1
|
Fan X, Wu L, Cheng T, Lv W, Tian J, Tao J, Tu S, Tan F, Wang Y. Oroxylin A may promote cell apoptosis and inhibit epithelial-mesenchymal transition in endometrial cancer, associated with the ERβ/PI3K/AKT pathway. Sci Rep 2025; 15:12225. [PMID: 40211010 PMCID: PMC11986019 DOI: 10.1038/s41598-025-97122-z] [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: 12/05/2024] [Accepted: 04/02/2025] [Indexed: 04/12/2025] Open
Abstract
Endometrial cancer (EC) is a prevalent gynecological cancer worldwide, often associated with poor prognosis after recurrence or metastasis. Oroxylin A (OA) is an active flavonoid compound with a strong anti-tumor function. However, the effects of OA on EC remain unknown. In this study, we planned to investigate the anti-EC effects of OA and explore its mechanisms. Five cell lines were used for in vitro experiments, and female BALB/c nude mice were applied for xenograft experiments. The cytotoxicity and experimental concentration of OA were detected by CCK-8. Wound healing, transwell, and colony formation assays were used to evaluate the anti-metastatic and anti-proliferative activities of OA on EC cells. TUNEL assay and flow cytometry were applied for the evaluation of apoptosis. Network pharmacology was used to explore potential targets, and molecular dynamics simulations and dockings were applied for the quantification of binding energy, and stability of OA. RT-qPCR, WB, and immunofluorescence were applied for the detection of localization and expression of correlated markers. The results showed that OA notably inhibited the proliferation, migration, and invasion of Ishikawa cells. Meanwhile, in vivo Ishikawa xenograft assays demonstrated that OA notably inhibited growth and promoted apoptosis of EC. Mechanistically, after treatment with OA, the expressions of Cleaved Caspase-3, BAX, E-cadherin, and ERβ were increased, while the expressions of Bcl-2, Vimentin, N-cadherin, MMP2, MMP9, PI3K and phospho-AKT (Ser473) were decreased. Therefore, OA may exhibit significant anti-EC effects by regulating the ERβ/PI3K/AKT pathway to promote apoptosis and inhibit epithelial-mesenchymal transition (EMT).
Collapse
Affiliation(s)
- Xue Fan
- Gansu International Scientific and Technological Cooperation Base of Reproductive Medicine Transformation Application and Key Laboratory for Reproductive Medicine and Embryo of Gansu Province, The First Hospital of Lanzhou University and The First School of Clinical Medicine, Lanzhou University, West Donggang Road 1, Lanzhou, 730000, China
| | - Luming Wu
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Tong Cheng
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Weilong Lv
- Gansu International Scientific and Technological Cooperation Base of Reproductive Medicine Transformation Application and Key Laboratory for Reproductive Medicine and Embryo of Gansu Province, The First Hospital of Lanzhou University and The First School of Clinical Medicine, Lanzhou University, West Donggang Road 1, Lanzhou, 730000, China
| | - Jiao Tian
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Jijun Tao
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Shiyan Tu
- Gansu International Scientific and Technological Cooperation Base of Reproductive Medicine Transformation Application and Key Laboratory for Reproductive Medicine and Embryo of Gansu Province, The First Hospital of Lanzhou University and The First School of Clinical Medicine, Lanzhou University, West Donggang Road 1, Lanzhou, 730000, China
| | - Fangjun Tan
- Gansu Province Third People's Hospital, Lanzhou, 730000, China
| | - Yiqing Wang
- Gansu International Scientific and Technological Cooperation Base of Reproductive Medicine Transformation Application and Key Laboratory for Reproductive Medicine and Embryo of Gansu Province, The First Hospital of Lanzhou University and The First School of Clinical Medicine, Lanzhou University, West Donggang Road 1, Lanzhou, 730000, China.
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
2
|
Soberanis Pina P, Clemens K, Bubie A, Grant B, Haynes G, Zhang N, Drusbosky L, Lheureux S. Genomic Landscape of ctDNA and Real-World Outcomes in Advanced Endometrial Cancer. Clin Cancer Res 2024; 30:5657-5665. [PMID: 39417689 PMCID: PMC11647206 DOI: 10.1158/1078-0432.ccr-24-2105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/04/2024] [Accepted: 10/15/2024] [Indexed: 10/19/2024]
Abstract
PURPOSE ctDNA is a novel technique extensively studied in solid tumors, although not currently well defined in endometrial cancer. EXPERIMENTAL DESIGN A de-identified retrospective analysis of 1,988 patients with advanced/recurrent endometrial cancer was performed. In addition, an analysis of a real-world evidence cohort was completed (n = 1,266). Patients underwent ctDNA testing using Guardant360 during routine clinical care. The objective was to describe and assess molecular landscape using ctDNA. RESULTS Among 1,988 ctDNA samples, at least one somatic alteration was detected in 91.6% (n = 1,821). Most frequently altered genes were TP53 (64%), PIK3CA (29%), PTEN (25%), ARID1A (20%), and KRAS (14%). Overall, 18.5% had amplifications, with the majority identified in CCNE1 (40.9%), PIK3CA (22%), and EGFR (19.3%). From the real-world evidence cohort, those with TP53 mutations had a worse overall survival (OS) versus those without TP53 mutations (P = 0.02) and those with TP53 comutations had an inferior OS in comparison with TP53-mutated only (P = 0.016). Amongst these, patients with a PIK3CA comutation (P = 0.012) and CCNE1 amplification (P = 0.01) had an inferior OS compared with those with only TP53 mutations. Fifty-seven patients with newly diagnosed endometrial cancer had at least two serial ctDNA samples showing evolution in detected variants compared with baseline samples, with TP53 being the most frequent change. CONCLUSIONS This study is one of the largest cohorts of ctDNA currently reported in endometrial cancer. The presence of TP53 mutation and other comutations detected by ctDNA have a negative effect on outcomes. This report suggests that ctDNA analysis is feasible and could become a useful biomarker for endometrial cancer.
Collapse
Affiliation(s)
- Pamela Soberanis Pina
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | | | | | - Brooke Grant
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | | | | | | | - Stephanie Lheureux
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| |
Collapse
|
3
|
Banerjee S, Ingles Russo Garces A, Garside J, Rahman T, Pearson C, Heffernan K. Real-world patient characteristics and survival outcomes in patients with advanced or recurrent endometrial cancer in England: a retrospective, population-based study. BMJ Open 2024; 14:e083540. [PMID: 39581729 PMCID: PMC11590861 DOI: 10.1136/bmjopen-2023-083540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 09/30/2024] [Indexed: 11/26/2024] Open
Abstract
OBJECTIVE This study defined a retrospective cohort of patients in England with primary advanced or recurrent (A/R) endometrial cancer (EC) who may have been eligible for clinical trials evaluating immune checkpoint inhibitors (ICIs) in the first-line (1L) setting within a real-world dataset, and described the characteristics, treatment patterns and outcomes within this cohort. DESIGN This was a retrospective, population-based study. SETTING Routine population-level data from the National Cancer Registration and Analysis Service in England were used. Patients diagnosed with A/R EC between 1 January 2013 and 31 December 2019 were included (follow-up until 23 August 2021). ICI-eligible patients who received any 1L therapy (defined as first systemic treatment for A/R EC with or without radiotherapy) and met key eligibility criteria for the RUBY trial (NCT03981796; 1L cohort) were included. A subpopulation who solely received carboplatin-paclitaxel at 1L (carboplatin-paclitaxel subcohort) was identified. METHODS Demographics, characteristics and therapy received were reported. Overall survival (OS), time to next treatment (TTNT) and time to treatment discontinuation (TTD) from 1L chemotherapy initiation were assessed using Kaplan-Meier methodology. RESULTS Of 13 954 patients identified, 2376 ICI-eligible patients were included in the 1L cohort (median [range] age: 67.9 [26.7-94.0] years); 902 patients received solely carboplatin-paclitaxel at 1L. Demographics and disease characteristics were generally similar between cohorts. Median (95% CI) OS, TTNT and TTD from 1L chemotherapy were 27.2 (24.7, 30.2), 16.9 (15.8, 18.5) and 3.4 (3.4, 3.4) months, respectively, in the 1L cohort, and 17.2 (15.5, 19.0), 12.4 (11.6, 13.5) and 3.4 (3.4, 3.4) months, respectively, in the carboplatin-paclitaxel subcohort. CONCLUSION Long-term outcomes were poor for both cohorts, particularly the carboplatin-paclitaxel subcohort, where patients did not receive radiotherapy and had predominantly metastatic disease. This reflects the unmet need for more durable treatment options to prevent relapse and prolong survival in this patient population. This real-world study will help contextualise outcomes from ongoing phase III clinical trials investigating 1L ICI treatments.
Collapse
Affiliation(s)
- Susana Banerjee
- The Royal Marsden NHS Foundation Trust, London, UK
- The Institute of Cancer Research, London, UK
| | | | | | - Tameera Rahman
- Health Data Insight CIC, Cambridge, UK
- NHS England, National Disease Registration Service, Leeds, UK
| | - Clare Pearson
- Health Data Insight CIC, Cambridge, UK
- NHS England, National Disease Registration Service, Leeds, UK
| | | |
Collapse
|
4
|
Li N, Li H, Wei L, Chen H, Wu Z, Yuwen S, Yang S. The Downregulation of MMP23B Facilitates the Suppression of Vitality and Induction of Apoptosis in Endometrial Cancer Cells. Reprod Sci 2024; 31:3452-3461. [PMID: 38782818 PMCID: PMC11527946 DOI: 10.1007/s43032-024-01581-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
Endometrial cancer is a malignant tumor that commonly occurs in the female reproductive system and its incidence is still increasing. The mechanism of the development of endometrial cancer has not yet been fully clarified, so we need to continuously study the relevant mechanisms of endometrial cancer and continue to explore its biomarkers in order to discover more precise and effective treatment methods for endometrial cancer. RT-qPCR (Real-Time quantitative Polymerase Chain Reaction) experiments were used to detect the expression level of MMP23B (Matrix Metalloproteinase 23B) in endometrial cancer cells; the clinical data of the TCGA (The Cancer Genome Atlas) database were downloaded, and gene expression profiles were analyzed to investigate the correlation between MMP23B (Matrix Metalloproteinase 23B) and the survival prognosis of endometrial cancer, and functional enrichment analysis was performed on MMP23B (Matrix Metalloproteinase 23B) related genes. After silencing MMP23B (Matrix Metalloproteinase 23B), CCK8 (Cell Counting Kit-8), RT-qPCR (Real-Time quantitative Polymerase Chain Reaction), scratch assay, and transwell assay were used to detect cell viability, levels of apoptotic factors, migration rate, and invasion number of endometrial cancer, respectively. MMP23B (Matrix Metalloproteinase 23B) was highly expressed in endometrial cancer, which is closely related to a poor survival prognosis for endometrial cancer, and may act on endometrial cancer through apoptosis-related functions. The downregulation of MMP23B (Matrix Metalloproteinase 23B) reduced the cell viability of endometrial cancer cells, upregulated the expression levels of CASP3 (Caspase-3), CASP8 (Caspase-8) and CASP9 (Caspase-9) in cells, and inhibited cell migration and invasion.
