 and 179 and the 5'-AG acceptor splice site of intron 6. The unique TP53 mutational signature for AA identified in this study can be used to explore the hypothesis that botanical products containing this human carcinogen and nephrotoxin are responsible, in part, for the high prevalence of UUC and chronic renal disease in countries where Aristolochia herbal remedies traditionally have been used for medicinal purposes.


Subject(s)
Aristolochic Acids/toxicity , Carcinogens, Environmental/toxicity , Diet , Genes, p53 , Mutation , Urothelium , Aristolochic Acids/administration & dosage , Carcinoma, Transitional Cell/genetics , Humans
17.
Mutat Res ; 663(1-2): 1-6, 2009 Apr 26.
Article in English | MEDLINE | ID: mdl-19428366

ABSTRACT

The activation of protooncogenes and inactivation of tumor suppressor genes are considered to be the main molecular events in the multistep process of carcinogenesis. Mutations of the TP53 tumor suppressor gene have been found in nearly all tumor types and are estimated to contribute to more than 50% of all cancers. Most mutations lead to the synthesis of highly stable, inactive proteins that accumulate in the nucleus of cancer cells. Among the 393 codons of the human p53 gene, 222 are targets of 698 different types of mutations. Alterations of codons 175, 248, 273 and 282 correspond to 19% of all mutations and are considered general hot spot mutations. Dietary exposure to aristolochic acid (AA), an established nephrotoxin and human carcinogen found in all Aristolochia species was shown to be the causative agent of aristolochic acid nephropathy (previously called Chinese herbs nephropathy). This syndrome is characterized by proximal tubular damage, renal interstitial fibrosis, slow progression to the end stage renal disease and a high prevalence of upper urinary tract urothelial carcinoma (otherwise a highly unusual location). AA preferentially binds to purines in DNA and is associated with a high frequency of A-->T transversions in the p53 gene. Rats treated with AA develop A:T-->T:A mutations in codon 61. The pathological and clinical features of endemic (Balkan) nephropathy closely resemble those associated with aristolochic acid nephropathy except for the slower progression to end stage renal disease and longer cumulative period before the appearance of urothelial cancer. Recently, we reported the presence of AA-DNA adducts in renal cortex and A-->T p53 mutations in tumor tissue of patients from Croatia and Bosnia with endemic nephropathy. These data support the hypothesis that dietary exposure to AA is a major risk factor for endemic (Balkan) nephropathy.


Subject(s)
Aristolochic Acids/toxicity , Balkan Nephropathy/chemically induced , Balkan Nephropathy/genetics , Carcinogens, Environmental/toxicity , Mutation/genetics , Tumor Suppressor Protein p53/genetics , Aristolochic Acids/chemistry , Carcinogens, Environmental/chemistry , Humans , Mutagens/chemistry , Mutagens/toxicity
18.
Proc Natl Acad Sci U S A ; 104(29): 12129-34, 2007 Jul 17.
Article in English | MEDLINE | ID: mdl-17620607

ABSTRACT

Endemic (Balkan) nephropathy (EN), a devastating renal disease affecting men and women living in rural areas of Bosnia, Bulgaria, Croatia, Romania, and Serbia, is characterized by its insidious onset, invariable progression to chronic renal failure and a strong association with transitional cell (urothelial) carcinoma of the upper urinary tract. Significant epidemiologic features of EN include its focal occurrence in certain villages and a familial, but not inherited, pattern of disease. Our experiments test the hypothesis that chronic dietary poisoning by aristolochic acid is responsible for EN and its associated urothelial cancer. Using (32)P-postlabeling/PAGE and authentic standards, we identified dA-aristolactam (AL) and dG-AL DNA adducts in the renal cortex of patients with EN but not in patients with other chronic renal diseases. In addition, urothelial cancer tissue was obtained from residents of endemic villages with upper urinary tract malignancies. The AmpliChip p53 microarray was then used to sequence exons 2-11 of the p53 gene where we identified 19 base substitutions. Mutations at A:T pairs accounted for 89% of all p53 mutations, with 78% of these being A:T --> T:A transversions. Our experimental results, namely, that (i) DNA adducts derived from aristolochic acid (AA) are present in renal tissues of patients with documented EN, (ii) these adducts can be detected in transitional cell cancers, and (iii) A:T --> T:A transversions dominate the p53 mutational spectrum in the upper urinary tract malignancies found in this population lead to the conclusion that dietary exposure to AA is a significant risk factor for EN and its attendant transitional cell cancer.


