1
|
Xu XS, Ma YS, Dai RH, Zhang HL, Yang QX, Fan QY, Liu XY, Liu JB, Feng WW, Meng H, Fu D, Yu H, Shen J. Identification of novel genomic hotspots and tumor-relevant genes via comprehensive analysis of HPV integration in Chinese patients of cervical cancer. Am J Cancer Res 2024; 14:4665-4682. [PMID: 39417198 PMCID: PMC11477843 DOI: 10.62347/kkle8602] [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: 04/03/2024] [Accepted: 09/04/2024] [Indexed: 10/19/2024] Open
Abstract
Cervical cancer accounts for 10-15% of cancer-related mortality among women globally. Infection with high-risk human papillomavirus (HPV) types constitutes a significant etiological factor in the development of cervical carcinoma. The integration of HPV DNA into the host genome is considered a pivotal event in cervical carcinogenesis. Nevertheless, the precise mechanisms underlying HPV integration and its role in promoting cancer progression remain inadequately understood. Therefore, this study aims to identify potential common denominators at HPV DNA integration sites and to analyze the adjacent cellular sequences. We conducted whole-genome sequencing on 13 primary cervical cancer samples, employing the chromosomal coordinates of 537 breakpoints to assess the statistical overrepresentation of integration sites in relation to various chromatin features. Our analysis, which encompassed all chromosomes, identified several integration hotspots within the human genome, notably at 14q32.2, 10p15, and 2q37. Additionally, our findings indicated a preferential integration of HPV DNA into intragenic and gene-dense regions of human chromosomes. A substantial number of host cellular genes impacted by the integration sites were associated with cancer, including IKZF2, IL26, AHRR, and PDCD6. Furthermore, the cellular genes targeted by integration were enriched in tumor-related terms and pathways, as demonstrated by gene ontology and KEGG analysis. In conclusion, these findings enhance our understanding of HPV integration sites and provide deeper insights into the molecular mechanisms underlying the pathogenesis of cervical carcinoma.
Collapse
Affiliation(s)
- Xiao-Sheng Xu
- Department of Obstetrics and Gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
| | - Yu-Shui Ma
- Institute of Oncology, Affiliated Tumor Hospital of Nantong UniversityNantong 226631, Jiangsu, China
- Department of Pathology, The Affiliated Taizhou People’s Hospital of Nanjing Medical UniversityTaizhou 225300, Jiangsu, China
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| | - Rong-Hua Dai
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong UniversityShanghai 200240, China
| | - Huan-Le Zhang
- Department of Radiotherapy, Suzhou Ninth People’s HospitalSuzhou 215299, Jiangsu, China
| | - Qin-Xin Yang
- Department of Pathology, The Affiliated Taizhou People’s Hospital of Nanjing Medical UniversityTaizhou 225300, Jiangsu, China
- Department of Pathology, Taizhou School of Clinical Medicine, Nanjing Medical UniversityTaizhou 225300, Jiangsu, China
| | - Qi-Yu Fan
- Institute of Oncology, Affiliated Tumor Hospital of Nantong UniversityNantong 226631, Jiangsu, China
| | - Xin-Yun Liu
- Department of Pathology, The Affiliated Taizhou People’s Hospital of Nanjing Medical UniversityTaizhou 225300, Jiangsu, China
| | - Ji-Bin Liu
- Institute of Oncology, Affiliated Tumor Hospital of Nantong UniversityNantong 226631, Jiangsu, China
| | - Wei-Wei Feng
- Department of Obstetrics and Gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
| | - He Meng
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong UniversityShanghai 200240, China
| | - Da Fu
- Institute of Oncology, Affiliated Tumor Hospital of Nantong UniversityNantong 226631, Jiangsu, China
- Department of Pathology, The Affiliated Taizhou People’s Hospital of Nanjing Medical UniversityTaizhou 225300, Jiangsu, China
| | - Hong Yu
- Department of Pathology, The Affiliated Taizhou People’s Hospital of Nanjing Medical UniversityTaizhou 225300, Jiangsu, China
- Department of Pathology, Taizhou School of Clinical Medicine, Nanjing Medical UniversityTaizhou 225300, Jiangsu, China
| | - Jian Shen
- Department of Obstetrics and Gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
| |
Collapse
|
2
|
Sisin NNT, Kong AR, Edinur HA, Jamil NIN, Che Mat NF. Silencing E6/E7 Oncoproteins in SiHa Cells Treated with siRNAs and Oroxylum indicum Extracts Induced Apoptosis by Upregulating p53/pRb Pathways. Appl Biochem Biotechnol 2024; 196:4234-4255. [PMID: 37922032 DOI: 10.1007/s12010-023-04762-w] [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] [Accepted: 10/19/2023] [Indexed: 11/05/2023]
Abstract
E6 and E7 human papillomavirus (HPV) oncoproteins play a significant role in the malignant transformation of infected cervical cancer cells via suppression of tumour suppressor pathways by targeting p53 and pRb, respectively. This study aimed to investigate the anticancer effects of Oroxylum indicum (OI) leaves' methanol extract on SiHa cervical cancer cells. Expression of apoptosis-related proteins (Bcl-2, caspase (cas)-3, and cas-9), viral oncoproteins (E6 and E7), and tumour suppressor proteins (p53 and pRb) were evaluated using western blot analysis before and after E6/E7 small interfering RNAs (siRNAs) transfection. In addition, the E6/E7 mRNA expression levels were assessed with real-time (RT)-PCR. The present study showed that the OI extract effectively hindered the proliferation of SiHa cells and instigated increments of cas-3 and cas-9 expressions but decreased the Bcl-2 expressions. The OI extract inhibited E6/E7 viral oncoproteins, leading to upregulation of p53 and pRb tumour suppressor genes in SiHa cells. Additionally, combinatorial treatment of OI extract and gossypin flavonoid induced restorations of p53 and pRb. Treatment with OI extract in siRNA-transfected cells also further suppressed E6/E7 expression levels and further upregulations of p53 and pRb proteins. In conclusion, OI extract treatment on siRNAs-transfected SiHa cells can additively and effectively block E6- and E7-dependent p53 and pRb degradations. All these data suggest that OI could be explored for its chemotherapeutic potential in cervical cancer cells with HPV-integrated genomes.