Collapse
Affiliation(s)
- Ning Li
- Department of Gynaecology and Obstetrics, Guangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Hua Li
- Department of Pathology, Guangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Lijuan Wei
- Institute of Basic Medical Science, Medicine and Health Research Institute of Guangxi Academy of Sciences, Nanning, Guangxi, China
| | - Hui Chen
- Institute of Basic Medical Science, Medicine and Health Research Institute of Guangxi Academy of Sciences, Nanning, Guangxi, China
| | - Zhaorong Wu
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Si Yuwen
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Sufang Yang
- Department of Reproductive Health and Infertility, Guangxi International Zhuang Medicine Hospital, Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China.
| |
Collapse
|
5
|
Yang X, Liu Z, Tang W, Pratap UP, Collier AB, Altwegg KA, Gopalam R, Li X, Yuan Y, Zhou D, Lai Z, Chen Y, Sareddy GR, Valente PT, Kost ER, Viswanadhapalli S, Vadlamudi RK. PELP1 inhibition by SMIP34 reduces endometrial cancer progression via attenuation of ribosomal biogenesis. Mol Oncol 2024; 18:2136-2156. [PMID: 37853941 PMCID: PMC11467795 DOI: 10.1002/1878-0261.13539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 10/20/2023] Open
Abstract
Endometrial carcinoma (ECa) is the fourth most common cancer among women. The oncogene PELP1 is frequently overexpressed in a variety of cancers, including ECa. We recently generated SMIP34, a small-molecule inhibitor of PELP1 that suppresses PELP1 oncogenic signaling. In this study, we assessed the effectiveness of SMIP34 in treating ECa. Treatment of established and primary patient-derived ECa cells with SMIP34 resulted in a significant reduction of cell viability, colony formation ability, and induction of apoptosis. RNA-seq analyses showed that SMIP34-regulated genes were negatively correlated with ribosome biogenesis and eukaryotic translation pathways. Mechanistic studies showed that the Rix complex, which is essential for ribosomal biogenesis, is disrupted upon SMIP34 binding to PELP1. Biochemical assays confirmed that SMIP34 reduced ribosomal biogenesis and new protein synthesis. Further, SMIP34 enhanced the efficacy of mTOR inhibitors in reducing viability of ECa cells. SMIP34 is also effective in reducing cell viability in ECa organoids in vitro and explants ex vivo. Importantly, SMIP34 treatment resulted in a significant reduction of the growth of ECa xenografts. Collectively, these findings underscore the potential of SMIP34 in treating ECa.
Collapse
Affiliation(s)
- Xue Yang
- Department of Obstetrics and GynecologyUniversity of Texas Health San AntonioTXUSA
- Department of Obstetrics and Gynecology, Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Zexuan Liu
- Department of Obstetrics and GynecologyUniversity of Texas Health San AntonioTXUSA
- Department of Oncology, Xiangya HospitalCentral South UniversityChangshaChina
| | - Weiwei Tang
- Department of Obstetrics and GynecologyUniversity of Texas Health San AntonioTXUSA
- Department of Obstetrics and Gynecology, Affiliated Hospital of Integrated Traditional Chinese and Western MedicineNanjing University of Chinese MedicineChina
| | - Uday P. Pratap
- Department of Obstetrics and GynecologyUniversity of Texas Health San AntonioTXUSA
| | - Alexia B. Collier
- Department of Obstetrics and GynecologyUniversity of Texas Health San AntonioTXUSA
| | - Kristin A. Altwegg
- Department of Obstetrics and GynecologyUniversity of Texas Health San AntonioTXUSA
- Mays Cancer CenterUniversity of Texas Health San AntonioTXUSA
| | - Rahul Gopalam
- Department of Obstetrics and GynecologyUniversity of Texas Health San AntonioTXUSA
| | - Xiaonan Li
- Department of Obstetrics and GynecologyUniversity of Texas Health San AntonioTXUSA
| | - Yaxia Yuan
- Department of Biochemistry & Structural BiologyUniversity of Texas Health San AntonioTXUSA
| | - Daohong Zhou
- Department of Biochemistry & Structural BiologyUniversity of Texas Health San AntonioTXUSA
| | - Zhao Lai
- Department of Molecular Medicine, Department of Population Sciences, and Greehey Children's Cancer Research InstituteUniversity of Texas Health San AntonioSan AntonioTXUSA
| | - Yidong Chen
- Department of Molecular Medicine, Department of Population Sciences, and Greehey Children's Cancer Research InstituteUniversity of Texas Health San AntonioSan AntonioTXUSA
| | - Gangadhara R. Sareddy
- Department of Obstetrics and GynecologyUniversity of Texas Health San AntonioTXUSA
- Mays Cancer CenterUniversity of Texas Health San AntonioTXUSA
| | | | - Edward R. Kost
- Department of Obstetrics and GynecologyUniversity of Texas Health San AntonioTXUSA
| | - Suryavathi Viswanadhapalli
- Department of Obstetrics and GynecologyUniversity of Texas Health San AntonioTXUSA
- Mays Cancer CenterUniversity of Texas Health San AntonioTXUSA
| | - Ratna K. Vadlamudi
- Department of Obstetrics and GynecologyUniversity of Texas Health San AntonioTXUSA
- Mays Cancer CenterUniversity of Texas Health San AntonioTXUSA
- Audie L. Murphy DivisionSouth Texas Veterans Health Care SystemSan AntonioTXUSA
| |
Collapse
|
6
|
Niebora J, Woźniak S, Domagała D, Data K, Farzaneh M, Zehtabi M, Dari MAG, Pour FK, Bryja A, Kulus M, Mozdziak P, Dzięgiel P, Kempisty B. The role of ncRNAs and exosomes in the development and progression of endometrial cancer. Front Oncol 2024; 14:1418005. [PMID: 39188680 PMCID: PMC11345653 DOI: 10.3389/fonc.2024.1418005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/26/2024] [Indexed: 08/28/2024] Open
Abstract
Endometrial cancer (EC) is one of the most common gynecologic cancers. In recent years, research has focused on the genetic characteristics of the tumors to detail their prognosis and tailor therapy. In the case of EC, genetic mutations have been shown to underlie their formation. It is very important to know the mechanisms of EC formation related to mutations induced by estrogen, among other things. Noncoding RNAs (ncRNAs), composed of nucleotide transcripts with very low protein-coding capacity, are proving to be important. Their expression patterns in many malignancies can inhibit tumor formation and progression. They also regulate protein coding at the epigenetic, transcriptional, and posttranscriptional levels. MicroRNAs (miRNAs), several varieties of which are associated with normal endometrium as well as its tumor, also play a particularly important role in gene expression. MiRNAs and long noncoding RNAs (lncRNAs) affect many pathways in EC tissues and play important roles in cancer development, invasion, and metastasis, as well as resistance to anticancer drugs through mechanisms such as suppression of apoptosis and progression of cancer stem cells. It is also worth noting that miRNAs are highly precise, sensitive, and robust, making them potential markers for diagnosing gynecologic cancers and their progression. Unfortunately, as the incidence of EC increases, treatment becomes challenging and is limited to invasive tools. The prospect of using microRNAs as potential candidates for diagnostic and therapeutic use in EC seems promising. Exosomes are extracellular vesicles that are released from many types of cells, including cancer cells. They contain proteins, DNA, and various types of RNA, such as miRNAs. The noncoding RNA components of exosomes vary widely, depending on the physiology of the tumor tissue and the cells from which they originate. Exosomes contain both DNA and RNA and have communication functions between cells. Exosomal miRNAs mediate communication between EC cells, tumor-associated fibroblasts (CAFs), and tumor-associated macrophages (TAMs) and play a key role in tumor cell proliferation and tumor microenvironment formation. Oncogenes carried by tumor exosomes induce malignant transformation of target cells. During the synthesis of exosomes, various factors, such as genetic and proteomic data are upregulated. Thus, they are considered an interesting therapeutic target for the diagnosis and prognosis of endometrial cancer by analyzing biomarkers contained in exosomes. Expression of miRNAs, particularly miR-15a-5p, was elevated in exosomes derived from the plasma of EC patients. This may suggest the important utility of this biomarker in the diagnosis of EC. In recent years, researchers have become interested in the topic of prognostic markers for EC, as there are still too few identified markers to support the limited treatment of endometrial cancer. Further research into the effects of ncRNAs and exosomes on EC may allow for cancer treatment breakthroughs.
Collapse
Affiliation(s)
- Julia Niebora
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Sławomir Woźniak
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Dominika Domagała
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Krzysztof Data
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Clinical Research Development Unit, Imam Khomeini Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mojtaba Zehtabi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahrokh Abouali Gale Dari
- Department of Obstetrics and Gynecology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Khojasteh Pour
- Department of Obstetrics and Gynecology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Artur Bryja
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Magdalena Kulus
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Paul Mozdziak
- Physiology Graduate Program, North Carolina State University, Raleigh, NC, United States
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Bartosz Kempisty
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland
- Physiology Graduate Program, North Carolina State University, Raleigh, NC, United States
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Brno, Czechia
| |
Collapse
|
7
|
Shen L, Zhang C, Cui K, Liang X, Zhu G, Hong L. Fer-mediated activation of the Ras-MAPK signaling pathway drives the proliferation, migration, and invasion of endometrial carcinoma cells. Mol Cell Biochem 2024; 479:1787-1799. [PMID: 38017327 DOI: 10.1007/s11010-023-04890-1] [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: 05/30/2023] [Accepted: 10/31/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND The role of Feline sarcoma-related protein (Fer) in various cancers has been extensively studied, but its specific involvement and underlying mechanisms in the progression of endometrial carcinoma (EC) are yet to be fully understood. METHODS The expression levels of Fer were assessed in EC tissues and cell lines using real-time quantitative PCR and western blot analysis. CCK-8 assay, Edu staining, transwell assays, and flow cytometry, were conducted to evaluate the impact of Fer on EC cells. Furthermore, a mice xenograft model and immunohistochemistry (IHC) staining were utilized for in vivo analysis. The levels of Ras, pMek1/2, and pErk1/2 were determined by western blot assay. Ras-MAPK signaling pathway inhibitor was utilized to study the regulatory role of Fer on EC cells. RESULTS Our findings revealed that Fer exhibited upregulation in both EC tissues and cell lines, concomitant with the activation of the Ras-MAPK signaling pathway. Silencing of Fer resulted in the suppression of cell proliferation, migration, invasion, and Ras-MAPK signaling pathway, while promoted hypoxia-induced apoptosis in RL95-2 and KLE cells. Fer overexpression stimulated cell proliferation, migration, invasion, and Ras-MAPK signaling pathway in Ishikawa and AN3-CA cells, which were reversed after treatment with either Ras or MAPK inhibitor. Moreover, silencing of Fer suppressed tumor growth and downregulated the expression of Ki-67, Ras, pMek1/2, and pErk1/2, but had no significant effect on Mek1/2 and Erk1/2, while upregulated caspase-3 expression in vivo. CONCLUSION In summary, the upregulation of Fer in EC cells resulted in the enhancement of cell proliferation, migration, and invasion through the activation of the Ras-MAPK signaling pathway.
Collapse
Affiliation(s)
- Lifan Shen
- Department of Gynecology, Surgery Building, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), 19Th Xiuhua Road, Xiuying District, Haikou, 570000, China
| | - Chen Zhang
- Department of Central Lab, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Kaiying Cui
- Department of Gynecology, Surgery Building, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), 19Th Xiuhua Road, Xiuying District, Haikou, 570000, China
| | - Xin Liang
- Department of Gynecology, Surgery Building, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), 19Th Xiuhua Road, Xiuying District, Haikou, 570000, China
| | - Genhai Zhu
- Department of Gynecology, Surgery Building, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), 19Th Xiuhua Road, Xiuying District, Haikou, 570000, China
| | - Lan Hong
- Department of Gynecology, Surgery Building, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), 19Th Xiuhua Road, Xiuying District, Haikou, 570000, China.
| |
Collapse
|
8
|
Solanki R, Zubbair Malik M, Alankar B, Ahmad FJ, Dohare R, Chauhan R, Kesharwani P, Kaur H. Identification of novel biomarkers and potential molecular targets for uterine cancer using network-based approach. Pathol Res Pract 2024; 260:155431. [PMID: 39029376 DOI: 10.1016/j.prp.2024.155431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 07/21/2024]
Abstract
A better understanding of incidences at the cellular level in uterine cancer is necessary for its effective treatment and favourable prognosis. Till date, it lacks appropriate molecular target-based treatment because of unknown molecular mechanisms that proceed to cancer and no drug has shown the required results of treatment with less severe side effects. Uterine Cancer is one of the top five cancer diagnoses and among the ten most common death-causing cancer in the United States of America. There is no FDA-approved drug for it yet. Therefore, it became necessary to identify the molecular targets for molecular targeted therapy of this widely prevalent cancer type. For this study, we used a network-based approach to the list of the deregulated (both up and down-regulated) genes taking adjacent p-Value ≤ 0.05 as significance cut off for the mRNA data of uterine cancer. We constructed the protein-protein interaction (PPI) network and analyzed the degree, closeness, and betweenness centrality-like topological properties of the PPI network. Then we traced the top 30 genes listed from each topological property to find the key regulators involved in the endometrial cancer (ECa) network. We then detected the communities and sub-communities from the PPI network using the Cytoscape network analyzer and Louvain modularity optimization method. A set of 26 (TOP2A, CENPE, RAD51, BUB1, BUB1B, KIF2C, KIF23, KIF11, KIF20A, ASPM, AURKA, AURKB, PLK1, CDC20, CDKN2A, EZH2, CCNA2, CCNB1, CDK1, FGF2, PRKCA, PGR, CAMK2A, HPGDS, and CDCA8) genes were found to be key genes of ECa regulatory network altered in disease state and might be playing the regulatory role in complex ECa network. Our study suggests that among these genes, KIF11 and H PGDS appeared to be novel key genes identified in our research. We also identified these key genes interactions with miRNAs.
