1
|
Subramani K, Huang HS, Chen PC, Ding DC, Chu TY. Ovulation sources ROS to confer mutagenic activities on the TP53 gene in the fallopian tube epithelium. Neoplasia 2025; 59:101085. [PMID: 39637685 DOI: 10.1016/j.neo.2024.101085] [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: 09/06/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Epidemiological studies have implicated ovulation as a risk factor for ovarian high-grade serous carcinoma (HGSC) at the initiation stage. Precancerous lesions of HGSC commonly exhibit TP53 mutations attributed to DNA deamination and are frequently localized in the fallopian tube epithelium (FTE), a site regularly exposed to ovulatory follicular fluid (FF). This study aimed to assess the mutagenic potential of FF and investigate the expression levels and functional role of activation-induced cytidine deaminase (AID) following ovulation, along with the resulting TP53 DNA deamination. METHODS The mutagenic activity of FF toward premalignant and malignant FTE cells was determined using the hypoxanthine phosphoribosyl transferase (HPRT) mutation assay with or without AID knockdown. The sequential activation of AID, including expressional induction, nuclear localization, DNA binding, and deamination, was determined. AID inducers in FF were identified, and the times of action and signaling pathways were determined. RESULTS FF induced AID activation and de novo FTE cell mutagenesis in two waves of activity in accordance with post-ovulation FF exposure. The ERK-mediated early activity started at 2 min and peaked at 45 min, and the NF-κB-mediated late activity started at 6 h and peaked at 8.5 h after exposure. ROS, TNF-α, and estradiol, which are abundant in FF, all induced the two activities, while all activities were abolished by antioxidant cotreatment. AID physically bound to and biochemically deaminated the TP53 gene, regardless of known mutational hotspots. It did not act on other prevalent tumor-suppressor genes of HGSC. CONCLUSION This study revealed the ROS-dependent AID-mediated mutagenic activity of the ovulatory FF. The results filled up the missing link between ovulation and the initial TP53 mutation and invited a strategy of antioxidation in prevention of HGSC.
Collapse
Affiliation(s)
- Kanchana Subramani
- Center for Prevention and Therapy of Gynecological Cancers, Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, ROC; Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan, ROC
| | - Hsuan-Shun Huang
- Center for Prevention and Therapy of Gynecological Cancers, Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, ROC
| | - Pao-Chu Chen
- Department of Obstetrics & Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, ROC
| | - Dah-Ching Ding
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan, ROC; Department of Obstetrics & Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, ROC
| | - Tang-Yuan Chu
- Center for Prevention and Therapy of Gynecological Cancers, Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, ROC; Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan, ROC; Department of Obstetrics & Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, ROC.
| |
Collapse
|
2
|
Pan-cancer landscape of AID-related mutations, composite mutations, and their potential role in the ICI response. NPJ Precis Oncol 2022; 6:89. [PMID: 36456685 PMCID: PMC9715662 DOI: 10.1038/s41698-022-00331-2] [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: 05/26/2022] [Accepted: 11/02/2022] [Indexed: 12/03/2022] Open
Abstract
Activation-induced cytidine deaminase, AICDA or AID, is a driver of somatic hypermutation and class-switch recombination in immunoglobulins. In addition, this deaminase belonging to the APOBEC family may have off-target effects genome-wide, but its effects at pan-cancer level are not well elucidated. Here, we used different pan-cancer datasets, totaling more than 50,000 samples analyzed by whole-genome, whole-exome, or targeted sequencing. AID mutations are present at pan-cancer level with higher frequency in hematological cancers and higher presence at transcriptionally active TAD domains. AID synergizes initial hotspot mutations by a second composite mutation. AID mutational load was found to be independently associated with a favorable outcome in immune-checkpoint inhibitors (ICI) treated patients across cancers after analyzing 2000 samples. Finally, we found that AID-related neoepitopes, resulting from mutations at more frequent hotspots if compared to other mutational signatures, enhance CXCL13/CCR5 expression, immunogenicity, and T-cell exhaustion, which may increase ICI sensitivity.
Collapse
|
3
|
Çakan E, Gunaydin G. Activation induced cytidine deaminase: An old friend with new faces. Front Immunol 2022; 13:965312. [PMID: 36405752 PMCID: PMC9670734 DOI: 10.3389/fimmu.2022.965312] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
Abstract
Activation induced cytidine deaminase (AID) protein is a member of APOBEC family. AID converts cytidine to uracil, which is a key step for somatic hypermutation (SHM) and class switch recombination (CSR). AID also plays critical roles in B cell precursor stages, removing polyreactive B cells from immune repertoire. Since the main function of AID is inducing point mutations, dysregulation can lead to increased mutation load, translocations, disturbed genomic integrity, and lymphomagenesis. As such, expression of AID as well as its function is controlled strictly at various molecular steps. Other members of the APOBEC family also play crucial roles during carcinogenesis. Considering all these functions, AID represents a bridge, linking chronic inflammation to carcinogenesis and immune deficiencies to autoimmune manifestations.
