Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Med Virol ; 95(12): e29280, 2023 12.
Article in English | MEDLINE | ID: mdl-38054507

ABSTRACT

Cycling hypoxia (cycH) is a prevalent form of tumor hypoxia that is characterized by exposure of tumor cells to recurrent phases of hypoxia and reoxygenation. CycH has been associated with a particularly aggressive cellular phenotype of tumor cells and increased therapy resistance. By performing comparative analyses under normoxia, physoxia, chronic hypoxia, and cycH, we here uncover distinct effects of cycH on the phenotype of human papillomavirus (HPV)-positive cervical cancer cells. We show that-other than under chronic hypoxia-viral E6/E7 oncogene expression is largely maintained under cycH as is the E6/E7-dependent regulation of p53 and retinoblastoma protein. Further, cycH enables HPV-positive cancer cells to evade prosenescent chemotherapy, similar to chronic hypoxia. Moreover, cells under cycH exhibit a particularly pronounced resistance to the proapoptotic effects of Cisplatin. Quantitative proteome analyses reveal that cycH induces a unique proteomic signature in cervical cancer cells, which includes a significant downregulation of luminal lysosomal proteins. These encompass the potentially proapoptotic cathepsins B and cathepsin L, which, however, appear not to affect the response to Cisplatin under any of the O2 conditions tested. Rather, we show that the proapoptotic Caspase 8/BH3-interacting domain death agonist (BID) cascade plays a pivotal role for the efficiency of Cisplatin-induced apoptosis in HPV-positive cancer cells under all investigated O2 conditions. In addition, we provide evidence that BID activation by Cisplatin is impaired under cycH, which could contribute to the high resistance to the proapoptotic effects of Cisplatin. Collectively, this study provides the first insights into the profound phenotypic alterations induced by cycH in HPV-positive cancer cells, with implications for their therapeutic susceptibility.


Subject(s)
Oncogene Proteins, Viral , Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , Cisplatin/pharmacology , Cisplatin/therapeutic use , Oncogene Proteins, Viral/genetics , Papillomavirus Infections/complications , Papillomavirus Infections/pathology , Proteomics , Repressor Proteins/genetics , Hypoxia , Papillomavirus E7 Proteins/genetics
2.
Int J Cancer ; 149(5): 1137-1149, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33844847

ABSTRACT

Oncogenic types of human papillomaviruses (HPVs) are major human carcinogens. The viral E6/E7 oncogenes maintain the malignant growth of HPV-positive cancer cells. Targeted E6/E7 inhibition results in efficient induction of cellular senescence, which could be exploited for therapeutic purposes. Here we show that viral E6/E7 expression is strongly downregulated by Metformin in HPV-positive cervical cancer and head and neck cancer cells, both at the transcript and protein level. Metformin-induced E6/E7 repression is glucose and PI3K-dependent but-other than E6/E7 repression under hypoxia-AKT-independent. Proteome analyses reveal that Metformin-induced HPV oncogene repression is linked to the downregulation of cellular factors associated with E6/E7 expression in HPV-positive cancer biopsies. Notably, despite efficient E6/E7 repression, Metformin induces only a reversible proliferative stop in HPV-positive cancer cells and enables them to evade senescence. Metformin also efficiently blocks senescence induction in HPV-positive cancer cells in response to targeted E6/E7 inhibition by RNA interference. Moreover, Metformin treatment enables HPV-positive cancer cells to escape from chemotherapy-induced senescence. These findings uncover profound effects of Metformin on the virus/host cell interactions and the phenotype of HPV-positive cancer cells with implications for therapy-induced senescence, for attempts to repurpose Metformin as an anticancer agent and for the development of E6/E7-inhibitory therapeutic strategies.


Subject(s)
Antineoplastic Agents/pharmacology , Cellular Senescence , Gene Expression Regulation, Neoplastic/drug effects , Metformin/pharmacology , Papillomaviridae/drug effects , Papillomavirus Infections/drug therapy , Uterine Cervical Neoplasms/drug therapy , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Female , Humans , Hypoglycemic Agents/pharmacology , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Proteome/drug effects , Tumor Cells, Cultured , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology
3.
mBio ; 10(1)2019 02 12.
Article in English | MEDLINE | ID: mdl-30755508

