Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Article in English | MEDLINE | ID: mdl-33436409

ABSTRACT

Long noncoding RNAs (lncRNAs) play diverse roles in biological processes, but their expression profiles and functions in cervical carcinogenesis remain unknown. By RNA-sequencing (RNA-seq) analyses of 18 clinical specimens and selective validation by RT-qPCR analyses of 72 clinical samples, we provide evidence that, relative to normal cervical tissues, 194 lncRNAs are differentially regulated in high-risk (HR)-HPV infection along with cervical lesion progression. One such lncRNA, lnc-FANCI-2, is extensively characterized because it is expressed from a genomic locus adjacent to the FANCI gene encoding an important DNA repair factor. Both genes are up-regulated in HPV lesions and in in vitro model systems of HR-HPV18 infection. We observe a moderate reciprocal regulation of lnc-FANCI-2 and FANCI in cervical cancer CaSki cells. In these cells, lnc-FANCI-2 is transcribed from two alternative promoters, alternatively spliced, and polyadenylated at one of two alternative poly(A) sites. About 10 copies of lnc-FANCI-2 per cell are detected preferentially in the cytoplasm. Mechanistically, HR-HPVs, but not low-risk (LR)-HPV oncogenes induce lnc-FANCI-2 in primary and immortalized human keratinocytes. The induction is mediated primarily by E7, and to a lesser extent by E6, mostly independent of p53/E6AP and pRb/E2F. We show that YY1 interacts with an E7 CR3 core motif and transactivates the promoter of lnc-FANCI-2 by binding to two critical YY1-binding motifs. Moreover, HPV18 increases YY1 expression by reducing miR-29a, which targets the 3' untranslated region of YY1 mRNA. These data have provided insights into the mechanisms of how HR-HPV infections contribute to cervical carcinogenesis.


Subject(s)
Fanconi Anemia Complementation Group Proteins/genetics , Human papillomavirus 16/genetics , MicroRNAs/genetics , Papillomavirus Infections/genetics , RNA, Long Noncoding/genetics , Uterine Cervical Neoplasms/genetics , YY1 Transcription Factor/genetics , Alternative Splicing , Base Sequence , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cervix Uteri/metabolism , Cervix Uteri/pathology , Cervix Uteri/virology , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Female , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Human papillomavirus 16/metabolism , Human papillomavirus 16/pathogenicity , Human papillomavirus 18/genetics , Human papillomavirus 18/metabolism , Human papillomavirus 18/pathogenicity , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Keratinocytes/virology , MicroRNAs/metabolism , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/metabolism , Papillomavirus Infections/metabolism , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Promoter Regions, Genetic , RNA, Long Noncoding/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , YY1 Transcription Factor/metabolism
3.
mBio ; 10(1)2019 01 29.
Article in English | MEDLINE | ID: mdl-30696738

ABSTRACT

RNA-binding proteins (RBPs) control mRNA processing, stability, transport, editing, and translation. We recently conducted transcriptome analyses comparing normal (i.e., healthy) cervical tissue samples with human papillomavirus (HPV)-positive cervical cancer tissue samples and identified 614 differentially expressed protein-coding transcripts which are enriched in cancer-related pathways and consist of 95 known RBPs. We verified the altered expression of 26 genes with a cohort of 72 cervical samples, including 24 normal cervical samples, 25 cervical intraepithelial neoplasia grade 2 (CIN2) and CIN3 samples, and 23 cervical cancer tissue samples. LY6K (lymphocyte antigen 6 complex locus K), FAM83A (family member with sequence similarity 83), CELSR3, ASF1B, IQGAP3, SEMA3F, CLDN10, MSX1, CXCL5, ASRGL1, ELAVL2, GRB7, KHSRP, NOVA1, PTBP1, and RNASEH2A were identified as novel candidate genes associated with cervical lesion progression and carcinogenesis. HPV16 or HPV18 infection was found to alter the expression of 8 RBP genes (CDKN2A, ELAVL2, GRB7, HSPB1, KHSRP, NOVA1, PTBP1, and RNASEH2A) in human vaginal and foreskin keratinocytes. Both viral E6 and E7 decreased NOVA1 expression, but only E7 increased the expression of RNASEH2A in an E2F1-dependent manner. Proliferating cell nuclear antigen (PCNA) directs RNASEH2 activity with respect to DNA replication by removing the RNA primers to promote Okazaki fragment maturation, and two factors are closely associated with neoplasia progression. Therefore, we predict that the induction of expression of RNASEH2A via viral E7 and E2F1 may promote DNA replication and cancer cell proliferation.IMPORTANCE High-risk HPV infections lead to development of cervical cancer. This study identified the differential expression of 16 novel genes (LY6K, FAM83A, CELSR3, ASF1B, IQGAP3, SEMA3F, CLDN10, MSX1, CXCL5, ASRGL1, ELAVL2, GRB7, KHSRP, NOVA1, PTBP1, and RNASEH2A) in HPV-infected cervical tissue samples and keratinocytes. Eight of these genes (CDKN2A, ELAVL2, GRB7, HSPB1, KHSRP, NOVA1, PTBP1, and RNASEH2A) encode RNA-binding proteins. Further studies indicated that both HPV16 and HPV18 infections lead to the aberrant expression of selected RBP-encoding genes. We found that viral E6 and E7 decrease NOVA1 expression but that E7 increases RNASEH2A expression via E2F1. The altered expression of these genes may be utilized as biomarkers for high-risk (HR)-HPV carcinogenesis and progression.


