Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 491
Filter
1.
Sci Rep ; 14(1): 2574, 2024 01 31.
Article in English | MEDLINE | ID: mdl-38296990

ABSTRACT

Esophageal carcinoma (ESCA) is a leading cause of cancer-related death worldwide, and certain oral and intestinal pathogens have been associated with cancer development and progression. We asked if esophageal microbiomes had shared alterations that could provide novel biomarkers for ESCA risk. We extracted DNA from tumor and non-tumor tissue of 212 patients in the NCI-MD case control study and sequenced the 16S rRNA gene (V3-4), with TCGA ESCA RNA-seq (n = 172) and WGS (n = 123) non-human reads used as validation. We identified four taxa, Campylobacter, Prevotella, Streptococcus, and Fusobacterium as highly enriched in esophageal cancer across all cohorts. Using SparCC, we discovered that Fusobacterium and Prevotella were also co-enriched across all cohorts. We then analyzed immune cell infiltration to determine if these dysbiotic taxa were associated with immune signatures. Using xCell to obtain predicted immune infiltrates, we identified a depletion of megakaryocyte-erythroid progenitor (MEP) cells in tumors with presence of any of the four taxa, along with enrichment of platelets in tumors with Campylobactor or Fusobacterium. Taken together, our results suggest that intratumoral presence of these co-occurring bacterial genera may confer tumor promoting immune alterations that allow disease progression in esophageal cancer.


Subject(s)
Esophageal Neoplasms , Humans , Case-Control Studies , RNA, Ribosomal, 16S/genetics , Fusobacterium/genetics , Blood Platelets
2.
Cell Death Differ ; 23(9): 1515-28, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27104929

ABSTRACT

Bidirectional interactions between astrocytes and neurons have physiological roles in the central nervous system and an altered state or dysfunction of such interactions may be associated with neurodegenerative diseases, such as Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS). Astrocytes exert structural, metabolic and functional effects on neurons, which can be either neurotoxic or neuroprotective. Their neurotoxic effect is mediated via the senescence-associated secretory phenotype (SASP) involving pro-inflammatory cytokines (e.g., IL-6), while their neuroprotective effect is attributed to neurotrophic growth factors (e.g., NGF). We here demonstrate that the p53 isoforms Δ133p53 and p53ß are expressed in astrocytes and regulate their toxic and protective effects on neurons. Primary human astrocytes undergoing cellular senescence upon serial passaging in vitro showed diminished expression of Δ133p53 and increased p53ß, which were attributed to the autophagic degradation and the SRSF3-mediated alternative RNA splicing, respectively. Early-passage astrocytes with Δ133p53 knockdown or p53ß overexpression were induced to show SASP and to exert neurotoxicity in co-culture with neurons. Restored expression of Δ133p53 in near-senescent, otherwise neurotoxic astrocytes conferred them with neuroprotective activity through repression of SASP and induction of neurotrophic growth factors. Brain tissues from AD and ALS patients possessed increased numbers of senescent astrocytes and, like senescent astrocytes in vitro, showed decreased Δ133p53 and increased p53ß expression, supporting that our in vitro findings recapitulate in vivo pathology of these neurodegenerative diseases. Our finding that Δ133p53 enhances the neuroprotective function of aged and senescent astrocytes suggests that the p53 isoforms and their regulatory mechanisms are potential targets for therapeutic intervention in neurodegenerative diseases.


Subject(s)
Tumor Suppressor Protein p53/metabolism , Alternative Splicing , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Autophagy/drug effects , Brain/metabolism , Brain/pathology , Cells, Cultured , Cellular Senescence , Coculture Techniques , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Leupeptins/pharmacology , Neurons/cytology , Neurons/metabolism , Neuroprotection/physiology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Sequestosome-1 Protein/antagonists & inhibitors , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Serine-Arginine Splicing Factors/antagonists & inhibitors , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
3.
Oncogene ; 34(44): 5548-59, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-25703328