Collapse
Affiliation(s)
- Elif Çakan
- Hacettepe University School of Medicine, Sihhiye, Ankara, Turkey
| | - Gurcan Gunaydin
- Department of Basic Oncology, Hacettepe University Cancer Institute, Sihhiye, Ankara, Turkey
| |
Collapse
|
4
|
Huang Q, Wei X, Li W, Ma Y, Chen G, Zhao L, Jiang Y, Xie S, Chen Q, Chen T. Endogenous Propionibacterium acnes Promotes Ovarian Cancer Progression via Regulating Hedgehog Signalling Pathway. Cancers (Basel) 2022; 14:5178. [PMID: 36358596 PMCID: PMC9658903 DOI: 10.3390/cancers14215178] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND The oncogenesis and progression of epithelial ovarian cancer (EOC) is a complicated process involving several key molecules and factors, yet whether microbiota are present in EOC, and their role in the development of EOC, remains greatly unknown. METHODS In this study, 20 patients were enrolled to compare the similarities and differences of intratumour microbiota among patients with epithelial benign ovarian tumours (EBOTs) and patients with EOC based on the high-throughput sequencing method. Subsequently, we further isolated the specific EOC-related bacteria and defined Propionibacterium acnes as a key strain in facilitating EOC progression. More importantly, we constructed a mouse EOC model to evaluate the effect of the P. acnes strain on EOC using immunohistochemistry, Western blotting, and RT-qPCR. RESULTS The high-throughput sequencing showed that the intratumour microbiota in EOC tissues had a higher microbial diversity and richness compared to EBOT tissues. The abundance of previously considered pathogens, Actinomycetales, Acinetobacter, Streptococcus, Ochrobacterium, and Pseudomonadaceae Pseudomonas, was increased in the EOC tissues. Meanwhile, we discovered the facilitating role of the P. acnes strain in the progression of EOC, which may be partially associated with the increased inflammatory response to activate the hedgehog (Hh) signalling pathway. This microbial-induced EOC progression mechanism is further confirmed using the inhibitor GANT61. CONCLUSIONS This study profiled the intratumour microbiota of EBOT and EOC tissues and demonstrated that the diversity and composition of the intratumour microbiota were significantly different. Furthermore, through in vivo and in vitro experiments, we confirmed the molecular mechanism of intratumour microbiota promotion of EOC progression in mice, which induces inflammation to activate the Hh signalling pathway. This could provide us clues for improving EOC treatment.
Collapse
Affiliation(s)
- Qifa Huang
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xin Wei
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Wenyu Li
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Yanbing Ma
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Guanxiang Chen
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Lu Zhao
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Ying Jiang
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Siqi Xie
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Qi Chen
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Tingtao Chen
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| |
Collapse
|
5
|
Konishi I, Abiko K, Hayashi T, Yamanoi K, Murakami R, Yamaguchi K, Hamanishi J, Baba T, Matsumura N, Mandai M. Peritoneal dissemination of high-grade serous ovarian cancer: pivotal roles of chromosomal instability and epigenetic dynamics. J Gynecol Oncol 2022; 33:e83. [PMID: 36032027 PMCID: PMC9428305 DOI: 10.3802/jgo.2022.33.e83] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 12/02/2022] Open
Abstract
Epithelial ovarian cancer remains the lethal gynecological malignancy in women. The representative histotype is high-grade serous carcinoma (HGSC), and most patients with HGSC present at advanced stages with peritoneal dissemination. Since the peritoneal dissemination is the most important factor for poor prognosis of the patients, complete exploration for its molecular mechanisms is mandatory. In this narrative review, being based on the clinical, pathologic, and genomic findings of HGSC, chromosomal instability and epigenetic dynamics have been discussed as the potential drivers for cancer development in the fallopian tube, acquisition of cancer stem cell (CSC)-like properties, and peritoneal metastasis of HGSC. The natural history of carcinogenesis with clonal evolution, and adaptation to microenvironment of peritoneal dissemination of HGSC should be targeted in the novel development of strategies for prevention, early detection, and precision treatment for patients with HGSC.
Collapse
Affiliation(s)
- Ikuo Konishi
- Department of Obstetrics and Gynecology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan.,Clinical Research Center, National Hospital Organization Kyoto Medical Center, Kyoto, Japan.,Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Kaoru Abiko
- Department of Obstetrics and Gynecology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Takuma Hayashi
- Clinical Research Center, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Koji Yamanoi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryusuke Murakami
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tsukasa Baba
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Morioka, Japan
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | |
Collapse
|
6
|
Inflammation accelerates BCR-ABL1+ B-ALL development through upregulation of AID. Blood Adv 2022; 6:4060-4072. [PMID: 35816360 PMCID: PMC9278295 DOI: 10.1182/bloodadvances.2021005017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 05/03/2022] [Indexed: 11/20/2022] Open
Abstract
Inflammatory stimulation promotes BCR-ABL1+ B-ALL disease progression by upregulating AID. Combination of imatinib and Hsp90 inhibitors significantly delays the inflammation-induced progression of BCR-ABL1+ B-ALL.
Inflammation contributes to the initiation and disease progression of several lymphoid malignancies. BCR-ABL1-positive B-cell acute lymphoblastic leukemia (BCR-ABL1+ B-ALL) is triggered by the malignant cloning of immature B cells promoted by the BCR-ABL1 fusion gene. However, it is unclear whether the mechanism driving the disease progression of BCR-ABL1+ B-ALL involves inflammatory stimulation. Here, we evaluate BCR-ABL1+ B-ALL cells’ response to inflammatory stimuli lipopolysaccharide (LPS) in vitro and in vivo. The results indicate that LPS promotes cell growth and genomic instability in cultured BCR-ABL1+ B-ALL cells and accelerates the BCR-ABL1+ B-ALL development in a mouse model. We show that the LPS-induced upregulation of activation-induced deaminase (AID) is required for the cell growth and disease progression of BCR-ABL1+ B-ALL. Moreover, AID modulates the expression of various genes that are dominated by suppressing apoptosis genes and upregulating DNA damage-repair genes. These genes lead to facilitation for BCR-ABL1+ B-ALL progression. The heat shock protein 90 (Hsp90) inhibitors significantly reduce AID protein level and delay the disease progression of BCR-ABL1+ B-ALL upon inflammatory stimulation. The present data demonstrate the causative role of AID in the development and progression of BCR-ABL1+ B-ALL during inflammation, thus highlighting potential therapeutic targets.