Collapse
Affiliation(s)
- Rubi Solanki
- School of Interdisciplinary Sciences and Technology, Jamia Hamdard, New Delhi 110062, India
| | - Md Zubbair Malik
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute Dasman 15462, Kuwait
| | - Bhavya Alankar
- Department of Computer Science and Engineering, School of Engineering Sciences and Technology, Jamia Hamdard, New Delhi 110062, India.
| | - Farhan Jalees Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ravins Dohare
- Centre for Interdisciplinary Research in Basic Sciences, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Ritu Chauhan
- Artificial Intelligence and IoT lab, Centre for Computational Biology and Bioinformatics, Amity University, Noida, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Harleen Kaur
- Department of Computer Science and Engineering, School of Engineering Sciences and Technology, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
9
|
Kelly SE, Wang X, Hsieh SC, Abdul-Wahid A, Derry M, Skidmore B, Wells GA. Additive toxicity arising from combined use of immune checkpoint inhibitors and tyrosine kinase inhibitors in patients with renal or endometrial carcinoma: Protocol for a rapid systematic review. MethodsX 2024; 12:102730. [PMID: 38779442 PMCID: PMC11109459 DOI: 10.1016/j.mex.2024.102730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/21/2024] [Indexed: 05/25/2024] Open
Abstract
The combined use of immune checkpoint inhibitors and tyrosine kinase inhibitors (ICI/TKI) is an effective treatment strategy for some cancers. A better understanding of the potential additive toxicity for ICI/TKI combinations is needed to inform patient and provider treatment decisions. We aim to evaluate the safety of ICI/TKI combinations for individuals with renal cell or endometrial carcinoma. This rapid systematic review (SR) protocol follows PRISMA guidelines. A systematic search will be designed, peer reviewed and executed by experienced information specialists (Cochrane Central, MEDLINE, Embase) to identify published SRs and primary studies published since the most recent SR search. Randomized, quasi- or non-randomized controlled trials and comparative cohort studies are eligible if they compare ICI/TKI combinations to monotherapy or standard of care in participants with renal cell or endometrial carcinoma. The primary outcome is grade ≥ 3 treatment-related adverse-effects. Studies will be screened, selected, extracted and assessed for risk of bias by a single reviewer and checked completely by a second. Where feasible and appropriate, we will pool studies separately by design and indication using meta-analysis and test robustness of effects using prespecified subgroup and sensitivity analyses. Results will be summarized descriptively and presented in tables and figures. (PROSPERO ID: CRD42023416388).•This will be a comprehensive systematic review of the additive toxicity arising from the combined use of ICI/TKIs in patients with renal-cell or endometrial carcinoma.•We will consider treatment-related, treatment-emergent adverse events (Grade 3 or higher).•Identified safety profile may be used to inform patient or provider treatment decisions.
Collapse
Affiliation(s)
- Shannon E. Kelly
- Cardiovascular Research Methods Centre, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Xiaoqin Wang
- Cardiovascular Research Methods Centre, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Shu-Ching Hsieh
- Cardiovascular Research Methods Centre, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Aws Abdul-Wahid
- Bureau of Biologics, Radiopharmaceuticals and Self-Care Products, Marketed Health Products Directorate, Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada
| | - Melanie Derry
- Bureau of Biologics, Radiopharmaceuticals and Self-Care Products, Marketed Health Products Directorate, Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada
| | - Becky Skidmore
- Independent Information Scientist, Ottawa, Ontario, Canada
| | - George A. Wells
- Cardiovascular Research Methods Centre, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
10
|
Soberanis Pina P, Lheureux S. Novel Molecular Targets in Endometrial Cancer: Mechanisms and Perspectives for Therapy. Biologics 2024; 18:79-93. [PMID: 38529411 PMCID: PMC10962462 DOI: 10.2147/btt.s369783] [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: 12/01/2023] [Accepted: 02/22/2024] [Indexed: 03/27/2024]
Abstract
Endometrial cancer (EC) has a high epidemiological impact with incidence and mortality rising worldwide. In recent years, the integration of the pathologic and molecular classification has provided relevant information to understand the heterogeneity in the biology of EC, which led to the evolution in the management of patients. Currently, therapeutic breakthroughs have been made in advanced EC to improve oncologic outcomes, with efforts to include patient reported outcomes. Precision and personalized medicine are under way in EC exploring different combination approaches to target cross-talk pathways, cancer cell microenvironment, and metabolic vulnerabilities and improve drug delivery. Yet, collaborative efforts are needed to face the challenges in practice by refining patient selection, ideal biomarker identification, and de-escalation of therapies according to emerging molecular and genomic features of EC.
Collapse
Affiliation(s)
- Pamela Soberanis Pina
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Stephanie Lheureux
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
11
|
Wang Y, Chen S, Wang C, Guo F. Nanocarrier-based targeting of metabolic pathways for endometrial cancer: Status and future perspectives. Biomed Pharmacother 2023; 166:115348. [PMID: 37639743 DOI: 10.1016/j.biopha.2023.115348] [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: 06/28/2023] [Revised: 08/10/2023] [Accepted: 08/19/2023] [Indexed: 08/31/2023] Open
Abstract
Cancer is the second-most lethal global disease, as per health reports, and is responsible for around 70% of deaths in low- and middle-income countries. Endometrial cancer is one of the emerging malignancies and has been predicted as a public health challenge for the future. Insulin resistance, obesity, and diabetes mellitus are the key metabolic factors that promote risks for the development of endometrial cancer. Various signaling pathways and associated genes are involved in the genesis of endometrial cancer, and any mutation or deletion in such related factors leads to the induction of endometrial cancer. The conventional way of drug delivery has been used for ages but is associated with poor management of cancer due to non-targeting of the endometrial cancer cells, low efficacy of the therapy, and toxicity issues as well. In this context, nanocarrier-based therapy for the management of endometrial cancer is an effective alternate choice that overcomes the problems associated with conventional therapy. In this review article, we highlighted the nanocarrier-based targeting of endometrial cancer, with a special focus on targeting various metabolic signaling pathways. Furthermore, the future perspectives of nanocarrier-based targeting of metabolic pathways in endometrial cancer were also underpinned. It is concluded that targeting metabolic signaling pathways in endometrial cancer via nanocarrier scaffolds is the future of pharmaceutical design for the significant management and treatment of endometrial cancer.
Collapse
Affiliation(s)
- Yichao Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130000, China
| | - Siyao Chen
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130000, China
| | - Chunling Wang
- Medical Affairs Department, The Second Hospital of Jilin University, Changchun 130000, China
| | - Fengjun Guo
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
12
|
Chen J, Dai S, Zhao L, Peng Y, Sun C, Peng H, Zhong Q, Quan Y, Li Y, Chen X, Pan X, Zhong A, Wang M, Zhang M, Yang S, Lu Y, Lian Z, Liu Y, Zhou S, Li Z, Na F, Chen C. A New Type of Endometrial Cancer Models in Mice Revealing the Functional Roles of Genetic Drivers and Exploring their Susceptibilities. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300383. [PMID: 37340596 PMCID: PMC10460855 DOI: 10.1002/advs.202300383] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/12/2023] [Indexed: 06/22/2023]
Abstract
Endometrial cancer (EC) is the most common female reproductive tract cancer and its incidence has been continuously increasing in recent years. The underlying mechanisms of EC tumorigenesis remain unclear, and efficient target therapies are lacking, for both of which feasible endometrial cancer animal models are essential but currently limited. Here, an organoid and genome editing-based strategy to generate primary, orthotopic, and driver-defined ECs in mice is reported. These models faithfully recapitulate the molecular and pathohistological characteristics of human diseases. The authors names these models and similar models for other cancers as organoid-initiated precision cancer models (OPCMs). Importantly, this approach can conveniently introduce any driver mutation or a combination of driver mutations. Using these models,it is shown that the mutations in Pik3ca and Pik3r1 cooperate with Pten loss to promote endometrial adenocarcinoma in mice. In contrast, the Kras G12D mutati led to endometrial squamous cell carcinoma. Then, tumor organoids are derived from these mouse EC models and performed high-throughput drug screening and validation. The results reveal distinct vulnerabilities of ECs with different mutations. Taken together, this study develops a multiplexing approach to model EC in mice and demonstrates its value for understanding the pathology of and exploring the potential treatments for this malignancy.
Collapse
Affiliation(s)
- Jingyao Chen
- Precision Medicine Research CenterState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Siqi Dai
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Lei Zhao
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Yiman Peng
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Chongen Sun
- West China Second HospitalSichuan UniversityChengdu610041China
| | - Hongling Peng
- West China Second HospitalSichuan UniversityChengdu610041China
| | - Qian Zhong
- West China Second HospitalSichuan UniversityChengdu610041China
| | - Yuan Quan
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Yue Li
- Department of DermatologyState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengdu610041China
| | - Xuelan Chen
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Xiangyu Pan
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Ailing Zhong
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Manli Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Mengsha Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Shengyong Yang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - You Lu
- Division of Thoracic Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengdu610041China
- Laboratory of Clinical Cell Therapy, West China HospitalSichuan UniversityChengdu610041China
| | - Zhong Lian
- Department of DermatologyState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengdu610041China
| | - Yu Liu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Shengtao Zhou
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
- West China Second HospitalSichuan UniversityChengdu610041China
| | - Zhengyu Li
- West China Second HospitalSichuan UniversityChengdu610041China
| | - Feifei Na
- Division of Thoracic Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Chong Chen
- Precision Medicine Research CenterState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| |
Collapse
|
13
|
Rafique A, Hichiwa G, Jatnika MF, Ito Y. A Novel Strategy for Screening Tumor-Specific Variable Domain of Heavy-Chain Antibodies. Int J Mol Sci 2023; 24:10804. [PMID: 37445977 DOI: 10.3390/ijms241310804] [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: 05/15/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
The properties of the variable domain of heavy-chain (VHH) antibodies are particularly relevant in cancer therapy. To isolate tumor cell-specific VHH antibodies, VHH phage libraries were constructed from multiple tumor cells. After enriching the libraries against particular tumor cell lines, a next-generation sequencer was used to screen the pooled phages of each library for potential antibody candidates. Based on high amplification folds, 50 sequences from each library were used to construct phylogenetic trees. Several clusters with identical CDR3 were observed. Groups X, Y, and Z were assigned as common sequences among the different trees. These identical groups over the trees were considered to be cross-reactive antibodies. To obtain monoclonal antibodies, we assembled 200 sequences (top 50 sequences from each library) and rebuilt a combined molecular phylogenetic tree. Groups were categorized as A-G. For each group, we constructed a phagemid and determined its binding specificity with tumor cells. The phage-binding results were consistent with the phylogenetic tree-generated groups, which indicated particular tumor-specific clusters; identical groups showed cross-reactivity. The strategy used in the current study is effective for screening and isolating monoclonal antibodies. Specific antibodies can be identified, even when the target markers of cancer cells are unknown.