ABSTRACT

Hypoxia is linked to therapeutic resistance and poor clinical prognosis for many tumor entities, including human papillomavirus (HPV)-positive cancers. Notably, HPV-positive cancer cells can induce a dormant state under hypoxia, characterized by a reversible growth arrest and strong repression of viral E6/E7 oncogene expression, which could contribute to therapy resistance, immune evasion and tumor recurrence. The present work aimed to gain mechanistic insights into the pathway(s) underlying HPV oncogene repression under hypoxia. We show that E6/E7 downregulation is mediated by hypoxia-induced stimulation of AKT signaling. Ablating AKT function in hypoxic HPV-positive cancer cells by using chemical inhibitors efficiently counteracts E6/E7 repression. Isoform-specific activation or downregulation of AKT1 and AKT2 reveals that both AKT isoforms contribute to hypoxic E6/E7 repression and act in a functionally redundant manner. Hypoxic AKT activation and consecutive E6/E7 repression is dependent on the activities of the canonical upstream AKT regulators phosphoinositide 3-kinase (PI3K) and mechanistic target of rapamycin (mTOR) complex 2 (mTORC2). Hypoxic downregulation of E6/E7 occurs, at least in part, at the transcriptional level. Modulation of E6/E7 expression by the PI3K/mTORC2/AKT cascade is hypoxia specific and not observed in normoxic HPV-positive cancer cells. Quantitative proteome analyses identify additional factors as candidates to be involved in hypoxia-induced activation of the PI3K/mTORC2/AKT signaling cascade and in the AKT-dependent repression of the E6/E7 oncogenes under hypoxia. Collectively, these data uncover a functional key role of the PI3K/mTORC2/AKT signaling cascade for viral oncogene repression in hypoxic HPV-positive cancer cells and provide new insights into the poorly understood cross talk between oncogenic HPVs and their host cells under hypoxia.IMPORTANCE Oncogenic HPV types are major human carcinogens. Under hypoxia, HPV-positive cancer cells can repress the viral E6/E7 oncogenes and induce a reversible growth arrest. This response could contribute to therapy resistance, immune evasion, and tumor recurrence upon reoxygenation. Here, we uncover evidence that HPV oncogene repression is mediated by hypoxia-induced activation of canonical PI3K/mTORC2/AKT signaling. AKT-dependent downregulation of E6/E7 is only observed under hypoxia and occurs, at least in part, at the transcriptional level. Quantitative proteome analyses identify additional factors as candidates to be involved in AKT-dependent E6/E7 repression and/or hypoxic PI3K/mTORC2/AKT activation. These results connect PI3K/mTORC2/AKT signaling with HPV oncogene regulation, providing new mechanistic insights into the cross talk between oncogenic HPVs and their host cells.


Subject(s)
Hypoxia , Mechanistic Target of Rapamycin Complex 2/metabolism , Oncogene Proteins, Viral/biosynthesis , Papillomaviridae/physiology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Cell Line, Tumor , Down-Regulation , Host-Pathogen Interactions , Humans
4.
Proc Natl Acad Sci U S A ; 114(6): E990-E998, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28115701

ABSTRACT

Oncogenic human papillomaviruses (HPVs) are closely linked to major human malignancies, including cervical and head and neck cancers. It is widely assumed that HPV-positive cancer cells are under selection pressure to continuously express the viral E6/E7 oncogenes, that their intracellular p53 levels are reconstituted on E6/E7 repression, and that E6/E7 inhibition phenotypically results in cellular senescence. Here we show that hypoxic conditions, as are often found in subregions of cervical and head and neck cancers, enable HPV-positive cancer cells to escape from these regulatory principles: E6/E7 is efficiently repressed, yet, p53 levels do not increase. Moreover, E6/E7 repression under hypoxia does not result in cellular senescence, owing to hypoxia-associated impaired mechanistic target of rapamycin (mTOR) signaling via the inhibitory REDD1/TSC2 axis. Instead, a reversible growth arrest is induced that can be overcome by reoxygenation. Impairment of mTOR signaling also interfered with the senescence response of hypoxic HPV-positive cancer cells toward prosenescent chemotherapy in vitro. Collectively, these findings indicate that hypoxic HPV-positive cancer cells can induce a reversible state of dormancy, with decreased viral antigen synthesis and increased therapeutic resistance, and may serve as reservoirs for tumor recurrence on reoxygenation.