Collapse
|
7
|
Ritch SJ, Telleria CM. The Transcoelomic Ecosystem and Epithelial Ovarian Cancer Dissemination. Front Endocrinol (Lausanne) 2022; 13:886533. [PMID: 35574025 PMCID: PMC9096207 DOI: 10.3389/fendo.2022.886533] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is considered the deadliest gynecological disease and is normally diagnosed at late stages, at which point metastasis has already occurred. Throughout disease progression, EOC will encounter various ecosystems and the communication between cancer cells and these microenvironments will promote the survival and dissemination of EOC. The primary tumor is thought to develop within the ovaries or the fallopian tubes, both of which provide a microenvironment with high risk of causing DNA damage and enhanced proliferation. EOC disseminates by direct extension from the primary tumors, as single cells or multicellular aggregates. Under the influence of cellular and non-cellular factors, EOC spheroids use the natural flow of peritoneal fluid to reach distant organs within the peritoneal cavity. These cells can then implant and seed distant organs or tissues, which develop rapidly into secondary tumor nodules. The peritoneal tissue and the omentum are two common sites of EOC metastasis, providing a microenvironment that supports EOC invasion and survival. Current treatment for EOC involves debulking surgery followed by platinum-taxane combination chemotherapy; however, most patients will relapse with a chemoresistant disease with tumors developed within the peritoneum. Therefore, understanding the role of the unique microenvironments that promote EOC transcoelomic dissemination is important in improving patient outcomes from this disease. In this review article, we address the process of ovarian cancer cellular fate at the site of its origin in the secretory cells of the fallopian tube or in the ovarian surface epithelial cells, their detachment process, how the cells survive in the peritoneal fluid avoiding cell death triggers, and how cancer- associated cells help them in the process. Finally, we report the mechanisms used by the ovarian cancer cells to adhere and migrate through the mesothelial monolayer lining the peritoneum. We also discuss the involvement of the transcoelomic ecosystem on the development of chemoresistance of EOC.
Collapse
Affiliation(s)
- Sabrina J. Ritch
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Carlos M. Telleria
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC, Canada
- *Correspondence: Carlos M. Telleria, ; orcid.org/0000-0003-1070-3538
| |
Collapse
|
8
|
Systematic Analysis of Long Noncoding RNA and mRNA in Granulosa Cells during the Hen Ovulatory Cycle. Animals (Basel) 2021; 11:ani11061533. [PMID: 34070248 PMCID: PMC8225051 DOI: 10.3390/ani11061533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Chicken is one of the most economically important farm poultry, and providing many food products, such as meat and eggs for human consumption. However, follicle transcriptome studies in chickens with timepoints relating to changes in luteinizing hormone level remain unknown. In this study, the largest preovulatory follicle of chicken underwent the early, middle, and terminal stages of ovulatory cycle. Our work provides a comprehensive analysis of lncRNAs and mRNAs in chicken granulosa cells during the ovulatory cycle. A total of 12,479 mRNAs and 7528 lncRNAs were identified among the three stages. Thousands of lncRNAs were annotated, and the most differentially abundant genes were detected in the luteinizing hormone surge stage. Functional features of the lncRNAs and mRNAs at each stage were revealed, which was also associated with the changes in serum luteinizing hormone level. Especially, genes related to oxidative stress, steroids regulation, and inflammatory process were enriched in the luteinizing hormone surge stage, The comprehensive data generated in this study provides the foundation for future investigations to improve the reproductive performance of chickens and explore the mechanisms responsible for female ovarian diseases. Abstract Long non-coding RNAs (lncRNAs) and mRNAs are temporally expressed during chicken follicle development. However, follicle transcriptome studies in chickens with timepoints relating to changes in luteinizing hormone (LH) levels are rare. In this study, gene expression in Rohman layers was investigated at three distinct stages of the ovulatory cycle: zeitgeber time 0 (ZT0, 9:00 a.m.), zeitgeber time 12 (ZT12, 9:00 p.m.), and zeitgeber time 20 (ZT20, 5:00 a.m.) representing the early, middle, and LH surge stages, respectively, of the ovulatory cycle. Gene expression profiles were explored during follicle development at ZT0, ZT12, and ZT20 using Ribo-Zero RNA sequencing. The three stages were separated into two major stages, including the pre-LH surge and the LH surge stages. A total of 12,479 mRNAs and 7528 lncRNAs were identified among the three stages, and 4531, 523 differentially expressed genes (DEGs) and 2367, 211 differentially expressed lncRNAs (DELs) were identified in the ZT20 vs. ZT12, and ZT12 vs. ZT0, comparisons. Functional enrichment analysis revealed that genes involved in cell proliferation and metabolism processes (lipid-related) were mainly enriched in the ZT0 and ZT12 stages, respectively, and genes related to oxidative stress, steroids regulation, and inflammatory process were enriched in the ZT20 stage. These findings provide the basis for further investigation of the specific genetic and molecular functions of follicle development in chickens.
Collapse
|
9
|
Brand H, Barnabas GD, Sapoznik S, Bahar-Shany K, Pozniak Y, Yung Y, Hourvitz A, Geiger T, Jacob-Hirsch J, Levanon K. NF-κB-miR-155 axis activation mediates ovulation-induced oncogenic effects in fallopian tube epithelium. Carcinogenesis 2021; 41:1703-1712. [PMID: 32614381 DOI: 10.1093/carcin/bgaa068] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/15/2020] [Accepted: 06/24/2020] [Indexed: 02/06/2023] Open
Abstract
The fallopian tube secretory epithelial cells (FTSECs) are the cell-of-origin of most high-grade serous ovarian carcinomas (HGSOC). FTSECs are repeatedly exposed to inflammation induced by follicular fluid (FF) that is released with every ovulation cycle throughout a woman's reproductive years. Uninterrupted ovulation cycles are an established risk factor for HGSOC. Stimuli present in the FF induce an inflammatory environment which may cause DNA damage eventually leading to serous tumorigenesis. With the aim of elucidating possible mechanistic pathways, we established an 'ex vivo persistent ovulation model' mimicking the repeated exposure of human benign fallopian tube epithelium (FTE) to FF. We performed mass spectrometry analysis of the secretome of the ex vivo cultures as well as confirmatory targeted expressional and functional analyses. We demonstrated activation of the NF-κB pathway and upregulation of miR-155 following short-term exposure of FTE to human FF. Increased expression of miR-155 was also detected in primary HGSOC tumors compared with benign primary human FTE and corresponded with changes in the expression of miR-155 target genes. The phenotype of miR-155 overexpression in FTSEC cell line is of increased migratory and altered adhesion capacities. Overall, activation of the NF-κB-miR-155 axis in FTE may represent a possible link between ovulation-induced inflammation, DNA damage, and transcriptional changes that may eventually lead to serious carcinogenesis.