Collapse
Affiliation(s)
- Abdur Rafique
- Graduate School of Science and Engineering, University of Kagoshima, Kagoshima 890-0065, Japan
| | - Genki Hichiwa
- Graduate School of Medical Sciences, Tottori University, Tottori 680-8550, Japan
| | - Muhammad Feisal Jatnika
- Graduate School of Science and Engineering, University of Kagoshima, Kagoshima 890-0065, Japan
| | - Yuji Ito
- Graduate School of Science and Engineering, University of Kagoshima, Kagoshima 890-0065, Japan
| |
Collapse
|
14
|
Tang TT, Jiang L, Zhong Q, Ni ZJ, Thakur K, Khan MR, Wei ZJ. Saikosaponin D exerts cytotoxicity on human endometrial cancer ishikawa cells by inducing apoptosis and inhibiting metastasis through MAPK pathways. Food Chem Toxicol 2023; 177:113815. [PMID: 37209937 DOI: 10.1016/j.fct.2023.113815] [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: 04/11/2023] [Revised: 05/01/2023] [Accepted: 05/07/2023] [Indexed: 05/22/2023]
Abstract
Saikosaponin D (SSD) is one of plant secondary metabolic active substance with effective anti-tumor ability; however, the toxicity of Saikosaponin D on human endometrial cancer Ishikawa cells is still unclear. Our results revealed that SSD displayed cytotoxicity on the Ishikawa cell with an IC50 = 15.69 μM, but was non-toxic to the human normal cell line HEK293. SSD could upregulate p21 and Cyclin B to keep cells in the G2/M stage. In addition, it activated the death receptor and mitochondrion routes to induce apoptosis in Ishikawa cells. The transwell chamber and wound healing results showed that SSD inhibited the cell migration and invasion. In addition, we found that it was closely related to the MAPK cascade pathway, and it could mediate the three classical MAPK pathways to block cell metastasis. In conclusion, SSD could be potentially beneficial as a natural secondary metabolite in preventing and treating endometrial carcinoma.
Collapse
Affiliation(s)
- Tong-Tong Tang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, People's Republic of China.
| | - Li Jiang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, People's Republic of China.
| | - Qian Zhong
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, People's Republic of China; School of Biological Science and Engineering, Ningxia Key Laboratory for the Development and Application of Microbial Resources in Extreme Environments, North Minzu University, Yinchuan, 750021, People's Republic of China.
| | - Zhi-Jing Ni
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, People's Republic of China; School of Biological Science and Engineering, Ningxia Key Laboratory for the Development and Application of Microbial Resources in Extreme Environments, North Minzu University, Yinchuan, 750021, People's Republic of China.
| | - Kiran Thakur
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, People's Republic of China; School of Biological Science and Engineering, Ningxia Key Laboratory for the Development and Application of Microbial Resources in Extreme Environments, North Minzu University, Yinchuan, 750021, People's Republic of China.
| | - Mohammad Rizwan Khan
- Department of Chemistry, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia.
| | - Zhao-Jun Wei
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, People's Republic of China; School of Biological Science and Engineering, Ningxia Key Laboratory for the Development and Application of Microbial Resources in Extreme Environments, North Minzu University, Yinchuan, 750021, People's Republic of China.
| |
Collapse
|
15
|
LncRNA BBOX1-AS1 Contributes to the Progression of Esophageal Carcinoma by Targeting the miR-361-3p/COL5A1 Axis. Biochem Genet 2022:10.1007/s10528-022-10307-3. [PMID: 36586008 DOI: 10.1007/s10528-022-10307-3] [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: 08/16/2022] [Accepted: 11/22/2022] [Indexed: 01/01/2023]
Abstract
Long noncoding RNAs (lncRNAs) are known to participate in the progression of several cancers, including esophageal carcinoma (EC), a common malignancy of the digestive system. Although the role of the lncRNA-miRNA-mRNA regulatory network is crucial for the growth and progression of EC, the regulation of lncRNA BBOX1-AS1 (BBOX1 antisense RNA1) remains unclear. We performed reverse transcription-quantitative PCR (RT-qPCR) and western blotting to evaluate miR-361-3p, collagen type V alpha 1 chain (COL5A1), and BBOX1-AS1 expression levels in EC cells and tissues. The colony formation assay (CFA) and Cell Counting Kit-8 (CCK-8) were employed to identify EC cell proliferation, while western blotting was used to examine EC cell apoptosis and Bax and Bcl-2 expression levels. The effect of BBOX1-AS1 on EC proliferation was determined using an in vivo carcinogenesis assay. Correlation between COL5A1, BBOX1-AS1, and miR-361-3p was examined using the luciferase reporter system and RNA immunoprecipitation assay (RIP). Herein, we observed that BBOX1-AS1 expression levels were upregulated in EC cells and tissues. BBOX1-AS1 knockdown inhibited EC cell proliferation and conferred a pro-apoptotic effect. These results indicated a positive interaction between BBOX1-AS1 and miR-361-3p in EC and a negative association with miR-361-3p. COL5A1 was recognized as a downstream miR-361-3p target and was inversely related to miR-361-3p in EC. Therefore, BBOX1-AS1 expression suppressed cell apoptosis and promoted cell proliferation via the downregulation of miR-361-3p and upregulation of COL5A1 expression. Overall, BBOX1-AS1 facilitates EC progression via the miR-361-3p or COL5A1 axis, indicating that BBOX1-AS1 might be a potential therapeutic target for EC therapy.
Collapse
|
16
|
XPO1-Mediated EIF1AX Cytoplasmic Relocation Promotes Tumor Migration and Invasion in Endometrial Carcinoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1361135. [PMID: 36589683 PMCID: PMC9800903 DOI: 10.1155/2022/1361135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/30/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022]
Abstract
Dysregulation of eukaryotic translation initiation factor 1A, X-linked (EIF1AX), has been implicated in the pathogenesis of some cancers. However, the role of EIF1AX in endometrial carcinoma (EC) remains unknown. We investigated the EIF1AX expression in EC patients and assessed its tumorigenesis-associated function and nucleocytoplasmic transport mechanism in vitro and in vivo. The results indicated that the cytoplasmic EIF1AX expression showed a gradual increase when going from endometrium normal tissue, simple endometrial hyperplasia, complex endometrial hyperplasia, and endometrial atypical hyperplasia to EC, while vice versa for the nuclear EIF1AX expression. In addition, the cytoplasmic EIF1AX expression was positively correlated with histologic type, high International Federation of Gynecology and Obstetrics (FIGO) grade, advanced FIGO stage, deeper infiltration, high Ki67 index, and shorter recurrence-free survival in EC patients. In vitro, short hairpin RNA-mediated EIF1AX depletion or SV40NLS-mediated EIF1AX import into the nucleus in multiple human EC cells potently suppressed cell migration and invasion, epithelial-mesenchymal transition, and lung metastasis. Moreover, exportin 1 induced the transport of EIF1AX from the nucleus to the cytoplasm that could be inhibited by leptomycin B treatment or the mutation in the EIF1AX location sequence. These results demonstrate that cytoplasmic EIF1AX may play a key role in the incidence and promotion of EC, and thus, targeting EIF1AX or its nucleocytoplasmic transport process may offer an effective new therapeutic approach to EC.
Collapse
|
17
|
MIG-6 Is Critical for Progesterone Responsiveness in Human Complex Atypical Hyperplasia and Early-Stage Endometrial Cancer. Int J Mol Sci 2022; 23:ijms232314596. [PMID: 36498921 PMCID: PMC9738720 DOI: 10.3390/ijms232314596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/14/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Women with complex atypical hyperplasia (CAH) or early-stage endometrioid endometrial cancer (EEC) are candidates for fertility preservation. The most common approach is progesterone (P4) therapy and deferral of hysterectomy until after completion of childbearing. However, P4 therapy response rates vary, and molecular mechanisms behind P4 resistance are poorly understood. One potential molecular cause of P4 resistance is a loss or attenuation of PGR expression. Mitogen-inducible gene 6 (MIG-6) is critical for P4 responsiveness. MIG-6 protein expression in the endometrial epithelial and stromal cells from women with CAH and EEC was significantly lower compared to women without CAH or EEC. The P4-responsive women (10/15) exhibited an increase of MIG-6 expression in epithelial and stromal cells compared to P4-resistant women (5/15). In addition, immunohistochemical analysis for PGR results showed that stromal PGR levels are significantly higher in P4-responsive women compared to P4-resistant women, whereas epithelial PGR expression was not different. A reverse correlation of MIG-6 and pAKT levels was observed in early-stage EEC patients. Studies strongly suggest that loss of MIG-6 and PGR and activation of pAKT lead to P4 resistance in CAH and EEC. These results will help to elucidate the molecular mechanism leading to P4 resistance in CAH and EEC.
Collapse
|
18
|
Luo F, Yang Q. Clinical Application of Digital 3D Reconstruction and 3D Printing Technology in Endometrial Cancer (EC) Surgery. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:9180216. [PMID: 36158121 PMCID: PMC9492336 DOI: 10.1155/2022/9180216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/03/2022] [Accepted: 08/27/2022] [Indexed: 11/17/2022]
Abstract
Aims We use CTA and magnetic resonance data to use digital three-dimensional reconstruction and 3D printing technology to reproduce the solid replication of the uterus and surrounding tissues in vitro, fully evaluate the adjacency of tumor tissues with surrounding important organs, blood vessels, and lymph nodes, and reduce the impact. The normal organ structure and function of the surgeon can shorten the operation time, reduce the bleeding during the operation, and reduce the perioperative complications of the patient to improve the prognosis of the patient. Materials and Methods Select 40 EC patients and divide them into group A (3D reconstruction data is transmitted to 3D printing equipment according to the results of CTA and MRI examination, and a 3D model is printed out according to the ratio of 1 : 1 for evaluation and judgment before surgery) and group B (according to MRI imaging examination, there were 20 cases each). Different surgical conditions, quality of life, adverse reactions, and clinical efficacy were evaluated in each group. Results The operation time, the time of the first anus exhaust, the hospitalization time after the operation, and the blood loss of the operation in group A were significantly lower than those in group B. Statistics showed that the difference was significant (P < 0.05). The quality of life scores of emotion, cognition, society, and overall health of group A were significantly higher than those of group B, while physical score, fatigue, nausea, vomiting, and pain were lower than those of group B, which were statistically significant (P < 0.05). Both groups of patients had complications after the operation, and they were asked to be followed up at the outpatient clinic 3 months after the operation. All patients recovered well. There were 19 and 18 patients in groups A and B, respectively, complaining of improvement in clinical symptoms, and the difference was not statistically significant (P < 0.05). Conclusion With the support of digital three-dimensional reconstruction and 3D printing technology, complex operations can be accurately performed, improving the efficacy and safety of patients after EC surgery, improving patient outcomes and quality of life, improving EC positioning accuracy, and reducing tumor residue.
Collapse
Affiliation(s)
- Fang Luo
- Department of Gynecology, Wuhan Puren Hospital, 430081, China
| | - Qin Yang
- Department of Gynecology, Wuhan Puren Hospital, 430081, China
| |
Collapse
|
19
|
Su P, Yu L, Mao X, Sun P. Role of HIF-1α/ERRα in Enhancing Cancer Cell Metabolism and Promoting Resistance of Endometrial Cancer Cells to Pyroptosis. Front Oncol 2022; 12:881252. [PMID: 35800058 PMCID: PMC9253301 DOI: 10.3389/fonc.2022.881252] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/25/2022] [Indexed: 12/24/2022] Open
Abstract
Oxygen is critical to energy metabolism, and tumors are often characterized by a hypoxic microenvironment. Owing to the high metabolic energy demand of malignant tumor cells, their survival is promoted by metabolic reprogramming in the hypoxic microenvironment, which can confer tumor cell resistance to pyroptosis. Pyroptosis resistance can inhibit anti-tumor immunity and promote the development of malignant tumors. Hypoxia inducible factor-1α (HIF-1α) is a key regulator of metabolic reprogramming in tumor cells, and estrogen-related receptor α (ERRα) plays a key role in regulating cellular energy metabolism. Therefore, the close interaction between HIF-1α and ERRα influences the metabolic and functional changes in cancer cells. In this review, we summarize the reprogramming of tumor metabolism involving HIF-1α/ERRα. We review our understanding of the role of HIF-1α/ERRα in promoting tumor growth adaptation and pyroptosis resistance, emphasize its key role in energy homeostasis, and explore the regulation of HIF-1α/ERRα in preventing and/or treating endometrial carcinoma patients. This review provides a new perspective for the study of the molecular mechanisms of metabolic changes in tumor progression.
