Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
Add more filters

Publication year range
1.
J Med Virol ; 95(4): e28681, 2023 04.
Article in English | MEDLINE | ID: mdl-36929719

ABSTRACT

Cervical cancer is the most frequent malignancy of the female genital tract and is associated with persistent infection of the uterine cervix with high-risk human papillomaviruses (HPV). The two HPV oncoproteins, E6 and E7, cooperatively immortalize cervical cells and are essential but insufficient for inducing tumorigenicity. During the progression of HPV-associated cervical dysplasia to carcinoma, the cellular telomerase reverse transcriptase (TERT) gene is activated and the TERC gene amplified. We questioned whether these increases in telomerase components might mediate the acquisition of the tumorigenic phenotype. We therefore transduced the TERT and TERC genes into E6/E7 immortalized keratinocytes that were anchorage-dependent and nontumorigenic. The resultant cells showed a profound morphological change characteristic of epithelial-mesenchymal transition as well as a corresponding increase in expression of vimentin, N-cadherin, Zinc finger E-Box binding homeobox 1, snail family transcriptional repressor 1 and matrix Metallopeptidase 2 and decrease in keratin and E-cadherin. More important, the transduced cells were now anchorage-independent and formed tumors in immunodeficient mice. Our findings indicate that overexpression of the telomerase holoenzyme in HPV-immortalized cells is sufficient to induce the complete transformed phenotype.


Subject(s)
Oncogene Proteins, Viral , Papillomavirus Infections , Telomerase , Uterine Cervical Neoplasms , Female , Humans , Animals , Mice , Oncogene Proteins, Viral/genetics , Telomerase/genetics , Telomerase/metabolism , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/complications , Papillomavirus Infections/genetics , Keratinocytes/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Uterine Cervical Neoplasms/genetics , Papillomaviridae/genetics
2.
J Med Virol ; 94(9): 4565-4575, 2022 09.
Article in English | MEDLINE | ID: mdl-35509176

ABSTRACT

The high-risk alpha human papillomaviruses (HPVs) are responsible for 99% of cervical cancers. While the biological functions of the HPV E6 and E7 oncoproteins are well-characterized, the function of E5 has remained elusive. Here, we examined gene expression changes induced by E5 proteins from high-risk HPV-16 and low-risk HPV-6b in multiple pools of primary human keratinocytes. Surprisingly, microarray analysis revealed that over 700 genes were significantly regulated by HPV-6b E5, while only 25 genes were consistently and significantly regulated by HPV-16 E5 in three biological replicates. However, we observed that more than thousand genes were altered in individual sample compared with vector. The gene expression profile induced by 16E5 in primary genital keratinocytes was very different from what has been previously published using immortalized HaCaT cells. Genes altered by HPV-16 E5 were unaffected by HPV-6b E5. Our data demonstrate that E5 proteins from the high- and low-risk HPVs have different functions in the HPV-host cell. Interestingly, conversion of two amino acids in HPV-16 E5 to the low-risk HPV-6b sequence eliminated the induction of high-risk related cellular genes.


Subject(s)
Oncogene Proteins, Viral , Papillomavirus Infections , Viroporin Proteins , Amino Acids , Female , Human papillomavirus 16/genetics , Humans , Oncogene Proteins, Viral/genetics , Viroporin Proteins/genetics
3.
J Med Virol ; 94(8): 3962-3977, 2022 08.
Article in English | MEDLINE | ID: mdl-35437795

ABSTRACT

The high-risk human papillomaviruses (HPV-16, -18) are critical etiologic agents in human malignancy, most importantly in cervical cancer. These oncogenic viruses encode the E6 and E7 proteins that are uniformly retained and expressed in cervical cancers and required for maintenance of the tumorigenic phenotype. The E6 and E7 proteins were first identified as targeting the p53 and pRB tumor suppressor pathways, respectively, in host cells, thereby leading to disruption of cell cycle controls. In addition to p53 degradation, a number of other functions and critical targets for E6 have been described, including telomerase, Myc, PDZ-containing proteins, Akt, Wnt, mTORC1, as well as others. In this study, we identified Amplified in Breast Cancer 1 (AIB1) as a new E6 target. We first found that E6 and hTERT altered similar profiling of gene expression in human foreskin keratinocytes (HFK), independent of telomerase activity. Importantly, AIB1 was a common transcriptional target of both E6 and hTERT. We then verified that high-risk E6 but not low-risk E6 expression led to increases in AIB1 transcript levels by real-time RT-PCR, suggesting that AIB1 upregulation may play an important role in cancer development. Western blots demonstrated that AIB1 expression increased in HPV-16 E6 and E7 expressing (E6E7) immortalized foreskin and cervical keratinocytes, and in three of four common cervical cancer cell lines as well. Then, we evaluated the expression of AIB1 in human cervical lesions and invasive carcinoma using immunohistochemical staining. Strikingly, AIB1 showed positivity in the nucleus of cells in the immediate suprabasal epithelium, while nuclei of the basal epithelium were negative, as evident in the Cervical Intraepithelial Neoplasia 1 (CIN1) samples. As the pathological grading of cervical lesions increased from CIN1, CIN2, CIN3 carcinoma in situ and invasive carcinoma, AIB1 staining increased progressively, suggesting that AIB1 may serve as a novel histological biomarker for cervical cancer development. For cases of invasive cervical carcinoma, AIB1 staining was specific to cancerous lesions. Increased expression of AIB1 was also observed in transgenic mouse cervical neoplasia and cancer models induced by E6E7 and estrogen. Knockdown of AIB1 expression in E6E7 immortalized human cervical cells significantly abolished cell proliferation. Taken together, these data support AIB1 as a novel target of HPV E6 and a biomarker of cervical cancer progression.


