1
|
Gbala I, Kavcic N, Banks L. The retinoblastoma protein contributes to maintaining the stability of HPV E7 in cervical cancer cells. J Virol 2025:e0220324. [PMID: 40130877 DOI: 10.1128/jvi.02203-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/25/2025] [Indexed: 03/26/2025] Open
Abstract
High-risk human papillomaviruses (HR HPV)-16 and -18, and other closely related subtypes, are associated with at least 90% of human cervical cancers. Cervical cancers and derived cell lines continuously express high levels of the HPV oncoprotein E7, known to degrade the tumor suppressor retinoblastoma protein (pRB). This E7-pRB interaction is important for the maintenance and progression of malignancy. In the case of HPV E6, substrate recognition has been reported to play an important role in stabilizing the viral oncoprotein; however, such regulation of E7 has so far not been investigated. Using biochemical, immunostaining, and clonogenic assays, we describe an intriguing role for pRB in the stabilization of E7 oncoprotein in HPV-positive cervical-cancer-derived cell lines. The knockdown of pRB expression by RNA interference results in a significant decrease in the levels of E7 protein in CaSki, SiHa, HeLa, and C-4 I cells. We show that pRB knockdown regulates HPV E7 at the transcription and protein levels, and significantly reduces the half-life of E7 protein by at least twofold in SiHa and HeLa cells. We also demonstrate that the destabilization of E7 caused by pRB knockdown results in significant inhibition of cell proliferation and colony formation of HPV-16 and -18 E7-positive SiHa and HeLa cells. Furthermore, the expression of wild-type pRB in pRB-depleted cells significantly restored E7 levels. Therefore, we propose that pRB, in addition to being a degradation target for HPV E7, is crucial for its stabilization.IMPORTANCEThe human papillomavirus (HPV) viral proteins E6 and E7 cooperatively contribute to tumorigenesis by disrupting cellular targets. These oncoproteins are degraded via the proteasome pathway; however, they are continuously expressed in cervical cancer cell lines. The retinoblastoma protein, pRB, is a degradation target of high-risk (HR) HPV E7 oncoprotein. Several studies have shown that the binding of E7 to pRB is important for its E7-mediated inactivation and demonstrated how pRB protein levels respond to the presence and absence of E7. However, the modulatory role of pRB on E7 protein levels has so far not been reported. Here, we report a novel regulatory relationship between E7 and pRB. We found that the continuous expression of pRB is critical for E7 stabilization. We demonstrate that this pRB-related E7 destabilization occurs in part through enhanced protein turnover. Thus, our findings provide new insights into the importance of the E7-pRB interaction in driving tumorigenesis.
Collapse
Affiliation(s)
- Ifeoluwa Gbala
- Tumour Virology Laboratory, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Nezka Kavcic
- Tumour Virology Laboratory, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Lawrence Banks
- Tumour Virology Laboratory, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| |
Collapse
|
2
|
Otálora-Otálora BA, Payán-Gómez C, López-Rivera JJ, Pedroza-Aconcha NB, Arboleda-Mojica SL, Aristizábal-Guzmán C, Isaza-Ruget MA, Álvarez-Moreno CA. Interplay of Transcriptomic Regulation, Microbiota, and Signaling Pathways in Lung and Gut Inflammation-Induced Tumorigenesis. Cells 2024; 14:1. [PMID: 39791702 PMCID: PMC11720097 DOI: 10.3390/cells14010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/15/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
Inflammation can positively and negatively affect tumorigenesis based on the duration, scope, and sequence of related events through the regulation of signaling pathways. A transcriptomic analysis of five pulmonary arterial hypertension, twelve Crohn's disease, and twelve ulcerative colitis high throughput sequencing datasets using R language specialized libraries and gene enrichment analyses identified a regulatory network in each inflammatory disease. IRF9 and LINC01089 in pulmonary arterial hypertension are related to the regulation of signaling pathways like MAPK, NOTCH, human papillomavirus, and hepatitis c infection. ZNF91 and TP53TG1 in Crohn's disease are related to the regulation of PPAR, MAPK, and metabolic signaling pathways. ZNF91, VDR, DLEU1, SATB2-AS1, and TP53TG1 in ulcerative colitis are related to the regulation of PPAR, AMPK, and metabolic signaling pathways. The activation of the transcriptomic network and signaling pathways might be related to the interaction of the characteristic microbiota of the inflammatory disease, with the lung and gut cell receptors present in membrane rafts and complexes. The transcriptomic analysis highlights the impact of several coding and non-coding RNAs, suggesting their relationship with the unlocking of cell phenotypic plasticity for the acquisition of the hallmarks of cancer during lung and gut cell adaptation to inflammatory phenotypes.
