Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
2.
Nat Cancer ; 5(4): 546-556, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38654103

ABSTRACT

The circadian clock regulates daily rhythms of numerous physiological activities through tightly coordinated modulation of gene expression and biochemical functions. Circadian disruption is associated with enhanced tumor formation and metastasis via dysregulation of key biological processes and modulation of cancer stem cells (CSCs) and their specialized microenvironment. Here, we review how the circadian clock influences CSCs and their local tumor niches in the context of different stages of tumor metastasis. Identifying circadian therapeutic targets could facilitate the development of new treatments that leverage circadian modulation to ablate tumor-resident CSCs, inhibit tumor metastasis and enhance response to current therapies.


Subject(s)
Circadian Clocks , Circadian Rhythm , Neoplasm Metastasis , Neoplasms , Neoplastic Stem Cells , Tumor Microenvironment , Humans , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/metabolism , Circadian Rhythm/physiology , Circadian Clocks/physiology , Animals , Neoplasms/pathology , Gene Expression Regulation, Neoplastic
3.
Lab Invest ; 104(3): 100303, 2024 03.
Article in English | MEDLINE | ID: mdl-38103870

ABSTRACT

Triple-negative breast cancer (TNBC) has a poor prognosis with limited therapeutic options available for affected patients. Efforts are ongoing to identify surrogate markers for tumor-specific CD8+ T cells that can predict the response to immune checkpoint inhibitor (ICI) therapies, such as programmed cell death protein 1 or programmed cell death ligand-1 blockade. We have previously identified tumor-specific CD39+CD8+ T cells in non-small cell lung cancer that might help predict patient responses to programmed cell death protein 1 or programmed cell death ligand-1 blockade. Based on this finding, we conducted a comparative interrogation of TNBC in an Asian cohort to evaluate the potential of CD39 as a surrogate marker of tumor-specific CD8+ T cells. Using ICI-treated TNBC mouse models (n = 24), flow cytometric analyses of peripheral blood mononuclear cells and tumor-infiltrating lymphocytes revealed that >99% of tumor-specific CD8+ T cells also expressed CD39. To investigate the relationship between CD39+CD8+ T-cell density and CD39 expression with disease prognosis, we performed multiplex immunohistochemistry staining on treatment-naive human TNBC tissues (n = 315). We saw that the proportion of CD39+CD8+ T cells in human TNBC tumors correlated with improved overall survival, as did the densities of other CD39+ immune cell infiltrates, such as CD39+CD68+ macrophages. Finally, increased CD39 expression on CD8+ T cells was also found to predict the response to ICI therapy (pembrolizumab) in a separate cohort of 11 TNBC patients. These findings support the potential of CD39+CD8+ T-cell density as a prognostic factor in Asian TNBC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Triple Negative Breast Neoplasms , Humans , Female , Animals , Mice , CD8-Positive T-Lymphocytes , Prognosis , Triple Negative Breast Neoplasms/metabolism , Programmed Cell Death 1 Receptor/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Leukocytes, Mononuclear/metabolism , Ligands , Lung Neoplasms/metabolism , Biomarkers/metabolism , Lymphocytes, Tumor-Infiltrating , B7-H1 Antigen/metabolism
4.
Proc Natl Acad Sci U S A ; 120(13): e2219978120, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36940336

ABSTRACT

We have previously shown that proteasome inhibitor bortezomib stabilizes p53 in stem and progenitor cells within gastrointestinal tissues. Here, we characterize the effect of bortezomib treatment on primary and secondary lymphoid tissues in mice. We find that bortezomib stabilizes p53 in significant fractions of hematopoietic stem and progenitor cells in the bone marrow, including common lymphoid and myeloid progenitors, granulocyte-monocyte progenitors, and dendritic cell progenitors. The stabilization of p53 is also observed in multipotent progenitors and hematopoietic stem cells, albeit at lower frequencies. In the thymus, bortezomib stabilizes p53 in CD4-CD8- T cells. Although there is less p53 stabilization in secondary lymphoid organs, cells in the germinal center of the spleen and Peyer's patch accumulate p53 in response to bortezomib. Bortezomib induces the upregulation of p53 target genes and p53 dependent/independent apoptosis in the bone marrow and thymus, suggesting that cells in these organs are robustly affected by proteasome inhibition. Comparative analysis of cell percentages in the bone marrow indicates expanded stem and multipotent progenitor pools in p53R172H mutant mice compared with p53 wild-type mice, suggesting a critical role for p53 in regulating the development and maturation of hematopoietic cells in the bone marrow. We propose that progenitors along the hematopoietic differentiation pathway express relatively high levels of p53 protein, which under steady-state conditions is constantly degraded by Mdm2 E3 ligase; however, these cells rapidly respond to stress to regulate stem cell renewal and consequently maintain the genomic integrity of hematopoietic stem/progenitor cell populations.


Subject(s)
Proteasome Inhibitors , Tumor Suppressor Protein p53 , Mice , Animals , Bortezomib/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/metabolism , Hematopoietic Stem Cells/metabolism , Myeloid Progenitor Cells/metabolism , Mice, Inbred C57BL
5.
Front Pharmacol ; 13: 1013740, 2022.
Article in English | MEDLINE | ID: mdl-36330092

ABSTRACT

Cardiovascular diseases are the leading cause of global mortality, in which myocardial infarction accounts for 46% of total deaths. Although good progress has been achieved in medication and interventional techniques, a proven method to repair the damaged myocardium has not yet been determined. Stem cell therapy for damaged myocardial repair has evolved into a promising treatment for ischemic heart disease. However, low retention and poor survival of the injected stem cells are the major obstacles to achieving the intended therapeutic effects. Chinese botanical and other natural drug substances are a rich source of effective treatment for various diseases. As such, numerous studies have revealed the role of Chinese medicine in stem cell therapy for myocardial infarction treatment, including promoting proliferation, survival, migration, angiogenesis, and differentiation of stem cells. Here, we discuss the potential and limitations of stem cell therapy, as well as the regulatory mechanism of Chinese medicines underlying stem cell therapy. We focus on the evidence from pre-clinical trials and clinical practices, and based on traditional Chinese medicine theories, we further summarize the mechanisms of Chinese medicine treatment in stem cell therapy by the commonly used prescriptions. Despite the pre-clinical evidence showing that traditional Chinese medicine is helpful in stem cell therapy, there are still some limitations of traditional Chinese medicine therapy. We also systematically assess the detailed experimental design and reliability of included pharmacological research in our review. Strictly controlled animal models with multi-perspective pharmacokinetic profiles and high-grade clinical evidence with multi-disciplinary efforts are highly demanded in the future.

7.
J Interv Card Electrophysiol ; 60(3): 535-542, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32430761

ABSTRACT

PURPOSE: This study evaluated the efficacy and safety of transcatheter radiofrequency ablation (RFCA) in treating ventricular premature contractions (PVCs) in children, summarized the countermeasures during intraoperative ventricular fibrillation (VF), and improved the safety of ventricular premature treatment. METHODS: A retrospective analysis was conducted on 75 children with PVCs who received RFCA in the Second Affiliated Hospital of Wenzhou Medical University from January 2010 to April 2019. Data including age, sex, body weight, ejection fraction, left ventricular end diastolic diameter, burden and number of PVCs/24 h, origin of PVCs, and its complications were collected. Paired t test was used to compare changes in cardiac function before and after surgery. RESULTS: Among the 75 cases treated with RFCA, 68 were successfully ablated, giving a success rate of 90.67%. After ablation, the left ventricular ejection fraction (LVEF) of the children was 69.13 ± 3.81%, which was significantly higher than that before surgery (69.13 ± 3.81% vs. 66.21 ± 3.22%, P = 0.012). One of the patients experienced VF during the operation, with no other complications. The initial locus of origin was the anterior septum of the right ventricular outflow tract, but VF occurred during the ablation process. Mean follow-up time was 39 ± 33 months, with two recurrent cases (2.94%). CONCLUSIONS: Performing RFCA in children is safe and effective, with a low recurrence rate and few complications. VF is not an indication to cease surgery; the key to eliminating complications is repositioning the catheter and finding a more accurate origin point.