Collapse
Affiliation(s)
- Pingping Su
- Laboratory of Gynecological Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Lirui Yu
- Laboratory of Gynecological Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiaodan Mao
- Laboratory of Gynecological Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Women and Children’s Critical Diseases Research, Fuzhou, China
| | - Pengming Sun
- Laboratory of Gynecological Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Women and Children’s Critical Diseases Research, Fuzhou, China
- *Correspondence: Pengming Sun,
| |
Collapse
|
20
|
Szabo Z, Juhasz E, Schally AV, Dezso B, Huga S, Hernadi Z, Halmos G, Kiss C. Expression of Growth Hormone-Releasing Hormone and Its Receptor Splice Variants in Primary Human Endometrial Carcinomas: Novel Therapeutic Approaches. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27092671. [PMID: 35566020 PMCID: PMC9101386 DOI: 10.3390/molecules27092671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 11/16/2022]
Abstract
Antagonists of growth hormone-releasing hormone (GHRH) inhibit the growth of various tumors, including endometrial carcinomas (EC). However, tumoral receptors that mediate the antiproliferative effects of GHRH antagonists in human ECs have not been fully characterized. In this study, we investigated the expression of mRNA for GHRH and splice variants (SVs) of GHRH receptors (GHRH-R) in 39 human ECs and in 7 normal endometrial tissue samples using RT-PCR. Primers designed for the PCR amplification of mRNA for the full length GHRH-R and SVs were utilized. The PCR products were sequenced, and their specificity was confirmed. Nine ECs cancers (23%) expressed mRNA for SV1, three (7.7%) showed SV2 and eight (20.5%) revealed mRNA for SV4. The presence of SVs for GHRH-Rs could not be detected in any of the normal endometrial tissue specimens. The presence of specific, high affinity GHRH-Rs was also demonstrated in EC specimens using radioligand binding studies. Twenty-four of the investigated thirty-nine tumor samples (61.5%) and three of the seven corresponding normal endometrial tissues (42.9%) expressed mRNA for GHRH ligand. Our findings suggest the possible existence of an autocrine loop in EC based on GHRH and its tumoral SV receptors. The antiproliferative effects of GHRH antagonists on EC are likely to be exerted in part by the local SVs and GHRH system.
Collapse
Affiliation(s)
- Zsuzsanna Szabo
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (G.H.)
| | - Eva Juhasz
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Andrew V. Schally
- Veterans Affairs Medical Center, Endocrine, Polypeptide and Cancer Institute, Miami, FL 33125, USA;
- Department of Pathology, Department of Medicine, Divisions of Hematology-Oncology and Endocrinology, Miller School of Medicine, University of Miami, Miami, FL 33101, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Balazs Dezso
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Sandor Huga
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (S.H.); (Z.H.)
| | - Zoltan Hernadi
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (S.H.); (Z.H.)
| | - Gabor Halmos
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (G.H.)
- Veterans Affairs Medical Center, Endocrine, Polypeptide and Cancer Institute, Miami, FL 33125, USA;
| | - Csongor Kiss
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
- Correspondence: ; Tel.: +36-52-452-747; Fax: +36-52-255-893
| |
Collapse
|
21
|
Venkata PP, Chen Y, Alejo S, He Y, Palacios BE, Loeffel I, Liu J, Pratap UP, Gray G, Achuthan Pillai SM, Zou Y, Lai Z, Suzuki T, Viswanadhapalli S, Palakurthi S, Tekmal RR, Vadlamudi RK, Kost E, Sareddy GR. KDM1A inhibition augments the efficacy of rapamycin for the treatment of endometrial cancer. Cancer Lett 2022; 524:219-231. [PMID: 34673129 PMCID: PMC10000284 DOI: 10.1016/j.canlet.2021.10.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/30/2021] [Accepted: 10/13/2021] [Indexed: 11/21/2022]
Abstract
Endometrial cancer (EC) often exhibit aberrant activation of PI3K/Akt/mTOR signaling and targeted therapies using mTOR inhibitors showed limited success. The epigenetic modifier, lysine-specific histone demethylase-1A (KDM1A/LSD1) is overexpressed in EC, however, the mechanistic and therapeutic implications of KDM1A in EC are poorly understood. Here, using 119 FDA-approved drugs screen, we identified that KDM1A inhibition is highly synergistic with mTOR inhibitors. Combination therapy of KDM1A and mTOR inhibitors potently reduced the cell viability, survival, and migration of EC cells. Mechanistic studies demonstrated that KDM1A inhibition attenuated the activation of mTOR signaling cascade and abolished rapamycin induced feedback activation of Akt. RNA-seq analysis identified that KDM1A inhibition downregulated the expression of genes involved in rapamycin induced activation of Akt, including the mTORC2 complex. Chromatin immunoprecipitation experiments confirmed KDM1A recruitment to the promoter regions of mTORC2 complex genes and that KDM1A inhibition promoted enrichment of repressive H3K9me2 marks at their promoters. Combination therapy of KDM1A inhibitor and rapamycin reduced the tumor growth in EC xenograft and patient derived xenograft models in vivo and patient derived tumor explants ex vivo. Importantly, in silico analysis of TCGA EC patients data sets revealed that KDM1A expression positively correlated with the levels of PI3K/Akt/mTOR genes. Collectively, our results provide compelling evidence that KDM1A inhibition potentiates the activity of mTOR inhibitors by attenuating the feedback activation of Akt survival signaling. Furthermore, the use of concurrent KDM1A and mTOR inhibitors may be an attractive targeted therapy for EC patients.
Collapse
Affiliation(s)
- Prabhakar Pitta Venkata
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Yihong Chen
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA; Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China
| | - Salvador Alejo
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Yi He
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA; Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China
| | - Bridgitte E Palacios
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Ilanna Loeffel
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Junhao Liu
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA; Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China
| | - Uday P Pratap
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Gabrielle Gray
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | | | - Yi Zou
- Greehey Children's Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Zhao Lai
- Greehey Children's Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX, 78229, USA; Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Takayoshi Suzuki
- The Institute of Scientific and Industrial Research, Osaka University, Japan
| | - Suryavathi Viswanadhapalli
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Srinath Palakurthi
- Department of Pharmaceutical Sciences, Texas A&M University, Kingsville, TX, 78363, USA
| | - Rajeshwar R Tekmal
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA; Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA; Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX, 78229, USA; Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| | - Edward Kost
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Gangadhara R Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA; Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
22
|
Español P, Luna R, Soler C, Caruana P, Altés-Arranz A, Rodríguez F, Porta O, Sanchez O, Llurba E, Rovira R, Céspedes MV. Neural plasticity of the uterus: New targets for endometrial cancer? WOMEN'S HEALTH (LONDON, ENGLAND) 2022; 18:17455057221095537. [PMID: 35465787 PMCID: PMC9047769 DOI: 10.1177/17455057221095537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Endometrial carcinoma is the most common gynecological malignancy in Western countries and is expected to increase in the following years because of the high index of obesity in the population. Recently, neural signaling has been recognized as part of the tumor microenvironment, playing an active role in tumor progression and invasion of different solid tumor types. The uterus stands out for the physiological plasticity of its peripheral nerves due to cyclic remodeling brought on by estrogen and progesterone hormones throughout the reproductive cycle. Therefore, a precise understanding of nerve-cancer crosstalk and the contribution of the organ-intrinsic neuroplasticity, mediated by estrogen and progesterone, of the uterine is urgently needed. The development of new and innovative medicines for patients with endometrial cancer would increase their quality of life and health. This review compiles information on the architecture and function of autonomous uterine neural innervations and the influence of hormone-dependent nerves in normal uterus and tumor progression. It also explores new therapeutic possibilities for endometrial cancer using these endocrine and neural advantages.
Collapse
Affiliation(s)
- Pia Español
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Rocio Luna
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Cristina Soler
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Pablo Caruana
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Amanda Altés-Arranz
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Francisco Rodríguez
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Oriol Porta
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Olga Sanchez
- Women and Perinatal Health Research Group, Obstetrics and Gynaecology Department, Hospital Sant Pau and Universitat Autònoma de Barcelona, Barcelona, Spain.,Maternal and Child Health and Development Network, Instituto Salud Carlos III, Madrid, Spain
| | - Elisa Llurba
- Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Women and Perinatal Health Research Group, Obstetrics and Gynaecology Department, Hospital Sant Pau and Universitat Autònoma de Barcelona, Barcelona, Spain.,Maternal and Child Health and Development Network, Instituto Salud Carlos III, Madrid, Spain
| | - Ramón Rovira
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - María Virtudes Céspedes
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| |
Collapse
|
23
|
Shimada H, Kohno T, Konno T, Okada T, Saito K, Shindo Y, Kikuchi S, Tsujiwaki M, Ogawa M, Matsuura M, Saito T, Kojima T. The Roles of Tricellular Tight Junction Protein Angulin-1/Lipolysis-Stimulated Lipoprotein Receptor (LSR) in Endometriosis and Endometrioid-Endometrial Carcinoma. Cancers (Basel) 2021; 13:6341. [PMID: 34944960 PMCID: PMC8699113 DOI: 10.3390/cancers13246341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/04/2021] [Accepted: 12/05/2021] [Indexed: 12/11/2022] Open
Abstract
Tight junction proteins play roles beyond permeability barriers functions and control cell proliferation and differentiation. The relation between tight junctions and the signal transduction pathways affects cell growth, invasion and migration. Abnormality of tight junction proteins closely contributes to epithelial mesenchymal transition (EMT) and malignancy of various cancers. Angulin-1/lipolysis-stimulated lipoprotein receptor (LSR) forms tricellular contacts that has a barrier function. Downregulation of angulin-1/LSR correlates with the malignancy in various cancers, including endometrioid-endometrial carcinoma (EEC). These alterations have been shown to link to not only multiple signaling pathways such as Hippo/YAP, HDAC, AMPK, but also cell metabolism in ECC cell line Sawano. Moreover, loss of angulin-1/LSR upregulates claudin-1, and loss of apoptosis stimulating p53 protein 2 (ASPP2) downregulates angulin-1/LSR. Angulin-1/LSR and ASPP2 concentrate at both midbody and centrosome in cytokinesis. In EEC tissues, angulin-1/LSR and ASPP2 are reduced and claudin-2 is overexpressed during malignancy, while in the tissues of endometriosis changes in localization of angulin-1/LSR and claudin-2 are seen. This review highlights how downregulation of angulin-1/LSR promotes development of endometriosis and EEC and discusses about the roles of angulin-1/LSR and its related proteins, including claudins and ASPP2.
Collapse
Affiliation(s)
- Hiroshi Shimada
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (H.S.); (T.K.); (T.O.); (K.S.); (Y.S.)
- Departments of Obstetrics, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (M.O.); (M.M.); (T.S.)
| | - Takayuki Kohno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (H.S.); (T.K.); (T.O.); (K.S.); (Y.S.)
| | - Takumi Konno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (H.S.); (T.K.); (T.O.); (K.S.); (Y.S.)
| | - Tadahi Okada
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (H.S.); (T.K.); (T.O.); (K.S.); (Y.S.)
- Departments of Obstetrics, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (M.O.); (M.M.); (T.S.)
| | - Kimihito Saito
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (H.S.); (T.K.); (T.O.); (K.S.); (Y.S.)