ABSTRACT

Although microRNAs (miRs) have been implicated in the pathogenesis of various human malignancies, limited information is available regarding mechanisms by which these noncoding RNAs contribute to initiation and progression of tobacco-induced esophageal cancers. In this study, array and quantitative reverse transcriptase-PCR techniques were used to examine miR expression in immortalized esophageal epithelia (IEE) and esophageal adenocarcinoma (EAC) cells cultured in normal media with or without cigarette smoke condensate (CSC). Under relevant exposure conditions, CSC significantly decreased miR-217 expression in these cells. Endogenous levels of miR-217 expression in cultured EAC cells (EACC)/primary EACs were significantly lower than those observed in IEE/ paired normal esophageal tissues. RNA crosslink immunoprecipitation, quantitative reverse transcriptase-PCR (qRT-PCR) and immunoblot experiments demonstrated direct interaction of miR-217 with kallikrein 7 (KLK7), encoding a putative oncogene not previously implicated in EAC. Repression of miR-217 correlated with increased levels of KLK7 in primary EACs, particularly those from smokers. Chromatin and methylated DNA immunoprecipitation experiments demonstrated that CSC-mediated repression of miR-217 coincided with DNMT3b-dependent hypermethylation and decreased occupancy of nuclear factor 1 within the miR-217 genomic locus. Deoxyazacytidine induced miR-217 expression and downregulated KLK7 in EACC; deoxyazacytidine also attenuated CSC-mediated miR-217 repression and upregulation of KLK7 in IEE and EACC. Overexpression of miR-217 significantly decreased, whereas overexpression of KLK7 increased proliferation, invasion and tumorigenicity of EACC. Collectively, these data demonstrate that epigenetic repression of miR-217 contributes to the pathogenesis of EAC via upregulation of KLK7 and suggest that restoration of miR-217 expression may be a novel treatment strategy for these malignancies.


Subject(s)
Adenocarcinoma/genetics , Carcinogenesis/genetics , Epigenetic Repression/genetics , Esophageal Neoplasms/genetics , MicroRNAs/genetics , Nicotiana/adverse effects , Smoking/genetics , Adenocarcinoma/pathology , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Chromatin/genetics , DNA Methylation/genetics , Down-Regulation/genetics , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/genetics , Humans , Kallikreins/genetics , NFI Transcription Factors/genetics , Neoplasm Invasiveness/genetics , Smoke/adverse effects , Up-Regulation/genetics
4.
Oncogene ; 33(13): 1640-8, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-23584481

ABSTRACT

LKB1/STK11 is a tumor suppressor gene responsible for Peutz-Jeghers syndrome, an inherited cancer disorder associated with genome instability. The LKB1 protein functions in the regulation of cell proliferation, polarization and differentiation. Here, we suggest a role of LKB1 in non-homologous end joining (NHEJ), a major DNA double-strand break (DSB) repair pathway. LKB1 localized to DNA ends upon the generation of micro-irradiation and I-SceI endonuclease-induced DSBs. LKB1 inactivation either by RNA interference or by kinase-dead mutation compromised NHEJ-mediated DNA repair by suppressing the accumulation of BRM, a catalytic subunit of the SWI/SNF complex, at DSB sites, which promotes the recruitment of an essential NHEJ factor, KU70. AMPK2, a major substrate of LKB1 and a histone H2B kinase, was recruited to DSBs in an LKB1-dependent manner. AMPK2 depletion and a mutation of H2B that disrupted the AMPK2 phoshorylation site impaired KU70 and BRM recruitment to DSB sites. LKB1 depletion induced the formation of chromosome breaks and radials. These results suggest that LKB1-AMPK signaling controls NHEJ and contributes to genome stability.


Subject(s)
AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , DNA End-Joining Repair , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Cell Line, Tumor , Chromatin Assembly and Disassembly , Genes, Tumor Suppressor , Genomic Instability , Humans , Signal Transduction , Transfection
5.
Br J Cancer ; 109(7): 1954-64, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24002594

ABSTRACT

BACKGROUND: The association between oral contraceptive (OC) use, hormone replacement therapy (HRT) and lung cancer risk in women is still debated. METHODS: We performed a pooled analysis of six case-control studies (1961 cases and 2609 controls) contributing to the International Lung Cancer Consortium. Potential associations were investigated with multivariable unconditional logistic regression and meta-analytic models. Multinomial logistic regressions were performed to investigate lung cancer risk across histologic types. RESULTS: A reduced lung cancer risk was found for OC (odds ratio (OR)=0.81; 95% confidence interval (CI): 0.68-0.97) and HRT ever users (OR=0.77; 95% CI: 0.66-0.90). Both oestrogen only and oestrogen+progestin HRT were associated with decreased risk (OR=0.76; 95% CI: 0.61-0.94, and OR=0.66; 95% CI: 0.49-0.88, respectively). No dose-response relationship was observed with years of OC/HRT use. The greatest risk reduction was seen for squamous cell carcinoma (OR=0.53; 95% CI: 0.37-0.76) in OC users and in both adenocarcinoma (OR=0.79; 95% CI: 0.66-0.95) and small cell carcinoma (OR=0.37; 95% CI: 0.19-0.71) in HRT users. No interaction with smoking status or BMI was observed. CONCLUSION: Our findings suggest that exogenous hormones can play a protective role in lung cancer aetiology. However, given inconsistencies with epidemiological evidence from cohort studies, further and larger investigations are needed for a more comprehensive view of lung cancer development in women.