Subject(s)
Catheter Ablation , Ventricular Premature Complexes , Child , Humans , Retrospective Studies , Stroke Volume , Treatment Outcome , Ventricular Function, Left , Ventricular Premature Complexes/diagnostic imaging , Ventricular Premature Complexes/surgery
8.
Chem Sci ; 10(26): 6457-6466, 2019 Jul 14.
Article in English | MEDLINE | ID: mdl-31316744

ABSTRACT

All-hydrocarbon, i, i+7 stapled peptide inhibitors of the p53-Mdm2 interaction have emerged as promising new leads for cancer therapy. Typical chemical synthesis via olefin metathesis results in the formation of both E- and Z-isomers, an observation that is rarely disclosed but may be of importance in targeting PPI. In this study, we evaluated the effect of staple geometry on the biological activity of five p53-reactivating peptides. We also present strategies for the modulation of the E/Z ratio and attainment of the hydrogenated adduct through repurposing of the metathesis catalyst.

9.
J Pathol ; 249(3): 274-285, 2019 11.
Article in English | MEDLINE | ID: mdl-31322742

ABSTRACT

Genomic alterations in different types of cancer have been identified by comprehensive sequencing methodologies, revealing TP53 as the most frequently mutated gene across the majority of human cancer types. Cytotoxic treatments are still major cancer therapy strategies but cancer recurrence due to therapy resistance is a major challenge. Resistant cell populations may be associated with TP53 mutant clones exhibiting abnormal p53 expression patterns in tumours. Given data that levels of mutant p53 influence cancer cell growth and survival, understanding the mechanisms underlying intratumour heterogeneity of p53 can be exploited to design strategies that improve patient survival. The patterns of p53 protein examined by immunohistochemistry of both premalignant and malignant tissues are complex, ranging from intense staining of all tumour cell nuclei to complete absence of staining and with many intermediate phenotypes. Animal models that express only mutant proteins and adoption of international standards for terminology have brought greater clarity to understanding the causes of variation and are at the same time demonstrating the utility of p53 in oncology. In addition to p53 mutation, MDM2 and chaperone activities, gene copy number and TP53 mRNA levels linked to proliferative activity and differentiation are all now established as causes of variation in p53 staining, with clinical implications. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Biomarkers, Tumor/metabolism , Immunohistochemistry , Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Biomarkers, Tumor/genetics , Cell Differentiation , Cell Proliferation , Gene Dosage , Gene Expression Regulation, Neoplastic , Humans , Mutation , Neoplasms/genetics , Neoplasms/pathology , Predictive Value of Tests , Protein Stability , Reproducibility of Results , Signal Transduction , Tumor Suppressor Protein p53/genetics
10.
Cancer Res ; 79(14): 3595-3607, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31138526

ABSTRACT

p53 protein, activated and stabilized by posttranslational modifications, performs its major functions by inducing DNA repair, cell-cycle arrest, or apoptosis through transcriptional activation. Here, we determined the ability of p53 protein stabilized via proteasome inhibition to perform similar functions as p53 induced by stresses such as DNA damage. Treating mice with the proteasome inhibitor bortezomib stabilized p53 in stem/progenitor cells of the intestine and stomach, in other proliferating tissues, and in intestinal tumors. Robust basal p53 mRNA levels were observed in the same compartments where p53 was stabilized. Spatial activation of p53 target genes in response to bortezomib in the small intestine demonstrated that CDKN1A and BAX were upregulated in the proliferative crypts but not in the differentiated villi of the small intestine; PUMA was specifically activated at the crypt base of p53 wild-type mice. Thus, cellular context determines the p53 transcriptional target selection. p53-dependent apoptosis was induced in Lgr5-expressing stem cells of the small intestine and high p53 transcriptional activity and apoptosis was induced in intestinal adenomas and in xenograft tumors. Bortezomib inhibited the growth of intestinal adenomas and xenograft tumors with wild-type p53, indicating the importance of p53 in the response to proteasome inhibitors in tissue homeostasis and in cancer therapy. SIGNIFICANCE: These findings show that bortezomib is less active in p53-defective tumors, yet its success in treating multiple myeloma suggests its use can be extended to p53-proficient solid tumors.