Collapse
Affiliation(s)
- Hadar Brand
- Sheba Cancer Research Center, Chaim Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | - Georgina D Barnabas
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | - Stav Sapoznik
- Sheba Cancer Research Center, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Keren Bahar-Shany
- Sheba Cancer Research Center, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Yair Pozniak
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | - Yuval Yung
- IVF Unit and Reproduction Lab, Department of Obstetrics and Gynecology, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Ariel Hourvitz
- Sackler Faculty of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel.,IVF Unit and Reproduction Lab, Department of Obstetrics and Gynecology, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Tamar Geiger
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | | | - Keren Levanon
- Sheba Cancer Research Center, Chaim Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel
| |
Collapse
|
10
|
Mei J, Tian H, Huang HS, Hsu CF, Liou Y, Wu N, Zhang W, Chu TY. Cellular models of development of ovarian high-grade serous carcinoma: A review of cell of origin and mechanisms of carcinogenesis. Cell Prolif 2021; 54:e13029. [PMID: 33768671 PMCID: PMC8088460 DOI: 10.1111/cpr.13029] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/19/2021] [Accepted: 03/03/2021] [Indexed: 12/11/2022] Open
Abstract
High-grade serous carcinoma (HGSC) is the most common and malignant histological type of epithelial ovarian cancer, the origin of which remains controversial. Currently, the secretory epithelial cells of the fallopian tube are regarded as the main origin and the ovarian surface epithelial cells as a minor origin. In tubal epithelium, these cells acquire TP53 mutations and expand to a morphologically normal 'p53 signature' lesion, transform to serous tubal intraepithelial carcinoma and metastasize to the ovaries and peritoneum where they develop into HGSC. This shifting paradigm of the main cell of origin has revolutionarily changed the focus of HGSC research. Various cell lines have been derived from the two cellular origins by acquiring immortalization via overexpression of hTERT plus disruption of TP53 and the CDK4/RB pathway. Malignant transformation was achieved by adding canonical driver mutations (such as gain of CCNE1) revealed by The Cancer Genome Atlas or by noncanonical gain of YAP and miR181a. Alternatively, because of the extreme chromosomal instability, spontaneous transformation can be achieved by long passage of murine immortalized cells, whereas in humans, it requires ovulatory follicular fluid, containing regenerating growth factors to facilitate spontaneous transformation. These artificially and spontaneously transformed cell systems in both humans and mice have been widely used to discover carcinogens, oncogenic pathways and malignant behaviours in the development of HGSC. Here, we review the origin, aetiology and carcinogenic mechanism of HGSC and comprehensively summarize the cell models used to study this fatal cancer having multiple cells of origin and overt genomic instability.
Collapse
Affiliation(s)
- Jie Mei
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Huixiang Tian
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Hsuan-Shun Huang
- Center for Prevention and Therapy of Gynecological Cancers, Department of Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan, ROC
| | - Che-Fang Hsu
- Center for Prevention and Therapy of Gynecological Cancers, Department of Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan, ROC
| | - Yuligh Liou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China
| | - Nayiyuan Wu
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Tang-Yuan Chu
- Center for Prevention and Therapy of Gynecological Cancers, Department of Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan, ROC.,Department of Obstetrics & Gynecology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan, ROC.,Department of Life Sciences, Tzu Chi University, Hualien, Taiwan, ROC
| |
Collapse
|
11
|
Activation-induced cytidine deaminase is a possible regulator of cross-talk between oocytes and granulosa cells through GDF-9 and SCF feedback system. Sci Rep 2021; 11:3833. [PMID: 33589683 PMCID: PMC7884688 DOI: 10.1038/s41598-021-83529-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/03/2021] [Indexed: 01/08/2023] Open
Abstract
Activation-induced cytidine deaminase (AID, Aicda) is a master gene regulating class switching of immunoglobulin genes. In this study, we investigated the significance of AID expression in the ovary. Immunohistological study and RT-PCR showed that AID was expressed in murine granulosa cells and oocytes. However, using the Aicda-Cre/Rosa-tdRFP reporter mouse, its transcriptional history in oocytes was not detected, suggesting that AID mRNA in oocytes has an exogenous origin. Microarray and qPCR validation revealed that mRNA expressions of growth differentiation factor-9 (GDF-9) in oocytes and stem cell factor (SCF) in granulosa cells were significantly decreased in AID-knockout mice compared with wild-type mice. A 6-h incubation of primary granuloma cells markedly reduced AID expression, whereas it was maintained by recombinant GDF-9. In contrast, SCF expression was induced by more than threefold, whereas GDF-9 completely inhibited its increase. In the presence of GDF-9, knockdown of AID by siRNA further decreased SCF expression. However, in AID-suppressed granulosa cells and ovarian tissues of AID-knockout mice, there were no differences in the methylation of SCF and GDF-9. These findings suggest that AID is a novel candidate that regulates cross-talk between oocytes and granulosa cells through a GDF-9 and SCF feedback system, probably in a methylation-independent manner.