Subject(s)
Aristolochic Acids/adverse effects , Balkan Nephropathy/chemically induced , Balkan Nephropathy/etiology , Aristolochic Acids/analysis , Aristolochic Acids/chemistry , Carcinoma, Transitional Cell/pathology , DNA Adducts/analysis , DNA Adducts/chemistry , Female , Humans , Kidney/chemistry , Kidney/pathology , Male , Mutation/genetics , Spectrometry, Mass, Electrospray Ionization , Tumor Suppressor Protein p53/genetics , Urologic Neoplasms/pathology
19.
Int J Oncol ; 29(4): 889-902, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16964385

ABSTRACT

The goal of this study was to determine whether patterns of expression profiles of p73 isoforms and of p53 mutational status are useful combinatorial biomarkers for predicting outcome in a gynecological cancer cohort. This is the first such study using matched tumor/normal tissue pairs from each patient. The median follow-up was over two years. The expression of all 5 N-terminal isoforms (TAp73, DeltaNp73, DeltaN'p73, Ex2p73 and Ex2/3p73) was measured by real-time RT-PCR and p53 status was analyzed by immunohistochemistry. TAp73, DeltaNp73 and DeltaN'p73 were significantly upregulated in tumors. Surprisingly, their range of overexpression was age-dependent, with the highest differences delta (tumor-normal) in the youngest age group. Correction of this age effect was important in further survival correlations. We used all 6 variables (five p73 isoform levels plus p53 status) as input into a principal component analysis with Varimax rotation (VrPCA) to filter out noise from non-disease related individual variability of p73 levels. Rationally selected and individually weighted principal components from each patient were then used to train a support vector machine (SVM) algorithm to predict clinical outcome. This SVM algorithm was able to predict correct outcome in 30 of the 35 patients. We use here a mathematical tool for pattern recognition that has been commonly used in e.g. microarray data mining and apply it for the first time in a prognostic model. We find that PCA/SVM is able to test a clinical hypothesis with robust statistics and show that p73 expression profiles and p53 status are useful prognostic biomarkers that differentiate patients with good vs. poor prognosis with gynecological cancers.


Subject(s)
DNA-Binding Proteins/metabolism , Endometrial Neoplasms/diagnosis , Nuclear Proteins/metabolism , Ovarian Neoplasms/diagnosis , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Proteins/metabolism , Age Factors , DNA-Binding Proteins/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Humans , Intracellular Signaling Peptides and Proteins , Nuclear Proteins/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Prognosis , Protein Isoforms/analysis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proteins/analysis , Proteins/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Tumor Protein p73 , Tumor Suppressor Proteins/genetics , Up-Regulation
20.
Mol Cancer Res ; 2(7): 371-86, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15280445

ABSTRACT

The tumor suppressor p53 is critically important in the cellular damage response and is the founding member of a family of proteins. All three genes regulate cell cycle and apoptosis after DNA damage. However, despite a remarkable structural and partly functional similarity among p53, p63, and p73, mouse knockout studies revealed an unexpected functional diversity among them. p63 and p73 knockouts exhibit severe developmental abnormalities but no increased cancer susceptibility, whereas this picture is reversed for p53 knockouts. Neither p63 nor p73 is the target of inactivating mutations in human cancers. Genomic organization is more complex in p63 and p73, largely the result of an alternative internal promoter generating NH2-terminally deleted dominant-negative proteins that engage in inhibitory circuits within the family. Deregulated dominant-negative p73 isoforms might play an active oncogenic role in some human cancers. Moreover, COOH-terminal extensions specific for p63 and p73 enable further unique protein-protein interactions with regulatory pathways involved in development, differentiation, proliferation, and damage response. Thus, p53 family proteins take on functions within a wide biological spectrum stretching from development (p63 and p73), DNA damage response via apoptosis and cell cycle arrest (p53, TAp63, and TAp73), chemosensitivity of tumors (p53 and TAp73), and immortalization and oncogenesis (DeltaNp73).


Subject(s)
DNA-Binding Proteins/metabolism , Embryonic Development , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Trans-Activators/metabolism , Amino Acid Sequence , Animals , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Expression Regulation , Genes, Tumor Suppressor , Humans , Molecular Sequence Data , Neoplasms/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Phosphoproteins/chemistry , Phosphoproteins/genetics , Trans-Activators/chemistry , Trans-Activators/genetics , Transcription Factors , Tumor Protein p73 , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...