- Departments of Obstetrics, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (M.O.); (M.M.); (T.S.)
| | - Yuma Shindo
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (H.S.); (T.K.); (T.O.); (K.S.); (Y.S.)
| | - Shin Kikuchi
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan;
| | - Mitsuhiro Tsujiwaki
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan;
| | - Marie Ogawa
- Departments of Obstetrics, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (M.O.); (M.M.); (T.S.)
| | - Motoki Matsuura
- Departments of Obstetrics, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (M.O.); (M.M.); (T.S.)
| | - Tsuyoshi Saito
- Departments of Obstetrics, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (M.O.); (M.M.); (T.S.)
| | - Takashi Kojima
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (H.S.); (T.K.); (T.O.); (K.S.); (Y.S.)
| |
Collapse
|
24
|
Fang X, Wang J, Chen L, Zhang X. circRNA circ_POLA2 increases microRNA-31 methylation to promote endometrial cancer cell proliferation. Oncol Lett 2021; 22:762. [PMID: 34539866 PMCID: PMC8436404 DOI: 10.3892/ol.2021.13023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 06/24/2021] [Indexed: 11/21/2022] Open
Abstract
Circular RNA (circRNA) circ_POLA2 is an oncogene in lung and cervical cancers. However, the role of circ_POLA2 in other types of cancer is unknown. The present study investigated the role of circ_POLA2 in endometrial cancer (EC). The mRNA expression levels of circ_POLA2 and microRNA (miR)-31 in EC and paired adjacent normal tissues were analyzed using reverse transcription-quantitative (RT-qPCR). Overexpression of circ_POLA2 was achieved in the EC cell lines, and its effects on miR-31 mRNA expression level and methylation were evaluated using RT-qPCR and methylation-specific PCR (MSP), respectively. Cell proliferation was assessed using a Cell Counting Kit-8 assay. The results indicated that circ_POLA2 was highly expressed in EC tissue and inversely correlated with miR-31 mRNA expression level. MSP analysis showed that circ_POLA2 overexpression increased miR-31 methylation and RT-qPCR analysis showed that circ_POLA2 overexpression decreased miR-31 mRNA expression level. Furthermore, circ_POLA2 overexpression also increased EC cell proliferation, while miR-31 overexpression decreased cell proliferation. Finally, circ_POLA2 overexpression reduced the effects of miR-31 overexpression. In conclusion, circ_POLA2 may increase miR-31 methylation of miR-31 in EC cells to promote cancer cell proliferation.
Collapse
Affiliation(s)
- Xia Fang
- Department of Gynecology, Beilun District People's Hospital, Ningbo, Zhejiang 315800, P.R. China
| | - Jinhua Wang
- Department of Neurology, Beilun District People's Hospital, Ningbo, Zhejiang 315800, P.R. China
| | - Lingying Chen
- Department of Gynecology, Beilun District People's Hospital, Ningbo, Zhejiang 315800, P.R. China
| | - Xiaochun Zhang
- Department of Gynecology, Beilun District People's Hospital, Ningbo, Zhejiang 315800, P.R. China
| |
Collapse
|
25
|
Yang P, Yun K, Zhang R. CircRNA circ-ATAD1 is downregulated in endometrial cancer and suppresses cell invasion and migration by downregulating miR-10a through methylation. Mamm Genome 2021; 32:488-494. [PMID: 34406448 DOI: 10.1007/s00335-021-09899-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/27/2021] [Indexed: 01/22/2023]
Abstract
It has been reported that circ-ATAD1 promotes the development of gastric cancer. This study was carried out to analyze the role of circ-ATAD1 in endometrial cancer (EC). EC and paired non-tumor tissues from EC patients (n = 60) were subjected to the isolation of total RNA and RT-qPCR to analyze the expression of circ-ATAD1 and miR-10a. Correlations between circ-ATAD1 and miR-10a were analyzed by Pearson's correlation coefficient. The effects of circ-ATAD1 overexpression on miR-10a expression and methylation were analyzed by RT-qPCR and methylation-specific PCR (MSP). The roles of circ-ATAD1 and miR-10a in regulating EC cell invasion and migration were analyzed by Transwell assays. We found that circ-ATAD1 was downregulated in EC, while miR-10a was upregulated in EC. An inverse correlation between circ-ATAD1 and miR-10a was observed across EC samples. In EC cells, circ-ATAD1 overexpression decreased miR-10a expression and increased miR-10a gene methylation. Transwell assay analysis showed that circ-ATAD1 overexpression totally reversed the enhancing effects of miR-10a on EC cell invasion and migration. Circ-ATAD1 is downregulated in EC and may suppress EC cell invasion and migration by downregulating miR-10a through methylation.
Collapse
Affiliation(s)
- Pengxia Yang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot City, 010050, Inner Mongolia, People's Republic of China
| | - Kunlun Yun
- Physical Examination Center, Inner Mongolia Autonomous Region People's Hospital, Hohhot City, 010050, Inner Mongolia, People's Republic of China
| | - Ruying Zhang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot City, 010050, Inner Mongolia, People's Republic of China.
| |
Collapse
|
26
|
Gut and Endometrial Microbiome Dysbiosis: A New Emergent Risk Factor for Endometrial Cancer. J Pers Med 2021; 11:jpm11070659. [PMID: 34357126 PMCID: PMC8304951 DOI: 10.3390/jpm11070659] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 12/14/2022] Open
Abstract
Endometrial cancer is one of the most common gynaecological malignancies worldwide. Histologically, two types of endometrial cancer with morphological and molecular differences and also therapeutic implications have been identified. Type I endometrial cancer has an endometrioid morphology and is estrogen-dependent, while Type II appears with non-endometrioid differentiation and follows an estrogen-unrelated pathway. Understanding the molecular biology and genetics of endometrial cancer is crucial for its prognosis and the development of novel therapies for its treatment. However, until now, scant attention has been paid to environmental components like the microbiome. Recently, due to emerging evidence that the uterus is not a sterile cavity, some studies have begun to investigate the composition of the endometrial microbiome and its role in endometrial cancer. In this review, we summarize the current state of this line of investigation, focusing on the relationship between gut and endometrial microbiome and inflammation, estrogen metabolism, and different endometrial cancer therapies.
Collapse
|
27
|
A ten-gene methylation signature as a novel biomarker for improving prediction of prognosis and indicating gene targets in endometrial cancer. Genomics 2021; 113:2032-2044. [PMID: 33915245 DOI: 10.1016/j.ygeno.2021.04.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 03/10/2021] [Accepted: 04/23/2021] [Indexed: 12/17/2022]
Abstract
Endometrial cancer (EC) is a common female reproductive tumor worldwide. Nonetheless, the pathogenesis of EC still remains ambiguous and associated epigenetic mechanism still to be explored. The goal of this study is to investigate whether gene methylation signature is associated with overall survival (OS) for EC patients. In this study, a 10-gene methylation risk model was built and the OS in high- and low-risk groups was significant different. The area under the ROC curve (AUC) of this model was 0.856 at 5 years survival. The nomogram could accurately predict the OS in EC patients, with concordance index and AUC at 5 year survival reached 0.796 and 0.792, respectively. Furthermore, we verified the nomogram with 24 patients in our center and the Kaplan-Meier survival curve also proved to be significantly different (p < 0.01). WGCNA revealed a key gene group for the model and further bioinformatics analysis indicated 6 genes as the hub genes in the module. Knockdown of MMP12 inhibited the proliferation, invasion and metastasis of EC cells. After all, a methylation signature and a nomogram based on this signature were constructed, and they could both predict survival in patients with EC. Moreover, WGCNA model identified MMP12 as a potential target for the treatment of EC.
Collapse
|
28
|
Zhang Z, He T, Huang L, Li J, Wang P. Immune gene prognostic signature for disease free survival of gastric cancer: Translational research of an artificial intelligence survival predictive system. Comput Struct Biotechnol J 2021; 19:2329-2346. [PMID: 34025929 PMCID: PMC8111455 DOI: 10.1016/j.csbj.2021.04.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 12/13/2022] Open
Abstract
The progress of artificial intelligence algorithms and massive data provide new ideas and choices for individual mortality risk prediction for cancer patients. The current research focused on depict immune gene related regulatory network and develop an artificial intelligence survival predictive system for disease free survival of gastric cancer. Multi-task logistic regression algorithm, Cox survival regression algorithm, and Random survival forest algorithm were used to develop the artificial intelligence survival predictive system. Nineteen transcription factors and seventy immune genes were identified to construct a transcription factor regulatory network of immune genes. Multivariate Cox regression identified fourteen immune genes as prognostic markers. These immune genes were used to construct a prognostic signature for gastric cancer. Concordance indexes were 0.800, 0.809, and 0.856 for 1-, 3- and 5- year survival. An interesting artificial intelligence survival predictive system was developed based on three artificial intelligence algorithms for gastric cancer. Gastric cancer patients with high risk score have poor survival than patients with low risk score. The current study constructed a transcription factor regulatory network and developed two artificial intelligence survival prediction tools for disease free survival of gastric cancer patients. These artificial intelligence survival prediction tools are helpful for individualized treatment decision.
Collapse
Key Words
- AJCC, the American Joint Committee on Cancer
- CI, confidence interval
- DCA, decision curve analysis
- DFS, disease free survival
- Disease free survival
- GC, gastric cancer
- GEO, the Gene Expression Omnibus
- Gastric cancer
- HR, hazard ratio
- Immune gene
- Prognostic signature
- ROC, receiver operating characteristic
- SD, standard deviation
- TCGA, The Cancer Genome Atlas
- Transcription factor
Collapse
Affiliation(s)
- Zhiqiao Zhang
- Department of Infectious Diseases, Shunde Hospital, Southern Medical University, Shunde, Guangdong, China
| | - Tingshan He
- Department of Infectious Diseases, Shunde Hospital, Southern Medical University, Shunde, Guangdong, China
| | - Liwen Huang
- Department of Infectious Diseases, Shunde Hospital, Southern Medical University, Shunde, Guangdong, China
| | - Jing Li
- Department of Infectious Diseases, Shunde Hospital, Southern Medical University, Shunde, Guangdong, China
| | - Peng Wang
- Department of Infectious Diseases, Shunde Hospital, Southern Medical University, Shunde, Guangdong, China
| |
Collapse
|
29
|
Howard D, James D, Murphy K, Garcia-Parra J, Pan-Castillo B, Rex S, Moul A, Jones E, Bilbao-Asensio M, Michue-Seijas S, Lutchman-Singh K, Margarit L, Francis LW, Rees P, Gonzalez D, Conlan RS. Dinaciclib, a Bimodal Agent Effective against Endometrial Cancer. Cancers (Basel) 2021; 13:1135. [PMID: 33800911 PMCID: PMC7962054 DOI: 10.3390/cancers13051135] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/19/2021] [Accepted: 03/01/2021] [Indexed: 12/24/2022] Open
Abstract
Endometrial cancer (EC) is the sixth most prevalent female cancer globally and although high rates of success are achieved when diagnosed at an early stage, the 5-year survival rate for cancers diagnosed at Stages II-IV is below 50%. Improving patient outcomes will necessitate the introduction of novel therapies to the clinic. Pan-cyclin-dependent kinase inhibitors (CDKis) have been explored as therapies for a range of cancers due to their ability to simultaneously target multiple key cellular processes, such as cell cycle progression, transcription, and DNA repair. Few studies, however, have reported on their potential for the treatment of EC. Herein, we examined the effects of the pan-CDKi dinaciclib in primary cells isolated directly from tumors and EC cell lines. Dinaciclib was shown to elicit a bimodal action in EC cell lines, disrupting both cell cycle progression and phosphorylation of the RNA polymerase carboxy terminal domain, with a concomitant reduction in Bcl-2 expression. Furthermore, the therapeutic potential of combining dinaciclib and cisplatin was explored, with the drugs demonstrating synergy at specific doses in Type I and Type II EC cell lines. Together, these results highlight the potential of dinaciclib for use as an effective EC therapy.