Subject(s)
Contraceptives, Oral/adverse effects , Hormone Replacement Therapy/adverse effects , Lung Neoplasms/epidemiology , Adenocarcinoma/epidemiology , Adenocarcinoma/etiology , Aged , Carcinoma, Squamous Cell/epidemiology , Carcinoma, Squamous Cell/etiology , Estrogens/pharmacology , Female , Humans , Lung Neoplasms/etiology , Middle Aged , Progestins/pharmacology , Risk , Small Cell Lung Carcinoma/epidemiology , Small Cell Lung Carcinoma/etiology
6.
Oncogene ; 32(22): 2792-8, 2013 May 30.
Article in English | MEDLINE | ID: mdl-22777358

ABSTRACT

Most human pre-mRNA transcripts are alternatively spliced, but the significance and fine-tuning of alternative splicing in different biological processes is only starting to be understood. SRSF3 (SRp20) is a member of a highly conserved family of splicing factors that have critical roles in key biological processes, including tumor progression. Here, we show that SRSF3 regulates cellular senescence, a p53-mediated process to suppress tumorigenesis, through TP53 alternative splicing. Downregulation of SRSF3 was observed in normal human fibroblasts undergoing replicative senescence, and was associated with the upregulation of p53ß, an alternatively spliced isoform of p53 that promotes p53-mediated senescence. Knockdown of SRSF3 by short interfering RNA (siRNA) in early-passage fibroblasts induced senescence, which was associated with elevated expression of p53ß at mRNA and protein levels. Knockdown of p53 partially rescued SRSF3-knockdown-induced senescence, suggesting that SRSF3 acts on p53-mediated cellular senescence. RNA pulldown assays demonstrated that SRSF3 binds to an alternatively spliced exon uniquely included in p53ß mRNA through the consensus SRSF3-binding sequences. RNA crosslinking and immunoprecipitation assays (CLIP) also showed that SRSF3 in vivo binds to endogenous p53 pre-mRNA at the region containing the p53ß-unique exon. Splicing assays using a transfected TP53 minigene in combination with siRNA knockdown of SRSF3 showed that SRSF3 functions to inhibit the inclusion of the p53ß-unique exon in splicing of p53 pre-mRNA. These data suggest that downregulation of SRSF3 represents an endogenous mechanism for cellular senescence that directly regulates the TP53 alternative splicing to generate p53ß. This study uncovers the role for general splicing machinery in tumorigenesis, and suggests that SRSF3 is a direct regulator of p53.


Subject(s)
Alternative Splicing/genetics , Protein Isoforms/metabolism , RNA-Binding Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Cell Line , Cell Transformation, Neoplastic , Cellular Senescence/genetics , Down-Regulation , Fibroblasts , Humans , Protein Isoforms/genetics , RNA Interference , RNA Precursors/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors , Up-Regulation
7.
Oncogene ; 30(35): 3784-91, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21532627

ABSTRACT

LKB1/STK11 is a tumor suppressor and a negative regulator of mammalian target of rapamycin signaling. It is inactivated in 30% of lung cancer cell lines but only 5-15% of primary lung adenocarcinomas. There is evidence that homozygous deletion (HD) of chromosome 19p at the LKB locus contributes to the inactivation of the gene in primary human lung cancers. Here, we used several complementary genetic approaches to assess the LKB1 locus in primary non-small cell lung cancers (NSCLCs). We first analyzed 124 NSCLC cases for allelic imbalance using eight microsatellite markers on chromosome 19p, which revealed an overall rate of 65% (80 of 124) loss of heterozygosity (LOH). We next used chromogenic in situ hybridization (CISH) to directly examine the chromosomal status of the LKB1 locus. In all, 65 of 124 LOH tested samples were available for CISH and 58 of those (89%) showed either loss of one copy of chromosome 19p (LOH, 40 of 65 cases, 62%) or both copies (HD 18 of 65 cases, 28%). The occurrence of HD was significantly more frequent in Caucasian (35%) than in African-American patients (6%) (P=0.04). A total of 62 of 124 samples with LOH at one or both markers immediately flanking the LKB1 gene were further analyzed by directly sequencing the complete coding region, which identified 7 of 62 (11%) tumors with somatic mutations in the gene. Jointly, our data identified total inactivation of the LKB1 gene by either HD or LOH with somatic mutation in 39% of tested samples, whereas loss of chromosome 19p region by HD or LOH at the LKB1 region occured in 90% of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Gene Deletion , Lung Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Adult , Aged , Aged, 80 and over , Chromosomes, Human, Pair 19/genetics , Female , Homozygote , Humans , Loss of Heterozygosity , Male , Microsatellite Repeats , Middle Aged
8.
Curr Drug Targets ; 10(5): 442-54, 2009 May.
Article in English | MEDLINE | ID: mdl-19442116