Subject(s)
Oncogene Proteins, Viral , Papillomavirus Infections , Telomerase , Uterine Cervical Dysplasia , Uterine Cervical Neoplasms , Animals , Biomarkers , Female , Humans , Mice , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/complications , Telomerase/genetics , Telomerase/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53
4.
Lab Invest ; 100(1): 38-51, 2020 01.
Article in English | MEDLINE | ID: mdl-31409888

ABSTRACT

Neuroblastoma (NB) is a pediatric tumor of the peripheral nervous system. Treatment of the disease represents an unsolved clinical problem, as survival of patients with aggressive form of NB remains below 50%. Despite recent identification of numerous potential therapeutic targets, clinical trials validating them are challenging due to the rarity of the disease and its high patient-to-patient heterogeneity. Hence, there is a need for the accurate preclinical models that would allow testing novel therapeutic approaches and prioritizing the clinical studies, preferentially in personalized way. Here, we propose using conditional reprogramming (CR) technology for rapid development of primary NB cell cultures that could become a new model for such tests. This newly established method allowed for indefinite propagation of normal and tumor cells of epithelial origin in an undifferentiated state by their culture in the presence of Rho-associated kinase (ROCK) inhibitor, Y-27632, and irradiated mouse feeder cells. Using a modification of this approach, we isolated cell lines from tumors arising in the TH-MYCN murine transgenic model of NB (CR-NB). The cells were positive for neuronal markers, including Phox2B and peripherin and consisted of two distinct populations: mesenchymal and adrenergic expressing corresponding markers of their specific lineage. This heterogeneity of the CR-NB cells mimicked the different tumor cell phenotypes in TH-MYCN tumor tissues. The CR-NB cells preserved anchorage-independent growth capability and were successfully passaged, frozen and biobanked. Further studies are required to determine the utility of this method for isolation of human NB cultures, which can become a novel model for basic, translational, and clinical research, including individualized drug testing.


Subject(s)
Cell Line, Tumor , Neuroblastoma/pathology , Animals , Biomarkers/metabolism , Cellular Reprogramming Techniques , Humans , Mice, Transgenic , Neoplasms, Experimental , Neuroblastoma/metabolism , Phenotype , Rats
5.
Br J Cancer ; 122(6): 868-884, 2020 03.
Article in English | MEDLINE | ID: mdl-31942031

ABSTRACT

BACKGROUND: Recent studies have suggested that fatty acid oxidation (FAO) is a key metabolic pathway for the growth of triple negative breast cancers (TNBCs), particularly those that have high expression of MYC. However, the underlying mechanism by which MYC promotes FAO remains poorly understood. METHODS: We used a combination of metabolomics, transcriptomics, bioinformatics, and microscopy to elucidate a potential mechanism by which MYC regulates FAO in TNBC. RESULTS: We propose that MYC induces a multigenic program that involves changes in intracellular calcium signalling and fatty acid metabolism. We determined key roles for fatty acid transporters (CD36), lipases (LPL), and kinases (PDGFRB, CAMKK2, and AMPK) that each contribute to promoting FAO in human mammary epithelial cells that express oncogenic levels of MYC. Bioinformatic analysis further showed that this multigenic program is highly expressed and predicts poor survival in the claudin-low molecular subtype of TNBC, but not other subtypes of TNBCs, suggesting that efforts to target FAO in the clinic may best serve claudin-low TNBC patients. CONCLUSION: We identified critical pieces of the FAO machinery that have the potential to be targeted for improved treatment of patients with TNBC, especially the claudin-low molecular subtype.