Subject(s)
E2F1 Transcription Factor/metabolism , Host-Pathogen Interactions , Human papillomavirus 16/physiology , Human papillomavirus 18/physiology , Papillomavirus E7 Proteins/metabolism , Ribonuclease H/metabolism , Uterine Cervical Neoplasms/pathology , Cells, Cultured , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Keratinocytes/pathology , Keratinocytes/virology , Papillomavirus Infections/pathology , Papillomavirus Infections/virology
6.
Vaccine ; 34(27): 3171-3177, 2016 06 08.
Article in English | MEDLINE | ID: mdl-27113165

ABSTRACT

The prevalent human papillomaviruses (HPVs) infect human epithelial tissues. Infections by the mucosotropic HPV genotypes cause hyperproliferative ano-genital lesions. Persistent infections by high-risk (HR) HPVs such as HPV-16, HPV-18 and related types can progress to high grade intraepithelial neoplasias and cancers. Prophylactic HPV vaccines are based on DNA-free virus-like particles (VLPs) composed of the major capsid protein L1 of HPV-16, -18, -6 and -11 (Gardasil) or HPV-16 and -18 (Cervarix). Sera from vaccinated animals effectively prevent HPV pseudovirions to infect cell lines and mouse cervical epithelia. Both vaccines have proven to be highly protective in people. HPV pseudovirions are assembled in HEK293TT cells from matched L1 and L2 capsid proteins to encapsidate a reporter gene. Pseudovirions and genuine virions have structural differences and they infect cell lines or primary human keratinocytes (PHKs) with different efficiencies. In this study, we show that sera and isolated IgG from women immunized with Gardasil prevent authentic HPV-18 virions from infecting PHKs, whereas non-immune sera and purified IgG thereof are uniformly ineffective. Using early passage PHKs, neutralization is achieved only if immune sera are added within 2-4h of infection. We attribute the timing effect to a conformational change in HPV virions, thought to occur upon initial binding to heparan sulfate proteoglycans (HSPG) on the cell surface. This interpretation is consistent with the inability of immune IgG bound to or taken up by PHKs to neutralize the virus. Interestingly, the window of neutralization increases to 12-16h in slow growing, late passage PHKs, suggestive of altered cell surface molecules. In vivo, this window might be further lengthened by the time required to activate the normally quiescent basal cells to become susceptible to infection. Our observations help explain the high efficacy of HPV vaccines.


Subject(s)
Antibodies, Viral/immunology , Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18/therapeutic use , Immune Sera/immunology , Keratinocytes/virology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Cells, Cultured , Female , Heparan Sulfate Proteoglycans/metabolism , Human papillomavirus 18 , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Neutralization Tests , Papillomavirus Infections/prevention & control
7.
Methods Mol Biol ; 1249: 93-109, 2015.
Article in English | MEDLINE | ID: mdl-25348300

ABSTRACT

The productive program of the human papillomaviruses takes place in terminally differentiating squamous epithelia. In this chapter, we provide the protocols for robust production of HPV-18 in organotypic cultures of early passages of primary human keratinocytes. A critical step is the generation of genomic HPV plasmids in vivo by using Cre-loxP-mediated excisional recombination from a vector plasmid. We discuss the rationale for this approach. This system produces high yields of infectious virus and facilitates genetic analyses of HPV protein functions and their regulation in the context of recapitulated host tissue environment.