Subject(s)
Cellular Senescence/genetics , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Papillomaviridae/genetics , Cell Hypoxia , Cell Line , Cell Line, Tumor , Female , HCT116 Cells , HeLa Cells , Hep G2 Cells , Host-Pathogen Interactions/genetics , Humans , Hypoxia , MCF-7 Cells , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/virology , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomaviridae/metabolism , Papillomaviridae/physiology , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/virology
5.
Oncotarget ; 8(63): 106342-106351, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29290953

ABSTRACT

Oncogenic types of human papillomaviruses (HPVs) are major human carcinogens. Cancer cells typically exhibit metabolic alterations which support their malignant growth. These include an enhanced rate of aerobic glycolysis ('Warburg effect') which in cancer cells is often linked to an increased expression of the rate-limiting glycolytic enzyme Hexokinase 2 (HK2). Intriguingly, recent studies indicate that the HPV E6/E7 oncogenes cause the metabolic reprogramming in HPV-positive cancer cells by directly upregulating HK2 expression. Notably, however, these results were obtained upon ectopic overexpression of E6/E7. Here, we investigated whether HK2 levels are affected by the endogenous E6/E7 amounts present in HPV-positive cancer cell lines. RNA interference analyses reveal that the sustained E6/E7 expression is critical to maintain HK2 expression levels in HeLa cells. Mechanistically, this effect is linked to the E6/E7-dependent upregulation of HK2-stimulatory MYC expression and the E6/E7-induced downregulation of the HK2-inhibitory micro(mi)RNA miR-143-3p. Importantly, however, a stimulatory effect of E6/E7 on HK2 expression was observed only in HeLa among a panel of 8 different HPV-positive cervical and head and neck cancer cell lines. Thus, whereas these results support the notion that E6/E7 can increase HK2 expression, they argue against the concept that the viral oncogenes, at endogenous expression levels, commonly induce the metabolic switch of HPV-positive cancer cells towards aerobic glycolysis by directly or indirectly stimulating HK2 expression.

6.
PLoS One ; 10(7): e0132339, 2015.
Article in English | MEDLINE | ID: mdl-26151636

ABSTRACT

Oncogenic types of human papillomaviruses (HPVs) cause cervical cancer and other malignancies in humans. The HPV E6 oncoprotein is considered to be an attractive therapeutic target since its inhibition can lead to the apoptotic cell death of HPV-positive cancer cells. The HPV type 16 (HPV16) E6-binding peptide pep11, and variants thereof, induce cell death specifically in HPV16-positive cancer cells. Although they do not encompass the LxxLL binding motif found in cellular HPV16 E6 interaction partners, such as E6AP, the pep11 variants strongly bind to HPV16 E6 by contacting the recently identified E6AP binding pocket. Thus, these peptides can serve as prototype E6-inhibitory molecules which target the E6AP pocket. We here analyzed their intracellular interaction with HPV16 E6. By comprehensive intracellular binding studies and GST pull-down assays, we show that E6-binding competent pep11 variants induce the formation of a trimeric complex, consisting of pep11, HPV16 E6 and p53. These findings indicate that peptides, which do not contain the LxxLL motif, can reshape E6 to enable its interaction with p53. The formation of the trimeric HPV16 E6 / peptide / p53 complex was associated with an increase of endogenous HPV16 E6 protein amounts. Yet, total cellular p53 amounts were also increased, indicating that the E6 / E6AP-mediated degradation of p53 is blocked. These findings suggest that inhibition of oncogenic activities by targeting the E6AP pocket on HPV16 E6 could be a strategy for therapeutic intervention.


Subject(s)
Cytoplasm/metabolism , Oncogene Proteins, Viral/metabolism , Peptides/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Binding Sites/genetics , Cell Line, Tumor , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Immunoblotting , Microscopy, Confocal , Molecular Sequence Data , Mutation , Oncogene Proteins, Viral/genetics , Peptides/genetics , Protein Binding , Repressor Proteins/genetics , Tumor Suppressor Protein p53/metabolism , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases/metabolism
7.
PLoS Pathog ; 10(3): e1003957, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24604027