Collapse
|
12
|
Pathak S, Wilczyński JR, Paradowska E. Factors in Oncogenesis: Viral Infections in Ovarian Cancer. Cancers (Basel) 2020; 12:E561. [PMID: 32121320 PMCID: PMC7139377 DOI: 10.3390/cancers12030561] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 02/16/2020] [Accepted: 02/25/2020] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer (OC) is one of the leading causes of cancer death in women, with high-grade serous ovarian cancer (HGSOC) being the most lethal gynecologic malignancy among women. This high fatality rate is the result of diagnosis of a high number of new cases when cancer implants have already spread. The poor prognosis is due to our inadequate understanding of the molecular mechanisms preceding ovarian malignancy. Knowledge about the site of origination has been improved recently by the discovery of tube intraepithelial cancer (TIC), but the potential risk factors are still obscure. Due to high tumoral heterogeneity in OC, the establishment of early stage biomarkers is still underway. Microbial infection may induce or result in chronic inflammatory infection and in the pathogenesis of cancers. Microbiome research has shed light on the relationships between the host and microbiota, as well as the direct roles of host pathogens in cancer development, progression, and drug efficacy. While controversial, the detection of viruses within ovarian malignancies and fallopian tube tissues suggests that these pathogens may play a role in the development of OC. Genomic and proteomic approaches have enhanced the methods for identifying candidates in early screening. This article summarizes the existing knowledge related to the molecular mechanisms that lead to tumorigenesis in the ovary, as well as the viruses detected in OC cases and how they may elevate this process.
Collapse
Affiliation(s)
- Sudipta Pathak
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, 93-232 Lodz, Poland;
| | - Jacek R. Wilczyński
- Department of Surgical and Oncological Gynecology, Medical University of Lodz, 90-419 Lodz, Poland;
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, 93-232 Lodz, Poland;
| |
Collapse
|
13
|
Bergsten TM, Burdette JE, Dean M. Fallopian tube initiation of high grade serous ovarian cancer and ovarian metastasis: Mechanisms and therapeutic implications. Cancer Lett 2020; 476:152-160. [PMID: 32067992 DOI: 10.1016/j.canlet.2020.02.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/31/2020] [Accepted: 02/13/2020] [Indexed: 01/09/2023]
Abstract
Ovarian cancer is the most lethal gynecologic malignancy and the fifth leading cause of cancer-related death in women. Although outcomes have improved in recent years, there remains an unmet clinical need to understand the early pathogenesis of ovarian cancer in order to identify new diagnostic approaches and agents of chemoprevention and chemotherapy. While high grade serous ovarian cancer (HGSOC), the most abundant histotype, was initially thought to arise from the ovarian surface epithelium, there is an increasing body of evidence suggesting that HGSOC originates in the fallopian tube. With this new understanding of cell of origin, understanding of disease development requires analysis with a novel perspective. Currently, factors that drive the initiation and migration of dysplastic tubal epithelial cells from the fallopian tube to the ovary are not yet fully defined. These factors include common mutations to fallopian tube epithelial cells, as well as factors originating from both the fallopian tube and ovary which are capable of inducing transformation and dissemination in said cells. Here, we review these changes, their causative agents, and various potential means of intervention.
Collapse
Affiliation(s)
- Tova M Bergsten
- Medical Scientist Training Program, University of Illinois at Chicago College of Medicine, Chicago, IL, USA; Department of Pharmaceutical Sciences, Center for Biomolecular Science, University of Illinois at Chicago, Chicago, IL, USA
| | - Joanna E Burdette
- Department of Pharmaceutical Sciences, Center for Biomolecular Science, University of Illinois at Chicago, Chicago, IL, USA
| | - Matthew Dean
- Department of Animal Science, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
14
|
Shi J, Huo R, Li N, Li H, Zhai T, Li H, Shen B, Ye J, Fu R, Di W. CYR61, a potential biomarker of tumor inflammatory response in epithelial ovarian cancer microenvironment of tumor progress. BMC Cancer 2019; 19:1140. [PMID: 31766991 PMCID: PMC6878653 DOI: 10.1186/s12885-019-6321-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 10/31/2019] [Indexed: 01/14/2023] Open
Abstract
Background Recent studies have found that inflammatory response is involved in the pathogenesis of ovarian cancer. Advanced ovarian cancer is often presented with ascites that is rich in cytokines, inflammatory factors or cancer cells. Therefore, it is important to study the microenvironment of ascites in order to further clarify the occurrence and progression of ovarian cancer. As a pro-inflammatory factor, the Cyr61 expression patterns are inconsistent in human tumors. Although it has been reported that Cyr61 is related to the progression of ovarian cancer, its specific mechanism is not yet clear. This study sought to evaluate the Cyr61 levels of ascites, serum and different tissues of ovarian cancer to explore the potential association of Cyr61with the tumor-associated inflammatory microenvironment of EOC. Methods Tumor specimens were procured from patients with ovarian serous cystadenocarcinoma and ovarian serous cystadenoma. Cyr61 and IL-6 levels of serum or ascites were determined by ELISA (Enzyme-Linked ImmunoSorbent Assay), while Cyr61 expressions of different ovarian tumor tissues were evaluated by IHC (Immunohistochemistry). Then the correlation of Cyr61 level in ascites with clinicopathologic features was analyzed. And other laboratory data were obtained from medical records. Results Both in ascites and serum, significantly higher Cyr61 levels were found in ovarian serous cystadenocarcinoma. In malignant ascites, higher Cyr61 level of ovarian serous cystadenocarcinoma was more closely associated with FIGO stage, initial tumor size > 10 cm and the residual tumor size. And the increased IL-6 level was linearly related to Cyr61 level. Moreover, the serum levels of Cyr61, IL-6 and CRP in advanced stage of ovarian cancer were much higher than those in early stage. Lastly, the IHC data demonstrate that Cyr61 expression of ovarian serous adenocarcinoma was higher than that of ovarian serous cystadenoma, but it was lower than the paired metastatic lesions. Conclusions As a pro-inflammatory factor, increased ascites Cyr61 level is associated with FIGO stage, initial tumor size > 10 cm and the residual tumor size. Moreover, serum Cyr61 may be used as a potential marker for EOC inflammatory response. Finally, Cyr61 may be involved in the process of tumor metastasis and progression by producing IL-6 and CRP in the EOC inflammatory microenvironment.