ABSTRACT

Recent emerging evidence suggests that ING family proteins play roles in carcinogenesis both as oncogenes and tumor suppressor genes depending on the family members and on cell status. Previous results from non-physiologic overexpression experiments showed that all five family members induce apoptosis or cell cycle arrest, thus it had been thought until very recently that all of the family members function as tumor suppressor genes. Therefore restoration of ING family proteins in cancer cells has been proposed as a treatment for cancers. However, ING2 knockdown experiments showed unexpected results: ING2 knockdown led to senescence in normal human fibroblast cells and suppressed cancer cell growth. ING2 is also overexpressed in colorectal cancer, and promotes cancer cell invasion through an MMP13 dependent pathway. Additionally, it was reported that ING2 has two isoforms, ING2a and ING2b. Although expression of ING2a predominates compared with ING2b, both isoforms confer resistance against cell cycle arrest or apoptosis to cancer cells, thus knockdown of both isoforms is critical to remove this resistance. Taken together, these results suggest that ING2 can function as an oncogene in some specific types of cancer cells, indicating restoration of this gene in cancer cells could cause cancer progression. Because knockdown of ING2 suppresses cancer cell invasion and induces apoptosis or cell cycle arrest, ING2 may be an anticancer drug target. In this brief review, we discuss possible clinical applications of ING2 with the latest knowledge of molecular targeted therapies.


Subject(s)
Drug Delivery Systems , Neoplasms/drug therapy , Tumor Suppressor Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Cancer Vaccines/pharmacology , Cell Cycle/physiology , Gene Expression Regulation , Humans , Neoplasms/physiopathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tumor Suppressor Proteins/genetics
9.
J R Army Med Corps ; 155(2): 157-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-20095186

ABSTRACT

Our recent experience of paediatric critical care during UK military operations in Afghanistan is discussed alongside consideration of the background to the paediatric critical care service on deployment. We describe the intensive care unit's capabilities, details of recent paediatric critical care admissions during July to September 2008 and some of the ethical issues arising. Some desirable future developments will be suggested.


Subject(s)
Critical Care , Hospitals, Military/organization & administration , Intensive Care Units/organization & administration , Mobile Health Units/organization & administration , Pediatrics/organization & administration , Adolescent , Afghan Campaign 2001- , Afghanistan , Child , Child, Preschool , Humans , Infant , Infant, Newborn , United Kingdom
10.
Mech Ageing Dev ; 129(11): 681-91, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19238688

ABSTRACT

Human cells express five DNA helicases that are paralogs of Escherichia coli RecQ and which constitute the family of human RecQ helicases. Disease-causing mutations in three of these five human DNA helicases, BLM, WRN, and RECQL4, cause rare severe human genetic diseases with distinct clinical phenotypes characterized by developmental defects, skin abnormalities, genomic instability, and cancer susceptibility. Although biochemical and genetic evidence support roles for all five human RecQ helicases in DNA replication, DNA recombination, and the biological responses to DNA damage, many questions concerning the various functions of the human RecQ helicases remain unanswered. Researchers investigating human and non-human RecQ helicases held a workshop on May 27-28, 2008, at the University of Chicago Gleacher Center, during which they shared insights, discussed recent progress in understanding the biochemistry, biology, and genetics of the RecQ helicases, and developed research strategies that might lead to therapeutic approaches to the human diseases that result from mutations in RecQ helicase genes. Some workshop sessions were held jointly with members of a recently formed advocacy and support group for persons with Bloom's syndrome and their families. This report describes the outcomes and main discussion points of the workshop.