Collapse
Affiliation(s)
| | - César Payán-Gómez
- Dirección Académica, Universidad Nacional de Colombia, Sede de La Paz, La Paz 202017, Colombia; (C.P.-G.); (N.B.P.-A.)
| | - Juan Javier López-Rivera
- Grupo de Investigación INPAC, Specialized Laboratory, Clinica Universitaria Colombia, Clínica Colsanitas S.A., Bogotá 111321, Colombia;
| | - Natalia Belén Pedroza-Aconcha
- Dirección Académica, Universidad Nacional de Colombia, Sede de La Paz, La Paz 202017, Colombia; (C.P.-G.); (N.B.P.-A.)
| | | | - Claudia Aristizábal-Guzmán
- Grupo de Investigación INPAC, Unidad de Investigación, Fundación Universitaria Sanitas, Bogotá 110131, Colombia;
| | - Mario Arturo Isaza-Ruget
- Keralty, Sanitas International Organization, Grupo de Investigación INPAC, Fundación Universitaria Sanitas, Bogotá 110131, Colombia;
| | - Carlos Arturo Álvarez-Moreno
- Infectious Diseases Department, Clinica Universitaria Colombia, Clínica Colsanitas S.A., Bogotá 111321, Colombia;
| |
Collapse
|
3
|
Broniarczyk J, Trejo-Cerro O, Massimi P, Kavčič N, Myers MP, Banks L. HPV-18 E6 enhances the interaction between EMILIN2 and SNX27 to promote WNT signaling. J Virol 2024; 98:e0073524. [PMID: 38874360 PMCID: PMC11265340 DOI: 10.1128/jvi.00735-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Abstract
Oncogenic HPV E6 proteins have a PDZ-binding motif (PBM) which plays important roles in both the viral life cycle and tumor development. The PBM confers interaction with a large number of different PDZ domain-containing substrates, one of which is Sorting Nexin 27. This protein is part of the retromer complex and plays an important role in endocytic sorting pathways. It has been shown that at least two SNX27 interacting partners, GLUT1 and TANC2, are aberrantly trafficked due to the E6 PBM-dependent interaction with SNX27. To investigate further which other components of the endocytic trafficking pathway might be affected by the SNX27-HPV E6 interaction, we analyzed the SNX27 proteome interaction profile in a previously described HeLa cell line expressing GFP-SNX27, both in the presence and absence of the HPV-18 E6 oncoprotein. In this study, we identify a novel interacting partner of SNX27, secreted glycoprotein EMILIN2, whose release is blocked by HPV18 E6 in a PBM-dependent manner. Mechanistically, E6 can block EMILIN2 interaction with the WNT1 ligand, thereby enhancing WNT1 signaling and promoting cell proliferation. IMPORTANCE This study demonstrates that HPV E6 blocks EMILIN2 inhibition of WNT1 signaling, thereby enhancing cell proliferation in HPV-positive tumor cells. This involves a novel mechanism whereby the E6 PBM actually contributes toward enhancing the interaction between SNX27 and EMILIN2, suggesting that the mode of recognition of SNX27 by E6 and EMILIN2 is different. This is the first example of the E6 PBM altering a PDZ domain-containing protein to enhance potential substrate recognition.