Subject(s)
Bortezomib/pharmacology , Colorectal Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Adenoma/drug therapy , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , HCT116 Cells , HT29 Cells , Humans , Intestine, Small/drug effects , Intestine, Small/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Stem Cells/drug effects , Stem Cells/metabolism , Tumor Suppressor Protein p53/genetics , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
11.
Cell Rep ; 22(1): 299-312, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29298430

ABSTRACT

The large number of mutations identified across all cancers represents an untapped reservoir of targets that can be useful for therapeutic targeting if highly selective, mutation-specific reagents are available. We report here our attempt to generate such reagents: monoclonal antibodies against the most common R175H, R248Q, and R273H hotspot mutants of the tumor suppressor p53. These antibodies recognize their intended specific alterations without any cross-reactivity against wild-type (WT) p53 or other p53 mutants, including at the same position (as exemplified by anti-R248Q antibody, which does not recognize the R248W mutation), evaluated by direct immunoblotting, immunoprecipitation, and immunofluorescence methods on transfected and endogenous proteins. Moreover, their clinical utility to diagnose the presence of specific p53 mutants in human tumor microarrays by immunohistochemistry is also shown. Together, the data demonstrate that antibodies against specific single-amino-acid alterations can be generated reproducibly and highlight their utility, which could potentially be extended to therapeutic settings.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/chemistry , Antibody Specificity , Mutation , Tumor Suppressor Protein p53/immunology , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Female , Humans , Mice , Mice, Inbred BALB C , Precision Medicine , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
12.
PLoS Pathog ; 12(12): e1006039, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27918748

ABSTRACT

The productive human papillomavirus (HPV) life cycle is tightly linked to the differentiation and cycling of keratinocytes. Deregulation of these processes and stimulation of cell proliferation by the action of viral oncoproteins and host cell factors underlies HPV-mediated carcinogenesis. Severe HPV infections characterize the wart, hypogammaglobulinemia, infection, and myelokathexis (WHIM) immunodeficiency syndrome, which is caused by gain-of-function mutations in the CXCR4 receptor for the CXCL12 chemokine, one of which is CXCR41013. We investigated whether CXCR41013 interferes in the HPV18 life cycle in epithelial organotypic cultures. Expression of CXCR41013 promoted stabilization of HPV oncoproteins, thus disturbing cell cycle progression and proliferation at the expense of the ordered expression of the viral genes required for virus production. Conversely, blocking CXCR41013 function restored virus production and limited HPV-induced carcinogenesis. Thus, CXCR4 and its potential activation by genetic alterations in the course of the carcinogenic process can be considered as an important host factor for HPV carcinogenesis.


Subject(s)
Cell Transformation, Viral/physiology , Chemokine CXCL12/metabolism , Papillomavirus Infections/genetics , Receptors, CXCR4/genetics , Signal Transduction , Skin Neoplasms/virology , Animals , Blotting, Western , Cell Line , Chemokine CXCL12/genetics , Genetic Predisposition to Disease/genetics , Heterografts , Human papillomavirus 18 , Keratinocytes/metabolism , Keratinocytes/virology , Mice , Mice, Nude , Receptors, CXCR4/metabolism , Signal Transduction/physiology , Skin Neoplasms/genetics
13.
Protein Eng Des Sel ; 29(1): 11-21, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26508747

ABSTRACT

The immunohistochemical (IHC) staining of mouse tissue sections using antibodies of mouse origin can result in high nonspecific background due to the staining of endogenous immunoglobulins (Igs) by enzyme-conjugated secondary antibodies. In order to obviate this issue, we developed a chimeric mouse-human anti-p53 monoclonal antibody (MH242) by grafting the variable regions of a known mouse antibody into a human Ig scaffold. This facilitated use of an anti-human secondary antibody, and resulted in near-zero background when compared with its parental mouse monoclonal antibody (PAb242). Furthermore, the chimeric antibody enabled reproducible detection of mutant p53 (homozygous R172H) expression in mouse tissue, an observation hitherto largely equivocal based on the use of existing antibodies. The approach we describe leads to the generation of tractable antibody reagents, whose integrity can be readily verified through DNA sequencing of expressor plasmids. The wide-spread adoption of such 'digitized' antibodies should reduce experimental disparities that can commonly arise through variations in antibody quality.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunohistochemistry/methods , Molecular Imaging/methods , Recombinant Fusion Proteins/chemistry , Animals , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Humans , Immunoglobulin G/genetics , Intestines/chemistry , Mice , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/immunology
14.
Oncotarget ; 6(33): 34979-91, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26474276