Subject(s)
Bloom Syndrome/enzymology , RecQ Helicases/metabolism , Aging/genetics , Aging/metabolism , Animals , Biomedical Research , Bloom Syndrome/genetics , DNA Damage , DNA Replication , Humans , Mutation , Neoplasms/enzymology , Neoplasms/genetics , RecQ Helicases/deficiency , RecQ Helicases/genetics , Recombination, Genetic , Self-Help Groups
11.
Carcinogenesis ; 28(7): 1552-60, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17404395

ABSTRACT

Hepatocellular carcinoma (HCC) is a fatal disease and hepatitis B and C viruses (HBV and HCV) are considered as major causative factors for the development of HCC. We have conducted gene expression profiling studies to search for potential target genes responsible for HCV-mediated HCC. Adenoviruses encoding core (HCV structural protein), NS3 and NS5A [HCV non-structural (NS) proteins] were generated and infected individually or together in freshly isolated primary human hepatocytes. An adenovirus harboring the oncogenic HBV protein, HBx, was included for comparison. A microarray platform of over 22,000 human oligos was analyzed to seek out significant differentially expressed genes among these viral proteins. We also compared these gene expression profiles with those obtained from HCV-infected liver samples from chronic liver disease (CLD) patients and HCV-related HCC. We found that HCV-related proteins largely induce unique genes when compared with HBx. In particular, interferon-inducible gene 27 (IFI27) was highly expressed in HCV or core-infected hepatocytes and HCV-related CLD or HCC, but was not significantly expressed in HBx-infected hepatocytes or HBV-related CLD or HCC, indicating that IFI27 may play a role in HCV-mediated HCC. In conclusion, our results suggest that HBV and HCV promote HCC development mainly through different mechanisms.


Subject(s)
Carcinoma, Hepatocellular/virology , Hepacivirus/metabolism , Liver Neoplasms/virology , Viral Nonstructural Proteins/physiology , Adenoviridae/genetics , Carcinoma, Hepatocellular/metabolism , Cells, Cultured , Hepatitis B virus/metabolism , Hepatocytes/metabolism , Humans , Liver Neoplasms/metabolism , Membrane Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Trans-Activators , Viral Nonstructural Proteins/genetics , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/physiology
12.
Oncogene ; 26(15): 2166-76, 2007 Apr 02.
Article in English | MEDLINE | ID: mdl-17401425

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide and the major risk factors include chronic infections with the hepatitis B (HBV) or C (HCV) virus, and exposure to dietary aflatoxin B(1) (AFB(1)) or alcohol consumption. Multiple genetic and epigenetic changes are involved in the molecular pathogenesis of HCC, for example, somatic mutations in the p53 tumor suppressor gene (TP53) and the activation of the WNT signal transduction pathway. AFB(1) frequently induces G:C to T:A transversions at the third base in codon 249 of TP53 and cooperates with HBV in causing p53 mutations in HCC. The detection of TP53 mutant DNA in plasma is a biomarker of both AFB(1) exposure and HCC risk. Chronic infection with HBV and HCV viruses, and oxyradical disorders including hemochromatosis, also generate reactive oxygen/nitrogen species that can both damage DNA and mutate cancer-related genes such as TP53. Certain mutant p53 proteins may exhibit a 'gain of oncogenic function'. The p53 biological network is a key responder to this oxidative and nitrosative stress. Depending on the extent of the DNA damage, p53 regulates the transcription of protective antioxidant genes and with extensive DNA damage, transactivates pro-oxidant genes that contribute to apoptosis. The X gene of HBV (HBx) is the most common open reading frame integrated into the host genome in HCC and the integrated HBx is frequently mutated. Mutant HBx proteins still retain their ability to bind to p53, and attenuate DNA repair and p53-mediated apoptosis. In summary, both viruses and chemicals are implicated in the etiology of TP53 mutations during the molecular pathogenesis of HCC.


Subject(s)
Carcinoma, Hepatocellular/etiology , Genetic Predisposition to Disease , Liver Neoplasms/etiology , Tumor Suppressor Protein p53/genetics , Aflatoxin B1/toxicity , Carcinoma, Hepatocellular/epidemiology , Carcinoma, Hepatocellular/genetics , Hepatitis Viruses , Hepatitis, Chronic/complications , Humans , Incidence , Liver Neoplasms/epidemiology , Liver Neoplasms/genetics , Mutagenesis , Mutation
13.
Oncogene ; 25(14): 2119-23, 2006 Mar 30.
Article in English | MEDLINE | ID: mdl-16288211