Collapse
Affiliation(s)
- Justyna Broniarczyk
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
- Department of Molecular Virology, Adam Mickiewicz University, Poznan, Poland
| | - Oscar Trejo-Cerro
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Paola Massimi
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Nežka Kavčič
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Michael P. Myers
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Lawrence Banks
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| |
Collapse
|
4
|
Catalán-Castorena O, Garibay-Cerdenares OL, Illades-Aguiar B, Castillo-Sánchez R, Zubillaga-Guerrero MI, Leyva-Vazquez MA, Encarnacion-Guevara S, Flores-Alfaro E, Ramirez-Ruano M, del Carmen Alarcón-Romero L. Bioinformatics Analysis of Human Papillomavirus 16 Integration in Cervical Cancer: Changes in MAGI-1 Expression in Premalignant Lesions and Invasive Carcinoma. Cancers (Basel) 2024; 16:2225. [PMID: 38927930 PMCID: PMC11202195 DOI: 10.3390/cancers16122225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/05/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
HPV 16 integration is crucial for the onset and progression of premalignant lesions to invasive squamous cell carcinoma (ISCC) because it promotes the amplification of proto-oncogenes and the silencing of tumor suppressor genes; some of these are proteins with PDZ domains involved in homeostasis and cell polarity. Through a bioinformatics approach based on interaction networks, a group of proteins associated with HPV 16 infection, PDZ domains, and direct physical interaction with E6 and related to different hallmarks of cancer were identified. MAGI-1 was selected to evaluate the expression profile and subcellular localization changes in premalignant lesions and ISCC with HPV 16 in an integrated state in cervical cytology; the profile expression of MAGI-1 diminished according to lesion grade. Surprisingly, in cell lines CaSki and SiHa, the protein localization was cytoplasmic and nuclear. In contrast, in histological samples, a change in subcellular localization from the cytoplasm in low-grade squamous intraepithelial lesions (LSIL) to the nucleus in the high-grade squamous intraepithelial lesion (HSIL) was observed; in in situ carcinomas and ISCC, MAGI-1 expression was absent. In conclusion, MAGI-1 expression could be a potential biomarker for distinguishing those cells with normal morphology but with HPV 16 integrated from those showing morphology-related uterine cervical lesions associated with tumor progression.
Collapse
Affiliation(s)
- Oscar Catalán-Castorena
- Cytopathology and Histochemistry Research Laboratory, Faculty of Chemical and Biological Sciences, Autonomous University of Guerrero, Chilpancingo 39070, Guerrero, Mexico; (O.C.-C.); (M.I.Z.-G.)
| | - Olga Lilia Garibay-Cerdenares
- Molecular Biomedicine Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo 39070, Guerrero, Mexico; (B.I.-A.); (M.A.L.-V.)
- CONAHCyT-Autonomous University of Guerrero, Chilpancingo 39070, Guerrero, Mexico
| | - Berenice Illades-Aguiar
- Molecular Biomedicine Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo 39070, Guerrero, Mexico; (B.I.-A.); (M.A.L.-V.)
| | - Rocio Castillo-Sánchez
- Cell Biology Department, CINVESTAV-IPN Research Institute, Ciudad de México 07360, Mexico;
| | - Ma. Isabel Zubillaga-Guerrero
- Cytopathology and Histochemistry Research Laboratory, Faculty of Chemical and Biological Sciences, Autonomous University of Guerrero, Chilpancingo 39070, Guerrero, Mexico; (O.C.-C.); (M.I.Z.-G.)
| | - Marco Antonio Leyva-Vazquez
- Molecular Biomedicine Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo 39070, Guerrero, Mexico; (B.I.-A.); (M.A.L.-V.)
| | - Sergio Encarnacion-Guevara
- Center for Genomic Sciences, National Autonomous University of Mexico, Cuernavaca 62210, Morelos, Mexico;
| | - Eugenia Flores-Alfaro
- Clinical and Molecular Epidemiology Research Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo 39070, Guerrero, Mexico;
| | - Mónica Ramirez-Ruano
- Functional Genomics and Proteomics Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo 39070, Guerrero, Mexico;
| | - Luz del Carmen Alarcón-Romero
- Cytopathology and Histochemistry Research Laboratory, Faculty of Chemical and Biological Sciences, Autonomous University of Guerrero, Chilpancingo 39070, Guerrero, Mexico; (O.C.-C.); (M.I.Z.-G.)
| |
Collapse
|
5
|
Trejo-Cerro O, Broniarczyk J, Kavcic N, Myers M, Banks L. Identification and characterisation of novel potential phospho-acceptor sites in HPV-16 E7. Tumour Virus Res 2023; 16:200270. [PMID: 37659653 PMCID: PMC10500460 DOI: 10.1016/j.tvr.2023.200270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/04/2023] Open
Abstract
Several studies have described functional regulation of high-risk human papillomaviruses (HPVs), E6 and E7 oncoproteins via posttranslational modifications (PTMs). However, how these PTMs modulate the activity of E6 and E7, particularly in their targeting of cellular proteins, is not completely understood. In this study, we show that HPV16 E7 can be phosphorylated by casein kinase I (CKI) and glycogen synthase kinase 3 (GSK3). This principal phosphorylation occurs at threonine residues 5 and 7 with a more minor role for residues 19-20 in the N-terminal region of 16 E7. Intriguingly, whilst mutational analyses suggest that residues 5 and 7 may be dispensable for the transformation of primary baby rat kidney cells by E7, intact residues 19 and 20 are required. Furthermore, negative charges at these residues (TT19-20DD) enhance the pRb-E7 interaction and cells display increased proliferation and invasion capacities. Using a proteomic approach with a phosphorylated peptide spanning the TT19-20 region of HPV16 E7, we have identified a panel of new, phospho-specific E7 interacting partners. These results shed new light on the complexity of N-terminal phosphorylation of E7 and how this can contribute towards expanding the repertoire of E7 targeted pathways.
Collapse
Affiliation(s)
- Oscar Trejo-Cerro
- International Centre for Genetic Engineering and Biotechnology, Padriciano 99, I-34149, Trieste, Italy.
| | - Justyna Broniarczyk
- International Centre for Genetic Engineering and Biotechnology, Padriciano 99, I-34149, Trieste, Italy; Department of Molecular Virology, Adam Mickiewicz University, Uniwersytetu Poznanskiego 6, 61-614, Poznan, Poland
| | - Nezka Kavcic
- International Centre for Genetic Engineering and Biotechnology, Padriciano 99, I-34149, Trieste, Italy
| | - Michael Myers
- International Centre for Genetic Engineering and Biotechnology, Padriciano 99, I-34149, Trieste, Italy
| | - Lawrence Banks
- International Centre for Genetic Engineering and Biotechnology, Padriciano 99, I-34149, Trieste, Italy.
| |
Collapse
|
6
|
Javorsky A, Humbert PO, Kvansakul M. Viral manipulation of cell polarity signalling. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119536. [PMID: 37437846 DOI: 10.1016/j.bbamcr.2023.119536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/24/2023] [Accepted: 07/04/2023] [Indexed: 07/14/2023]
Abstract
Cell polarity refers to the asymmetric distribution of biomacromolecules that enable the correct orientation of a cell in a particular direction. It is thus an essential component for appropriate tissue development and function. Viral infections can lead to dysregulation of polarity. This is associated with a poor prognosis due to viral interference with core cell polarity regulatory scaffolding proteins that often feature PDZ (PSD-95, DLG, and ZO-1) domains including Scrib, Dlg, Pals1, PatJ, Par3 and Par6. PDZ domains are also promiscuous, binding to several different partners through their C-terminal region which contain PDZ-binding motifs (PBM). Numerous viruses encode viral effector proteins that target cell polarity regulators for their benefit and include papillomaviruses, flaviviruses and coronaviruses. A better understanding of the mechanisms of action utilised by viral effector proteins to subvert host cell polarity sigalling will provide avenues for future therapeutic intervention, while at the same time enhance our understanding of cell polarity regulation and its role tissue homeostasis.
Collapse
Affiliation(s)
- Airah Javorsky
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Patrick O Humbert
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia; Research Centre for Molecular Cancer Prevention, La Trobe University, Melbourne, Victoria 3086, Australia; Department of Biochemistry & Pharmacology, University of Melbourne, Melbourne, Victoria 3010, Australia; Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Marc Kvansakul
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia; Research Centre for Molecular Cancer Prevention, La Trobe University, Melbourne, Victoria 3086, Australia.
| |
Collapse
|
7
|
Hu J, Ji Y, Miao T, Zheng S, Cui X, Hu J, Yang L, Li F. HPV 16 E6 promotes growth and metastasis of esophageal squamous cell carcinoma cells in vitro. Mol Biol Rep 2023; 50:1181-1190. [PMID: 36435921 DOI: 10.1007/s11033-022-07952-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 09/16/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the most lethal malignancies worldwide. Increasing evidence suggests that human papillomavirus (HPV) infection may be associated with the etiology of ESCC. However, the precise role of HPV in ESCC remains unclear. METHODS AND RESULTS Proliferation and apoptosis of ESCC cells upon infection with HPV16 E6 were detected using CCK-8 assays and Western blot analyses. The migration rate was measured with a wound healing assay, and a Transwell Matrigel invasion assay was used to detect the invasive ability. RT-qPCR was performed to detect the expression of E6AP, p53, and miR-34a. The proliferation rates were significantly higher in HPV16E6-transfected cell groups compared with the negative control groups. Bax protein expression was downregulated in HPV16E6-treated groups compared to the controls. The wound healing and Transwell Matrigel invasion assays indicated that HPV16 E6 infection could increase ESCC cell migration and invasion. Furthermore, E6AP, p53 and miR-34a expression were decreased in HPV16 E6-transfected cell lines. CONCLUSION Our results not only provide evidence that HPV16 E6 promotes cell proliferation, migration, and invasion in ESCC, but also suggests a correlation between HPV infection and E6AP, p53 and miR-34a expression. Consequently, HPV16 E6 may play an important role in ESCC development.
Collapse
Affiliation(s)
- JiaoJiao Hu
- Department of Pathology, Shihezi University School of Medicine, 832000, Shihezi, Xinjiang, People's Republic of China
| | - Yu Ji
- Department of Pathology, Shihezi University School of Medicine, 832000, Shihezi, Xinjiang, People's Republic of China
- Pathology Department, Jiangmen Maternity and Child Health Care Hospital, Guangdong, 529000, Jiangmen, People's Republic of China
| | - TingTing Miao
- Department of Pathology, Shihezi University School of Medicine, 832000, Shihezi, Xinjiang, People's Republic of China
| | - ShiYao Zheng
- Department of Pathology, Shihezi University School of Medicine, 832000, Shihezi, Xinjiang, People's Republic of China
- Department of Pathology, The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi University School of Medicine, 832000, Shihezi, Xinjiang, China
| | - XiaoBin Cui
- Department of Pathology, Shihezi University School of Medicine, 832000, Shihezi, Xinjiang, People's Republic of China
- Department of Pathology, The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi University School of Medicine, 832000, Shihezi, Xinjiang, China
| | - JianMing Hu
- Department of Pathology, Shihezi University School of Medicine, 832000, Shihezi, Xinjiang, People's Republic of China
- Department of Pathology, The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi University School of Medicine, 832000, Shihezi, Xinjiang, China
| | - Lan Yang
- Department of Pathology, Shihezi University School of Medicine, 832000, Shihezi, Xinjiang, People's Republic of China.
- Department of Pathology, The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi University School of Medicine, 832000, Shihezi, Xinjiang, China.
| | - Feng Li
- Department of Pathology, Shihezi University School of Medicine, 832000, Shihezi, Xinjiang, People's Republic of China.
- Department of Pathology, The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi University School of Medicine, 832000, Shihezi, Xinjiang, China.
- Department of Pathology and Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, 100020, Beijing, People's Republic of China.
| |
Collapse
|
8
|
Hashemi L, Ormsbee ME, Patel PJ, Nielson JA, Ahlander J, Padash Barmchi M. A Drosophila model of HPV16-induced cancer reveals conserved disease mechanism. PLoS One 2022; 17:e0278058. [PMID: 36508448 PMCID: PMC9744332 DOI: 10.1371/journal.pone.0278058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/09/2022] [Indexed: 12/14/2022] Open
Abstract
High-risk human papillomaviruses (HR-HPVs) cause almost all cervical cancers and a significant number of vaginal, vulvar, penile, anal, and oropharyngeal cancers. HPV16 and 18 are the most prevalent types among HR-HPVs and together cause more than 70% of all cervical cancers. Low vaccination rate and lack of molecularly-targeted therapeutics for primary therapy have led to a slow reduction in cervical cancer incidence and high mortality rate. Hence, creating new models of HPV-induced cancer that can facilitate understanding of the disease mechanism and identification of key cellular targets of HPV oncogenes are important for development of new interventions. Here in this study, we used the tissue-specific expression technique, Gal4-UAS, to establish the first Drosophila model of HPV16-induced cancer. Using this technique, we expressed HPV16 oncogenes E5, E6, E7 and the human E3 ligase (hUBE3A) specifically in the epithelia of Drosophila eye, which allows simple phenotype scoring without affecting the viability of the organism. We found that, as in human cells, hUBE3A is essential for cellular abnormalities caused by HPV16 oncogenes in flies. Several proteins targeted for degradation by HPV16 oncoproteins in human cells were also reduced in the Drosophila epithelial cells. Cell polarity and adhesion were compromised, resulting in impaired epithelial integrity. Cells did not differentiate to the specific cell types of ommatidia, but instead were transformed into neuron-like cells. These cells extended axon-like structures to connect to each other and exhibited malignant behavior, migrating away to distant sites. Our findings suggest that given the high conservation of genes and signaling pathways between humans and flies, the Drosophila model of HPV16- induced cancer could serve as an excellent model for understanding the disease mechanism and discovery of novel molecularly-targeted therapeutics.
Collapse
Affiliation(s)
- Lydia Hashemi
- Department of Biology, University of Oklahoma, Norman, OK, United States of America
| | - McKenzi E. Ormsbee
- Department of Biology, University of Oklahoma, Norman, OK, United States of America
| | - Prashant J. Patel
- Department of Biology, University of Oklahoma, Norman, OK, United States of America
| | - Jacquelyn A. Nielson
- Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA, United States of America
| | - Joseph Ahlander
- Department of Natural Sciences, Northeastern State University, Broken Arrow, OK, United States of America
| | - Mojgan Padash Barmchi
- Department of Biology, University of Oklahoma, Norman, OK, United States of America
- * E-mail:
| |
Collapse
|
9
|
Human DLG1 and SCRIB Are Distinctly Regulated Independently of HPV-16 during the Progression of Oropharyngeal Squamous Cell Carcinomas: A Preliminary Analysis. Cancers (Basel) 2021; 13:cancers13174461. [PMID: 34503271 PMCID: PMC8430552 DOI: 10.3390/cancers13174461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 11/30/2022] Open
Abstract
Simple Summary The process of HPV-mediated oncogenesis in HNSCCs is not fully understood. DLG1 and SCRIB protein expression levels and localization changes were evaluated in a number of HPV16-positive and HPV-negative OPSCCs and seem to be associated with malignant transformation. Moreover, loss of SCRIB expression inversely correlates with higher grade tumors, and this is much more evident in the presence of HPV16 E6. This could serve as a potential marker in predicting development of OPSCCs. Abstract The major causative agents of head and neck squamous cell carcinomas (HNSCCs) are either environmental factors, such as tobacco and alcohol consumption, or infection with oncogenic human papillomaviruses (HPVs). An important aspect of HPV-induced oncogenesis is the targeting by the E6 oncoprotein of PDZ domain-containing substrates for proteasomal destruction. Tumor suppressors DLG1 and SCRIB are two of the principal PDZ domain-containing E6 targets. Both have been shown to play critical roles in the regulation of cell growth and polarity and in maintaining the structural integrity of the epithelia. We investigated how modifications in the cellular localization and protein expression of DLG1 and SCRIB in HPV16-positive and HPV-negative histologic oropharyngeal squamous cell carcinomas (OPSCC) might reflect disease progression. HPV presence was determined by p16 staining and HPV genotyping. Whilst DLG1 expression levels did not differ markedly between HPV-negative and HPV16-positive OPSCCs, it appeared to be relocated from cell–cell contacts to the cytoplasm in most samples, regardless of HPV16 positivity. This indicates that alterations in DLG1 distribution could contribute to malignant progression in OPSCCs. Interestingly, SCRIB was also relocated from cell–cell contacts to the cytoplasm in the tumor samples in comparison with normal tissue, regardless of HPV16 status, but in addition there was an obvious reduction in SCRIB expression in higher grade tumors. Strikingly, loss of SCRIB was even more pronounced in HPV16-positive OPSCCs. These alterations in SCRIB levels may contribute to transformation and loss of tissue architecture in the process of carcinogenesis and could potentially serve as markers in the development of OPSCCs.