ABSTRACT

Cervical intraepithelial neoplasia (CIN) is caused by human papillomavirus (HPV) infection and is the precursor to cervical carcinoma. The completion of the HPV productive life cycle depends on the expression of viral proteins which further determines the severity of the cervical neoplasia. Initiation of the viral productive replication requires expression of the E2 viral protein that cooperates with the E1 viral DNA helicase. A decrease in the viral DNA replication ability and increase in the severity of cervical neoplasia is accompanied by simultaneous elevated expression of E6 and E7 oncoproteins. Here we reveal a novel and important role for the HPV16-E2 protein in controlling host cell cycle during malignant transformation. We showed that cells expressing HPV16-E2 in vitro are arrested in prophase alongside activation of a sustained DDR signal. We uncovered evidence that HPV16-E2 protein is present in vivo in cells that express both mitotic and DDR signals specifically in CIN3 lesions, immediate precursors of cancer, suggesting that E2 may be one of the drivers of genomic instability and carcinogenesis in vivo.


Subject(s)
Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/metabolism , Oncogene Proteins, Viral/metabolism , Precancerous Conditions/virology , Uterine Cervical Dysplasia/virology , Uterine Cervical Neoplasms/virology , Blotting, Western , Cell Cycle Checkpoints , DNA Damage/physiology , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization , Papillomavirus Infections/complications , Papillomavirus Infections/virology , Precancerous Conditions/pathology , Prophase , Uterine Cervical Neoplasms/pathology , Uterine Cervical Dysplasia/pathology
15.
Oncotarget ; 6(20): 17968-80, 2015 Jul 20.
Article in English | MEDLINE | ID: mdl-26255629

ABSTRACT

The tumour suppressor p53 is regulated primarily at the protein level. In normal tissues its levels are maintained at a very low level by the action of specific E3 ligases and the ubiquitin proteosome pathway. The mutant p53 protein contributes to transformation, metastasis and drug resistance. High levels of mutant p53 can be found in tumours and the accumulation of mutant p53 has previously been reported in pathologically normal cells in human skin. We show for the first time that similarly elevated levels of mutant p53 can be detected in apparently normal cells in a mutant p53 knock-in mouse model. In fact, in the small intestine, mutant p53 spontaneously accumulates in a manner dependent on gene dosage and cell type. Mutant p53 protein is regulated similarly to wild type p53, which can accumulate rapidly after induction by ionising radiation or Mdm2 inhibitors, however, the clearance of mutant p53 protein is much slower than wild type p53. The accumulation of the protein in the murine small intestine is limited to the cycling, crypt base columnar cells and proliferative zone and is lost as the cells differentiate and exit the cell cycle. Loss of Mdm2 results in even higher levels of p53 expression but p53 is still restricted to proliferating cells in the small intestine. Therefore, the small intestine of these p53 mutant mice is an experimental system in which we can dissect the molecular pathways leading to p53 accumulation, which has important implications for cancer prevention and therapy.


Subject(s)
Cell Cycle , Cell Proliferation , Intestine, Small/metabolism , Mutation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Age Factors , Animals , Cell Differentiation , DNA Damage , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Genotype , Intestine, Small/diagnostic imaging , Intestine, Small/drug effects , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/metabolism , Radiography , Time Factors , Tissue Culture Techniques
16.
Virology ; 462-463: 14-24, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25092457

ABSTRACT

Two classes of Human papillomaviruses (HPV) infect the anogenital track: high risk viruses that are associated with risk of cervical cancer and low risk types that drive development of benign lesions, such as condylomas. In the present study, we established quantitative transcriptional maps of the viral genome in clinical lesions associated with high risk HPV16 or low risk HPV6b. Marked qualitative and quantitative changes in the HPV16 transcriptome were associated with progression from low to high grade lesions. Specific transcripts encoding essential regulatory proteins such as E7, E2, E1^E4 and E5 were identified. We also identified intrinsic differences between the HPV6b-associated condyloma transcript map and that of the HPV16-associated low grade CIN specifically regarding promoter usage. Characterization and quantification of HPV transcripts in patient samples thus establish the impact of viral transcriptional regulation on the status of HPV-associated lesions and may therefore help in defining new biologically-relevant prognosis markers.