ABSTRACT

A subset of DNA helicases, the RecQ family, has been found to be associated with the p53-mediated apoptotic pathway and is involved in maintaining genomic integrity. This family contains the BLM and WRN helicases, in which germline mutations are responsible for Bloom and Werner syndromes, respectively. TFIIH DNA helicases, XPB and XPD, are also components in this apoptotic pathway. We hypothesized that there may be some redundancy between helicases in their ability to complement the attenuated p53-mediated apoptotic levels seen in cells from individuals with diseases associated with these defective helicase genes. The attenuated apoptotic phenotype in Bloom syndrome cells was rescued not only by ectopic expression of BLM, but also by WRN or XPB, both 3' --> 5' helicases, but not expression of the 5' --> 3' helicase XPD. Overexpression of Sgs1, a WRN/BLM yeast homolog, corrected the reduction in BS cells only, which is consistent with Sgs1 being evolutionarily most homologous to BLM. A restoration of apoptotic levels in cells from WS, XPB or XPD patients was attained only by overexpression of the specific helicase. Our data suggest a limited redundancy in the pathways of these RecQ helicases in p53-induced apoptosis.


Subject(s)
Apoptosis/physiology , DNA Helicases/metabolism , Tumor Suppressor Protein p53/physiology , Bloom Syndrome/enzymology , Germ-Line Mutation , Humans , Werner Syndrome/enzymology
15.
Lung Cancer ; 34 Suppl 2: S7-15, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11720736

ABSTRACT

The p53 tumor suppressor gene is mutated in about half of all human cancer cases. The p53 protein modulates multiple cellular functions, such as gene transcription, DNA synthesis and repair, cell cycle arrest, senescence, and apoptosis. Mutations in the p53 gene can abrogate these functions and may lead to genetic instability and progress to cancer. The molecular archeology of the p53 mutation spectrum generates hypotheses concerning the etiology and molecular pathogenesis of cancer. The spectrum of somatic mutations in the p53 gene implicates environmental carcinogens and endogenous processes in the etiology of human cancer. The presence of a characteristic p53 mutation also can manifest a molecular link between exposure to a particular carcinogen and a specific type of human cancer, e.g. aflatoxin B1 (AFB1) exposure and codon 249ser mutations in hepatocellular carcinoma, ultraviolet (UV) exposure and CC to TT tandem mutations in skin cancer, and cigarette smoke and the prevalence of G to T transversions in lung cancer. Although several different exogenous carcinogens have been shown to selectively target p53, evidence supporting the endogenous insult of p53 from oxyradical and nitrogen-oxyradicals is accumulating. p53 mutations can be a biomarker of carcinogen effect. Determining the characteristic p53 mutation load in nontumorous tissue, with a highly sensitive mutation assay, can indicate a specific carcinogen exposure and also may help in identifying individuals at an increased risk of cancer.


Subject(s)
Carcinogens/adverse effects , Cell Transformation, Neoplastic , Genes, Tumor Suppressor , Genes, p53/genetics , Genetic Predisposition to Disease , Genetic Testing , Lung Neoplasms/genetics , Free Radical Scavengers/adverse effects , Humans , Lung Neoplasms/etiology , Nitric Oxide/adverse effects , Oxidative Stress , Point Mutation , Risk Assessment , Skin Neoplasms/etiology , Skin Neoplasms/genetics , Smoking/adverse effects
16.
Cancer Res ; 61(20): 7430-4, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11606376

ABSTRACT

Recent molecular epidemiological studies have identified polymorphisms in the XPD gene that are associated with increased risk of brain gliomas and head, neck, lung, and skin cancers. However, the functional significance of these polymorphic variants in altering cell processes such as cell cycle checkpoints, DNA repair, and apoptosis is uncertain. We have cloned the XPD variants Lys751Gln, Asp312Asn, and Lys751Gln-Asp312Asn into a pcDNA-3.1-expression vector. Using these constructs, we did not find any detectable difference in either in vitro binding with wild-type p53 or in DNA repair proficiency as measured by host cell reactivation assay. We then genotyped 34 different lymphoblastoid cell lines from six Centre d'Etude du Polymorphisme Humaine (CEPH)/Utah pedigree families and a CEPH/French pedigree family for polymorphisms at codons 751 and 312 and assessed their apoptotic response after either UV or ionized radiation exposure. The lymphoblastoid cell lines with homozygous or heterozygous Asp at codon 312 have similar apoptotic rates, whereas cell lines with homozygous Asn at codon 312 showed a 2.5-fold increased response to UV (P = 0.005; Student's t test). This is the first report known to us of a functional polymorphism in a gene involved in DNA damage-induced apoptosis. However, the presence of Lys or Gln at codon 751 did not influence the apoptotic response to UV. The diminished apoptotic response of cells containing the 312 Asp allele could both allow the survival and selective clonal expansion of carcinogen-damaged cells and be a mechanistic explanation for the increased risk of cancer at diverse tissue sites.