Collapse
Affiliation(s)
- Jun Shi
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, People's Republic of China
| | - Rongfen Huo
- Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Ningli Li
- Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Haichuan Li
- Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Tianhang Zhai
- Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Huidan Li
- Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Baihua Shen
- Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Jing Ye
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, People's Republic of China
| | - Ruojin Fu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, People's Republic of China
| | - Wen Di
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China. .,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, People's Republic of China.
| |
Collapse
|
15
|
Li L, Su N, Cui M, Li H, Zhang Q, Yu N, Wu S, Cao Z. Activation-induced cytidine deaminase expression in colorectal cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:4119-4124. [PMID: 31933808 PMCID: PMC6949800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 09/27/2019] [Indexed: 06/10/2023]
Abstract
Activation-induced cytidine deaminase (AID) produces immune-diversity by inducing somatic hypermutations and class-switch recombinations in human immunoglobulin genes. This role of AID in causing genomic mutations, also can potentially cause somatic mutations in various host genes of non-lymphoid tissues, and contribute to tumorigenesis. The goal of the present study was to investigate whether AID expression was involved in the development or progression of colorectal cancer, and the nuclear expression of p53 protein in cancer cells. We examined the pattern of expression of AID and p53 proteins in 71 colorectal adenomas and 122 sporadic colorectal cancers by immunohistochemistry. AID and p53 expression was detected in 57 (46.7%) and 78 (63.9%) out of 122 colorectal cancers, respectively. Statistically, the expression of the AID protein was not associated with the 5-year survival or clinical and pathological parameters, including tumor stage, location, size, and lymph node metastasis (P > 0.05). However, the expression of the AID protein was associated with tumor differentiation (P = 0.004). In addition, a significant association was observed between AID and the nuclear expression of p53 in colorectal cancers (P = 0.0357). Only 3 (4.2%) of the 71 colorectal adenomas showed immunopostivity for AID, resulting in a significant difference between total colorectal cancers and adenomas (P < 0.001). The p53 expression was detected in 7 (9.9%) out of 71 colorectal adenomas. Statistically, AID protein was not associated with the degree of dysplasia and the nuclear expression of p53 in colorectal adenomas (P > 0.05). These results suggest that aberrant expression of the AID protein might play a role in the development of colorectal cancers.
Collapse
Affiliation(s)
- Lanlan Li
- Department of Pathology, Binzhou Medical UniversityYantai, Shandong, China
| | - Nana Su
- Department of Pathology, Binzhou Medical UniversityYantai, Shandong, China
| | - Min Cui
- Department of Pediatrics, Binzhou City People’s HospitalBinzhou, Shandong, China
| | - Hong Li
- Department of Pathology, Binzhou Medical UniversityYantai, Shandong, China
| | - Qian Zhang
- Department of Pathology, Binzhou Medical UniversityYantai, Shandong, China
| | - Ning Yu
- Department of Pathology, Binzhou Medical UniversityYantai, Shandong, China
| | - Shuhua Wu
- Department of Pathology, Binzhou Medical UniversityYantai, Shandong, China
| | - Zhang Cao
- Department of Pathology, Binzhou Medical UniversityYantai, Shandong, China
| |
Collapse
|
16
|
Wilson JJ, Chow KH, Labrie NJ, Branca JA, Sproule TJ, Perkins BRA, Wolf EE, Costa M, Stafford G, Rosales C, Mills KD, Roopenian DC, Hasham MG. Enhancing the efficacy of glycolytic blockade in cancer cells via RAD51 inhibition. Cancer Biol Ther 2018; 20:169-182. [PMID: 30183475 PMCID: PMC6343731 DOI: 10.1080/15384047.2018.1507666] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Targeting the early steps of the glycolysis pathway in cancers is a well-established therapeutic strategy; however, the doses required to elicit a therapeutic effect on the cancer can be toxic to the patient. Consequently, numerous preclinical and clinical studies have combined glycolytic blockade with other therapies. However, most of these other therapies do not specifically target cancer cells, and thus adversely affect normal tissue. Here we first show that a diverse number of cancer models – spontaneous, patient-derived xenografted tumor samples, and xenografted human cancer cells – can be efficiently targeted by 2-deoxy-D-Glucose (2DG), a well-known glycolytic inhibitor. Next, we tested the cancer-cell specificity of a therapeutic compound using the MEC1 cell line, a chronic lymphocytic leukemia (CLL) cell line that expresses activation induced cytidine deaminase (AID). We show that MEC1 cells, are susceptible to 4,4ʹ-Diisothiocyano-2,2ʹ-stilbenedisulfonic acid (DIDS), a specific RAD51 inhibitor. We then combine 2DG and DIDS, each at a lower dose and demonstrate that this combination is more efficacious than fludarabine, the current standard- of- care treatment for CLL. This suggests that the therapeutic blockade of glycolysis together with the therapeutic inhibition of RAD51-dependent homologous recombination can be a potentially beneficial combination for targeting AID positive cancer cells with minimal adverse effects on normal tissue. Implications: Combination therapy targeting glycolysis and specific RAD51 function shows increased efficacy as compared to standard of care treatments in leukemias.