Subject(s)
Cell Culture Techniques/methods , Human papillomavirus 18/growth & development , Keratinocytes/virology , Animals , Attachment Sites, Microbiological/genetics , Capsid Proteins/metabolism , Cell Separation , Cells, Cultured , DNA, Viral/genetics , DNA, Viral/isolation & purification , Genome, Viral , Humans , Mice , Plasmids/metabolism , Real-Time Polymerase Chain Reaction , Transfection , Virion/metabolism
8.
Diabetes ; 63(9): 2949-61, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24722250

ABSTRACT

Brown adipose tissue oxidizes chemical energy for heat generation and energy expenditure. Promoting brown-like transformation in white adipose tissue (WAT) is a promising strategy for combating obesity. Here, we find that targeted deletion of KH-type splicing regulatory protein (KSRP), an RNA-binding protein that regulates gene expression at multiple levels, causes a reduction in body adiposity. The expression of brown fat-selective genes is increased in subcutaneous/inguinal WAT (iWAT) of Ksrp(-/-) mice because of the elevated expression of PR domain containing 16 and peroxisome proliferator-activated receptor gamma coactivator 1α, which are key regulators promoting the brown fat gene program. The expression of microRNA (miR)-150 in iWAT is decreased due to impaired primary miR-150 processing in the absence of KSRP. We show that miR-150 directly targets and represses Prdm16 and Ppargc1a, and that forced expression of miR-150 attenuates the elevated expression of brown fat genes caused by KSRP deletion. This study reveals the in vivo function of KSRP in controlling brown-like transformation of iWAT through post-transcriptional regulation of miR-150 expression.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , MicroRNAs/biosynthesis , Trans-Activators/deficiency , Adiposity/genetics , Animals , DNA-Binding Proteins/biosynthesis , Diet, High-Fat , Down-Regulation , Gene Expression Regulation , Male , Mice , MicroRNAs/genetics , Obesity/genetics , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA-Binding Proteins/physiology , Trans-Activators/physiology , Transcription Factors/biosynthesis , Up-Regulation
9.
Proc Natl Acad Sci U S A ; 111(11): 4262-7, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24591631

ABSTRACT

Cellular and viral microRNAs (miRNAs) are the transcriptional products of RNA polymerase II and are regulated by transcriptional factors for their differential expression. The altered expression of miRNAs in many cancer types has been explored as a marker for possible diagnosis and therapy. We report in this study that oncogenic human papillomaviruses (HPVs) induce aberrant expression of many cellular miRNAs and that HPV18 infection produces no detectable viral miRNA. Thirteen abundant host miRNAs were specifically regulated by HPV16 and HPV18 in organotypic raft cultures of foreskin and vaginal keratinocytes as determined by miRNA array in combination with small RNA sequencing. The increase of miR-16, miR-25, miR-92a, and miR-378 and the decrease of miR-22, miR-27a, miR-29a, and miR-100 could be attributed to viral oncoprotein E6, E7, or both, all of which are known to target many cellular transcription factors. The examination of 158 cervical specimens, including 38 normal, 52 cervical intraepithelial neoplasia (CIN), and 68 cervical cancer (CC) tissues, for the expression of these eight miRNAs showed a remarkable increase of miR-25, miR-92a, and miR-378 with lesion progression but no obvious change of miR-22, miR-29a, and miR-100 among the HPV-infected tissues. Further analyses indicate that an expression ratio ≥1.5 of miR-25/92a group over miR-22/29a group could serve as a cutoff value to distinguish normal cervix from CIN and from CIN to CC.


Subject(s)
Biomarkers/metabolism , Human papillomavirus 16 , Human papillomavirus 18 , MicroRNAs/metabolism , Oncogenic Viruses/genetics , Papillomavirus Infections/genetics , Uterine Cervical Neoplasms/virology , Base Sequence , Blotting, Northern , DNA Primers/genetics , Female , Humans , MicroRNAs/genetics , Molecular Sequence Data , Oligonucleotides, Antisense/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA , Uterine Cervical Neoplasms/genetics
10.
Proc Natl Acad Sci U S A ; 110(19): 7542-9, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23572574