Subject(s)
Apoptosis/genetics , DNA Helicases , DNA-Binding Proteins , Polymorphism, Genetic , Proteins/genetics , Transcription Factors , Amino Acid Substitution/physiology , Apoptosis/radiation effects , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/pathology , Cell Line , Codon/genetics , DNA Repair , Humans , Lymphocytes/pathology , Lymphocytes/physiology , Protein Binding , Proteins/metabolism , Proteins/physiology , Tumor Suppressor Protein p53/metabolism , Xeroderma Pigmentosum Group D Protein
17.
Cancer Res ; 61(18): 6660-4, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11559530

ABSTRACT

The expression of genes involved in p53-mediated apoptosis was studied using cDNA microarray after treating isogenic cell lines with either ionizing radiation or doxorubicin. Most of the known p53 transcriptional activation target genes clustered in a functional category defined by early and p53-dependent induction, regardless of the type of stress. Apoptotic protease activating factor-1 (APAF-1) emerged from this analysis as a novel p53 target gene. Genomic sequences upstream of the APAF-1 transcription start site contain a classic p53-responsive element that bound to p53. Consistently, p53 directly induced APAF-1 gene expression. Furthermore, DNA damage-mediated induction of APAF-1 mRNA and protein expression, accompanied by apoptosis, were strictly dependent on wild-type p53 function. These data are consistent with the hypothesis that APAF-1 is an essential downstream effector of p53-mediated apoptosis.


Subject(s)
Apoptosis/genetics , DNA Damage , Proteins/genetics , Tumor Suppressor Protein p53/genetics , Apoptotic Protease-Activating Factor 1 , Cell Line , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Profiling , Humans , Lymphocytes/drug effects , Lymphocytes/physiology , Lymphocytes/radiation effects , Multigene Family , Oligonucleotide Array Sequence Analysis , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Transcriptional Activation/radiation effects , Tumor Cells, Cultured
18.
Cancer Res ; 61(17): 6350-5, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11522624

ABSTRACT

p53 mutations are common in lung cancer. In smoking-associated lung cancer,the occurrence of G:C to T:A transversions at hotspot codons, e.g., 157, 248, 249,and 273, has been linked to the presence of carcinogenic chemicalsin tobacco smoke including polycyclic aromatic hydrocarbons suchas benzo(a)pyrene (BP). In the present study, we have used a highly sensitive mutation assay to determine the p53 mutation load in nontumorous human lung and to study the mutability of p53 codons 157, 248, 249, and 250 to benzo(a)pyrene-diol-epoxide (BPDE), an active metabolite of BP in human bronchial epithelial BEAS-2B cells. We determined the p53 mutational load at codons 157, 248, 249, and 250 in nontumorous peripheral lung tissue either from lung cancer cases among smokers or noncancer controls among smokers and nonsmokers. A 5-25-fold higher frequency of GTC(val) to TTC(phe) transversions at codon 157 was found in nontumorous samples (57%) from cancer cases (n = 14) when compared with noncancer controls (n = 8; P < 0.01). Fifty percent (7/14) of the nontumorous samples from lung cancer cases showed a high frequency of codon 249 AGG(arg) to AGT(ser) mutations (P < 0.02). Four of these seven samples with AGT(ser) mutations also showed a high frequency of codon 249 AGG(arg) to ATG(met) mutations, whereas only one sample showed a codon 250 CCC to ACC transversion. Tumor tissue from these lung cancer cases (38%) contained p53 mutations but were different from the above mutations found in the nontumorous pair. Noncancer control samples from smokers or nonsmokers did not contain any detectable mutations at codons 248, 249, or 250. BEAS-2B bronchial epithelial cells exposed to doses of 0.125, 0.5, and 1.0 microM BPDE, showed G:C to T:A transversions at codon 157 at a frequency of 3.5 x 10(-7), 4.4 x 10(-7), and 8.9 x 10(-7), respectively. No mutations at codon 157 were found in the DMSO-treated controls. These doses of BPDE induced higher frequencies, ranging from 4-12-fold, of G:C to T:A transversions at codon 248, G:C to T:A transversions and G:C to A:T transitions at codon 249, and C:G to T:A transitions at codon 250 when compared with the DMSO-treated controls. These data are consistent with the hypothesis that chemical carcinogens such as BP in cigarette smoke cause G:C to T:A transversions at p53 codons 157, 248, and 249 and that nontumorous lung tissues from smokers with lung cancer carry a high p53 mutational load at these codons.