Collapse
Affiliation(s)
- John J Wilson
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Kin-Hoe Chow
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Nathan J Labrie
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Jane A Branca
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Thomas J Sproule
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Bryant R A Perkins
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Elise E Wolf
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Mauro Costa
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Grace Stafford
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Christine Rosales
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | | | - Derry C Roopenian
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Muneer G Hasham
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| |
Collapse
|
17
|
The Role of Inflammation and Inflammatory Mediators in the Development, Progression, Metastasis, and Chemoresistance of Epithelial Ovarian Cancer. Cancers (Basel) 2018; 10:cancers10080251. [PMID: 30061485 PMCID: PMC6116184 DOI: 10.3390/cancers10080251] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/20/2018] [Accepted: 07/24/2018] [Indexed: 12/12/2022] Open
Abstract
Inflammation plays a role in the initiation and development of many types of cancers, including epithelial ovarian cancer (EOC) and high grade serous ovarian cancer (HGSC), a type of EOC. There are connections between EOC and both peritoneal and ovulation-induced inflammation. Additionally, EOCs have an inflammatory component that contributes to their progression. At sites of inflammation, epithelial cells are exposed to increased levels of inflammatory mediators such as reactive oxygen species, cytokines, prostaglandins, and growth factors that contribute to increased cell division, and genetic and epigenetic changes. These exposure-induced changes promote excessive cell proliferation, increased survival, malignant transformation, and cancer development. Furthermore, the pro-inflammatory tumor microenvironment environment (TME) contributes to EOC metastasis and chemoresistance. In this review we will discuss the roles inflammation and inflammatory mediators play in the development, progression, metastasis, and chemoresistance of EOC.
Collapse
|
18
|
Caldwell RB, Braselmann H, Heuer S, Schötz U, Zitzelsberger H. Gain-of-function analysis of cis-acting diversification elements in DT40 cells. Immunol Cell Biol 2018; 96:948-957. [PMID: 29665088 DOI: 10.1111/imcb.12158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 04/09/2018] [Accepted: 04/09/2018] [Indexed: 02/02/2023]
Abstract
Activation-induced cytidine deaminase (AID) is required for the immunoglobulin diversification processes of somatic hypermutation, gene conversion and class-switch recombination. The targeting of AID's deamination activity is thought to be a combination of cis- and trans-acting elements, but has not been fully elucidated. Deletion analysis of putative proximal cis-regulatory motifs, while helpful, fails to identify additive versus cumulative effects, redundancy, and may create new motifs where none previously existed. In contrast, gain-of-function analysis can be more insightful with fewer of the same drawbacks and the output is a positive result. Here, we show five defined DNA regions of the avian Igλ locus that are sufficient to confer events of hypermutation to a target gene. In our analysis, the essential cis-targeting elements fully reconstituted diversification of a transgene under heterologous promotion in the avian B-cell line DT40. Furthermore, to the best of our knowledge two of the five regions we report on here have not previously been described as individually having an influence on somatic hypermutation.
Collapse
Affiliation(s)
- Randolph B Caldwell
- Department of Radiation Sciences - Research Unit Radiation Cytogenetics, Helmholtz Center Munich - German Research Center for Environmental Health (GmbH), Neuherberg, 85764, Germany
| | - Herbert Braselmann
- Department of Radiation Sciences - Research Unit Radiation Cytogenetics, Helmholtz Center Munich - German Research Center for Environmental Health (GmbH), Neuherberg, 85764, Germany
| | - Steffen Heuer
- Department of Radiation Sciences - Research Unit Radiation Cytogenetics, Helmholtz Center Munich - German Research Center for Environmental Health (GmbH), Neuherberg, 85764, Germany
| | - Ulrike Schötz
- Department of Radiotherapy and Radiooncology, Philipps-University Marburg, University Hospital Gießen and Marburg, Marburg, 35043, Germany
| | - Horst Zitzelsberger
- Department of Radiation Sciences - Research Unit Radiation Cytogenetics, Helmholtz Center Munich - German Research Center for Environmental Health (GmbH), Neuherberg, 85764, Germany.,Helmholtz Center Munich, Clinical Cooperation Group 'Personalized Radiotherapy of Head and Neck Cancer', Neuherberg, 85764, Germany.,Department of Radiotherapy and Radiation Oncology, Ludwig-Maximilians-Universitaet, University Hospital Munich, Munich, 81377, Germany
| |
Collapse
|
19
|
Kobayashi H, Ogawa K, Kawahara N, Iwai K, Niiro E, Morioka S, Yamada Y. Sequential molecular changes and dynamic oxidative stress in high-grade serous ovarian carcinogenesis. Free Radic Res 2017; 51:755-764. [PMID: 28931330 DOI: 10.1080/10715762.2017.1383605] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The mechanism of high-grade serous ovarian cancer (HGSC) development remains elusive. This review outlines recent advances in the understanding of sequential molecular changes associated with the development of HGSC, as well as describes oxidative stress-induced genomic instability and carcinogenesis. This article reviews the English language literature between 2005 and 2017. Clinicopathological features analysis provides a sequential progression of fallopian tubal epithelium to precursor lesions to type 2 HGSC. HGSC may develop over a long time after incessant ovulation and repeated retrograde menstruation via stepwise accumulation of genetic alterations, including PAX2, ALDH1A1, STMN1, EZH2 and CCNE1, which confer positive selection of cells with growth advantages through acquiring driver mutations such as BRCA1/2, p53 or PTEN/PIK3CA. Haemoglobin and iron-induced oxidative stress leads to the emergence of genetic alterations in fallopian tubal epithelium via increased DNA damage and impaired DNA repair. Serous tubal intraepithelial carcinoma (STIC), the likely precursor of HGSC, may be susceptible to DNA double-strand breaks, exhibit DNA replication stress and increase genomic instability. The induction of genomic instability is considered to be a driving mechanism of reactive oxygen species (ROS)-induced carcinogenesis. HGSC exemplifies the view of stepwise cancer development. We describe how genetic alterations emerge during HGSC carcinogenesis related to oxidative stress.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- a Department of Obstetrics and Gynecology , Nara Medical University , Nara , Japan