Collapse
|
10
|
Expression of Retroelements in Cervical Cancer and Their Interplay with HPV Infection and Host Gene Expression. Cancers (Basel) 2021; 13:cancers13143513. [PMID: 34298727 PMCID: PMC8306386 DOI: 10.3390/cancers13143513] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/02/2021] [Accepted: 07/02/2021] [Indexed: 12/11/2022] Open
Abstract
Retroelements are expressed in diverse types of cancer and are related to tumorigenesis and to cancer progression. We characterized the expression of retroelements in cervical cancer and explored their interplay with HPV infection and their association with expression of neighboring genes. Forty biopsies of invasive cervical carcinoma (squamous cell carcinomas and adenocarcinomas) with genotyped HPV were selected and analyzed for human endogenous retrovirus (HERV) and long interspersed nuclear element 1 (L1) expression through RNA-seq data. We found 8060 retroelements expressed in the samples and a negative correlation of DNA methyltransferase 1 expression with the two most expressed L1 elements. A total of 103 retroelements were found differentially expressed between tumor histological types and between HPV types, including several HERV families (HERV-K, HERV-H, HERV-E, HERV-I and HERV-L). The comparison between HPV mono- and co-infections showed the highest proportion of differentially expressed L1 elements. The location of retroelements affected neighboring gene expression, such as shown for the interleukin-20 gene family. Three HERVs and seven L1 were located close to this gene family and two L1 showed a positive association with IL20RB expression. This study describes the expression of retroelements in cervical cancer and shows their association with HPV status and host gene expression.
Collapse
|
11
|
Chen Y, Hu X, Yang S. Clinical significance of focal adhesion kinase (FAK) in cervical cancer progression and metastasis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:2586-2592. [PMID: 33165433 PMCID: PMC7642699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/20/2020] [Indexed: 06/11/2023]
Abstract
Focal adhesion kinase is a non-receptor, tyrosine kinase of cells whose key functions are cell adhesion, migration, and invasion. Aberrant expression and regulation of FAK-mediated intracellular signaling pathways has been reported in several cancers and they are involved in cancer cell migration and apoptosis resistance. By RT-PCR, we found that cervical cancer cells showed a 4-fold increase of relative mRNA expression of FAK compared to control cells. In parallel, the FAK protein expression level was also elevated in cervical cancer cells. Interestingly, knockdown of FAK in cervical cancer cells showed attenuated cell proliferation and migration. Further, the FAK RNAi cells became more sensitive to chemotherapeutic drugs such as 5-FU and docetaxel and therefore the rate of cell survival is declined. The significant over-expression of FAK in cervical cancer cells might involve in cervical carcinogenesis and prolonged cell survival. This FAK overexpression might be a potential target for anti-cancer drugs to attenuate rapid cell proliferation and invasion by inducing apoptosis.
Collapse
Affiliation(s)
- Yanxian Chen
- Department of Obstetrics and Gynecology, Suzhou Hospital of Integrated Traditional Chinese and Western MedicineSuzhou, Jiangsu, China
| | - Xiaofeng Hu
- Department of Obstetrics and Gynecology, Suzhou Hospital of Integrated Traditional Chinese and Western MedicineSuzhou, Jiangsu, China
| | - Shu Yang
- Department of Abdominal Oncology, The Second Affiliated Gospital of Zunyi Medical UniversityZunyi 563000, Guizhou, China
| |
Collapse
|