Subject(s)
7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/toxicity , Genes, p53/drug effects , Genes, p53/genetics , Lung/drug effects , Mutagenesis, Site-Directed/genetics , Mutagens/toxicity , Adolescent , Adult , Aged , Carcinogens/toxicity , Cells, Cultured , Child , Child, Preschool , Codon/drug effects , Codon/genetics , Humans , Infant , Lung/physiology , Lung Neoplasms/chemically induced , Lung Neoplasms/genetics , Middle Aged , Mutation , Smoking/adverse effects , Smoking/genetics
19.
Proc Natl Acad Sci U S A ; 98(17): 9671-6, 2001 Aug 14.
Article in English | MEDLINE | ID: mdl-11481424

ABSTRACT

The p33ING1 protein is a regulator of cell cycle, senescence, and apoptosis. Three alternatively spliced transcripts of p33ING1 encode p47ING1a, p33ING1b, and p24ING1c. We cloned an additional ING family member, p33ING2/ING1L. Unlike p33ING1b, p33ING2 is induced by the DNA-damaging agents etoposide and neocarzinostatin. p33ING1b and p33ING2 negatively regulate cell growth and survival in a p53-dependent manner through induction of G(1)-phase cell-cycle arrest and apoptosis. p33ING2 strongly enhances the transcriptional-transactivation activity of p53. Furthermore, p33ING2 expression increases the acetylation of p53 at Lys-382. Taken together, p33ING2 is a DNA damage-inducible gene that negatively regulates cell proliferation through activation of p53 by enhancing its acetylation.


Subject(s)
Apoptosis/physiology , DNA Damage , Homeodomain Proteins/physiology , Protein Processing, Post-Translational , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins , Acetylation , Amino Acid Sequence , Apoptosis/drug effects , Bleomycin/pharmacology , Cell Division , Cells, Cultured/drug effects , Cells, Cultured/radiation effects , Cisplatin/pharmacology , Cloning, Molecular , Doxorubicin/pharmacology , Etoposide/pharmacology , G1 Phase , Gamma Rays , Gene Expression Regulation/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/isolation & purification , Humans , Molecular Sequence Data , Receptors, Cytoplasmic and Nuclear , Transcription, Genetic , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/radiation effects , Tumor Stem Cell Assay , Zinostatin/pharmacology
20.
Oncogene ; 20(28): 3620-8, 2001 Jun 21.
Article in English | MEDLINE | ID: mdl-11439325

ABSTRACT

Chronic hepatitis B virus (HBV) infection and the integration of its X gene (HBx) are closely associated with the development of hepatocellular carcinoma (HCC). The integrated HBx frequently is truncated or contains point mutations. Previous studies indicated that these HBx mutants have a diminished co-transactivational activity. We have compared the effects of wild-type (wt) HBx and its naturally occurring mutants derived from human HCCs on transcriptional co-transactivation, apoptosis and interactive effects with p53. We demonstrated that overexpression of mutant, but not wt HBx, is defective in transcriptional co-transactivation of the NF-kappaB-driven luciferase reporter. By using a microinjection technique, the HBx mutants were shown to have an attenuated pro-apoptotic activity. This deficiency may be attributed to multiple mutations in the co-transactivation domain of HBx, that leads to decreased stability of the translated product. However, wt or mutant HBx bind to p53 in vitro and retain their ability to block p53-mediated apoptosis in vivo, which has been implicated as its major tumor suppressor function. The abrogation of p53-mediated apoptosis by integrated HBx mutants may provide a selective clonal advantage for preneoplastic or neoplastic hepatocytes and contribute to hepatocellular carcinogenesis.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular/virology , Hepatitis B virus/metabolism , Hepatitis B, Chronic/virology , Liver Neoplasms/virology , Trans-Activators/metabolism , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Genes, Reporter , Glutathione Transferase/metabolism , Hepatitis B virus/genetics , Humans , Luciferases/genetics , Molecular Sequence Data , Mutation , NF-kappa B/metabolism , Protein Biosynthesis , Trans-Activators/genetics , Transcriptional Activation , Viral Regulatory and Accessory Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...