Collapse
Affiliation(s)
- David Howard
- Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, UK; (D.H.); (D.J.); (J.G.-P.); (B.P.-C.); (L.W.F.); (D.G.)
| | - David James
- Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, UK; (D.H.); (D.J.); (J.G.-P.); (B.P.-C.); (L.W.F.); (D.G.)
| | - Kate Murphy
- Department of Pathology, Singleton Hospital, Swansea Bay University Health Board, Swansea SA2 8QA, UK; (K.M.); (S.R.); (A.M.); (E.J.)
| | - Jezabel Garcia-Parra
- Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, UK; (D.H.); (D.J.); (J.G.-P.); (B.P.-C.); (L.W.F.); (D.G.)
| | - Belen Pan-Castillo
- Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, UK; (D.H.); (D.J.); (J.G.-P.); (B.P.-C.); (L.W.F.); (D.G.)
| | - Stuart Rex
- Department of Pathology, Singleton Hospital, Swansea Bay University Health Board, Swansea SA2 8QA, UK; (K.M.); (S.R.); (A.M.); (E.J.)
| | - Annemarie Moul
- Department of Pathology, Singleton Hospital, Swansea Bay University Health Board, Swansea SA2 8QA, UK; (K.M.); (S.R.); (A.M.); (E.J.)
| | - Eilir Jones
- Department of Pathology, Singleton Hospital, Swansea Bay University Health Board, Swansea SA2 8QA, UK; (K.M.); (S.R.); (A.M.); (E.J.)
| | - Marc Bilbao-Asensio
- Department of Chemistry, College of Science, Swansea University, Singleton Park, Swansea SA2 8PP, UK; (M.B.-A.); (S.M.-S.)
| | - Saul Michue-Seijas
- Department of Chemistry, College of Science, Swansea University, Singleton Park, Swansea SA2 8PP, UK; (M.B.-A.); (S.M.-S.)
| | - Kerryn Lutchman-Singh
- Department of Gynaecology Oncology, Singleton Hospital, Swansea Bay University Health Board, Swansea SA2 8QA, UK;
| | - Lavinia Margarit
- Department of Obstetrics and Gynaecology, Princess of Wales Hospital, Cwm Taf Morgannwg University Health Board, Bridgend CF31 1RQ, UK;
| | - Lewis W. Francis
- Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, UK; (D.H.); (D.J.); (J.G.-P.); (B.P.-C.); (L.W.F.); (D.G.)
| | - Paul Rees
- College of Engineering, Swansea University, Bay Campus, Swansea SA1 8EN, UK;
| | - Deyarina Gonzalez
- Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, UK; (D.H.); (D.J.); (J.G.-P.); (B.P.-C.); (L.W.F.); (D.G.)
| | - R. Steven Conlan
- Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, UK; (D.H.); (D.J.); (J.G.-P.); (B.P.-C.); (L.W.F.); (D.G.)
| |
Collapse
|
30
|
Kurimchak AM, Kumar V, Herrera-Montávez C, Johnson KJ, Srivastava N, Davarajan K, Peri S, Cai KQ, Mantia-Smaldone GM, Duncan JS. Kinome Profiling of Primary Endometrial Tumors Using Multiplexed Inhibitor Beads and Mass Spectrometry Identifies SRPK1 as Candidate Therapeutic Target. Mol Cell Proteomics 2020; 19:2068-2090. [PMID: 32994315 PMCID: PMC7710141 DOI: 10.1074/mcp.ra120.002012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 09/15/2020] [Indexed: 12/11/2022] Open
Abstract
Endometrial carcinoma (EC) is the most common gynecologic malignancy in the United States, with limited effective targeted therapies. Endometrial tumors exhibit frequent alterations in protein kinases, yet only a small fraction of the kinome has been therapeutically explored. To identify kinase therapeutic avenues for EC, we profiled the kinome of endometrial tumors and normal endometrial tissues using Multiplexed Inhibitor Beads and Mass Spectrometry (MIB-MS). Our proteomics analysis identified a network of kinases overexpressed in tumors, including Serine/Arginine-Rich Splicing Factor Kinase 1 (SRPK1). Immunohistochemical (IHC) analysis of endometrial tumors confirmed MIB-MS findings and showed SRPK1 protein levels were highly expressed in endometrioid and uterine serous cancer (USC) histological subtypes. Moreover, querying large-scale genomics studies of EC tumors revealed high expression of SRPK1 correlated with poor survival. Loss-of-function studies targeting SRPK1 in an established USC cell line demonstrated SRPK1 was integral for RNA splicing, as well as cell cycle progression and survival under nutrient deficient conditions. Profiling of USC cells identified a compensatory response to SRPK1 inhibition that involved EGFR and the up-regulation of IGF1R and downstream AKT signaling. Co-targeting SRPK1 and EGFR or IGF1R synergistically enhanced growth inhibition in serous and endometrioid cell lines, representing a promising combination therapy for EC.
Collapse
Affiliation(s)
- Alison M Kurimchak
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Vikas Kumar
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | | | - Katherine J Johnson
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Nishi Srivastava
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Karthik Davarajan
- Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Suraj Peri
- Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Kathy Q Cai
- Histopathology Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Gina M Mantia-Smaldone
- Division of Gynecologic Oncology, Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - James S Duncan
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
31
|
Key factors mediated by PI3K signaling pathway and related genes in endometrial carcinoma. J Bioenerg Biomembr 2020; 52:465-473. [PMID: 33159265 DOI: 10.1007/s10863-020-09854-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 09/30/2020] [Indexed: 12/16/2022]
Abstract
By analyzing the gene expression of endometrial carcinoma (EC) patients, the key factors in PI3K signaling pathway and its related genes mediating EC were explored. The EC samples and normal endometrial samples were downloaded from TCGA database and GTEx database. The R language "limma" package was used for differential analysis, and the expression level of genes in each tissue was analyzed by "gganatogram" package. Functional enrichment analysis of differential genes was carried out by KOBAS, an online bioinformatics website. The correlation between key genes and differential genes was evaluated using TCGA data and GTEx combined gene expression data. The corresponding clinical data were downloaded from TCGA database and GTEx database, and the R language "survival" package was used to assess the potential of candidate differential genes as a key factor of EC. Based on the combined differential analysis of TCGA and GTEx databases, 299 genes with significant differential in expression were finally got. Functional enrichment analysis revealed that genes were predominantly enriched in the entry of "Pathways in cancer", including RAC2 and PIK3R3 genes which were related with the abnormal PI3K pathway in cancer. PIK3R3, a key gene in the PI3K signaling pathway, was highly-expressed in EC. SPDEF, GCNT2, KIAA1324, C9orf152, MARVELD3, and APEX2 genes were found to be positively correlated with PIK3R3 in EC, all of which were highly expressed in EC. KM survival analysis showed that SPDEF, GCNT2, KIAA1324 and C9orf152 were significantly correlated with patients' survival. ROC analysis showed that SPDEF, GCNT2, KIAA1324 and C9orf152 gene could be used as potential markers for prognosis and survival of EC patients. It was found that PIK3R3, a key gene in the PI3K signaling pathway, was highly expressed in EC. The SPDEF, GCNT2, KIAA1324 and C9orf152 genes were also highly expressed in EC, and were positively correlated with PIK3R3 in EC. Moreover, they are significantly correlated with the patients' survival, suggesting that they may be potential markers for the prognosis of patients with EC.
Collapse
|
32
|
Wang Y, Yin L, Sun X. CircRNA hsa_circ_0002577 accelerates endometrial cancer progression through activating IGF1R/PI3K/Akt pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:169. [PMID: 32847606 PMCID: PMC7450704 DOI: 10.1186/s13046-020-01679-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 08/17/2020] [Indexed: 12/30/2022]
Abstract
Background Endometrial cancer (EC) is a common gynecologic malignancy worldwide. This study investigated the regulatory effects of circular RNA (circRNA) hsa_circ_0002577 on the tumorigenesis of EC. Methods Tumor samples and adjacent normal tissues were obtained from 84 EC patients. Recombinant lentiviral vectors expressing hsa_circ_0002577 (Lv-circRNA), short hairpin RNAs against hsa_circ_0002577 (sh-circRNA), miR-625-5p mimics, miR-625-5p inhibitor, lentiviral vectors expressing insulin-like growth factor 1 receptor (IGF1R) and their corresponding controls were transfected into EC cells as designated. A mouse xenograft model was established in BALB/c mice by inoculating Ishikawa cells transfected with sh-circRNA or control sequence. Results Hsa_circ_0002577 was upregulated in EC tissue samples and cells as compared to normal controls. EC patients with higher expression of hsa_circ_0002577 showed poorer overall survival and more advanced tumor stage. EC cells transfected with Lv-circRNA showed promoted proliferation, migration, and invasion, whereas the delivery of sh-circRNA exerted an opposite effect. Further analyses showed that hsa_circ_0002577 acted as a miR-625-5p sponge in EC cells. IGF1R was a potential downstream target of miR-625-5p. The expression of IGF1R in EC tissues was significantly higher than that in matched controls. Hsa_circ_0002577 accelerated EC development by inducing IGF1R expression and activating PI3K/Akt signaling pathway. Also, the knockdown of hsa_circ_0002577 delayed tumor growth and metastasis in the inoculated mice. Conclusion Our study showed that circRNA hsa_circ_002577 accelerated EC progression by acting as a miR-625-5p sponge, upregulating IGF1R and activating the PI3K/Akt pathway, suggesting the potential therapeutic use of hsa_circ_002577 in EC treatment. Trial registration Not Applicable.
Collapse
Affiliation(s)
- Yu Wang
- Department of Obstetrics & Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang City, 110004, Liaoning Province, China.
| | - Lili Yin
- Department of Obstetrics & Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang City, 110004, Liaoning Province, China
| | - Xiaofei Sun
- Department of Obstetrics & Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang City, 110004, Liaoning Province, China
| |
Collapse
|
33
|
Endometrial Cancer as a Metabolic Disease with Dysregulated PI3K Signaling: Shedding Light on Novel Therapeutic Strategies. Int J Mol Sci 2020; 21:ijms21176073. [PMID: 32842547 PMCID: PMC7504460 DOI: 10.3390/ijms21176073] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 12/15/2022] Open
Abstract
Endometrial cancer (EC) is one of the most common malignancies of the female reproductive organs. The most characteristic feature of EC is the frequent association with metabolic disorders. However, the components of these disorders that are involved in carcinogenesis remain unclear. Accumulating epidemiological studies have clearly revealed that hyperinsulinemia, which accompanies these disorders, plays central roles in the development of EC via the insulin-phosphoinositide 3 kinase (PI3K) signaling pathway as a metabolic driver. Recent comprehensive genomic analyses showed that over 90% of ECs have genomic alterations in this pathway, resulting in enhanced insulin signaling and production of optimal tumor microenvironments (TMEs). Targeting PI3K signaling is therefore an attractive treatment strategy. Several clinical trials for recurrent or advanced ECs have been attempted using PI3K-serine/threonine kinase (AKT) inhibitors. However, these agents exhibited far lower efficacy than expected, possibly due to activation of alternative pathways that compensate for the PIK3-AKT pathway and allow tumor growth, or due to adaptive mechanisms including the insulin feedback pathway that limits the efficacy of agents. Overcoming these responses with careful management of insulin levels is key to successful treatment. Further interest in specific TMEs via the insulin PI3K-pathway in obese women will provide insight into not only novel therapeutic strategies but also preventive strategies against EC.
Collapse
|
34
|
Aoki Y, Kanao H, Wang X, Yunokawa M, Omatsu K, Fusegi A, Takeshima N. Adjuvant treatment of endometrial cancer today. Jpn J Clin Oncol 2020; 50:753-765. [PMID: 32463094 DOI: 10.1093/jjco/hyaa071] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 01/15/2020] [Accepted: 04/30/2020] [Indexed: 01/22/2023] Open
Abstract
Endometrial cancer frequently occurs in post-menopausal women, and the endometrium is a well-known site of cancer affecting women. Endometrial cancer is found with genital bleeding and often at an early stage. However, there are some risks of recurrence after hysterectomy. As a medical treatment after the diagnosis of endometrial cancer, appropriate adjuvant therapy is considered to lead to a decrease in the rate of recurrence and improvement of prognosis according to the determination of the cancer stage from the surgical and histopathological results. In this review, we describe post-operative adjuvant therapy administered for endometrial cancer and advanced disease, focusing on chemotherapy, radiation therapy and the combination of both. These treatments are divided according to the risk of recurrence as based primarily on the reported evidence.