ABSTRACT

Human papillomaviruses (HPVs) amplify in differentiated strata of a squamous epithelium. The HPV E7 protein destabilizes the p130/retinoblastoma susceptibility protein family of tumor suppressors and reactivates S-phase reentry, thereby facilitating viral DNA amplification. The high-risk HPV E6 protein destabilizes the p53 tumor suppressor and many other host proteins. However, the critical E6 targets relevant to viral DNA amplification have not been identified, because functionally significant E6 mutants are not stably maintained in transfected cells. Using Cre-loxP recombination, which efficiently generates HPV genomic plasmids in transfected primary human keratinocytes, we have recapitulated a highly productive infection of HPV-18 in organotypic epithelial cultures. By using this system, we now report the characterization of four HPV-18 E6 mutations. An E6 null mutant accumulated high levels of p53 and amplified very poorly. p53 siRNA or ectopic WT E6 partially restored amplification, whereas three missense E6 mutations that did not effectively destabilize p53 complemented the null mutant poorly. Unexpectedly, in cis, two of the missense mutants amplified, albeit to a lower extent than the WT and only in cells with undetectable p53. These observations and others implicate p53 and additional host proteins in regulating viral DNA amplification and also suggest an inhibitory effect of E6 overexpression. We show that high levels of viral DNA amplification are critical for late protein expression and report several previously undescribed viral RNAs, including bicistronic transcripts predicted to encode E5 and L2 or an alternative form of E1^E4 and L1.


Subject(s)
DNA, Viral/metabolism , DNA-Binding Proteins/genetics , Genes, p53 , Human papillomavirus 18/genetics , Mutation , Oncogene Proteins, Viral/genetics , Cells, Cultured , Genes, Tumor Suppressor , Genetic Complementation Test , Genome, Viral , Humans , Integrases/metabolism , Keratinocytes/cytology , Mutation, Missense , Phenotype , Plasmids/metabolism , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/metabolism
11.
J Virol ; 87(11): 6127-36, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23536682

ABSTRACT

While the oncogenic human papillomavirus (HPV) types with the greatest medical impact are clustered within the α9 and α7 species, a significant fraction of cervical cancers are caused by α5, α6, and α11 viruses. Benign genital warts are caused principally by the α10 viruses HPV6 and HPV11. In an effort to achieve broad protection against both cervical cancer- and genital wart-associated types, we produced at high levels in bacteria a multimeric protein (α11-88x8) fusing eight polypeptides corresponding to a protective domain comprising L2 residues ∼11 to 88 derived from HPV6 (α10), HPV16 (α9), HPV18 (α7), HPV31 (α9), HPV39 (α7), HPV51 (α5), HPV56 (α6), and HPV73 (α11) and a truncated derivative with the last three units deleted (α11-88x5). Mice were immunized three times with α11-88x8 or α11-88x5 adjuvanted with alum or the licensed HPV vaccines and challenged intravaginally with HPV6, HPV16, HPV26, HPV31, HPV33, HPV35, HPV45, HPV51, HPV56, HPV58, or HPV59 pseudovirions. The α11-88x5 and α11-88x8 vaccines induced similarly robust protection against each HPV type tested and indistinguishable HPV16-neutralizing antibody titers. Passive transfer of α11-88x8 antisera was protective. Further, rabbit antisera to α11-88x8 and α11-88x5 similarly neutralized native HPV18 virions. These findings suggest that immunologic competition between units is not a significant issue and that it is not necessary to include a unit of L2 derived from each species to achieve broader protection against diverse medically significant HPV types than is achieved with the licensed HPV vaccines.


Subject(s)
Alphapapillomavirus/classification , Alphapapillomavirus/genetics , Capsid Proteins/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Phylogeny , Alphapapillomavirus/immunology , Animals , Antibodies, Viral/immunology , Capsid Proteins/administration & dosage , Capsid Proteins/genetics , Female , Genotype , Humans , Mice , Mice, Inbred BALB C , Papillomavirus Infections/immunology , Papillomavirus Vaccines/genetics , Papillomavirus Vaccines/immunology , Rabbits
12.
Hum Genomics ; 5(6): 569-76, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22155604