ABSTRACT

The expression of the human papillomavirus (HPV) E6/E7 oncogenes is crucial for HPV-induced malignant cell transformation. The identification of cellular targets attacked by the HPV oncogenes is critical for our understanding of the molecular mechanisms of HPV-associated carcinogenesis and may open novel therapeutic opportunities. Here, we identify the Lens Epithelial-Derived Growth Factor (LEDGF) gene as a novel cellular target gene for the HPV oncogenes. Elevated LEDGF expression has been recently linked to human carcinogenesis and can protect tumor cells towards different forms of cellular stress. We show that intracellular LEDGF mRNA and protein levels in HPV-positive cancer cells are critically dependent on the maintenance of viral oncogene expression. Ectopic E6/E7 expression stimulates LEDGF transcription in primary keratinocytes, at least in part via activation of the LEDGF promoter. Repression of endogenous LEDGF expression by RNA interference results in an increased sensitivity of HPV-positive cancer cells towards genotoxic agents. Immunohistochemical analyses of cervical tissue specimens reveal a highly significant increase of LEDGF protein levels in HPV-positive lesions compared to histologically normal cervical epithelium. Taken together, these results indicate that the E6/E7-dependent maintenance of intracellular LEDGF expression is critical for protecting HPV-positive cancer cells against various forms of cellular stress, including DNA damage. This could support tumor cell survival and contribute to the therapeutic resistance of cervical cancers towards genotoxic treatment strategies in the clinic.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/genetics , Intercellular Signaling Peptides and Proteins/genetics , Papillomavirus Infections/genetics , Uterine Cervical Neoplasms/genetics , Cell Line , Female , Humans , Immunoblotting , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/biosynthesis , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/metabolism , RNA, Small Interfering , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Uterine Cervical Neoplasms/metabolism
8.
Hepatology ; 56(1): 9-16, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22271223

ABSTRACT

UNLABELLED: The inhibitors of pyrimidine synthesis, leflunomide and FK778, have been reported to exert broad antiviral effects, in addition to their immunosuppressive activities. Their possible therapeutic benefit for transplantation medicine is currently discussed, because they also block the replication of human cytomegalovirus and human polyomavirus BK, which both cause important complications in transplant recipients. Here, we show that leflunomide and FK778 strongly enhance hepatitis B virus (HBV) replication in vitro. This activity is shared by mycophenolic acid (MPA), an inhibitor of purine biosynthesis. Stimulation of HBV replication by these agents was linked to their inhibitory effects on de novo nucleotide biosynthesis because it could be efficiently counteracted by external nucleoside supply. Mechanistically, we found that mitogen-activated protein kinase p38 played a key role for the enhancement of HBV replication by leflunomide, FK778, and MPA. All three HBV-activating compounds increased p38 phosphorylation, in contrast to the HBV inhibitors, telbivudine and cyclosporine A. Moreover, silencing of p38 expression through RNA interference efficiently counteracted the stimulatory effect of leflunomide, FK778, and MPA on HBV replication. CONCLUSION: Our data indicate that, in contrast to their reported inhibitory effects on other viruses, both leflunomide and FK778 can augment HBV replication. Treatment with leflunomide, FK778, or MPA may bear the risk to enhance HBV replication in infected patients.


Subject(s)
Alkynes/pharmacology , Hepatitis B virus/drug effects , Hepatitis B virus/physiology , Isoxazoles/pharmacology , Mycophenolic Acid/pharmacology , Nitriles/pharmacology , Virus Replication/drug effects , Cells, Cultured/drug effects , Cells, Cultured/virology , Enzyme Inhibitors/pharmacology , HEK293 Cells , Hep G2 Cells , Humans , Immunosuppressive Agents/pharmacology , In Vitro Techniques , Leflunomide , RNA, Viral/analysis , Real-Time Polymerase Chain Reaction , Sensitivity and Specificity
9.
Int J Cancer ; 125(9): 2014-20, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19551855

ABSTRACT

Human papillomavirus (HPV)-induced carcinogenesis is critically dependent on the activities of the viral E6 and E7 oncogenes. Here, we demonstrate that expression of the putative tumor suppressor gene B-cell translocation gene-2 (BTG2) is reinduced in HPV16- and HPV18-positive cancer cells on silencing of viral oncogene expression, indicating that BTG2 is repressed by oncogenic HPVs. Inhibition of BTG2 expression was mediated by the HPV E6 oncogene and occurred in a p53-dependent manner. Luciferase reporter gene analyses revealed that BTG2 repression takes place at the transcriptional level and is dependent on the integrity of the major p53-response element within the BTG2 promoter. Ectopic expression of BTG2 acted antiproliferative in cervical cancer cells. Tissue specimens commonly exhibited reduced BTG2 protein levels in HPV-positive high-grade lesions (CIN2/3) and cervical carcinomas, when compared with normal cervical epithelium. These findings identify the antiproliferative BTG2 gene as a novel cellular target blocked by the HPV E6 oncoprotein.