Subject(s)
Cell Transformation, Viral , Gene Expression Profiling , Gene Expression Regulation, Viral , Papillomaviridae/physiology , Papillomavirus Infections/virology , Humans , Papillomaviridae/genetics
17.
Open Virol J ; 6: 163-72, 2012.
Article in English | MEDLINE | ID: mdl-23341852

ABSTRACT

Integration of the viral DNA in the cellular genome has been suggested to be critical in carcinogenic progression of HPV-associated cervical neoplasia. This event can be accompanied by disruption of the open reading frame (ORF) encoding the E2 repressor, thus leading to transcriptional up-regulation of the E6 and E7 viral oncogenes. At this stage, it is unclear whether disruption of the E2 ORF is mandatory for carcinogenic progression. We measured E2 RNA and protein expression in clinical samples of various grades of HPV16-associated cervical neoplasia and compared it with the status of the viral genome. RNA extracted from paraffin embedded tissues was hybridized to specific probes and quantified by the NanoString technology. Protein expression was appreciated by immunohistochemistry and the status of viral DNA was determined by in situ hybridization, all performed on serial sections of the same samples. E2 protein was found highly expressed in CIN1, CIN2 lesions where the HPV DNA was highly replicative, while it was decreased in more advanced grade lesions where replication is decreased or lost (CIN3 and SCC). In contrast, E2 transcripts could be elevated even in conditions of no or low expression of the protein, as found in the Caski cell line. Our data demonstrate that integration of the viral DNA in the cellular genome does not always lead to disruption of the E2 ORF and drastic reduction of E2 transcripts, while in contrast, expression of the E2 protein is always drastically reduced.

18.
Cancer Res ; 70(13): 5316-25, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20530671

ABSTRACT

The viral E2 gene product plays a crucial role in the human papillomavirus (HPV) vegetative cycle by regulating both transcription and replication of the viral genome. E2 is a transcriptional repressor of the E6 and E7 viral oncogenes for HPV types 16 and 18, which are involved in cervical cancers. Using new polyclonal antibodies against the HPV16 E2 protein, we showed that E2 is expressed at various precursor stages of cervical carcinoma by immunohistochemistry on paraffin-embedded clinical samples. E2 was found to be highly expressed in the nuclei and cytoplasm of cells forming the intermediate and upper layers of cervical intraepithelial neoplasia (CIN). We could show that the expressions of E2 and p16(INK4a) (surrogate marker for oncogenic E7 expression) were exclusive in most of the cases, thus implying that E2 is not expressed together with high levels of E7. Moreover, we found that E2 is expressed in a subset of columnar cells adjacent to the CIN. We could show that expression of E2 is topologically distinct from the proliferation markers p63 and Ki67, whereas it coincides with the expression of cytokeratin K13, a marker of squamous cell differentiation. Expression of E2 also topologically coincides with episomal amplification of viral genomes in the upper layers of CIN1. These in vivo data thus validate previous assumptions of the crucial role of E2 in the early steps of HPV infection and of its negative link with expression of the viral E6 and E7 oncogenes.