ABSTRACT

The genes encoding the enzymes for metabolising alcohol dehydrogenase 1B (ADH1B) and aldehyde dehydrogenase 2 (ALDH2) - exhibit genetic polymorphism and ethnic variations. Although the ALDH2*2 variant allele has been widely accepted as protecting against the development of alcoholism in Asians, the association of the ADH1B*2 variant allele with drinking behaviour remains inconclusive. The goal of this study was to determine whether the polymorphic ADH1B and ALDH2 genes are associated with stroke in male Han Chinese with high alcohol consumption. Sixty-five stroke patients with a history of heavy drinking (HDS) and 83 stroke patients without such a history (NHDS) were recruited for analysis of the ADH1B and ALDH2 genotypes from the stroke registry in the Tri-Service General Hospital, Taipei, Taiwan, between January 2000 and December 2001. The allelotypes of ADH1B and ALDH2 were determined using the polymerase chain reaction-restriction fragment length polymorphism method. The HDS patients (3 per cent) showed a significantly lower ALDH2*2 allele frequency than NHDS patients (27 per cent) (p < 0.001). After controlling for age, patients with HDS were associated with a significantly higher occurrence of cigarette smoking (p < 0.01) and liver dysfunction (p < 0.01). Multiple logistic regression analyses revealed that the ALDH2*2 variant allele was an independent variable exhibiting strong protection (odds ratio 0.072; 95 per cent confidence interval 0.02-0.26) against HDS after adjustment for hypertension, diabetes mellitus, smoking status and liver dysfunction. By contrast, allelic variations in ADH1B exerted no significant effect on HDS. The present study indicated that, unlike ALDH2*2, ADH1B*2 appears not to be a significant negative risk factor for high alcohol consumption in male Han Chinese with stroke.


Subject(s)
Alcohol Dehydrogenase/genetics , Alcohol Drinking/adverse effects , Aldehyde Dehydrogenase/genetics , Polymorphism, Genetic/genetics , Stroke/chemically induced , Stroke/genetics , Adult , Aged , Aged, 80 and over , Aldehyde Dehydrogenase, Mitochondrial , Case-Control Studies , China , Follow-Up Studies , Genotype , Humans , Male , Middle Aged , Prognosis , Risk Factors
13.
J Virol ; 85(16): 8080-92, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21680515

ABSTRACT

Human papillomavirus type 18 (HPV18) is the second most common oncogenic HPV genotype, responsible for ∼15% of cervical cancers worldwide. In this study, we constructed a full HPV18 transcription map using HPV18-infected raft tissues derived from primary human vaginal or foreskin keratinocytes. By using 5' rapid amplification of cDNA ends (RACE), we mapped two HPV18 transcription start sites (TSS) for early transcripts at nucleotide (nt) 55 and nt 102 and the HPV18 late TSS frequently at nt 811, 765, or 829 within the E7 open reading frame (ORF) of the virus genome. HPV18 polyadenylation cleavage sites for early and late transcripts were mapped to nt 4270 and mainly to nt 7299 or 7307, respectively, by using 3' RACE. Although all early transcripts were cleaved exclusively at a single cleavage site, HPV18 late transcripts displayed the heterogeneity of 3' ends, with multiple minor cleavage sites for late RNA polyadenylation. HPV18 splice sites/splice junctions for both early and late transcripts were identified by 5' RACE and primer walking techniques. Five 5' splice sites (donor sites) and six 3' splice sites (acceptor sites) that are highly conserved in other papillomaviruses were identified in the HPV18 genome. HPV18 L1 mRNA translates a L1 protein of 507 amino acids (aa), smaller than the 568 aa residues previously predicted. Collectively, a full HPV18 transcription map constructed from this report will lead us to further understand HPV18 gene expression and virus oncogenesis.


Subject(s)
Genome, Viral , Human papillomavirus 18/genetics , Keratinocytes/virology , Transcription, Genetic , Capsid Proteins/genetics , Cells, Cultured , DNA, Viral/genetics , Gene Expression , Humans , Open Reading Frames , Polyadenylation , RNA Splice Sites , RNA Splicing , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Initiation Site
14.
J Biol Chem ; 286(17): 15473-82, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21321122