Subject(s)
Claudins/metabolism , Fatty Acids/metabolism , Metabolomics/methods , Proto-Oncogene Proteins c-myc/genetics , Triple Negative Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Humans , Transfection
6.
Biochem Biophys Res Commun ; 517(1): 49-56, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31303270

ABSTRACT

Patient-derived xenografts (PDXs) are widely recognised as a more physiologically relevant preclinical model than standard cell lines, but are expensive and low throughput, have low engraftment rate and take a long time to develop. Our newly developed conditional reprogramming (CR) technology addresses many PDX drawbacks, but lacks many in vivo factors. Here we determined whether PDXs and CRCs of the same cancer origin maintain the biological fidelity and complement each for translational research and drug development. Four CRC lines were generated from bladder cancer PDXs. Short tandem repeat (STR) analyses revealed that CRCs and their corresponding parental PDXs shared the same STRs, suggesting common cancer origins. CRCs and their corresponding parental PDXs contained the same genetic alterations. Importantly, CRCs retained the same drug sensitivity with the corresponding downstream signalling activity as their corresponding parental PDXs. This suggests that CRCs and PDXs can complement each other, and that CRCs can be used for in vitro fast, high throughput and low cost screening while PDXs can be used for in vivo validation and study of the in vivo factors during translational research and drug development.


Subject(s)
Urinary Bladder Neoplasms/drug therapy , Xenograft Model Antitumor Assays/methods , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Culture Techniques/economics , Cell Culture Techniques/methods , Disease Models, Animal , Drug Development , Drug Resistance, Neoplasm , Humans , Mice , Mutation , Translational Research, Biomedical , Tumor Cells, Cultured , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays/economics
7.
Am J Respir Cell Mol Biol ; 56(5): 568-574, 2017 05.
Article in English | MEDLINE | ID: mdl-27983869

ABSTRACT

Well-differentiated primary human bronchial epithelial (HBE) cell cultures are vital for cystic fibrosis (CF) research, particularly for the development of cystic fibrosis transmembrane conductance regulator (CFTR) modulator drugs. Culturing of epithelial cells with irradiated 3T3 fibroblast feeder cells plus the RhoA kinase inhibitor Y-27632 (Y), termed conditionally reprogrammed cell (CRC) technology, enhances cell growth and lifespan while preserving cell-of-origin functionality. We initially determined the electrophysiological and morphological characteristics of conventional versus CRC-expanded non-CF HBE cells. On the basis of these findings, we then created six CF cell CRC populations, three from sequentially obtained CF lungs and three from F508 del homozygous donors previously obtained and cryopreserved using conventional culture methods. Growth curves were plotted, and cells were subcultured, without irradiated feeders plus Y, into air-liquid interface conditions in nonproprietary and proprietary Ultroser G-containing media and were allowed to differentiate. Ussing chamber studies were performed after treatment of F508 del homozygous CF cells with the CFTR modulator VX-809. Bronchial epithelial cells grew exponentially in feeders plus Y, dramatically surpassing the numbers of conventionally grown cells. Passage 5 and 10 CRC HBE cells formed confluent mucociliary air-liquid interface cultures. There were differences in cell morphology and current magnitude as a function of extended passage, but the effect of VX-809 in increasing CFTR function was significant in CRC-expanded F508 del HBE cells. Thus, CRC technology expands the supply of functional primary CF HBE cells for testing CFTR modulators in Ussing chambers.


Subject(s)
Bronchi/pathology , Cellular Reprogramming , Cystic Fibrosis/pathology , Epithelial Cells/pathology , Animals , Cell Line , Cell Proliferation , Cell Shape , Cystic Fibrosis/physiopathology , Electrophysiological Phenomena , Humans , Mice
8.
N Engl J Med ; 367(13): 1220-7, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-23013073

ABSTRACT

A patient with a 20-year history of recurrent respiratory papillomatosis had progressive, bilateral tumor invasion of the lung parenchyma. We used conditional reprogramming to generate cell cultures from the patient's normal and tumorous lung tissue. Analysis revealed that the laryngeal tumor cells contained a wild-type 7.9-kb human papillomavirus virus type 11 (HPV-11) genome, whereas the pulmonary tumor cells contained a 10.4-kb genome. The increased size of the latter viral genome was due to duplication of the promoter and oncogene regions. Chemosensitivity testing identified vorinostat as a potential therapeutic agent. At 3 months after treatment initiation, tumor sizes had stabilized, with durable effects at 15 months.