Subject(s)
Genes, Tumor Suppressor , Human papillomavirus 16/genetics , Human papillomavirus 18/genetics , Immediate-Early Proteins/genetics , Cell Proliferation , DNA-Binding Proteins/genetics , HeLa Cells , Human papillomavirus 16/pathogenicity , Human papillomavirus 18/pathogenicity , Humans , Oncogene Proteins, Viral/genetics , Repressor Proteins/genetics , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins
10.
Int J Mol Med ; 23(3): 415-20, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19212661

ABSTRACT

The malononitrilamide FK778 is a novel immunosuppressive agent with antiproliferative activities. To gain insight into the molecular mechanism of FK778-mediated growth inhibition, we analyzed cells which differ in their p53 status and functionality of retinoblastoma protein (pRb). FK778 acted as a broad inhibitor of cell proliferation independent of the p53 or pRb status. However, the mechanism of FK778-mediated growth inhibition differed, leading either to cell cycle arrest in G1, or cell accumulation in S phase. This differential response was linked to the phosphorylation status of pRb. In addition, since FK778 was reported to exhibit antiviral activities, we analyzed the effect of FK778 on the growth stimulatory human papillomavirus (HPV)-16 and -18 E7 genes. Although growth of HPV-positive cells was strongly inhibited by FK778, we did not observe significant effects on viral E7 expression, indicating that the antiproliferative effect is not linked to an antiviral activity of FK778.


Subject(s)
Alkynes/pharmacology , Antiviral Agents/pharmacology , G1 Phase/drug effects , Isoxazoles/pharmacology , Nitriles/pharmacology , Retinoblastoma Protein/metabolism , S Phase/drug effects , Tumor Suppressor Protein p53/metabolism , DNA-Binding Proteins/biosynthesis , HeLa Cells , Human papillomavirus 16/metabolism , Human papillomavirus 18/metabolism , Humans , Oncogene Proteins, Viral/biosynthesis , Papillomavirus E7 Proteins , Phosphorylation/drug effects
11.
J Mol Med (Berl) ; 87(3): 321-31, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19099279

ABSTRACT

Specific types of human papillomaviruses (HPVs) cause cervical cancer. The viral E6 oncogene is a critical factor for maintaining the malignant phenotype of HPV-positive tumour cells. By yeast two-hybrid screening of a randomised peptide expression library, we isolated linear short peptides, which specifically bind to the HPV16 E6 oncoprotein. Sequence alignments and mutational analyses of the peptides identified a hitherto undiscovered E6-binding motif. Intracellular expression of a peptide containing the novel E6-binding motif resulted in inhibition of colony formation capacity, specifically of HPV16-positive cancer cells. A solubility-optimised variant of the peptide was created, which binds to HPV16 E6 with high affinity. Its intracellular expression efficiently induced apoptosis in HPV16-positive cancer cells. This was linked to restoration of intracellular p53 activities. Thus, this newly identified E6-binding motif could form a novel basis for the development of rational strategies for the treatment of HPV16-positive preneoplastic and neoplastic lesions.


Subject(s)
Oncogene Proteins, Viral/metabolism , Peptides/metabolism , Repressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Binding Sites/genetics , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Discs Large Homolog 1 Protein , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , In Situ Nick-End Labeling , Kinetics , Membrane Proteins/metabolism , Molecular Sequence Data , Oncogene Proteins, Viral/genetics , Peptide Library , Peptides/genetics , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Transfection , Tumor Suppressor Protein p53/metabolism , Two-Hybrid System Techniques , Ubiquitination
12.
Cancer Res ; 68(23): 9964-72, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19047178

ABSTRACT

The malignant phenotype of human papillomavirus (HPV)-positive cancer cells is maintained by the activity of the viral E6 and E7 genes. Here, we identified the polycomb group gene enhancer of zeste homologue 2 (EZH2) as a novel downstream target for the viral oncogenes in HPV-transformed cells. EZH2 expression was activated by HPV16 E7 at the transcriptional level via E7-mediated release of E2F from pocket proteins. RNA interference analyses showed that continuous EZH2 expression is required for the proliferation of HPV-positive tumor cells by stimulating cell cycle progression at the G1-S boundary. In addition to its growth-promoting activity, EZH2 also contributed to the apoptotic resistance of cervical cancer cells. Furthermore, we found that HPV-positive dysplastic and tumorigenic cervical lesions were characterized by high levels of EZH2 protein in vivo. We conclude that the E7 target gene EZH2 is a major determinant for the proliferation of HPV-positive cancer cells and contributes to their apoptotic resistance. Moreover, EZH2 may serve as a novel therapeutic target for the treatment of cervical cancer.


Subject(s)
DNA-Binding Proteins/genetics , Neoplasms/genetics , Neoplasms/virology , Oncogene Proteins, Viral/genetics , Transcription Factors/genetics , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/virology , Cell Line, Tumor , DNA-Binding Proteins/biosynthesis , Enhancer of Zeste Homolog 2 Protein , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , HeLa Cells , Human papillomavirus 16/genetics , Human papillomavirus 18/genetics , Humans , Neoplasms/pathology , Osteosarcoma/genetics , Osteosarcoma/pathology , Osteosarcoma/virology , Papillomavirus E7 Proteins , Polycomb Repressive Complex 2 , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Repressor Proteins/genetics , Transcription Factors/biosynthesis , Transcription, Genetic , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology
13.
J Biol Chem ; 281(30): 21345-21352, 2006 Jul 28.
Article in English | MEDLINE | ID: mdl-16717089

ABSTRACT

Peptide aptamers (PAs) can be employed to block the intracellular function of target proteins. Little is known about the mechanism of PA-mediated protein inhibition. Here, we generated PAs that specifically bound to the duck hepatitis B virus (HBV) core protein. Among them, PA34 strongly blocked duck HBV replication by inhibiting viral capsid formation. We found that PA34 led to a dramatic intracellular redistribution of its target protein into perinuclear inclusion bodies, which exhibit the typical characteristics of aggresomes. As a result, the core protein is efficiently removed from the viral life cycle. Corresponding findings were obtained for bioactive PAs that bind to the HBV core protein or to the human papillomavirus-16 (HPV16) E6 protein, respectively. The observation that PAs induce the specific sequestration of bound proteins into aggresomes defines a novel mechanism as to how this new class of intracellular inhibitors blocks the function of their target proteins.


Subject(s)
Aptamers, Peptide/chemistry , Hepatitis B virus/genetics , Proteins/antagonists & inhibitors , Amino Acid Sequence , Antiviral Agents/pharmacology , Cell Line, Tumor , Humans , Molecular Sequence Data , Oncogene Proteins, Viral/metabolism , Peptides/chemistry , Plasmids/metabolism , Protein Binding , Repressor Proteins/metabolism , Vimentin/chemistry , Virus Replication
14.
Cancer Lett ; 239(1): 64-70, 2006 Jul 28.
Article in English | MEDLINE | ID: mdl-16135399

ABSTRACT

Infections with human papillomaviruses are divided basically into three different infection types: those producing specific clinically visible lesions, those remaining subclinical, and those being latent. The assumed infection type thought to be present in tissue specimens has influence on the conclusions that can be made from an analysis, i.e. whether or not the HPV infection has a causal relationship with other epidemiological or molecular investigation observations. To determine whether HPV DNA detection in different entities of the upper aerodigestive tract represents a coincidental, persistent/latent or specific infection, 20 clinically intact mucosa specimens of the upper aerodigestive tract, 20 sinonasal polyps, 26 inverted papillomas, and 20 squamous cell carcinomas of the paranasal sinuses were investigated. HPV DNA was not detectable in specimens derived from clinically intact mucosa or in nasal polyps. Yet, three out of 26 inverted papillomas were HPV-positive, each showing double infection with HPV6 and 11. Four out of 20 squamous cell carcinomas were HPV16 positive. To our knowledge, we are presenting the first study contemporaneously analyzing benign as well as malignant non-proliferative and proliferative mucosal entities whilst applying identical methodical standards. The data corroborate the hypothesis that HPV DNA demonstration in tissue specimens represents a specific infection of the mucosa of the upper aerodigestive tract. It can thus be assumed that there is a causative involvement of HPV infections in the alteration of cell proliferation and in the case of infection with high risk HPV types even on progression to malignant transformation.


Subject(s)
Carcinoma, Squamous Cell/virology , DNA, Viral/analysis , Papilloma, Inverted/virology , Papillomaviridae/genetics , Papillomavirus Infections/virology , Paranasal Sinus Neoplasms/virology , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/pathology , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Mucous Membrane , Nasal Cavity/pathology , Nasal Cavity/virology , Nasal Polyps/pathology , Nasal Polyps/virology , Papilloma, Inverted/pathology , Papillomavirus Infections/pathology , Paranasal Sinus Neoplasms/pathology , Paranasal Sinuses/pathology , Paranasal Sinuses/virology , Polymerase Chain Reaction
15.
Acta Otolaryngol ; 125(4): 415-21, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15823814

ABSTRACT

CONCLUSION: The results of this study corroborate earlier findings that human papillomavirus (HPV)16 is the most prevalent type of HPV in squamous cell carcinomas of the head and neck (SCCHNs) and reinforce a possible influence of HPV on SCCHN progression by showing that the majority of HPV-positive patients harbor HPV16 (or HPV33) both in their primary tumors and in lymph node neck metastases (LNNMs). OBJECTIVE: HPVs are causally associated with carcinomas of the uterine cervix and have also been linked to a subset of SCCHNs. In order to further investigate the predicted causative role of HPV in SCCHNs, we analyzed pairs of primary tumors and LNNMs or LNNMs alone for the presence of HPV DNA using polymerase chain reaction (PCR). MATERIAL AND METHODS: DNA was extracted from fresh frozen tissue samples of primary tumors and the corresponding LNNMs of 18 patients and from LNNMs alone in 17 patients. For the detection and typing of HPV, PCR was performed using both type-specific and consensus primer pairs, followed by Southern hybridization and, in selected cases, sequencing of the PCR products. RESULTS: Of the 35 patients investigated, 22 (63%) were found to have HPV DNA in their tumors: HPV16 DNA in 21 cases and HPV33 in 1. The highest HPV prevalence was detected in tumors of Waldeyer's tonsillar ring (8/9 patients; 89%). Of the 18 patients in whom primary tumors and LNNMs were analyzed, 7 (39%) were HPV-positive in both samples (HPV16, n = 6; HPV33, n = 1), in 3 (17%) the primary tumors were HPV-negative and the LNNMs HPV16-positive and in 1 (5.5%) the primary tumor contained HPV16 and the LNNM was negative. Interestingly, of the 7 patients in whom LNNMs had been detected only several months after diagnosis and treatment of the primary tumors, only 1 showed infection with HPV (HPV33).


Subject(s)
Carcinoma, Squamous Cell/virology , Lymphatic Metastasis/pathology , Otorhinolaryngologic Neoplasms/virology , Papillomaviridae/pathogenicity , Papillomavirus Infections/virology , Adult , Aged , Aged, 80 and over , Blotting, Southern , Carcinoma, Squamous Cell/pathology , Combined Modality Therapy , DNA Probes, HPV , Disease Progression , Female , Humans , Lymph Nodes/pathology , Lymph Nodes/virology , Male , Middle Aged , Neoplasm Staging , Otorhinolaryngologic Neoplasms/diagnosis , Otorhinolaryngologic Neoplasms/pathology , Palatine Tonsil/pathology , Palatine Tonsil/virology , Papillomavirus Infections/diagnosis , Papillomavirus Infections/pathology , Tonsillar Neoplasms/pathology , Tonsillar Neoplasms/virology
16.
Cancer Lett ; 218(2): 199-206, 2005 Feb 10.
Article in English | MEDLINE | ID: mdl-15670897

ABSTRACT

Depending on the primary tumour's anatomical location, squamous cell carcinoma of the head and neck (HNSCC) shows HPV prevalences between 20 and 30% for oro-, hypopharyngeal as well as laryngeal SCC and up to over 50% for SCC of the Waldeyer's tonsillar ring. There is persistent controversy on the role of HPV infection in HNSCC-progression, and on the influence of these infections on the final clinical outcome. To evaluate the possible relevance of HPV infection on survival and prognosis, 73 patients with HNSCC were investigated statistically with a median follow-up time of 28 (0.3-94) months. The statistical analysis revealed no differences in the overall survival of HPV-positive and HPV-negative cancer patients. A correlation between decreased survival and increased lymph node status was expected. Patients with carcinomas of the Waldeyer's tonsillar ring with a high HPV prevalence rate as compared to tumours of other anatomical locations revealed a better survival. Moreover, an association between HPV positivity and higher lymph node status at time of first diagnosis, and a better survival of HPV-positive patients compared to HPV-negative patients given the same initial nodal status (N0 vs. N1-N2b vs. N2c-N3) could be demonstrated. The influence of HPV on the patient's survival can only be observed statistically in combination with other prognostic factors, as the lymph nodal status of the patients. The better prognosis of survival of HPV-positive vs. the HPV-negative patients with lymph node neck metastasis is attributable to a better response of the HPV-positive group to therapy, especially radiotherapy.


Subject(s)
Carcinoma, Squamous Cell/mortality , Head and Neck Neoplasms/mortality , Papillomaviridae/isolation & purification , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/virology , Female , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/virology , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Recurrence, Local
17.
Oncogene ; 24(5): 810-9, 2005 Jan 27.
Article in English | MEDLINE | ID: mdl-15531914

ABSTRACT

Squamous cell carcinoma of the uterine cervix is one of the most frequent cancers affecting women worldwide. Carcinomas arise from cervical intraepithelial lesions, in which infection with high-risk human papillomavirus types has led to deregulated growth control through the actions of the viral E6 and E7 oncoproteins. The molecular mechanisms underlying progression to invasive tumor growth are poorly understood. One important feature, however, is the escape from growth inhibition by transforming growth factor beta (TGF-beta). Loss of chromosomal arm 18q is among the most frequent cytogenetic alterations in cervical cancers and has been associated with poor prognosis. Since the TGF-beta response is mediated by Smad proteins and the tumor suppressor gene Smad4 resides at 18q21, we have analysed the Smad4 gene for cervical cancer-associated alterations in cell lines and primary carcinomas. Here, we report Smad4 deficiency in four out of 13 cervical cancer cell lines which is due to an intronic rearrangement or deletions of 3' exons. All cell lines, however, showed either absent or moderate responsiveness to TGF-beta irrespective of their Smad4 status. In 41 primary squamous cervical carcinomas analysed, 10 samples showed loss of Smad4 protein expression and 26 samples a reduced expression. Altogether, our results strongly suggest that Smad4 gene alterations are involved in cervical carcinogenesis.


Subject(s)
Chromosomes, Human, Pair 18 , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Loss of Heterozygosity , Trans-Activators/deficiency , Trans-Activators/genetics , Uterine Cervical Neoplasms/genetics , Base Sequence , Cell Division/drug effects , Cell Line, Tumor , Chromosome Mapping , DNA Primers , Female , Genes, Tumor Suppressor , Humans , Reverse Transcriptase Polymerase Chain Reaction , Smad4 Protein , Transforming Growth Factor beta/pharmacology , Uterine Cervical Neoplasms/pathology
18.
Oral Oncol ; 40(5): 520-4, 2004 May.
Article in English | MEDLINE | ID: mdl-15006625

ABSTRACT

Human papillomavirus type 16 (HPV16) is associated with squamous cell carcinomas of the head and neck (HNSCC) particularly from the Waldeyer's tonsillar ring. A causal role of HPV16 in carcinogenesis is linked to the activity of the viral oncoproteins E6 and E7 which inactivate the cellular tumor suppressors p53 and pRB, respectively. Lack of E6 expression in HPV16-positive HNSCC has been reported, in some cases caused by disruption of the E6 gene. We have examined the status of the HPV16 E6-E7 gene region in tumor and metastasis samples of 24 HNSCC patients employing genomic PCR. No cases with a disrupted E6-E7 region could be identified. Sequence analysis of the E6-E7 segments revealed three different HPV16 E6-E7 genotypes: the HPV16 prototype (6 of 21 cases), the E6 variant T350G (8 of 21 cases), and the E6-E7 variant A131G/C712A (7 of 21 cases). The E6 variants T350G and A131G have been associated with increased oncogenic potential in cervical cancer patients depending on host genetic factors. Their high prevalence in the HNSCC samples studied indicates that they may be important also in HNSCC development.


Subject(s)
Carcinoma, Squamous Cell/virology , Head and Neck Neoplasms/virology , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Repressor Proteins , Base Sequence , Carcinoma, Squamous Cell/genetics , DNA, Neoplasm/analysis , DNA, Viral/analysis , Head and Neck Neoplasms/genetics , Humans , Lymphatic Metastasis , Mutation , Oligonucleotide Array Sequence Analysis/methods , Papillomavirus E7 Proteins , Polymerase Chain Reaction/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...