ABSTRACT

The productive program of human papillomaviruses occurs in differentiated squamous keratinocytes. We have previously shown that HPV-18 DNA amplification initiates in spinous cells in organotypic cultures of primary human keratinocytes during prolonged G(2) phase, as signified by abundant cytoplasmic cyclin B1 (Wang, H. K., Duffy, A. A., Broker, T. R., and Chow, L. T. (2009) Genes Dev. 23, 181-194). In this study, we demonstrated that the E7 protein, which induces S phase reentry in suprabasal cells by destabilizing the p130 pocket protein (Genovese, N. J., Banerjee, N. S., Broker, T. R., and Chow, L. T. (2008) J. Virol. 82, 4862-4873), also elicited extensive G(2) responses. Western blots and indirect immunofluorescence assays were used to probe for host proteins known to control G(2)/M progression. E7 expression induced cytoplasmic accumulation of cyclin B1 and cdc2 in the suprabasal cells. The elevated cdc2 had inactivating phosphorylation on Thr(14) or Tyr(15), and possibly both, due to an increase in the responsible Wee1 and Myt1 kinases. In cells that harbored cytoplasmic cyclin B1 or cdc2, there was also an accumulation of the phosphatase-inactive cdc25C phosphorylated on Ser(216), unable to activate cdc2. Moreover, E7 expression induced elevated expression of phosphorylated ATM (Ser(1981)) and the downstream phosphorylated Chk1, Chk2, and JNKs, kinases known to inactivate cdc25C. Similar results were observed in primary human keratinocyte raft cultures in which the productive program of HPV-18 took place. Collectively, this study has revealed the mechanisms by which E7 induces prolonged G(2) phase in the differentiated cells following S phase induction.


Subject(s)
G2 Phase , Keratinocytes/pathology , Keratinocytes/virology , Papillomavirus E7 Proteins/physiology , S Phase , Cell Cycle Proteins/metabolism , Cell Differentiation , DNA-Binding Proteins/physiology , Humans , Oncogene Proteins, Viral/physiology , Phosphorylation
15.
Cell Cycle ; 8(9): 1319-23, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19342877

ABSTRACT

A simple, efficient system has been developed to produce high titers of infectious human papillomavirus type 18 (HPV-18) in organotypic raft cultures of primary human keratinocytes (PHKs). Molecular characterization elucidated key early and late events in the reproductive program. The system obviates the need for immortalized cells and allows the analyses of mutant HPV genomes not previously possible. An E6 deletion mutant incapable of causing p53 degradation is defective in viral DNA amplification and capsid protein production. The high levels of p53 protein which accumulated in numerous cells did not lead to apoptosis over a prolonged duration. Time course and metabolic labeling experiments revealed novel interactions with the host. Notably, post-mitotic, differentiated cells are induced by HPV E7 expression to reenter S phase, whereupon host chromosomes replicate, but HPV DNA does not amplify until the cells have progressed to and are arrested in G(2) phase. Here, we present data that strongly suggest that the abundant cytoplasmic viral E1;E4 protein is not responsible for this G(2) arrest, as described in the literature upon ectopic expression in cell lines. We provide additional insights into the viral life cycle and contrast them to conclusions derived from experiments in cell lines.


Subject(s)
Genetic Techniques , Host-Pathogen Interactions , Papillomaviridae/pathogenicity , DNA, Viral/metabolism , Human papillomavirus 18/physiology , Humans , Papillomaviridae/physiology , Viral Proteins/metabolism , Virion/ultrastructure , Virus Replication
16.
RNA ; 15(4): 637-47, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19258450

ABSTRACT

MicroRNAs (miRNA) play pivotal roles in controlling cell proliferation and differentiation. Aberrant miRNA expression in human is becoming recognized as a new molecular mechanism of carcinogenesis. However, the causes for alterations in miRNA expression remain largely unknown. Infection with oncogenic human papillomavirus types 16 (HPV16) and 18 (HPV18) can lead to cervical and other ano-genital cancers. Here, we have demonstrated that cervical cancer tissues and cervical cancer-derived cell lines containing oncogenic HPVs display reduced expression of tumor-suppressive miR-34a. The reduction of miR-34a expression in organotypic tissues derived from HPV-containing primary human keratinocytes correlates with the early productive phase and is attributed to the expression of viral E6, which destabilizes the tumor suppressor p53, a known miR-34a transactivator. Knockdown of viral E6 expression in HPV16(+) and HPV18(+) cervical cancer cell lines by siRNAs leads to an increased expression of p53 and miR-34a and accumulation of miR-34a in G(0)/G(1) phase cells. Ectopic expression of miR-34a in HPV18(+) HeLa cells and HPV(-) HCT116 cells results in a substantial induction of cell growth retardation and a moderate cell apoptosis. Together, this is the first time a viral oncoprotein has been shown to regulate cellular miRNA expression. Our data have provided new insights into mechanisms by which high-risk HPVs contribute to the development of cervical cancer.