Collapse
Affiliation(s)
- Yoichi Aoki
- Department of Gynecologic Oncology, Cancer Institute Hospital, Tokyo, Japan
| | - Hiroyuki Kanao
- Department of Gynecologic Oncology, Cancer Institute Hospital, Tokyo, Japan
| | - Xipeng Wang
- Department of Gynecology and Obstetrics, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mayu Yunokawa
- Department of Gynecologic Oncology, Cancer Institute Hospital, Tokyo, Japan
| | - Kohei Omatsu
- Department of Gynecologic Oncology, Cancer Institute Hospital, Tokyo, Japan
| | - Atsushi Fusegi
- Department of Gynecologic Oncology, Cancer Institute Hospital, Tokyo, Japan.,Department of Perinatal and Women's Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Nobuhiro Takeshima
- Department of Gynecologic Oncology, Cancer Institute Hospital, Tokyo, Japan
| |
Collapse
|
35
|
Liu L, Yu ZY, Yu TT, Cui SH, Yang L, Chang H, Qu YH, Lv XF, Zhang XA, Ren CC. A Slug-dependent mechanism is responsible for tumor suppression of p53-stabilizing compound CP-31398 in p53-mutated endometrial carcinoma. J Cell Physiol 2020; 235:8768-8778. [PMID: 32633026 DOI: 10.1002/jcp.29720] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 03/30/2020] [Accepted: 04/03/2020] [Indexed: 12/19/2022]
Abstract
Mutation in the tumor suppressor gene p53 is the most frequent molecular defect in endometrial carcinoma (EC). Recently, CP-31398, a p53-stabilizing compound, has been indicated to possess the ability to alter the expression of non-p53 target genes in addition to p53 downstream genes in tumor cells. Herein, we explore the alternative mechanisms underlying the restoration of EC tumor suppressor function in mutant p53 by CP-31398. A p53-mutated EC cell was constructed in AN3CA cells with restored or partial loss of Slug using lentiviral vectors, followed by treatment with 25 μM CP-31398. A p53-independent mechanism of CP-31398 was confirmed by the interaction between mouse double minute 2 homolog (MDM2) and Slug AN3CA cells treated with IWR-1 (inhibitor of Wnt response 1). Furthermore, the AN3CA cells were treated with short hairpin RNA against Slug, Wnt-specific activators (LiCl) or inhibitors (XAV-939) followed by CP-31398 treatment. Moreover, AN3CA cell proliferation and apoptosis were examined. A tumorigenicity assay was conducted in nude mice. CP-31398 could promote the apoptosis of p53-mutated EC cells, while Slug reversed this effect. Slug ubiquitination was found to occur via binding of Slug to MDM2 in AN3CA cells. We found that CP-31398 increased the GSK-3ß, p-Slug, Puma, Wtp53, and Bax expressions whereas Wnt, Mtp-53, Slug, Bcl-2, and Ki-67 expressions were decreased. However, these findings were reversed following the activation of the Wnt pathway and overexpression of Slug. Finally, the in vivo experimental evidence confirmed that CP-31398 with depleted Slug suppressed tumor growth by downregulating the Slug. Collectively, CP-31398-regulated Slug downregulation represses the p53-mutated EC via the p53/Wnt/Puma pathway.
Collapse
Affiliation(s)
- Ling Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Prenatal Diagnosis and Fetal Therapy, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhi-Ying Yu
- Department of Gynecology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Tan-Tan Yu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Prenatal Diagnosis and Fetal Therapy, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shi-Hong Cui
- Department of Prenatal Diagnosis and Fetal Therapy, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Yang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Chang
- Department of Prenatal Diagnosis and Fetal Therapy, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan-Hong Qu
- Department of Prenatal Diagnosis and Fetal Therapy, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao-Feng Lv
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao-An Zhang
- Department of Imaging, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chen-Chen Ren
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
36
|
Coll-de la Rubia E, Martinez-Garcia E, Dittmar G, Gil-Moreno A, Cabrera S, Colas E. Prognostic Biomarkers in Endometrial Cancer: A Systematic Review and Meta-Analysis. J Clin Med 2020; 9:E1900. [PMID: 32560580 PMCID: PMC7356541 DOI: 10.3390/jcm9061900] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 12/23/2022] Open
Abstract
Endometrial cancer (EC) is the sixth most common cancer in women worldwide and its mortality is directly associated with the presence of poor prognostic factors driving tumor recurrence. Stratification systems are based on few molecular, and mostly clinical and pathological parameters, but these systems remain inaccurate. Therefore, identifying prognostic EC biomarkers is crucial for improving risk assessment pre- and postoperatively and to guide treatment decisions. This systematic review gathers all protein biomarkers associated with clinical prognostic factors of EC, recurrence and survival. Relevant studies were identified by searching the PubMed database from 1991 to February 2020. A total number of 398 studies matched our criteria, which compiled 255 proteins associated with the prognosis of EC. MUC16, ESR1, PGR, TP53, WFDC2, MKI67, ERBB2, L1CAM, CDH1, PTEN and MMR proteins are the most validated biomarkers. On the basis of our meta-analysis ESR1, TP53 and WFDC2 showed potential usefulness for predicting overall survival in EC. Limitations of the published studies in terms of appropriate study design, lack of high-throughput measurements, and statistical deficiencies are highlighted, and new approaches and perspectives for the identification and validation of clinically valuable EC prognostic biomarkers are discussed.
Collapse
Affiliation(s)
- Eva Coll-de la Rubia
- Biomedical Research Group in Gynecology, Vall Hebron Institute of Research, Universitat Autònoma de Barcelona, CIBERONC, 08035 Barcelona, Spain;
| | - Elena Martinez-Garcia
- Quantitative Biology Unit, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg; (E.M.-G.); (G.D.)
| | - Gunnar Dittmar
- Quantitative Biology Unit, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg; (E.M.-G.); (G.D.)
| | - Antonio Gil-Moreno
- Biomedical Research Group in Gynecology, Vall Hebron Institute of Research, Universitat Autònoma de Barcelona, CIBERONC, 08035 Barcelona, Spain;
- Gynecological Department, Vall Hebron University Hospital, CIBERONC, 08035 Barcelona, Spain
| | - Silvia Cabrera
- Biomedical Research Group in Gynecology, Vall Hebron Institute of Research, Universitat Autònoma de Barcelona, CIBERONC, 08035 Barcelona, Spain;
- Gynecological Department, Vall Hebron University Hospital, CIBERONC, 08035 Barcelona, Spain
| | - Eva Colas
- Biomedical Research Group in Gynecology, Vall Hebron Institute of Research, Universitat Autònoma de Barcelona, CIBERONC, 08035 Barcelona, Spain;
| |
Collapse
|
37
|
Zhang N, Wang Y, Liu H, Shen W. Extracellular vesicle encapsulated microRNA-320a inhibits endometrial cancer by suppression of the HIF1α/VEGFA axis. Exp Cell Res 2020; 394:112113. [PMID: 32473223 DOI: 10.1016/j.yexcr.2020.112113] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/22/2020] [Accepted: 05/24/2020] [Indexed: 01/02/2023]
Abstract
Accumulating evidence indicates that cancer-associated fibroblasts (CAFs) play a crucial role in endometrial cancer (EC) pathogenesis. The present study investigated the clinical significance and biological function of extracellular vesicle (EV) encapsulated miR-320a released from CAFs in EC. EC-related microarray data was obtained from the GSE25405 database and differential analysis was performed. Expression of miR-320a in CAFs and normal endometrial fibroblasts (NFs) as well as CAF-delivered EVs was detected; also, delivery of miR-320a from CAFs to EC cells was observed. In addition we confirmed that miR-320a targets HIF1α via a dual-luciferase reporter assay. Phenotypic analysis was used to study the functional significance of EV delivered miR-320a and its downstream effects. miR-320a was found to have low expression in EC cells and tissues. CAF-secreted EVs were successfully isolated and miR-320a was found also be expressed at low levels in these EVs. Finally, we found direct transfer of CAF-secreted exosomal miR-320a to EC cells, which inhibited their proliferation. Mechanistically, we found this is due to downregulation of HIF1α by miR-320a, which led to lowered VEGFA expression in vitro. Accordingly, we overexpressed HIF1α also showed that the inhibitory effect of miR-320a overexpression in EC cells could be reversed. These results point to CAF-derived EVs carrying overexpressed miR-320a as a novel direction for therapeutic strategies for EC.
Collapse
Affiliation(s)
- Na Zhang
- Department of Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, 110001, PR China
| | - Yuehong Wang
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, 110001, PR China
| | - Hongbo Liu
- Department of Health Statistics, School of Public Health, China Medical University, Shenyang, 110001, PR China
| | - Wenjing Shen
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, 110001, PR China.
| |
Collapse
|
38
|
Lightfoot M, Montemorano L, Bixel K. PARP Inhibitors in Gynecologic Cancers: What Is the Next Big Development? Curr Oncol Rep 2020; 22:29. [PMID: 32067102 DOI: 10.1007/s11912-020-0873-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Conventional and novel applications of Poly(adenosine diphosphate [ADP]-ribose) polymerase (PARP) inhibitors (PARPi) are reviewed in the context of recently published clinical trials and preclinical data supporting rapidly expanding uses of this class of chemotherapy. RECENT FINDINGS PARPi block a pathway of DNA repair and target defects in homologous recombination repair (HRR), a pathway responsible for high-fidelity repair of double-strand breaks in DNA. BRCA1/2 proteins are essential to this pathway. Approximately 15-30% of women with ovarian cancer will have a germline or somatic BRCA mutation, and PARPi have shown promise in this population in a variety of settings. With growing understanding of the HRR pathway and its role in gynecologic malignancies, the potential applications of PARPi continue to expand. While the role of PARPi in gynecologic malignancies is most established in ovarian cancer, there are also promising applications in uterine and cervical cancer. We review current indications for PARPi use and promising applications of these medications in gynecologic malignancies.
Collapse
Affiliation(s)
- Michelle Lightfoot
- Ohio State Wexner Medical Center, M210 Starling Loving Hall, 320 West 10th Avenue, Columbus, OH-43210, USA
| | - Lauren Montemorano
- Ohio State Wexner Medical Center, M210 Starling Loving Hall, 320 West 10th Avenue, Columbus, OH-43210, USA
| | - Kristin Bixel
- Ohio State Wexner Medical Center, M210 Starling Loving Hall, 320 West 10th Avenue, Columbus, OH-43210, USA.
| |
Collapse
|
39
|
Nagesh PKB, Chowdhury P, Hatami E, Jain S, Dan N, Kashyap VK, Chauhan SC, Jaggi M, Yallapu MM. Tannic acid inhibits lipid metabolism and induce ROS in prostate cancer cells. Sci Rep 2020; 10:980. [PMID: 31969643 PMCID: PMC6976712 DOI: 10.1038/s41598-020-57932-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/29/2019] [Indexed: 12/27/2022] Open
Abstract
Prostate cancer (PCa) cells exploit the aberrant lipid signaling and metabolism as their survival advantage. Also, intracellular storage lipids act as fuel for the PCa proliferation. However, few studies were available that addressed the topic of targeting lipid metabolism in PCa. Here, we assessed the tannic acid (TA) lipid-targeting ability and its capability to induce endoplasmic reticulum (ER) stress by reactive oxygen species (ROS) in PCa cells. TA exhibited dual effects by inhibiting lipogenic signaling and suppression of lipid metabolic pathways. The expression of proteins responsible for lipogenesis was down regulated. The membrane permeability and functionality of PCa were severely affected and caused nuclear disorganization during drug exposure. Finally, these consolidated events shifted the cell's survival balance towards apoptosis. These results suggest that TA distinctly interferes with the lipid signaling and metabolism of PCa cells.
Collapse
Affiliation(s)
- Prashanth K B Nagesh
- Department of Microbiology and Immunology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Pallabita Chowdhury
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Elham Hatami
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Shashi Jain
- Tumor Initiation and Maintenance, Sanford-Burnham Medical Research Institute, La Jolla, California, 92037, USA
- Department of Pathology, Moores UCSD Cancer Center, and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Nirnoy Dan
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Vivek Kumar Kashyap
- Department of Microbiology and Immunology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Subhash C Chauhan
- Department of Microbiology and Immunology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Meena Jaggi
- Department of Microbiology and Immunology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Murali M Yallapu
- Department of Microbiology and Immunology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA.
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|