Collapse
Affiliation(s)
| | - Aaron Raphael Kong
- School of Health Sciences, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Hisham Atan Edinur
- School of Health Sciences, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Noor Izani Noor Jamil
- School of Health Sciences, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Nor Fazila Che Mat
- School of Health Sciences, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia.
| |
Collapse
|
3
|
Wang Y, Wang X, Liu Y, He Y, Duan X, Li Q, Huang Y, Xu G, Lu Q. HPV16-miRNAs exert oncogenic effects through enhancers in human cervical cancer. Cancer Cell Int 2024; 24:172. [PMID: 38750489 PMCID: PMC11097496 DOI: 10.1186/s12935-024-03364-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 05/08/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Cervical cancer is a human papillomavirus (HPV)-related disease. HPV type 16 (HPV16), which is the predominant cause of cervical cancer, can encode miRNAs (HPV16-miRNAs). However, the role of HPV16-miRNAs in the pathogenesis of cervical cancer remains unclear. METHODS Human cervical cancer cell lines SiHa (HPV16-positive) and C33A (HPV-negative), and cervical cancer tissues were collected to investigate the expression levels of two HPV16-miRNAs (HPV16-miR-H1 and HPV16-miR-H6). The overexpression and knockdown of HPV16-miR-H1 and HPV16-miR-H6 were performed using the lentiviral vector system and miRNA inhibitors, respectively. RNA-sequencing (RNA-seq) analysis and H3K27ac chromatin immunoprecipitation and sequencing (CHIP-seq) experiments were utilized to explore the roles of HPV16-miR-H1 and HPV16-miR-H6 facilitated by enhancers. CCK8, EdU, transwell, and wound healing assays were performed to verify the effects of HPV16-miR-H1 and HPV16-miR-H6 on cell proliferation and migration. RESULTS HPV16-miR-H1 and HPV16-miR-H6 were highly expressed in both SiHa cells and tissue samples from HPV16-positive cervical cancer patients. RNA-seq analysis showed that HPV16-miR-H1 and HPV16-miR-H6 induced the upregulation of numerous tumor progression-associated genes. H3K27ac CHIP-seq experiments further revealed that HPV16-miR-H1 and HPV16-miR-H6 modulated the expression of critical genes by regulating their enhancer activity. The functional study demonstrated that HPV16-miR-H1 and HPV16-miR-H6 increased the migratory capacity of SiHa cells. CONCLUSIONS Our data shed light on the role of HPV16-encoded miRNAs in cervical cancer, particularly emphasizing their involvement in the miRNA-enhancer-target gene system. This novel regulatory mechanism of HPV16-miRNAs provides new insights and approaches for the development of therapeutic strategies by targeting HPV16-positive cervical cancer.
Collapse
Affiliation(s)
- Yunuan Wang
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai, 201508, China
- Department of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xueying Wang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Clinical Research Center for Mental Health, Shanghai Institute of Traditional Chinese Medicine for Mental Health, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiting Liu
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai, 201508, China
- Department of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuxin He
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Xiaoling Duan
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Qinmei Li
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Yanchun Huang
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Guoxiong Xu
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Qi Lu
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai, 201508, China.
| |
Collapse
|
4
|
Wang Z, Liu C, Liu W, Lv X, Hu T, Yang F, Yang W, He L, Huang X. Long-read sequencing reveals the structural complexity of genomic integration of HPV DNA in cervical cancer cell lines. BMC Genomics 2024; 25:198. [PMID: 38378450 PMCID: PMC10877919 DOI: 10.1186/s12864-024-10101-y] [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: 06/16/2023] [Accepted: 02/08/2024] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Cervical cancer (CC) causes more than 311,000 deaths annually worldwide. The integration of human papillomavirus (HPV) is a crucial genetic event that contributes to cervical carcinogenesis. Despite HPV DNA integration is known to disrupt the genomic architecture of both the host and viral genomes in CC, the complexity of this process remains largely unexplored. RESULTS In this study, we conducted whole-genome sequencing (WGS) at 55-65X coverage utilizing the PacBio long-read sequencing platform in SiHa and HeLa cells, followed by comprehensive analyses of the sequence data to elucidate the complexity of HPV integration. Firstly, our results demonstrated that PacBio long-read sequencing effectively identifies HPV integration breakpoints with comparable accuracy to targeted-capture Next-generation sequencing (NGS) methods. Secondly, we constructed detailed models of complex integrated genome structures that included both the HPV genome and nearby regions of the human genome by utilizing PacBio long-read WGS. Thirdly, our sequencing results revealed the occurrence of a wide variety of genome-wide structural variations (SVs) in SiHa and HeLa cells. Additionally, our analysis further revealed a potential correlation between changes in gene expression levels and SVs on chromosome 13 in the genome of SiHa cells. CONCLUSIONS Using PacBio long-read sequencing, we have successfully constructed complex models illustrating HPV integrated genome structures in SiHa and HeLa cells. This accomplishment serves as a compelling demonstration of the valuable capabilities of long-read sequencing in detecting and characterizing HPV genomic integration structures within human cells. Furthermore, these findings offer critical insights into the complex process of HPV16 and HPV18 integration and their potential contribution to the development of cervical cancer.
Collapse
Affiliation(s)
- Zhijie Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chen Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Wanxin Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Xinyi Lv
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Ting Hu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Fan Yang
- Wuhan Kandwise Biotechnology, Inc. Wuhan, Hubei, China
| | - Wenhui Yang
- Wuhan Kandwise Biotechnology, Inc. Wuhan, Hubei, China
| | - Liang He
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Xiaoyuan Huang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
5
|
Rosendo-Chalma P, Antonio-Véjar V, Ortiz Tejedor JG, Ortiz Segarra J, Vega Crespo B, Bigoni-Ordóñez GD. The Hallmarks of Cervical Cancer: Molecular Mechanisms Induced by Human Papillomavirus. BIOLOGY 2024; 13:77. [PMID: 38392296 PMCID: PMC10886769 DOI: 10.3390/biology13020077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/24/2024]
Abstract
Human papillomaviruses (HPVs) and, specifically, high-risk HPVs (HR-HPVs) are identified as necessary factors in the development of cancer of the lower genital tract, with CaCU standing out as the most prevalent tumor. This review summarizes ten mechanisms activated by HR-HPVs during cervical carcinogenesis, which are broadly associated with at least seven of the fourteen distinctive physiological capacities of cancer in the newly established model by Hanahan in 2022. These mechanisms involve infection by human papillomavirus, cellular tropism, genetic predisposition to uterine cervical cancer (CaCU), viral load, viral physical state, regulation of epigenetic mechanisms, loss of function of the E2 protein, deregulated expression of E6/E7 oncogenes, regulation of host cell protein function, and acquisition of the mesenchymal phenotype.
Collapse
Affiliation(s)
- Pedro Rosendo-Chalma
- Laboratorio de Virus y Cáncer, Unidad de Investigación Biomédica en Cáncer of Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (IIB-UNAM), Mexico City 14080, Mexico
- Unidad Académica de Posgrado, Universidad Católica de Cuenca, Cuenca 010101, Ecuador
| | - Verónica Antonio-Véjar
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Guerrero, Mexico
| | - Jonnathan Gerardo Ortiz Tejedor
- Unidad Académica de Posgrado, Universidad Católica de Cuenca, Cuenca 010101, Ecuador
- Carrera de Bioquímica y Farmacia, Universidad Católica de Cuenca, Cuenca 010101, Ecuador
| | - Jose Ortiz Segarra
- Carrera de Medicina, Facultad de Ciencias Médicas, Universidad de Cuenca, Cuenca 010107, Ecuador
| | - Bernardo Vega Crespo
- Carrera de Medicina, Facultad de Ciencias Médicas, Universidad de Cuenca, Cuenca 010107, Ecuador
| | | |
Collapse
|
6
|
Zheng X, Zhao D, Liu Y, Jin Y, Liu T, Li H, Liu D. Regeneration and anti-inflammatory effects of stem cells and their extracellular vesicles in gynecological diseases. Biomed Pharmacother 2023; 168:115739. [PMID: 37862976 DOI: 10.1016/j.biopha.2023.115739] [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: 08/25/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023] Open
Abstract
There are many gynecological diseases, among which breast cancer (BC), cervical cancer (CC), endometriosis (EMs), and polycystic ovary syndrome (PCOS) are common and difficult to cure. Stem cells (SCs) are a focus of regenerative medicine. They are commonly used to treat organ damage and difficult diseases because of their potential for self-renewal and multidirectional differentiation. SCs are also commonly used for difficult-to-treat gynecological diseases because of their strong directional differentiation ability with unlimited possibilities, their tendency to adhere to the diseased tissue site, and their use as carriers for drug delivery. SCs can produce exosomes in a paracrine manner. Exosomes can be produced in large quantities and have the advantage of easy storage. Their safety and efficacy are superior to those of SCs, which have considerable potential in gynecological treatment, such as inhibiting endometrial senescence, promoting vascular reconstruction, and improving anti-inflammatory and immune functions. In this paper, we review the mechanisms of the regenerative and anti-inflammatory capacity of SCs and exosomes in incurable gynecological diseases and the current progress in their application in genetic engineering to provide a foundation for further research.