Subject(s)
DNA-Binding Proteins/metabolism , Human papillomavirus 16/metabolism , Human papillomavirus 18/metabolism , MicroRNAs/metabolism , Oncogene Proteins, Viral/metabolism , Papillomavirus Infections/metabolism , Repressor Proteins/metabolism , Adult , Cell Line, Tumor , Female , Gene Knockdown Techniques , Humans , Middle Aged , Papillomavirus Infections/virology
17.
Genes Dev ; 23(2): 181-94, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19131434

ABSTRACT

Using Cre-loxP-mediated recombination, we established a highly efficient and reproducible system that generates autonomous HPV-18 genomes in primary human keratinocytes (PHKs), the organotypic raft cultures of which recapitulated a robust productive program. While E7 promoted S-phase re-entry in numerous suprabasal differentiated cells, HPV DNA unexpectedly amplified following a prolonged G2 arrest in mid- and upper spinous cells. As viral DNA levels intensified, E7 activity diminished and then extinguished. These cells then exited the cell cycle to undergo virion morphogenesis. High titers of progeny virus generated an indistinguishable productive infection in naïve PHK raft cultures as before, never before achieved until now. An immortalization-defective HPV-18 E6 mutant genome was also characterized for the first time. Numerous cells accumulated p53 protein, without inducing apoptosis, but the productive program was severely curtailed. Complementation of mutant genomes by E6-expressing retrovirus restored proper degradation of p53 as well as viral DNA amplification and L1 production. This system will be invaluable for HPV genetic dissection and serves as a faithful ex vivo model for investigating infections and interventions.


Subject(s)
Cell Culture Techniques/methods , Human papillomavirus 18/physiology , Keratinocytes/virology , Papillomavirus Infections/virology , Virus Replication/physiology , Apoptosis , Cells, Cultured , DNA Replication , DNA-Binding Proteins/metabolism , Epithelium/virology , G2 Phase , Genome, Viral/genetics , Human papillomavirus 18/genetics , Human papillomavirus 18/ultrastructure , Humans , Keratinocytes/cytology , Microscopy, Electron, Transmission , Oncogene Proteins, Viral/metabolism , Plasmids/genetics , Time Factors , Tumor Suppressor Protein p53/metabolism , Virion/metabolism , Virion/ultrastructure
18.
Pharm Res ; 21(8): 1480-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15359585

ABSTRACT

PURPOSE: To determine whether a vaccine consisting of an idiotope peptide mimic of the third complementarity-determining region of the immunoglobulin heavy chain (CDR-H3) is an effective substitute for its parent idiotype. Such peptide vaccines could ultimately be used for targeting pathological B lymphocytes. METHODS: Hen egg lysozyme (HEL) conjugates of the Fab' fragment of monoclonal anti-fluorescein antibody 9-40 (Fab'-HEL) or a peptide mimic of the 9-40 CDR-H3 (referred to as the "B epitope" or "Bep," the conjugate is referred to as "Bep-HEL") were injected into separate cohorts of B10.A mice. Two additional control cohorts were injected with either the Bep peptide alone or a noncovalent mixture of Bep and HEL. Sera were assayed for both anti-idiotope and anti-idiotype activity by enzyme-linked immunosorbant assay (ELISA). Primary, secondary, and tertiary immune responses were examined. RESULTS: Both the Bep-HEL idiotope and the Fab-HEL idiotype immunogens elicited homologous, allogenic immune responses. No cross-reactivity was observed between anti-idiotope and anti-idiotype responses after primary immunization. With secondary immunization, 50% of mice immunized with the Bep-HEL conjugate exhibited a cross-reactive anti-idiotype response. Conversely, 100% of mice immunized with the Fab'-HEL conjugate exhibited a marginal, but statistically significant cross-reactive anti-idiotope response. Upon tertiary immunization, 100% of mice immunized with Bep-HEL exhibited a cross-reactive anti-idiotype response, and 55.6% of mice immunized with the Fab'-HEL conjugate exhibited a cross-reactive anti-idiotope response. CONCLUSIONS: Covalent coupling of a xenogenic carrier protein to an idiotype immunogen or its peptide mimic significantly enhances the intensity of homologous, allogenic anti-idiotype or anti-idiotope immune responses. Multiple immunizations are necessary to induce cross-reactivity between the peptide mimic and its parent idiotype.


Subject(s)
Complementarity Determining Regions/immunology , Immunoglobulin Idiotypes/immunology , Peptides/immunology , Vaccines/immunology , Animals , Antibodies, Anti-Idiotypic/immunology , B-Lymphocytes/immunology , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Female , Immunization , Mice , Mice, Inbred Strains , Muramidase/immunology , Peptides/chemical synthesis , Peptides/pharmacology , Vaccines/chemical synthesis
19.
Pharm Res ; 20(4): 591-6, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12739766

ABSTRACT

PURPOSE: This work examines the effectiveness of synthetic peptide immunogens derived from immunodominant T-cell epitopes as replacements for their intact parent protein in vaccines. METHODS: Fluorescein was conjugated to hen egg lysozyme (FL-HEL, positive control) and three synthetic peptide immunogens: (a) murine B10.A (H-2a) immunodominant T-cell epitope of HEL [FL-(T-cell epitope)]; (b) multiple antigenic peptide (MAP) multimer of this epitope ([FL-(T epitope)]n-MAP, n = 2-4); and (c) negative control MAP with T-cell epitope residues replaced with glycine [(FL-Gly18)4-MAP]. The dose response of each immunogen was examined over a 300-fold range in B10.A mice. The immune response was monitored using antifluorescein ELISA assays. RESULTS: FL-(T epitope)'s immune response correlated positively with dose, with maximum response comparable to that of [FL-(T epitope)]n-MAP, or FL-HEL. This trend was consistent across 1 degrees, 2 degrees, and 3 degrees responses, although interanimal variability was higher in the latter two because of an all-or-none response in mice immunized with this peptide. [FL-(T epitope)]n-MAP's immune response was consistently high and nearly dose independent, a trend observed across 1 degrees, 2 degrees, and 3 degrees responses. FL-HEL's immune response correlated negatively to dose in the 1 degrees response but was nearly dose independent in the 2 degrees and 3 degrees responses. The magnitude of these latter responses was comparable to that observed for [FL-(T epitope)]n-MAP. (FL-Gly18)4-MAP did not elicit an immune response except at the highest dose. This trend was consistent across 1 degrees, 2 degrees, and 3 degrees responses. CONCLUSIONS: The monomeric epitope was 300-fold less potent than its parent carrier protein, but increasing immunogen valency using MAP technology compensated totally for reduced potency. (FL-Gly18)4-MAP's lack of response at all but the highest dose strongly suggests that a specific immunodominant T-cell epitope sequence for HEL is necessary for successful peptide mimicry of HEL. This work also demonstrates the importance of quality assessment of commercial MAP core resins.


Subject(s)
Carrier Proteins/immunology , Dose-Response Relationship, Immunologic , Epitopes, T-Lymphocyte/immunology , Immunodominant Epitopes/immunology , Animals , Antigens/administration & dosage , Antigens/immunology , Enzyme-Linked Immunosorbent Assay , Fluorescein/administration & dosage , Male , Mice , Muramidase/administration & dosage , Peptides/administration & dosage , Peptides/immunology
20.
Anal Biochem ; 315(2): 223-37, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12689832

ABSTRACT

Rapid detection of single nucleotide polymorphisms (SNPs) has potential applications in both genetic screening and pharmacogenomics. Planar waveguide fluorescent biosensor technology was employed to detect SNPs using a simple hybridization assay with the complementary strand ("capture oligo") immobilized on the waveguide. This technology allows real-time measurements of DNA hybridization kinetics. Under normal conditions, both the wild-type sequence and the SNP-containing sequence will hybridize with the capture oligo, but with different reaction kinetics and equilibrium duplex concentrations. A "design of experiments" approach was used to maximize the differences in the kinetics profiles of the two. Nearly perfect discrimination can be achieved at short times (2 min) with temperatures that destabilize or melt the heteroduplex while maintaining the stability of the homoduplex. The counter ion content of the solvent was shown to have significant effect not only on the melting point of the heteroduplex and the homoduplex but also on the hybridization rate. Changes in both the stability and the difference between the hybridization rates of the hetero- and homoduplex were observed with varying concentrations of three different cations (Na(+), K(+), Mg(2+)). With the difference in hybridization rates maximized, discrimination between the hetero- and the homoduplex can be obtained at lower, less rigorous temperatures at hybridization times of 7.5 min or longer.


Subject(s)
Biosensing Techniques/methods , Long QT Syndrome/genetics , Polymorphism, Single Nucleotide/genetics , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Fluorescence , Ions/chemistry , KCNQ Potassium Channels , Kinetics , Nucleic Acid Denaturation , Nucleic Acid Hybridization , Sensitivity and Specificity , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...