Subject(s)
Biomarkers, Tumor/analysis , DNA-Binding Proteins/analysis , Human papillomavirus 16/metabolism , Oncogene Proteins, Viral/analysis , Papillomavirus E7 Proteins/analysis , Papillomavirus Infections/virology , Uterine Cervical Dysplasia/virology , Uterine Cervical Neoplasms/virology , Antibodies/chemistry , Antibody Specificity , DNA Replication , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/immunology , Female , Human papillomavirus 16/genetics , Human papillomavirus 16/isolation & purification , Humans , Immunohistochemistry , Oncogene Proteins, Viral/biosynthesis , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins/biosynthesis , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/diagnosis , Paraffin Embedding , Uterine Cervical Neoplasms/diagnosis , Virus Replication , Uterine Cervical Dysplasia/diagnosis
19.
Eur J Obstet Gynecol Reprod Biol ; 145(2): 214-8, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19525056

ABSTRACT

OBJECTIVE: The purpose of the present study was to estimate the prevalence of high-risk human papillomavirus infection and the viral load in different age groups, to describe the distribution of human papillomavirus prevalence in women enrolled in two hospitals. STUDY DESIGN: We retrospectively investigated 17,148 cases undergoing hybrid capture II between January 2005 and February 2007. The prevalence of human papillomavirus infection and the level of viral load were estimated in different age groups to describe the distribution in the cases. RESULTS: Human papillomavirus was detected in 5173 of 17,148 women (30.2%) aged 17-79 years. The highest prevalence appeared in the or=61-year group. The mean viral load was 294.12+/-511.66 relative light units/positive control in total human papillomavirus positive cases. The viral load of the 21-30-year age group was the lowest (271.99+/-499.24 relative light units/positive control) and the highest was found in the >or=61-year interval (560.30+/-672.87 relative light units/positive control). No significant correlation was found between viral load and the severity of cervical lesions. CONCLUSIONS: Our study showed a "U" shape of age-specific prevalence of high-risk human papillomavirus infection occurring in women attending hospitals in Shanghai, China, similar to worldwide figures.


Subject(s)
Papillomavirus Infections/epidemiology , Uterine Cervical Dysplasia/epidemiology , Uterine Cervical Neoplasms/epidemiology , Adolescent , Adult , Age Factors , Aged , China/epidemiology , DNA, Viral/analysis , Female , Humans , Middle Aged , Papillomaviridae/genetics , Papillomavirus Infections/virology , Prevalence , Retrospective Studies , Uterine Cervical Neoplasms/virology , Uterine Cervical Dysplasia/virology
20.
Chin Med J (Engl) ; 121(16): 1578-82, 2008 Aug 20.
Article in English | MEDLINE | ID: mdl-18982872

ABSTRACT

BACKGROUND: Cervical cancer is the second most common cause of death from cancer among women worldwide. Human papillomavirus (HPV) plays a central role in the etiology of cervical cancer. It is important to describe the prevalence of HPV infection in different types of cervical lesions and to explore the relation between HPV viral load and the severity of cervical lesions. METHODS: To describe the HPV infection prevalence and viral load in different age groups, we retrospectively investigated 6405 cases of women who were organized by their units to take health-examination. They were given Hybrid Capture II tests between January 2005 and December 2006. The correlation between HPV viral load and pathology was assessed. RESULTS: Overall HPV infection prevalence was 29.1% (1864/6405), while in women 18-20 years old it was 54.4% (31/57), the highest among all age groups. After declining rapidly, HPV prevalence stabilized at about 30.0% in women aged 30 and older. Of the 6405 women, 1483 women had a colposcopic biopsy and 33.2% (492/1483) were positive for HPV DNA. Twenty-one percent of women with a normal diagnosis (238/1095) had HPV infection, a statistically significantly lower prevalence than in women with cervical lesions, including those with cervical intraepithelial neoplasia (68.8% in CIN1, 66.7% in CIN2, and 76.5% in CIN3) or with cervical cancer (94.1%). The correlation coefficient between viral load and cervical lesion severity was 0.134, which was not statistically significant (P = 0.075). Viral load values in women with CINs and cervical cancer were calculated, and no significant differences were identified. CONCLUSIONS: The prevalence of high-risk HPV infection among women attending hospitals for health-examination in Shanghai is similar to the worldwide rate. HPV viral load can distinguish cervical lesions from normal individuals but cannot adequately predict the severity of cervical lesions.


Subject(s)
Papillomavirus Infections/epidemiology , Uterine Cervical Dysplasia/virology , Uterine Cervical Neoplasms/virology , Adolescent , Adult , Aged , DNA, Viral/analysis , Female , Humans , Middle Aged , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL
...