Collapse
Affiliation(s)
- Xu Zheng
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Dan Zhao
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun 130000, China
| | - Yang Liu
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun 130000, China
| | - Ye Jin
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Tianjia Liu
- Changchun University of Chinese Medicine, Changchun 130117, China; Baicheng Medical College, Baicheng 137000, China.
| | - Huijing Li
- Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Da Liu
- Changchun University of Chinese Medicine, Changchun 130117, China.
| |
Collapse
|
7
|
Benítez-Arvizu G, Castro-Jácome TP, Tovar-Pérez E, Alcántara-Quintana LE. [Antiproliferative, apoptotic, and antimigratory activities of kafirins on cervical cancer-derived cell lines]. REVISTA MEDICA DEL INSTITUTO MEXICANO DEL SEGURO SOCIAL 2023; 61:S4-S11. [PMID: 36378016 PMCID: PMC10395951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/27/2022] [Indexed: 06/16/2023]
Abstract
Background Cervical cancer is one of the leading causes of death in women worldwide, both in developed and developing countries. Therefore, effective treatment of cervical cancer with potential anti-tumor drugs is important. However, new treatments inspired by nutritional medicine are needed. Objective To use the human cervical cancer cell lines HeLa and SiHa to evaluate the antiproliferative, apoptotic, and migratory activity of sorghum (kafirins). Materials and methods The anticancer effects of the kafirins were examined by counting cells, MTT assays, apoptosis, and migration assays. Results This investigation showed that sorghum induced growth inhibition of HeLa and SiHa cells at a significant level. The growth inhibition is dose-dependent and irreversible. When HeLa and SiHa cells were treated with sorghum due to the activity of kafirins, morphological changes were observed, which were identified through the formation of apoptopic bodies. And the kafirins at concentrations of 37.5, 75, 150, and 300 μg/mL decreased the migration of HeLa cells and SiHa cells. Conclusion This paper demonstrates the induction of antiproliferative, apoptotic, and anti-migratory activity in HeLa and SiHa cells by kafirins. Sorghum may be used as a nutraceutical with potential cancer-prevention benefits.
Collapse
Affiliation(s)
- Gamaliel Benítez-Arvizu
- Instituto Mexicano del Seguro Social, Centro Médico Nacional Siglo XXI, Hospital de Especialidades “Dr. Bernardo Sepúlveda Gutiérrez”, Unidad Complementaria Banco de Sangre. Ciudad de México, MéxicoInstituto Mexicano del Seguro SocialMéxico
| | - Tania Patricia Castro-Jácome
- Instituto Tecnológico de Tepic, Laboratorio Integral de Investigación en Alimentos. Tepic, Nayarit, MéxicoInstituto Tecnológico de TepicMéxico
| | - Erik Tovar-Pérez
- Universidad Autónoma de Querétaro, Campus Amazcala, Facultad de Ingeniería. El Marqués, Querétaro, MéxicoUniversidad Autónoma de QuerétaroMéxico
| | - Luz Eugenia Alcántara-Quintana
- Universidad Autónoma de San Luis Potosí, Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología. San Luis Potosí, San Luis Potosí, México Universidad Autónoma de San Luis PotosíMéxico
| |
Collapse
|
8
|
Yuan W, Li S, Jia J, Wang L, Huang Y, Wang M, Xie F, Li J, Hao Y. Human papillomavirus is an important risk factor for esophageal carcinoma in a Chinese population. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04322-5. [PMID: 36394699 PMCID: PMC10349784 DOI: 10.1007/s00432-022-04322-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/22/2022] [Indexed: 11/18/2022]
Abstract
Abstract
Purpose
Different types of HPV have been associated with cancer in humans, but the role of HPV in esophageal cancer (EC) is controversial. The purpose of this study was to evaluate the correlation between HPV infection and EC in the Chinese population and to provide the scientific basis for the future prevention, control, early diagnosis, and treatment strategies of EC in China.
Methods
PCR detected HPV infection in 1112 esophageal cancer tissue samples, and 89 HPV-positive samples were detected by genotyping. Proximity ligation assays (PLAs) and immunohistochemistry were used to detect the expression of HPV E6 and E7 proteins. Real-time fluorescent quantitative PCR was used to detect the integration of HPV16 E6. The level of HPV-specific antibody IgG in serum was detected by ELISA and PLA.
Results
The positive rates of HPV L1, HPV16, HPV18, hpv16 + 18 E6 and hpv16/18 E6 in 1,112 EC tissue samples were 77.6%, 41.4%, 27.2%, 14.2% and 55.4% respectively. Multiple HPV subtypes were detected in HPV-positive EC samples. PLA showed that E6 and E7 were expressed in EC109 and formed complexes with p53 and pRb, respectively. Immunohistochemistry showed that the positive rates of hpv16 + 18 E6 and E7 in HPV-positive EC samples were 56.4% and 37.0%, respectively. HPV-DNA integration rate in HPV-positive EC tissues (88.79%) was higher than that in adjacent tissues (54.17%). HPV antibody was found in the serum of EC patients by a serological test.
Conclusion
The study suggests that HPV, especially HPV16 and HPV18, the infection may be a risk factor for EC in the Chinese population and that the E6 protein may play a key role in HPV-associated malignancies. These results may be important for the prevention and treatment of HPV-positive EC in China.
Collapse
|
9
|
Antonio-Véjar V, Ortiz-Sánchez E, Rosendo-Chalma P, Patiño-Morales CC, Guido-Jiménez MC, Alvarado-Ortiz E, Hernández G, García-Carrancá A. New insights into the interactions of HPV-16 E6*I and E6*II with p53 isoforms and induction of apoptosis in cancer-derived cell lines. Pathol Res Pract 2022; 234:153890. [PMID: 35487028 DOI: 10.1016/j.prp.2022.153890] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 02/07/2023]
Abstract
An important characteristic of cancers associated with high-risk human papillomaviruses (HR-HPV) is the inability of p53 to activate apoptosis due to the effect of the oncoprotein E6. However, the effect of HPV-16 E6 splice variant isoforms (namely E6*I and E6*II), their interaction with the existing p53 isoforms, and their influence on apoptosis is unclear. Here, we report the outcome of ectopic expression of HPV-16 E6, E6*I, and E6*II on the relative levels of p53 and p53 isoforms Δ40p53 and Δ133p53 and their interactions with these proteins. Additionally, we evaluated the effect of ectopic expression of p53, Δ40p53, and Δ133p53 on apoptosis in a p53 null pulmonary cell line (H1299) co-transfected with E6 isoforms and p53+/+ cell lines with HR-HPV (SiHa and HeLa), transfected with p53 isoforms and treated with cisplatin, a conventional drug used to treat cervical cancer. Our results show that E6 and E6*II induced a significant decrease in p53, but only E6 triggered a Δ40p53 decrease and that E6*II interacts with p53 but not with Δ40p53 and Δ133p53. On the other hand, E6*I did not show any effect or interaction with the p53 isoforms. We found that apoptosis was elevated in H1299 cells transfected with p53 (p = 0.0001) and Δ40p53 (p = 0.0001). A weak apoptotic effect was observed when Δ133p53 was ectopically expressed (p = 0.0195). We observed that both p53 (p = 0.0006) and Δ40p53 (p = 0.0014) induced apoptosis in cisplatin-treated SiHa cells; however in cisplatin-treated HeLa cells, only p53 induced apoptosis (p = 0.0029). No significant differences in apoptosis were observed upon ectopic expression of p53, Δ40p53, and Δ133p53 in SiHa and HeLa cells. Our findings suggest a possible therapeutic application for the combining of p53 or Δ40p53 with cisplatin to induce an increased apoptosis of cancer cells expressing E6 isoforms from HPV-16.
Collapse
Affiliation(s)
- Verónica Antonio-Véjar
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, 10450, Mexico; Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, 39090, Guerrero, Mexico; Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cancerología, Ciudad de México, 14080, Mexico.
| | - Elizabeth Ortiz-Sánchez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México, 14080, Mexico.
| | - Pedro Rosendo-Chalma
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, 10450, Mexico; Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cancerología, Ciudad de México, 14080, Mexico.
| | - Carlos C Patiño-Morales
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cancerología, Ciudad de México, 14080, Mexico.
| | - Miriam C Guido-Jiménez
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cancerología, Ciudad de México, 14080, Mexico.
| | - Eduardo Alvarado-Ortiz
- Programa de Posgrado en Ciencias Biológicas. Universidad Nacional Autónoma de México (UNAM), Ciudad de México, 04510, Mexico; Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cancerología, Ciudad de México, 14080, Mexico.
| | - Greco Hernández
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México, 14080, Mexico.
| | - Alejandro García-Carrancá
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cancerología, Ciudad de México, 14080, Mexico.
| |
Collapse
|
10
|
Xu X, Han Z, Ruan Y, Liu M, Cao G, Li C, Li F. HPV16-LINC00393 Integration Alters Local 3D Genome Architecture in Cervical Cancer Cells. Front Cell Infect Microbiol 2021; 11:785169. [PMID: 34950609 PMCID: PMC8691139 DOI: 10.3389/fcimb.2021.785169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/04/2021] [Indexed: 11/30/2022] Open
Abstract
High-risk human papillomavirus (hrHPV) infection and integration were considered as essential onset factors for the development of cervical cancer. However, the mechanism on how hrHPV integration influences the host genome structure remains not fully understood. In this study, we performed in situ high-throughput chromosome conformation capture (Hi-C) sequencing, chromatin immunoprecipitation and sequencing (ChIP-seq), and RNA-sequencing (RNA-seq) in two cervical cells, 1) NHEK normal human epidermal keratinocyte; and 2) HPV16-integrated SiHa tumorigenic cervical cancer cells. Our results reveal that the HPV-LINC00393 integrated chromosome 13 exhibited significant genomic variation and differential gene expression, which was verified by calibrated CTCF and H3K27ac ChIP-Seq chromatin restructuring. Importantly, HPV16 integration led to differential responses in topologically associated domain (TAD) boundaries, with a decrease in the tumor suppressor KLF12 expression downstream of LINC00393. Overall, this study provides significant insight into the understanding of HPV16 integration induced 3D structural changes and their contributions on tumorigenesis, which supplements the theory basis for the cervical carcinogenic mechanism of HPV16 integration.
Collapse
Affiliation(s)
- Xinxin Xu
- Department of Obstetrics and Gynecology, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhiqiang Han
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yetian Ruan
- Department of Obstetrics and Gynecology, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Min Liu
- Department of Obstetrics and Gynecology, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guangxu Cao
- Department of Obstetrics and Gynecology, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chao Li
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fang Li
- Department of Obstetrics and Gynecology, East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Abstract
C6 deamination of adenosine (A) to inosine (I) in double-stranded RNA (dsRNA) is catalyzed by a family of enzymes known as ADARs (adenosine deaminases acting on RNA) encoded by three genes in mammals. Alternative promoters and splicing produce two ADAR1 proteins, an interferon-inducible cytoplasmic p150 and a constitutively expressed p110 that like ADAR2 is a nuclear enzyme. ADAR3 lacks deaminase activity. A-to-I editing occurs with both viral and cellular RNAs. Deamination activity is dependent on dsRNA substrate structure and regulatory RNA-binding proteins and ranges from highly site selective with hepatitis D RNA and glutamate receptor precursor messenger RNA (pre-mRNA) to hyperediting of measles virus and polyomavirus transcripts and cellular inverted Alu elements. Because I base-pairs as guanosine instead of A, editing can alter mRNA decoding, pre-mRNA splicing, and microRNA silencing. Editing also alters dsRNA structure, thereby suppressing innate immune responses including interferon production and action. Expected final online publication date for the Annual Review of Virology, Volume 8 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Christian K Pfaller
- Division of Veterinary Medicine, Paul-Ehrlich-Institute, Langen 63225, Germany
| | - Cyril X George
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, California 93106, USA;
| | - Charles E Samuel
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, California 93106, USA;
| |
Collapse
|
12
|
Fan Q, Huang T, Sun X, Wang YW, Wang J, Liu Y, Ni T, Gu SL, Li YH, Wang YD. HPV-16/18 E6-induced APOBEC3B expression associates with proliferation of cervical cancer cells and hypomethylation of Cyclin D1. Mol Carcinog 2021; 60:313-330. [PMID: 33631046 DOI: 10.1002/mc.23292] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 12/13/2022]
Abstract
Oncogenic high-risk human papillomavirus (HR-HPV) infection causes a majority of cases of cervical cancer and pre-cancerous cervical lesions. However, the mechanisms underlying the direct evolution from HPV-16/18-infected epithelium to cervical intraepithelial neoplasia (CIN) III, which can progress to cervical cancer, remain poorly identified. Here, we performed RNA-seq after laser capture microdissection, and found that APOBEC3B was highly expressed in cervical cancer specimens compared with CIN III with HPV-16/18 infection. Furthermore, immunohistochemical analysis confirmed that high levels of APOBEC3B were correlated with lymph node metastasis in cervical cancer. Subsequent experiments revealed that HPV-16 E6 could upregulate APOBEC3B through direct binding to the promoter of APOBEC3B in cervical cancer cells. Silencing of APOBEC3B by stable short hairpin RNA-mediated knockdown reduced the proliferative capacity of Caski and HeLa cells in vitro and in vivo, but had only a small effect on the migration and invasion of two cervical cancer cell lines. Finally, we identified the changes in gene expression following APOBEC3B silencing in Caski cells by microarray, demonstrating a biological link between APOBEC3B and CCND1 in cervical cancer cells. Importantly, through methyl-capture sequencing and pyrosequencing, APOBEC3B was found to affect the levels of the downstream protein Cyclin D1 (which is encoded by the CCND1 gene) through hypomethylation of the CCND1 promoter. In conclusion, our study supports HPV-16 E6-induced APOBEC3B expression associates with proliferation of cervical cancer cells and hypomethylation of Cyclin D1. Thus, APOBEC3B may be a potential therapeutic target in human cervical cancer.
Collapse
Affiliation(s)
- Qiong Fan
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Municipal Key Clinical Specialty, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Ting Huang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Municipal Key Clinical Specialty, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Xiao Sun
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Municipal Key Clinical Specialty, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Yi-Wei Wang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Wang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yao Liu
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ting Ni
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sheng-Lan Gu
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yu-Hong Li
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yu-Dong Wang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Municipal Key Clinical Specialty, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| |
Collapse
|
13
|
Tolomeo D, Agostini A, Visci G, Traversa D, Storlazzi CT. PVT1: A long non-coding RNA recurrently involved in neoplasia-associated fusion transcripts. Gene 2021; 779:145497. [PMID: 33600954 DOI: 10.1016/j.gene.2021.145497] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/04/2021] [Indexed: 12/12/2022]
Abstract
NGS technologies and bioinformatics tools allow the rapid identification of chimeric transcripts in cancer. More than 40,000 fusions are so far reported in the literature; however, for most of them, the role in oncogenesis is still not fully understood. This is the case for fusions involving the long non-coding RNA (lncRNA) Plasmacytoma variant translocation 1 (PVT1) (8q24.21). This lncRNA displays oncogenic functions in several cancer types interacting with microRNAs and proteins, but the role of PVT1 fusion transcripts is more obscure. These chimeras have been identified in both hematological malignancies and solid tumors, mainly arising from rearrangements and/or amplification of the 8q24 chromosomal region. In this review, we detail the full spectrum of PVT1 fusions in cancer, summarizing current knowledge about their genesis, function, and role as biomarkers.
Collapse
Affiliation(s)
- Doron Tolomeo
- Department of Biology, University of Bari, Via Orabona no.4, 70125 Bari, Italy.
| | - Antonio Agostini
- Department of Biomedical Sciences and Human Oncology, Unit of Internal Medicine "Guido Baccelli", University of Bari Medical School, Piazza Giulio Cesare 11, 70124 Bari, Italy.
| | - Grazia Visci
- Department of Biology, University of Bari, Via Orabona no.4, 70125 Bari, Italy.
| | - Debora Traversa
- Department of Biology, University of Bari, Via Orabona no.4, 70125 Bari, Italy.
| | | |
Collapse
|
14
|
Rotondo JC, Oton-Gonzalez L, Mazziotta C, Lanzillotti C, Iaquinta MR, Tognon M, Martini F. Simultaneous Detection and Viral DNA Load Quantification of Different Human Papillomavirus Types in Clinical Specimens by the High Analytical Droplet Digital PCR Method. Front Microbiol 2020; 11:591452. [PMID: 33329471 PMCID: PMC7710522 DOI: 10.3389/fmicb.2020.591452] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/19/2020] [Indexed: 12/31/2022] Open
Abstract
Human papillomaviruses (HPVs) are small DNA tumor viruses that mainly infect mucosal epithelia of anogenital and upper respiratory tracts. There has been progressive demand for more analytical assays for HPV DNA quantification. A novel droplet digital PCR (ddPCR) method was developed to simultaneously detect and quantify HPV DNA from different HPV types. DdPCR was initially tested for assay sensitivity, accuracy, specificity as well as intra- and inter-run assay variation employing four recombinant plasmids containing HPV16, HPV18, HPV11, and HPV45 DNAs. The assay was extended to investigate/quantify HPV DNA in Cervical Intraepithelial Neoplasia (CIN, n = 45) specimens and human cell lines (n = 4). DdPCR and qPCR data from clinical samples were compared. The assay showed high accuracy, sensitivity and specificity, with low intra-/inter- run variations, in detecting/quantifying HPV16/18/11/45 DNAs. HPV DNA was detected in 51.1% (23/45) CIN DNA samples by ddPCR, whereas 40% (18/45) CIN tested HPV-positive by qPCR. Five CIN, tested positive by ddPCR, were found to be negative by qPCR. In CIN specimens, the mean HPV DNA loads determined by ddPCR were 3.81 copy/cell (range 0.002-51.02 copy/cell), whereas 8.04 copy/cell (range 0.003-78.73 copy/cell) by qPCR. DdPCR and qPCR concordantly detected HPV DNA in SiHa, CaSki and Hela cells, whereas HaCaT tested HPV-negative. The correlation between HPV DNA loads simultaneously detected by ddPCR/qPCR in CINs/cell lines was good (R 2 = 0.9706, p < 0.0001). Our data indicate that ddPCR is a valuable technique in quantifying HPV DNA load in CIN specimens and human cell lines, thereby improving clinical applications, such as patient management after primary diagnosis of HPV-related lesions with HPV-type specific assays.
Collapse
Affiliation(s)
| | | | | | | | | | - Mauro Tognon
- Laboratories of Cell Biology and Molecular Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Fernanda Martini
- Laboratories of Cell Biology and Molecular Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
15
|
Rosendo-Chalma P, Antonio-Vejar V, Bigoni-Ordóñez GD, Patiño-Morales CC, Cano-García A, García-Carrancá A. CDH1 and SNAI1 are regulated by E7 from human papillomavirus types 16 and 18. Int J Oncol 2020; 57:301-313. [PMID: 32319591 DOI: 10.3892/ijo.2020.5039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 10/24/2019] [Indexed: 11/05/2022] Open
Abstract
A common characteristic of cancer types associated with viruses is the dysregulated expression of the CDH1 gene, which encodes E‑cadherin, in general by activation of DNA methyltransferases (Dnmts). In cervical cancer, E7 protein from high risk human papillomaviruses (HPVs) has been demonstrated to interact with Dnmt1 and histone deacetylase type 1 (HDAC1). The present study proposed that E7 may regulate the expression of CDH1 through two pathways: i) Epigenetic, including DNA methylation; and ii) Epigenetic‑independent, including the induction of negative regulators of CDH1 expression, such as Snail family transcriptional repressor Snai1 and Snai2. To test this hypothesis, HPV16‑ and HPV18‑positive cell lines were used to determine the methylation pattern of the CDH1 promoter and its expression in association with its negative regulators. Different methylation frequencies were identified in the CDH1 promoter in HeLa (88.24%) compared with SiHa (17.65%) and Ca Ski (0%) cell lines. Significant differences in the expression of SNAI1 were observed between these cell lines, and an inverse association was identified between the expression levels of SNAI1 and CDH1. In addition, suppressing E7 not only increased the expression of CDH1, but notably decreased the expression of SNAI1 and modified the methylation pattern of the CDH1 promoter. These results suggested that the expression of CDH1 was dependent on the expression of SNAI1 and was inversely associated with the expression of E7. The present results indicated that E7 from HPV16/18 regulated the expression of CDH1 by the two following pathways in which Snai1 is involved: i) Hypermethylation of the CDH1 promoter region and increasing expression of SNAI1, as observed in HeLa; and ii) Hypomethylation of the CDH1 promoter region and expression of SNAI1, as observed in SiHa. Therefore, the suppression of CDH1 and expression of SNAI1 may be considered to be biomarkers of metastasis in uterine cervical cancer.
Collapse
Affiliation(s)
- Pedro Rosendo-Chalma
- Programa de Doctorado en Ciencias Biomédicas, Instituto de Investigaciones Biomédicas (IIB), Universidad Nacional Autónoma de México (UNAM), Mexico City 10450, Mexico
| | - Verónica Antonio-Vejar
- Laboratorio de Virus y Cáncer, Unidad de Investigación Biomédica en Cáncer of Instituto de Investigaciones Biomédicas‑Universidad Nacional Autónoma de México (IIB‑UNAM) and División de Investigación Básica of Instituto Nacional de Cancerología‑Secretaría de Salud (INCan‑SSA), Mexico City 14080, Mexico
| | - Gabriele Davide Bigoni-Ordóñez
- Laboratorio de Virus y Cáncer, Unidad de Investigación Biomédica en Cáncer of Instituto de Investigaciones Biomédicas‑Universidad Nacional Autónoma de México (IIB‑UNAM) and División de Investigación Básica of Instituto Nacional de Cancerología‑Secretaría de Salud (INCan‑SSA), Mexico City 14080, Mexico
| | - Carlos César Patiño-Morales
- Laboratorio de Virus y Cáncer, Unidad de Investigación Biomédica en Cáncer of Instituto de Investigaciones Biomédicas‑Universidad Nacional Autónoma de México (IIB‑UNAM) and División de Investigación Básica of Instituto Nacional de Cancerología‑Secretaría de Salud (INCan‑SSA), Mexico City 14080, Mexico
| | - Amparo Cano-García
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC‑UAM), Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid 28029, Spain
| | - Alejandro García-Carrancá
- Laboratorio de Virus y Cáncer, Unidad de Investigación Biomédica en Cáncer of Instituto de Investigaciones Biomédicas‑Universidad Nacional Autónoma de México (IIB‑UNAM) and División de Investigación Básica of Instituto Nacional de Cancerología‑Secretaría de Salud (INCan‑SSA), Mexico City 14080, Mexico
| |
Collapse
|
16
|
Gu Y, Lin X, Kapoor A, Chow MJ, Jiang Y, Zhao K, Tang D. The Oncogenic Potential of the Centromeric Border Protein FAM84B of the 8q24.21 Gene Desert. Genes (Basel) 2020; 11:genes11030312. [PMID: 32183428 PMCID: PMC7140883 DOI: 10.3390/genes11030312] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/09/2020] [Accepted: 03/13/2020] [Indexed: 12/14/2022] Open
Abstract
FAM84B is a risk gene in breast and prostate cancers. Its upregulation is associated with poor prognosis of prostate cancer, breast cancer, and esophageal squamous cell carcinoma. FAM84B facilitates cancer cell proliferation and invasion in vitro, and xenograft growth in vivo. The FAM84B and Myc genes border a 1.2 Mb gene desert at 8q24.21. Co-amplification of both occurs in 20 cancer types. Mice deficient of a 430 Kb fragment within the 1.2 Mb gene desert have downregulated FAM84B and Myc expressions concurrent with reduced breast cancer growth. Intriguingly, Myc works in partnership with other oncogenes, including Ras. FAM84B shares similarities with the H-Ras-like suppressor (HRASLS) family over their typical LRAT (lecithin:retinal acyltransferase) domain. This domain contains a catalytic triad, H23, H35, and C113, which constitutes the phospholipase A1/2 and O-acyltransferase activities of HRASLS1-5. These enzymatic activities underlie their suppression of Ras. FAM84B conserves H23 and H35 but not C113 with both histidine residues residing within a highly conserved motif that FAM84B shares with HRASLS1-5. Deletion of this motif abolishes FAM84B oncogenic activities. These properties suggest a collaboration of FAM84B with Myc, consistent with the role of the gene desert in strengthening Myc functions. Here, we will discuss recent research on FAM84B-derived oncogenic potential.
Collapse
Affiliation(s)
- Yan Gu
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph’s Hospital, Hamilton, ON L8N 4A6, Canada; (Y.G.); (X.L.); (M.J.C.); (Y.J.); (K.Z.)
- Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada;
- The Research Institute of St Joe’s Hamilton, St Joseph’s Hospital, Hamilton, ON L8N 4A6, Canada
| | - Xiaozeng Lin
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph’s Hospital, Hamilton, ON L8N 4A6, Canada; (Y.G.); (X.L.); (M.J.C.); (Y.J.); (K.Z.)
- Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada;
- The Research Institute of St Joe’s Hamilton, St Joseph’s Hospital, Hamilton, ON L8N 4A6, Canada
| | - Anil Kapoor
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph’s Hospital, Hamilton, ON L8N 4A6, Canada; (Y.G.); (X.L.); (M.J.C.); (Y.J.); (K.Z.)
- Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada;
- Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Mathilda Jing Chow
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph’s Hospital, Hamilton, ON L8N 4A6, Canada; (Y.G.); (X.L.); (M.J.C.); (Y.J.); (K.Z.)
- Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada;
- The Research Institute of St Joe’s Hamilton, St Joseph’s Hospital, Hamilton, ON L8N 4A6, Canada
| | - Yanzhi Jiang
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph’s Hospital, Hamilton, ON L8N 4A6, Canada; (Y.G.); (X.L.); (M.J.C.); (Y.J.); (K.Z.)
- Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada;
- The Research Institute of St Joe’s Hamilton, St Joseph’s Hospital, Hamilton, ON L8N 4A6, Canada
| | - Kuncheng Zhao
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph’s Hospital, Hamilton, ON L8N 4A6, Canada; (Y.G.); (X.L.); (M.J.C.); (Y.J.); (K.Z.)
- Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada;
- The Research Institute of St Joe’s Hamilton, St Joseph’s Hospital, Hamilton, ON L8N 4A6, Canada
| | - Damu Tang
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph’s Hospital, Hamilton, ON L8N 4A6, Canada; (Y.G.); (X.L.); (M.J.C.); (Y.J.); (K.Z.)
- Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada;
- The Research Institute of St Joe’s Hamilton, St Joseph’s Hospital, Hamilton, ON L8N 4A6, Canada
- Correspondence: ; Tel.: +(905)-522-1155 (ext. 35168)
| |
Collapse
|
17
|
Jiang N, Xie F, Chen L, Chen F, Sui L. The effect of TLR4 on the growth and local inflammatory microenvironment of HPV-related cervical cancer in vivo. Infect Agent Cancer 2020; 15:12. [PMID: 32095158 PMCID: PMC7027303 DOI: 10.1186/s13027-020-0279-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 02/04/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Cervical cancer is the most common malignancy of the female lower genital tract. In our previous study, we found that TLR4 promotes cervical cancer cell growth in vitro. The aim of this study was to further explore the role of TLR4 in HPV-related cervical cancer in vivo by using a nude mouse xenograft model. METHODS Cervical cancer-derived HeLa and CaSki cells (5 × 107/mL) were either stimulated with an optimal concentration of LPS for the appropriate time (HeLa cells were treated with 1 μg/mL LPS for 1 h, and CaSki cells were treated with 2 μg/mL LPS for 1.5 h) or transfected with TLR4 shRNA and then injected subcutaneously into the dorsal right posterior side of nude mice. The shortest width and longest diameter of the transplanted tumors in the nude mice were measured every 3 days.TLR4, IL-6,iNOS, IL-8,COX-2, MIP-3α, TGF-β1 and VEGF expression levels in the transplanted tumor tissue were detected by immunohistochemistry. RESULTS The tumor formation rate was 100% in both HeLa and CaSki nude mouse groups. The tumors grew faster, and the cachexia symptoms were more serious in the LPS groups than in the control group. In contrast, the tumors grew slower, and the cachexia symptoms were milder in the TLR4-silenced groups. TLR4, iNOS, IL-6, MIP-3α and VEGF were highly expressed in the transplanted tumor tissues from the LPS groups, and their expression levels were decreased in the TLR4-silenced groups. CONCLUSION TLR4 expression is closely associated with the tumorigenesis and growth of HPV-positive cervical cancer; TLR4 promotes HPV-positive cervical tumor growth and facilitates the formation of a local immunosuppressive microenvironment. Eventually, these conditions may lead to cervical cancer development.
Collapse
Affiliation(s)
- Ninghong Jiang
- Medical center for diagnosis and treatment of cervical disease, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011 China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011 China
| | - Feng Xie
- Medical center for diagnosis and treatment of cervical disease, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011 China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011 China
| | - Limei Chen
- Medical center for diagnosis and treatment of cervical disease, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011 China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011 China
| | - Fang Chen
- Medical center for diagnosis and treatment of cervical disease, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011 China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011 China
| | - Long Sui
- Medical center for diagnosis and treatment of cervical disease, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011 China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011 China
| |
Collapse
|
18
|
Pujantell M, Badia R, Galván-Femenía I, Garcia-Vidal E, de Cid R, Alcalde C, Tarrats A, Piñol M, Garcia F, Chamorro AM, Revollo B, Videla S, Parés D, Corral J, Tural C, Sirera G, Esté JA, Ballana E, Riveira-Muñoz E. ADAR1 function affects HPV replication and is associated to recurrent human papillomavirus-induced dysplasia in HIV coinfected individuals. Sci Rep 2019; 9:19848. [PMID: 31882741 PMCID: PMC6934649 DOI: 10.1038/s41598-019-56422-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 12/11/2019] [Indexed: 12/18/2022] Open
Abstract
Infection by human papillomavirus (HPV) alters the microenvironment of keratinocytes as a mechanism to evade the immune system. A-to-I editing by ADAR1 has been reported to regulate innate immunity in response to viral infections. Here, we evaluated the role of ADAR1 in HPV infection in vitro and in vivo. Innate immune activation was characterized in human keratinocyte cell lines constitutively infected or not with HPV. ADAR1 knockdown induced an innate immune response through enhanced expression of RIG-I-like receptors (RLR) signaling cascade, over-production of type-I IFNs and pro-inflammatory cytokines. ADAR1 knockdown enhanced expression of HPV proteins, a process dependent on innate immune function as no A-to-I editing could be identified in HPV transcripts. A genetic association study was performed in a cohort of HPV/HIV infected individuals followed for a median of 6 years (range 0.1-24). We identified the low frequency haplotype AACCAT significantly associated with recurrent HPV dysplasia, suggesting a role of ADAR1 in the outcome of HPV infection in HIV+ individuals. In summary, our results suggest that ADAR1-mediated innate immune activation may influence HPV disease outcome, therefore indicating that modification of innate immune effectors regulated by ADAR1 could be a therapeutic strategy against HPV infection.
Collapse
Affiliation(s)
- Maria Pujantell
- AIDS Research Institute-IrsiCaixa, Badalona, Spain
- Health Research Institute Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Roger Badia
- AIDS Research Institute-IrsiCaixa, Badalona, Spain
- Health Research Institute Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Iván Galván-Femenía
- Genomes for Life-GCAT Lab Group - Program of Predictive and Personalized Medicine of Cancer (PMPPC), Badalona, Spain
- Health Research Institute Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Edurne Garcia-Vidal
- AIDS Research Institute-IrsiCaixa, Badalona, Spain
- Health Research Institute Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Rafael de Cid
- Genomes for Life-GCAT Lab Group - Program of Predictive and Personalized Medicine of Cancer (PMPPC), Badalona, Spain
- Health Research Institute Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Carmen Alcalde
- Fundació Lluita contra la Sida, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Antonio Tarrats
- Department of Gynecology, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Marta Piñol
- Department of Surgery, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Francesc Garcia
- Fundació Lluita contra la Sida, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Ana M Chamorro
- Fundació Lluita contra la Sida, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Boris Revollo
- Fundació Lluita contra la Sida, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Sebastian Videla
- Fundació Lluita contra la Sida, Hospital Germans Trias i Pujol, Badalona, Spain
| | - David Parés
- Fundació Lluita contra la Sida, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Javier Corral
- Fundació Lluita contra la Sida, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Cristina Tural
- Department of Internal Medicine, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Guillem Sirera
- Fundació Lluita contra la Sida, Hospital Germans Trias i Pujol, Badalona, Spain
| | - José A Esté
- AIDS Research Institute-IrsiCaixa, Badalona, Spain.
- Health Research Institute Germans Trias i Pujol (IGTP), Badalona, Spain.
| | - Ester Ballana
- AIDS Research Institute-IrsiCaixa, Badalona, Spain.
- Health Research Institute Germans Trias i Pujol (IGTP), Badalona, Spain.
| | - Eva Riveira-Muñoz
- AIDS Research Institute-IrsiCaixa, Badalona, Spain
- Health Research Institute Germans Trias i Pujol (IGTP), Badalona, Spain
| |
Collapse
|
19
|
Brant AC, Menezes AN, Felix SP, de Almeida LM, Sammeth M, Moreira MAM. Characterization of HPV integration, viral gene expression and E6E7 alternative transcripts by RNA-Seq: A descriptive study in invasive cervical cancer. Genomics 2018; 111:1853-1861. [PMID: 30552977 DOI: 10.1016/j.ygeno.2018.12.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/06/2018] [Accepted: 12/07/2018] [Indexed: 12/27/2022]
Abstract
Scarce data are available on the expression of papillomavirus genome and the frequency of alternatively spliced E6E7 mRNAs in invasive cervical cancer. We carried out a comprehensive characterization of HPV expression by RNA-Seq analysis in 22 invasive cervical cancer with HPV16 or HPV18, characterizing the presence of integrated/episomal viral DNA, the integration sites in human genome and the proportion of alternative splicing products of E6 and E7 genes. The expression patterns suggested the presence of episomal and/or integrated viral DNA, with integration detected in most tumors, frequently occurring within human genes in HPV18+ and in intergenic regions in HPV16+ tumors. Alternative splicing of E6E7 transcripts showed E6*I as the most frequent isoform for both viral types, followed by E6*II and E6/E7 (unspliced) transcripts in HPV16+, and by E6/E7 in HPV18+ tumors. Previously described E6*VI and E6*V transcript isoforms for HPV16, and E6*X for HPV18, were rare or not detected.
Collapse
Affiliation(s)
- Ayslan C Brant
- Genetics Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil; Post-Graduate Program in Genetics, Universidade Federal do Rio de Janeiro (UFRJ), Brazil
| | - Albert N Menezes
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, UK
| | - Shayany P Felix
- Genetics Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| | - Liz M de Almeida
- Department of Population Research, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil.
| | - Michael Sammeth
- Department of Bioinformatics, Transcriptomics and Functional Genomics, Federal University of Rio de Janeiro (UFRJ), Brazil.
| | - Miguel A M Moreira
- Genetics Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil.
| |
Collapse
|
20
|
Wolf B, Diop F, Ferraris P, Wichit S, Busso C, Missé D, Gönczy P. Zika virus causes supernumerary foci with centriolar proteins and impaired spindle positioning. Open Biol 2017; 7:rsob.160231. [PMID: 28100662 PMCID: PMC5303270 DOI: 10.1098/rsob.160231] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/15/2016] [Indexed: 12/30/2022] Open
Abstract
Zika virus (ZIKV) causes congenital microcephaly. Although ZIKV can impair cell cycle progression and provoke apoptosis, which probably contributes to disease aetiology through depletion of neural progenitor cells, additional cellular mechanisms may be important. Here, we investigated whether ZIKV infection alters centrosome number and spindle positioning, because such defects are thought to be at the root of inherited primary autosomal recessive microcephaly (MCPH). In addition to HeLa cells, in which centrosome number and spindle positioning can be well monitored, we analysed retinal epithelial cells (RPE-1), as well as brain-derived microglial (CHME-5) and neural progenitor (ReN) cells, using immunofluorescence. We established that ZIKV infection leads to supernumerary foci containing centriolar proteins that in some cases drive multipolar spindle assembly, as well as spindle positioning defects in HeLa, RPE-1 and CHME-5 cells, but not in ReN cells. We uncovered similar phenotypes in HeLa cells upon infection with dengue virus (DENV-2), another flavivirus that does not target brain cells and does not cause microcephaly. We conclude that infection with Flaviviridae can increase centrosome numbers and impair spindle positioning, thus potentially contributing to microcephaly in the case of Zika.
Collapse
Affiliation(s)
- Benita Wolf
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Fodé Diop
- Laboratoire MIVEGEC, UMR 224 IRD/CNRS/UM1, 34394 Montpellier, France
| | - Pauline Ferraris
- Laboratoire MIVEGEC, UMR 224 IRD/CNRS/UM1, 34394 Montpellier, France
| | | | - Coralie Busso
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Dorothée Missé
- Laboratoire MIVEGEC, UMR 224 IRD/CNRS/UM1, 34394 Montpellier, France
| | - Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015, Lausanne, Switzerland
| |
Collapse
|
21
|
Wimardhani YS, Sasanti H, Wardhany II, Sarsito AS, Pradono SA, Subita GP, Soegyanto AI, Rahmayanti F, Chamusri N, Iamaroon A. Saliva-Based Screening of High-Risk Human Papillomavirus Strains: Detection in Female Indonesian and Thai Dental Students. Asian Pac J Cancer Prev 2016. [PMID: 26225705 DOI: 10.7314/apjcp.2015.16.13.5525] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Currently it is believed that human papillomaviruses (HPV) are associated with the development of some oral/oropharyngeal cancers. It has been suggested that these viruses influence carcinogenesis in both smokers and non-smokers. Data on the prevalence of HPV in healthy adults are thus needed to estimate the risk of oral/oropharyngeal cancer. The aim of this study was to assess the prevalence of oral HPV in healthy female adults in Indonesia and Thailand. MATERIALS AND METHODS Healthy female students from the Faculties of Dentistry of Universitas Indonesia and Chiang Mai University were asked to participate in this pilot study. DNA was extracted from saliva specimens and screened for HPV16 and HPV18 using PCR. RESULTS The age, marital status and sexual experience of the subjects between the two countries were not significantly different. Eight (4%) and 4 (2%) samples were positive for HPV16 and HPV18, respectively. Fisher's Exact test found a significant difference between HPV16 positivity in subjects who were married and had sexual intercourse but not for HPV18. CONCLUSIONS This study successfully detected presence of HPV16 and HPV18 DNA in a number of saliva samples from female dental school students. Marital status, experience of sexual intercourse and safe sexual practice are related to the possibility of finding HPV DNA finding in saliva. Dentists, physicians and other health care professionals may gain significant value from the findings of this study, which provide an understanding of the nature of HPV infection and its risk to patient health and disease.
Collapse
|
22
|
Ding S, Qian SY, Zhang Y, Wu W, Lu G, Lu Y, Feng X, Li L, Shen P. Establishment of immunoassay for detecting HPV16 E6 and E7 RNA. Sci Rep 2015; 5:13686. [PMID: 26333509 PMCID: PMC4558577 DOI: 10.1038/srep13686] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 08/03/2015] [Indexed: 11/09/2022] Open
Abstract
Cervical carcinoma is the most prevalent malignancy second only to breast cancer among women worldwide. Since more than 99% of cervical cancers are caused by human papilloma virus (HPV), measurement of HPV (HPV test) was commonly used in screening risk and/or early stage of cervical cancer as well as assessing the efficacies of the treatments that can decrease the incidence of cervical cancer. Many approaches that diagnose HPV infections have been developed, while most of them have distinct shortcomings. We here established a novel immunoassay method in which the pairs of unlabeled DNA probes firstly bind to HPV16 E6 and E7 RNAs to form the DNA-RNA hybrids, and the hybrids will subsequently be identified by S9.6 antibody. The sensitivity of this highly specific method can reach ~0.923 pg/mL and ~0.424 pg/mL of in vitro transcribed HPV16 E6 and E7 RNA, respectively, and reverse transcription and polymerase chain reaction (PCR) amplification were no longer needed. Thus, our immunoassay approaches can precisely reflect the actually viral load that is related to the course of HPV infection. In addition, it has also fast and low cost characteristic feature.
Collapse
Affiliation(s)
- Sen Ding
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210023, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210061, China
| | - Steven Y. Qian
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58108-6050, United States
| | - Yang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210023, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210061, China
| | - Wenlei Wu
- Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing, 210093, China
| | - Gensheng Lu
- Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Yan Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210023, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210061, China
| | - Xiujing Feng
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210023, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210061, China
| | - Li Li
- School of Chemistry & Life Sciences, Nanjing University Jinling College, Nanjing, 210089, China
| | - Pingping Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210023, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210061, China
| |
Collapse
|