| | - Kenji Ogawa
- a Department of Obstetrics and Gynecology , Nara Medical University , Nara , Japan
| | - Naoki Kawahara
- a Department of Obstetrics and Gynecology , Nara Medical University , Nara , Japan
| | - Kana Iwai
- a Department of Obstetrics and Gynecology , Nara Medical University , Nara , Japan
| | - Emiko Niiro
- a Department of Obstetrics and Gynecology , Nara Medical University , Nara , Japan
| | - Sachiko Morioka
- a Department of Obstetrics and Gynecology , Nara Medical University , Nara , Japan
| | - Yuki Yamada
- a Department of Obstetrics and Gynecology , Nara Medical University , Nara , Japan
| |
Collapse
|
20
|
Choudhary M, Tamrakar A, Singh AK, Jain M, Jaiswal A, Kodgire P. AID Biology: A pathological and clinical perspective. Int Rev Immunol 2017; 37:37-56. [PMID: 28933967 DOI: 10.1080/08830185.2017.1369980] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Activation-induced cytidine deaminase (AID), primarily expressed in activated mature B lymphocytes in germinal centers, is the key factor in adaptive immune response against foreign antigens. AID is responsible for producing high-affinity and high-specificity antibodies against an infectious agent, through the physiological DNA alteration processes of antibody genes by somatic hypermutation (SHM) and class-switch recombination (CSR) and functions by deaminating deoxycytidines (dC) to deoxyuridines (dU), thereby introducing point mutations and double-stranded chromosomal breaks (DSBs). The beneficial physiological role of AID in antibody diversification is outweighed by its detrimental role in the genesis of several chronic immune diseases, under non-physiological conditions. This review offers a comprehensive and better understanding of AID biology and its pathological aspects, as well as addresses the challenges involved in AID-related cancer therapeutics, based on various recent advances and evidence available in the literature till date. In this article, we discuss ways through which our interpretation of AID biology may reflect upon novel clinical insights, which could be successfully translated into designing clinical trials and improving patient prognosis and disease management.
Collapse
Affiliation(s)
- Meenal Choudhary
- a Centre for Biosciences and Biomedical Engineering , Indian Institute of Technology Indore , Simrol , Indore , Madhya Pradesh , India
| | - Anubhav Tamrakar
- a Centre for Biosciences and Biomedical Engineering , Indian Institute of Technology Indore , Simrol , Indore , Madhya Pradesh , India
| | - Amit Kumar Singh
- a Centre for Biosciences and Biomedical Engineering , Indian Institute of Technology Indore , Simrol , Indore , Madhya Pradesh , India
| | - Monika Jain
- a Centre for Biosciences and Biomedical Engineering , Indian Institute of Technology Indore , Simrol , Indore , Madhya Pradesh , India
| | - Ankit Jaiswal
- a Centre for Biosciences and Biomedical Engineering , Indian Institute of Technology Indore , Simrol , Indore , Madhya Pradesh , India
| | - Prashant Kodgire
- a Centre for Biosciences and Biomedical Engineering , Indian Institute of Technology Indore , Simrol , Indore , Madhya Pradesh , India
| |
Collapse
|
21
|
Khajeh M, Rahbarghazi R, Nouri M, Darabi M. Potential role of polyunsaturated fatty acids, with particular regard to the signaling pathways of arachidonic acid and its derivatives in the process of maturation of the oocytes: Contemporary review. Biomed Pharmacother 2017; 94:458-467. [PMID: 28779707 DOI: 10.1016/j.biopha.2017.07.140] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 01/20/2023] Open
Abstract
Oocyte meiotic maturation is one of the significant physiological requirements for ovulation and fertility. It is believed that Cyclic Adenosine Monophosphate, protein kinase A and protein kinase C pathways along with eicosanoids, particularly prostaglandin E2, and steroids are the key factors regulating mammalian oocyte maturation. The aim of the current study was to highlight the molecular events triggered by arachidonic acid during oocyte meiotic arrest and resumption at the time of gonadotrophin surge. It should be noted that arachidonic acid release is tightly regulated by Follicle-stimulating and Luteinizing hormones during oocyte development.
Collapse
Affiliation(s)
- Masoumeh Khajeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Nouri
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Darabi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
22
|
Cytidine deaminase Apobec3a induction in fallopian epithelium after exposure to follicular fluid. Gynecol Oncol 2017; 145:577-583. [PMID: 28215840 DOI: 10.1016/j.ygyno.2017.02.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 02/07/2017] [Accepted: 02/08/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Ovarian carcinomas that originate from fallopian epithelial cells are suggested to arise due to repeated exposure to ovulatory follicular fluid (FF). Mechanistic explanation(s) for how this occurs are unknown. Here, we sought to understand if FF exposure to fallopian epithelial cells could induce DNA damage and expression of a known family of DNA mutators, apolipoprotein B mRNA editing enzyme, catalytic polypeptide (APOBEC) cytidine deaminases. METHODS Follicular fluid and matched patient plasma samples were obtained from donors. Fallopian epithelial cells (FT33-TAg, FT189, FT190, and FT194) were cultured with FF or plasma for 24h, and cell proliferation and DNA damage were assessed. Effects of FF on Apobec gene expression were determined by qRT-PCR and western blot analyses. Fallopian epithelial cells were transfected with an APOBEC3A expression vector and DNA damage was assessed. RESULTS Follicular fluid exposure increased epithelial cell proliferation as measured by three independent methods, and DNA damage accumulation as assessed using three independent measures. This effect was specific to FF, as matched patient plasma did not have the same effects. Increased expression of Apobec3a was observed in fallopian epithelial cells following exposure to 5 of 8 patient FF samples, and transient overexpression of APOBEC3A was sufficient to induce double strand DNA breaks. CONCLUSIONS Follicular fluid can induce cell proliferation and DNA damage accumulation in cultured fallopian epithelial cells. Increased expression of APOBEC3A, a known DNA mutator, may explain the high incidence of DNA damage after FF exposure. The role of Apobec3a in ovulation-induced inflammation warrants further investigation.
Collapse
|