Subject(s)
Antineoplastic Agents/therapeutic use , Hydroxamic Acids/therapeutic use , Lung Neoplasms/pathology , Lung/cytology , Papillomavirus Infections/drug therapy , Respiratory Tract Infections/drug therapy , Cells, Cultured , DNA, Viral/isolation & purification , Gene Expression , Genome, Viral , Human papillomavirus 11/genetics , Humans , Laryngeal Neoplasms/surgery , Laryngeal Neoplasms/virology , Lung Neoplasms/drug therapy , Lung Neoplasms/virology , Male , Mutation , Papillomavirus Infections/complications , Papillomavirus Infections/pathology , Papillomavirus Infections/surgery , RNA, Messenger/metabolism , RNA, Viral/analysis , Respiratory Tract Infections/complications , Respiratory Tract Infections/pathology , Respiratory Tract Infections/surgery , Tumor Cells, Cultured , Vorinostat , Young Adult
9.
PLoS Pathog ; 9(4): e1003284, 2013.
Article in English | MEDLINE | ID: mdl-23592995

ABSTRACT

Previous studies have shown that wild-type human telomerase reverse transcriptase (hTERT) protein can functionally replace the human papillomavirus type 16 (HPV-16) E6 protein, which cooperates with the viral E7 protein in the immortalization of primary keratinocytes. In the current study, we made the surprising finding that catalytically inactive hTERT (hTERT-D868A), elongation-defective hTERT (hTERT-HA), and telomere recruitment-defective hTERT (hTERT N+T) also cooperate with E7 in mediating bypass of the senescence blockade and effecting cell immortalization. This suggests that hTERT has activities independent of its telomere maintenance functions that mediate transit across this restriction point. Since hTERT has been shown to have a role in gene activation, we performed microarray studies and discovered that E6, hTERT and mutant hTERT proteins altered the expression of highly overlapping sets of cellular genes. Most important, the E6 and hTERT proteins induced mRNA and protein levels of Bmi1, the core subunit of the Polycomb Group (PcG) complex 1. We show further that Bmi1 substitutes for E6 or hTERT in cell immortalization. Finally, tissue array studies demonstrated that expression of Bmi1 increased with the severity of cervical dysplasia, suggesting a potential role in the progression of cervical cancer. Together, these data demonstrate that hTERT has extra-telomeric activities that facilitate cell immortalization and that its induction of Bmi1 is one potential mechanism for mediating this activity.


Subject(s)
Cell Transformation, Viral , Human papillomavirus 16/metabolism , Keratinocytes/physiology , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Polycomb Repressive Complex 1/metabolism , Repressor Proteins/metabolism , Telomerase/metabolism , Human papillomavirus 16/genetics , Humans , Keratinocytes/cytology , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Polycomb Repressive Complex 1/genetics , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Telomerase/genetics , Telomere/genetics , Telomere/physiology
10.
Proc Natl Acad Sci U S A ; 109(49): 20035-40, 2012 Dec 04.
Article in English | MEDLINE | ID: mdl-23169653

ABSTRACT

The combination of irradiated fibroblast feeder cells and Rho kinase inhibitor, Y-27632, conditionally induces an indefinite proliferative state in primary mammalian epithelial cells. These conditionally reprogrammed cells (CRCs) are karyotype-stable and nontumorigenic. Because self-renewal is a recognized property of stem cells, we investigated whether Y-27632 and feeder cells induced a stem-like phenotype. We found that CRCs share characteristics of adult stem cells and exhibit up-regulated expression of α6 and ß1 integrins, ΔNp63α, CD44, and telomerase reverse transcriptase, as well as decreased Notch signaling and an increased level of nuclear ß-catenin. The induction of CRCs is rapid (occurs within 2 d) and results from reprogramming of the entire cell population rather than the selection of a minor subpopulation. CRCs do not overexpress the transcription factor sets characteristic of embryonic or induced pluripotent stem cells (e.g., Sox2, Oct4, Nanog, or Klf4). The induction of CRCs is also reversible, and removal of Y-27632 and feeders allows the cells to differentiate normally. Thus, when CRCs from ectocervical epithelium or tracheal epithelium are placed in an air-liquid interface culture system, the cervical cells form a well differentiated stratified squamous epithelium, whereas the tracheal cells form a ciliated airway epithelium. We discuss the diagnostic and therapeutic opportunities afforded by a method that can generate adult stem-like cells in vitro without genetic manipulation.


Subject(s)
Adult Stem Cells/cytology , Amides/pharmacology , Cell Proliferation/drug effects , Cellular Reprogramming/physiology , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Pyridines/pharmacology , Adult Stem Cells/drug effects , Antigens, Surface/metabolism , Blotting, Western , Cellular Reprogramming/drug effects , Epithelial Cells/drug effects , Feeder Cells , Flow Cytometry , Humans , Hyaluronan Receptors/metabolism , Immunohistochemistry , Integrin beta1/metabolism , Karyotyping , Kruppel-Like Factor 4 , Real-Time Polymerase Chain Reaction , Telomerase/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL