1
|
Semertzidou A, Whelan E, Smith A, Ng S, Roberts L, Brosens JJ, Marchesi JR, Bennett PR, MacIntyre DA, Kyrgiou M. Microbial signatures and continuum in endometrial cancer and benign patients. MICROBIOME 2024; 12:118. [PMID: 38951935 PMCID: PMC11218081 DOI: 10.1186/s40168-024-01821-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 04/22/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND Endometrial cancer is a multifactorial disease with inflammatory, metabolic and potentially microbial cues involved in disease pathogenesis. The endometrial cancer microbiome has been poorly characterised so far and studies have often overestimated bacterial biomass due to lack of integration of appropriate contamination controls. There is also a scarcity of evidence on the functionality of microbial microenvironments in endometrial cancer. This work addresses that knowledge gap by interrogating the genuine, contamination-free microbial signatures in the female genital tract and rectum of women with endometrial cancer and the mechanistic role of microbiome on carcinogenic processes. RESULTS Here we sampled different regions of the reproductive tract (vagina, cervix, endometrium, fallopian tubes and ovaries) and rectum of 61 patients (37 endometrial cancer; 24 benign controls). We performed 16S rRNA gene sequencing of the V1-V2 hypervariable regions and qPCR of the 16S rRNA gene to qualitatively and quantitatively assess microbial communities and used 3D benign and endometrial cancer organoids to evaluate the effect of microbial products of L. crispatus, which was found depleted in endometrial cancer patients following primary analysis, on endometrial cell proliferation and inflammation. We found that the upper genital tract of a subset of women with and without endometrial cancer harbour microbiota quantitatively and compositionally distinguishable from background contaminants. Endometrial cancer was associated with reduced cervicovaginal and rectal bacterial load together with depletion of Lactobacillus species relative abundance, including L. crispatus, increased bacterial diversity and enrichment of Porphyromonas, Prevotella, Peptoniphilus and Anaerococcus in the lower genital tract and endometrium. Treatment of benign and malignant endometrial organoids with L. crispatus conditioned media exerted an anti-proliferative effect at high concentrations but had minimal impact on cytokine and chemokine profiles. CONCLUSIONS Our findings provide evidence that the upper female reproductive tract of some women contains detectable levels of bacteria, the composition of which is associated with endometrial cancer. Whether this is a cause or consequence of cancer pathophysiology and what is the functional significance of this finding remain to be elucidated to guide future screening tools and microbiome-based therapeutics. Video Abstract.
Collapse
Affiliation(s)
- Anita Semertzidou
- Institute of Reproductive and Developmental Biology, Department of Digestion, Metabolism and Reproduction, Department of Surgery and Cancer, Imperial College Faculty of Medicine, Room 3006, 3rd Floor, Du Cane Road, London, W12 0NN, UK
- Department of Obstetrics & Gynaecology, Imperial College Healthcare NHS Trust, London, W12 0HS, UK
| | - Eilbhe Whelan
- Institute of Reproductive and Developmental Biology, Department of Digestion, Metabolism and Reproduction, Department of Surgery and Cancer, Imperial College Faculty of Medicine, Room 3006, 3rd Floor, Du Cane Road, London, W12 0NN, UK
- Department of Obstetrics & Gynaecology, Imperial College Healthcare NHS Trust, London, W12 0HS, UK
| | - Ann Smith
- Faculty of Health and Applied Sciences, University West of England, Glenside Campus, Bristol, BS16 1DD, UK
| | - Sherrianne Ng
- Institute of Reproductive and Developmental Biology, Department of Digestion, Metabolism and Reproduction, Department of Surgery and Cancer, Imperial College Faculty of Medicine, Room 3006, 3rd Floor, Du Cane Road, London, W12 0NN, UK
| | - Lauren Roberts
- Institute of Reproductive and Developmental Biology, Department of Digestion, Metabolism and Reproduction, Department of Surgery and Cancer, Imperial College Faculty of Medicine, Room 3006, 3rd Floor, Du Cane Road, London, W12 0NN, UK
| | - Jan J Brosens
- Division of Reproductive Health, Warwick Medical School, Clinical Sciences Research Laboratories, University Hospital, Coventry, CV2 2DX, UK
| | - Julian R Marchesi
- Division of Digestive Diseases, Department of Digestion, Metabolism and Reproduction, Imperial College London, London, W2 1NY, UK
| | - Phillip R Bennett
- Institute of Reproductive and Developmental Biology, Department of Digestion, Metabolism and Reproduction, Department of Surgery and Cancer, Imperial College Faculty of Medicine, Room 3006, 3rd Floor, Du Cane Road, London, W12 0NN, UK
- Department of Obstetrics & Gynaecology, Imperial College Healthcare NHS Trust, London, W12 0HS, UK
| | - David A MacIntyre
- Institute of Reproductive and Developmental Biology, Department of Digestion, Metabolism and Reproduction, Department of Surgery and Cancer, Imperial College Faculty of Medicine, Room 3006, 3rd Floor, Du Cane Road, London, W12 0NN, UK
| | - Maria Kyrgiou
- Institute of Reproductive and Developmental Biology, Department of Digestion, Metabolism and Reproduction, Department of Surgery and Cancer, Imperial College Faculty of Medicine, Room 3006, 3rd Floor, Du Cane Road, London, W12 0NN, UK.
- Department of Obstetrics & Gynaecology, Imperial College Healthcare NHS Trust, London, W12 0HS, UK.
| |
Collapse
|
2
|
Anazco D, Acosta A, Cathcart-Rake EJ, D'Andre SD, Hurtado MD. Weight-centric prevention of cancer. OBESITY PILLARS 2024; 10:100106. [PMID: 38495815 PMCID: PMC10943063 DOI: 10.1016/j.obpill.2024.100106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/19/2024]
Abstract
Background The link between excess adiposity and carcinogenesis has been well established for multiple malignancies, and cancer is one of the main contributors to obesity-related mortality. The potential role of different weight-loss interventions on cancer risk modification has been assessed, however, its clinical implications remain to be determined. In this clinical review, we present the data assessing the effect of weight loss interventions on cancer risk. Methods In this clinical review, we conducted a comprehensive search of relevant literature using MEDLINE, Embase, Web of Science, and Google Scholar databases for relevant studies from inception to January 20, 2024. In this clinical review, we present systematic reviews and meta-analysis, randomized clinical trials, and prospective and retrospective observational studies that address the effect of different treatment modalities for obesity in cancer risk. In addition, we incorporate the opinions from experts in the field of obesity medicine and oncology regarding the potential of weight loss as a preventative intervention for cancer. Results Intentional weight loss achieved through different modalities has been associated with a reduced cancer incidence. To date, the effect of weight loss on the postmenopausal women population has been more widely studied, with multiple reports indicating a protective effect of weight loss on hormone-dependent malignancies. The effect of bariatric interventions as a protective intervention for cancer has been studied extensively, showing a significant reduction in cancer incidence and mortality, however, data for the effect of bariatric surgery on certain specific types of cancer is conflicting or limited. Conclusion Medical nutrition therapy, exercise, antiobesity medication, and bariatric interventions, might lead to a reduction in cancer risk through weight loss-dependent and independent factors. Further evidence is needed to better determine which population might benefit the most, and the amount of weight loss required to provide a clinically significant preventative effect.
Collapse
Affiliation(s)
- Diego Anazco
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | - Maria D. Hurtado
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
3
|
Ahmadi K, Reiisi S, Habibi Z. Comparison of the gene expression profiles of endometrial and trophoblastic cells in women with recurrent miscarriage: A bioinformatics approach. Int J Reprod Biomed 2024; 22:495-506. [PMID: 39205919 PMCID: PMC11347766 DOI: 10.18502/ijrm.v22i6.16800] [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: 05/11/2023] [Revised: 11/04/2023] [Accepted: 05/11/2024] [Indexed: 09/04/2024] Open
Abstract
Background Recurrent miscarriage (RM) remains unsolved in > 50% of patients and causes physical and psychological problems in women without specific risk factors for miscarriage. For a successful pregnancy, acceptance of the endometrium and invasion of trophoblast cells into the endometrium is necessary. Objective This study aimed to use computational analysis to identify key genes and related pathways in endometrial and trophoblast cells derived from RM samples. Materials and Methods In this bioinformatics study, we explored the differential expression of genes in endometrial and trophoblast cells by analyzing the GSE165004 and GSE76862 datasets, respectively with the limma package in R software. Subsequently, overlapped genes between 2 datasets were selected, gene ontology and Kyoto Encyclopedia of Genes and Genomes analyses were performed. The overlapped genes were integrated to construct a protein-protein interaction network and hub genes selection. Results We observed 41 overlapped genes between endometrial and trophoblast cells, and future analysis was accomplished in overlapped and nonoverlapped genes. Kyoto Encyclopedia of Genes and Genomes analysis indicated that overlapped genes were significantly enriched in the complement and coagulation cascades, pluripotency of stem cells, and synthesis and degradation of ketone bodies. Gene ontology analysis suggested that the genes were enriched in the cell cycle, apoptosis, and cell division. The top 10 genes included: IRS1, FGF2, MAPK6, MAPK1, MAPK3, MAPK8, MAPK9, PLK1, PRKACA, and PRKCA were identified from the PPI network. Conclusion This study identified the key genes and potential molecular pathways underlying the development of RM. This could provide novel insights to determine the possible mechanisms and interventional strategies associated with miscarriage.
Collapse
Affiliation(s)
- Kambiz Ahmadi
- Department of Computer Sciences, Faculty of Mathematical Sciences, Shahrekord University, Shahrekord, Iran
| | - Somayeh Reiisi
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Zahra Habibi
- Department of Women and Family Affairs, Chaharmahal and Bakhtiari Governorate, Shahrekord, Iran
| |
Collapse
|
4
|
Bukato K, Kostrzewa T, Gammazza AM, Gorska-Ponikowska M, Sawicki S. Endogenous estrogen metabolites as oxidative stress mediators and endometrial cancer biomarkers. Cell Commun Signal 2024; 22:205. [PMID: 38566107 PMCID: PMC10985914 DOI: 10.1186/s12964-024-01583-0] [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: 11/29/2023] [Accepted: 03/23/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Endometrial cancer is the most common gynecologic malignancy found in developed countries. Because therapy can be curative at first, early detection and diagnosis are crucial for successful treatment. Early diagnosis allows patients to avoid radical therapies and offers conservative management options. There are currently no proven biomarkers that predict the risk of disease occurrence, enable early identification or support prognostic evaluation. Consequently, there is increasing interest in discovering sensitive and specific biomarkers for the detection of endometrial cancer using noninvasive approaches. CONTENT Hormonal imbalance caused by unopposed estrogen affects the expression of genes involved in cell proliferation and apoptosis, which can lead to uncontrolled cell growth and carcinogenesis. In addition, due to their ability to cause oxidative stress, estradiol metabolites have both carcinogenic and anticarcinogenic properties. Catechol estrogens are converted to reactive quinones, resulting in oxidative DNA damage that can initiate the carcinogenic process. The molecular anticancer mechanisms are still not fully understood, but it has been established that some estradiol metabolites generate reactive oxygen species and reactive nitrogen species, resulting in nitro-oxidative stress that causes cancer cell cycle arrest or cell death. Therefore, identifying biomarkers that reflect this hormonal imbalance and the presence of endometrial cancer in minimally invasive or noninvasive samples such as blood or urine could significantly improve early detection and treatment outcomes.
Collapse
Affiliation(s)
- Katarzyna Bukato
- Department of Obstetrics and Gynecology, Oncological Gynecology and Gynecological Endocrinology, Medical University of Gdansk, Smoluchowskiego 17, Gdańsk, 80-214, Poland
| | - Tomasz Kostrzewa
- Department of Medical Chemistry, Faculty of Medicine, Medical University of Gdansk, Dębinki 1, Gdansk, 80-211, Poland
| | - Antonella Marino Gammazza
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, 90127, Italy
| | - Magdalena Gorska-Ponikowska
- Department of Medical Chemistry, Faculty of Medicine, Medical University of Gdansk, Dębinki 1, Gdansk, 80-211, Poland.
- IEMEST Istituto Euro-Mediterraneo di Scienza e Tecnologia, Palermo, 90127, Italy.
- Department of Biophysics, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, 70174, Stuttgart, Germany.
| | - Sambor Sawicki
- Department of Obstetrics and Gynecology, Oncological Gynecology and Gynecological Endocrinology, Medical University of Gdansk, Smoluchowskiego 17, Gdańsk, 80-214, Poland.
| |
Collapse
|
5
|
Puspitaningtyas H, Hutajulu SH, Fachiroh J, Anggorowati N, Sanjaya GY, Lazuardi L, Sripan P. Diverging likelihood of colon and rectal cancer in Yogyakarta, Indonesia: A cross sectional study. PLoS One 2024; 19:e0301191. [PMID: 38547083 PMCID: PMC10977797 DOI: 10.1371/journal.pone.0301191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/11/2024] [Indexed: 04/02/2024] Open
Abstract
OBJECTIVES Colon and rectal cancer are associated with different risk factors and prognostic. However, this discrepancy has not been widely explored in the local population. This study aimed to investigate the site-specific likelihood of colorectal cancer (CRC) incidence in the Yogyakarta province, Indonesia. METHODS This cross-sectional study analyses 1,295 CRC cases diagnosed in 2008-2019 registered in the Yogyakarta population-based cancer registry (PBCR) database. Cases were grouped into colon and rectal cancer. Log-binomial regression was used to determine the relative risk of either colon or rectal cancer across different gender, age group, and rurality of residence. The age-specific rates were calculated by age group and temporal trend for each group were analyzed using joinpoint regression. RESULTS Females displayed higher odds of colon cancer (relative risk/RR = 1.20, 95%CI = 1.02-1.41) and lower odds of rectal cancer (RR = 0.92, 95%CI = 0.85-0.99). Elevated odds of colon cancer were observed in younger age group, especially 30-39 (RR = 1.87, 95%CI = 1.10-3.19), while decreased odds of rectal cancer was apparent in age group 30-39 and 40-49 (RR = 0.75, 95%CI = 0.60-0.93 and RR = 0.82, 95%CI = 0.69-0.98, respectively). Living in urban or rural areas did not significantly influence the odds of either having colon (RR = 0.98, 95%CI = 0.82-1.17) or rectal cancer (RR = 1.01, 95%CI = 0.93-1.10). During 2008-2019, trends of colon cancer in age <50 increased by 8.15% annually while rectal cancer displayed a 9.71% increase annually prior to 2017, followed by a 17.23% decrease until 2019. CONCLUSIONS Yogyakarta population shows higher odds of young-onset colon cancer, especially between age 30-39 years old. Overall observation of trend shows increasing incidence in young-onset colon cancer, and non-significant decrease in rectal cancer.
Collapse
Affiliation(s)
- Herindita Puspitaningtyas
- Faculty of Medicine, Public Health and Nursing, Doctorate Program of Health and Medical Science, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Susanna Hilda Hutajulu
- Faculty of Medicine, Public Health and Nursing, Department of Internal Medicine, Division of Hematology and Medical Oncology, Universitas Gadjah Mada/Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Jajah Fachiroh
- Faculty of Medicine, Public Health and Nursing, Department of Histology and Cell Biology, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Nungki Anggorowati
- Faculty of Medicine, Public Health and Nursing, of Anatomical Pathology, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Guardian Yoki Sanjaya
- Faculty of Medicine, Public Health and Nursing, of Health Policy and Management, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Lutfan Lazuardi
- Faculty of Medicine, Public Health and Nursing, of Health Policy and Management, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Patumrat Sripan
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
6
|
Song J, Han S, Choi S, Lee J, Jeong Y, Lee HM, Son J, Jeong DY, Yu SS, Lee W. A mixture of Pueraria lobata and Platycodon grandiflorum extracts ameliorates RANKL-induced osteoclast differentiation and ovariectomy-induced bone loss by regulating Src- PI3K-AKT and JNK/p38 signaling pathways. Heliyon 2024; 10:e24842. [PMID: 38312605 PMCID: PMC10835310 DOI: 10.1016/j.heliyon.2024.e24842] [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: 07/30/2023] [Revised: 01/15/2024] [Accepted: 01/15/2024] [Indexed: 02/06/2024] Open
Abstract
Osteoporosis is caused by increased bone resorption due to the excessive activity of osteoclasts. Pueraria lobata has demonstrated the ability to improve bone density in ovariectomized mice, and Platycodon grandiflorum can suppress osteolysis biomarkers such as collagen content in cartilage and alkaline phosphatase activity. In this study, we examined whether HX112, a mixture of Pueraria lobata and Platycodon grandiflorum extracts, could inhibit the receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL)-induced osteoclast differentiation to alleviate osteoporosis. To induce the differentiation of osteoclasts, RAW 264.7 cell were cultured with RANKL and HX112. Osteoclasts differentiation was evaluated by TRAP activity and TRAP staining. Bone resorption as osteoclasts major function was assessed by pit formation assay. As a result, HX112 suppressed osteoclast differentiation and bone resorptive function. Additionally, HX112 reduced the expression of osteoclastogenic genes including NFATc1 and c-Fos, and these effects of HX112 were mediated by inhibiting Src-phosphoinositide 3-kinase (PI3K)- Protein kinase B (Akt) and c-Jun N-terminal kinase (JNK)/p38 signaling pathways. Furthermore, ICR mice were ovariectomized to induce osteoporosis and bone mineral density of femur was measured using micro-CT. Consequently, oral administration of HX112 to ovariectomized mice significantly improved bone microstructure and bone mineral density. Collectively, these findings indicate that the mixed extract of Pueraria lobata and Platycodon grandiflorum may be useful as therapeutics for osteoporosis.
Collapse
Affiliation(s)
- Jisun Song
- R&D Center for Innovative Medicines, Helixmith Co., Ltd., Seoul 07794, South Korea
| | - Suhyun Han
- R&D Center for Innovative Medicines, Helixmith Co., Ltd., Seoul 07794, South Korea
| | - Sooyeon Choi
- R&D Center for Innovative Medicines, Helixmith Co., Ltd., Seoul 07794, South Korea
| | - Jungkyu Lee
- R&D Center for Innovative Medicines, Helixmith Co., Ltd., Seoul 07794, South Korea
| | - Yoonseon Jeong
- R&D Center for Innovative Medicines, Helixmith Co., Ltd., Seoul 07794, South Korea
| | - Hyun Myung Lee
- R&D Center for Innovative Medicines, Helixmith Co., Ltd., Seoul 07794, South Korea
| | - JongDai Son
- R&D Center for Innovative Medicines, Helixmith Co., Ltd., Seoul 07794, South Korea
| | - Dam Yeon Jeong
- R&D Center for Innovative Medicines, Helixmith Co., Ltd., Seoul 07794, South Korea
| | - Seung-Shin Yu
- R&D Center for Innovative Medicines, Helixmith Co., Ltd., Seoul 07794, South Korea
| | - Wonwoo Lee
- R&D Center for Innovative Medicines, Helixmith Co., Ltd., Seoul 07794, South Korea
| |
Collapse
|
7
|
Ge Y, Ni X, Li J, Ye M, Jin X. Roles of estrogen receptor α in endometrial carcinoma (Review). Oncol Lett 2023; 26:530. [PMID: 38020303 PMCID: PMC10644365 DOI: 10.3892/ol.2023.14117] [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: 08/10/2023] [Accepted: 10/06/2023] [Indexed: 12/01/2023] Open
Abstract
Endometrial carcinoma (EC) is a group of endometrial epithelial malignancies, most of which are adenocarcinomas and occur in perimenopausal and postmenopausal women. It is one of the most common carcinomas of the female reproductive system. It has been shown that the occurrence and development of EC is closely associated with the interaction between estrogen (estradiol, E2) and estrogen receptors (ERs), particularly ERα. As a key nuclear transcription factor, ERα is a carcinogenic factor in EC. Its interactions with upstream and downstream effectors and co-regulators have important implications for the proliferation, metastasis, invasion and inhibition of apoptosis of EC. In the present review, the structure of ERα and the regulation of ERα in multiple dimensions are described. In addition, the classical E2/ERα signaling pathway and the crosstalk between ERα and other EC regulators are elucidated, as well as the therapeutic targeting of ERα, which may provide a new direction for clinical applications of ERα in the future.
Collapse
Affiliation(s)
- Yidong Ge
- Department of Medical Oncology, The First Hospital of Ningbo University, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Zhejiang Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xiaoqi Ni
- Department of Medical Oncology, The First Hospital of Ningbo University, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Zhejiang Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jingyun Li
- Department of Medical Oncology, The First Hospital of Ningbo University, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Zhejiang Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Meng Ye
- Department of Medical Oncology, The First Hospital of Ningbo University, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Zhejiang Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xiaofeng Jin
- Department of Medical Oncology, The First Hospital of Ningbo University, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Zhejiang Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
8
|
Satpathi S, Gaurkar SS, Potdukhe A, Wanjari MB. Unveiling the Role of Hormonal Imbalance in Breast Cancer Development: A Comprehensive Review. Cureus 2023; 15:e41737. [PMID: 37575755 PMCID: PMC10415229 DOI: 10.7759/cureus.41737] [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: 06/28/2023] [Accepted: 07/11/2023] [Indexed: 08/15/2023] Open
Abstract
Breast cancer is a complex and multifactorial disease with a significant global impact. Hormonal imbalance has emerged as a crucial factor in breast cancer development, highlighting the importance of understanding the intricate interplay between hormones and breast tissue. This comprehensive review aims to unveil the role of hormonal imbalance in breast cancer by exploring the involvement of key hormones, including estrogen and progesterone, and their receptors in tumor development. The review delves into how hormonal imbalance impacts breast tissue, emphasizing the significance of hormone receptor status in guiding treatment decisions. Furthermore, the review investigates the influence of other hormones, such as insulin and growth factors, and their cross-talk with hormone pathways in breast cancer progression. The implications of hormonal imbalance assessment in breast cancer risk assessment and the importance of hormone testing in diagnosis and treatment decisions are also discussed. Moreover, the review provides an overview of the various hormonal therapies used in breast cancer treatment, their benefits, limitations, and ongoing research efforts to optimize their efficacy and overcome resistance. Future directions in hormonal therapy research, including developing novel therapies and personalized medicine approaches, are explored. This review underscores the need for a comprehensive understanding of hormonal imbalance in breast cancer to enhance prevention, diagnosis, and treatment strategies, ultimately improving outcomes for individuals affected by this disease.
Collapse
Affiliation(s)
- Shweta Satpathi
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sagar S Gaurkar
- Otolaryngology and Head and Neck Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Ashwini Potdukhe
- Medical Surgical Nursing, Srimati Radhikabai Meghe Memorial College of Nursing, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Mayur B Wanjari
- Research and Development, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
9
|
Huang P, Fan X, Yu H, Zhang K, Li H, Wang Y, Xue F. Glucose metabolic reprogramming and its therapeutic potential in obesity-associated endometrial cancer. J Transl Med 2023; 21:94. [PMID: 36750868 PMCID: PMC9906873 DOI: 10.1186/s12967-022-03851-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/24/2022] [Indexed: 02/09/2023] Open
Abstract
Endometrial cancer (EC) is a common gynecological cancer that endangers women health. Although substantial progresses of EC management have been achieved in recent years, the incidence of EC still remains high. Obesity has been a common phenomenon worldwide that increases the risk of EC. However, the mechanism associating obesity and EC has not been fully understood. Metabolic reprogramming as a remarkable characteristic of EC is currently emerging. As the primary factor of metabolic syndrome, obesity promotes insulin resistance, hyperinsulinemia and hyperglycaemia. This metabolic disorder remodels systemic status, which increases EC risk and is related with poor prognosis. Glucose metabolism in EC cells is complex and mediated by glycolysis and mitochondria to ensure energy requirement. Factors that affect glucose metabolism may have an impact on EC initiation and progression. In this study, we review the glucose metabolic reprogramming of EC not only systemic metabolism but also inherent tumor cell metabolism. In particular, the role of glucose metabolic regulation in malignant properties of EC will be focused. Understanding of metabolic profile and glucose metabolism-associated regulation mechanism in EC may provide novel perspective for treatment.
Collapse
Affiliation(s)
- Pengzhu Huang
- grid.412645.00000 0004 1757 9434Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052 China ,grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiangqin Fan
- grid.412645.00000 0004 1757 9434Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052 China ,grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongfei Yu
- grid.412645.00000 0004 1757 9434Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052 China ,grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Kaiwen Zhang
- grid.412645.00000 0004 1757 9434Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052 China ,grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Huanrong Li
- grid.412645.00000 0004 1757 9434Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052 China ,grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yingmei Wang
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China. .,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China.
| | - Fengxia Xue
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China. .,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
10
|
Thakur L, Thakur S. The interplay of sex steroid hormones and microRNAs in endometrial cancer: current understanding and future directions. Front Endocrinol (Lausanne) 2023; 14:1166948. [PMID: 37152960 PMCID: PMC10161733 DOI: 10.3389/fendo.2023.1166948] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/07/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction Endometrial cancer is a hormone-dependent malignancy, and sex steroid hormones play a crucial role in its pathogenesis. Recent studies have demonstrated that microRNAs (miRNAs) can regulate the expression of sex steroid hormone receptors and modulate hormone signaling pathways. Our aim is to provide an overview of the current understanding of the role of miRNAs in endometrial cancer regulated by sex steroid hormone pathways. Methods A thorough literature search was carried out in the PubMed database. The articles published from 2018 to the present were included. Keywords related to miRNAs, endometrial cancer, and sex steroid hormones were used in the search. Results Dysregulation of miRNAs has been linked to abnormal sex steroid hormone signaling and the development of endometrial cancer. Various miRNAs have been identified as modulators of estrogen and progesterone receptor expression, and the miRNA expression profile has been shown to be a predictor of response to hormone therapy. Additionally, specific miRNAs have been implicated in the regulation of genes involved in hormone-related signaling pathways, such as the PI3K/Akt/mTOR and MAPK/ERK pathways. Conclusion The regulation of sex steroid hormones by miRNAs is a promising area of research in endometrial cancer. Future studies should focus on elucidating the functional roles of specific miRNAs in sex steroid hormone signaling and identifying novel miRNA targets for hormone therapy in endometrial cancer management.
Collapse
Affiliation(s)
- Lovlesh Thakur
- Department of Medical Microbiology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Sunil Thakur
- Origin LIFE Healthcare Solutions and Research Center, Chandigarh, India
- *Correspondence: Sunil Thakur,
| |
Collapse
|
11
|
Lin Z, Sui X, Jiao W, Wang Y, Zhao J. Exploring the mechanism and experimental verification of puerarin in the treatment of endometrial carcinoma based on network pharmacology and bioinformatics analysis. BMC Complement Med Ther 2022; 22:150. [PMID: 35672846 PMCID: PMC9175360 DOI: 10.1186/s12906-022-03623-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/12/2022] [Indexed: 11/30/2022] Open
Abstract
Endometrial carcinoma is one of the two cancers with rising mortality and morbidity in recent years. In the light of many controversies about its treatment, it is urgent to construct a new prognostic model and to find out new therapeutic directions. As a small drug molecule widely used in clinical treatment and experimental research in China, puerarin has recently been proven to have obvious anti-cancer effects in multiple cancer cells. In this study, bioinformatics analysis and experimental validation were used to explore the potential mechanism of puerarin for endometrial carcinoma and construct a prognostic model. A total of 22 drug-related differential genes were found by constructing a database of drug targets and disease genes. The protein–protein interaction network was constructed for GO and KEGG enrichment analysis to initially explore the potential mechanism of its therapeutic effects. To construct the prognostic model, validation was performed by risk regression analysis and LASSO analysis. Finally, two prognostic genes—PIM1 and BIRC5 were determined to establish high and low risk groups. Kaplan–Meier analysis displayed a higher survival rate in the low-risk group than in the high-risk group. ROC curves indicated the stable and good effect in prediction (one-year AUC is 0.626; two-year AUC is 0.620; three-year AUC is 0.623). The interrelationship between immunity and its disease was explored by immune infiltration analysis. Finally, the potential effect of puerarin on endometrial carcinoma cells was further verified by experiments.
Collapse
|
12
|
Cheng T, Bai Y, Huang S, Wang Y, Zhou S, Liu H, Zhang R, Luo X, Yu P. Estrogen receptor 1 inhibits the progression of hepatocellular carcinoma via positively regulating lncRNA maternally expressed gene 3 under high glucose conditions. J Gastrointest Oncol 2022; 13:2485-2496. [PMID: 36388662 PMCID: PMC9660067 DOI: 10.21037/jgo-22-825] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/14/2022] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Long noncoding RNA (lncRNA) maternally expressed gene 3 (MEG3) is crucial in the association of diabetes mellitus (DM) and hepatocellular carcinoma (HCC), and estrogen receptor 1 (ESR1) plays an essential role in various cancers. However, the underlying regulatory effect of ESR1/lncRNA MEG3 on HCC with DM remains unclear. This study explored the regulatory effect of ESR1/lncRNA MEG3 on HCC cell progression. METHODS Bioinformatics analysis was used to predict the promoter sequence of lncRNA MEG3 using UCSC (http://genome.ucsc.edu/), followed by luciferase reporter and RNA immunoprecipitation (RIP) assays to verify the specific combination between ESR1 and lncRNA MEG3 promoter. After cotransfection with ESR1, ESR1 siRNA or lncRNA MEG3 RNA, CCK-8, 5-ethynyl-2'-deoxyuridine (EdU) and colony formation assays were used to evaluate the cell proliferation capacity. Cell apoptosis was assessed using flow cytometry analysis. Next, wound healing and Transwell assays were conducted to examine cell invasiveness and migration. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blot analysis were performed to quantify the expression of ESR1 or lncRNA MEG3. RESULTS ESR1 might be the transcription factor (TF) of lncRNA MEG3, and ESR1 bound with lncRNA MEG3 promoter. Overexpression of ESR1 repressed the proliferation, migration and invasion of HepG2 cells, and promoted apoptosis of HepG2 cells under high glucose conditions. Silencing ESR1 decreased lncRNA MEG3 expression in HepG2 cells but enhanced proliferation, migration and invasion. Meanwhile, a rescue assay demonstrated that silencing lncRNA MEG3 reversed the inhibitory effect of ESR1 on HepG2 cell progression. CONCLUSIONS ESR1 inhibits HCC cell progression through positively regulating lncRNA MEG3, and the results provide a promising strategy for HCC management.
Collapse
Affiliation(s)
- Ting Cheng
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Yu Bai
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Shuai Huang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Yao Wang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Saijun Zhou
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Hongyan Liu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Rui Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Xi Luo
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Pei Yu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
13
|
Ross RC, Akinde YM, Schauer PR, le Roux CW, Brennan D, Jernigan AM, Bueter M, Albaugh VL. The role of bariatric and metabolic surgery in the development, diagnosis, and treatment of endometrial cancer. Front Surg 2022; 9:943544. [PMID: 36117808 PMCID: PMC9470773 DOI: 10.3389/fsurg.2022.943544] [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: 05/13/2022] [Accepted: 07/25/2022] [Indexed: 12/13/2022] Open
Abstract
The obesity pandemic continues to contribute to a worsening burden of disease worldwide. The link between obesity and diseases such as diabetes, cardiovascular disease, and cancer has been well established, yet most patients living with obesity remain untreated or undertreated. Metabolic and bariatric surgery is the most effective and durable treatment for obesity, is safe, and may have a protective benefit with respect to cancer incidence. In this review, an overview of the link between obesity, metabolic surgery, and cancer is discussed with emphasis on indications for endometrial cancer, the malignancy most strongly associated with obesity. Considerable evidence from retrospective and prospective cohort studies supports a decreased risk of endometrial cancer in patients with obesity who undergo bariatric surgery compared with nonsurgical controls. Survivors of endometrial cancer are at increased risk of poor health outcomes associated with obesity, and women with endometrial cancer are more likely to die of cardiovascular disease and other obesity-related illnesses than of the malignancy itself. Recent advances in anticancer drug therapies have targeted pathways that may also be therapeutically altered with metabolic surgery. Metabolic surgery has significant potential to enter the treatment paradigm for endometrial cancer, and gynecologic oncologist visits present an opportunity to identify patients who may benefit the most.
Collapse
Affiliation(s)
- Robert C. Ross
- Translational and Integrative Gastrointestinal and Endocrine Research Laboratory, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| | - Yetunde M. Akinde
- Department of Obstetrics and Gynecology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Philip R. Schauer
- Metamor Institute, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| | - Carel W. le Roux
- School of Medicine, St. Vincent's University Hospital and University College Dublin, Dublin, Ireland
| | - Donal Brennan
- UCD Gynecological Oncology Group, UCD School of Medicine, Catherine McAuley Research Centre, Mater Misericordiae University Hospital, Belfield, Dublin, Ireland
| | - Amelia M. Jernigan
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Marco Bueter
- Department of Visceral and Transplantation Surgery, University Hospital of Zürich, Zürich, Switzerland
| | - Vance L. Albaugh
- Translational and Integrative Gastrointestinal and Endocrine Research Laboratory, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
- Metamor Institute, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
- Correspondence: Vance L. Albaugh
| |
Collapse
|
14
|
Chimento A, De Luca A, Avena P, De Amicis F, Casaburi I, Sirianni R, Pezzi V. Estrogen Receptors-Mediated Apoptosis in Hormone-Dependent Cancers. Int J Mol Sci 2022; 23:1242. [PMID: 35163166 PMCID: PMC8835409 DOI: 10.3390/ijms23031242] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 02/04/2023] Open
Abstract
It is known that estrogen stimulates growth and inhibits apoptosis through estrogen receptor(ER)-mediated mechanisms in many cancer cell types. Interestingly, there is strong evidence that estrogens can also induce apoptosis, activating different ER isoforms in cancer cells. It has been observed that E2/ERα complex activates multiple pathways involved in both cell cycle progression and apoptotic cascade prevention, while E2/ERβ complex in many cases directs the cells to apoptosis. However, the exact mechanism of estrogen-induced tumor regression is not completely known. Nevertheless, ERs expression levels of specific splice variants and their cellular localization differentially affect outcome of estrogen-dependent tumors. The goal of this review is to provide a general overview of current knowledge on ERs-mediated apoptosis that occurs in main hormone dependent-cancers. Understanding the molecular mechanisms underlying the induction of ER-mediated cell death will be useful for the development of specific ligands capable of triggering apoptosis to counteract estrogen-dependent tumor growth.
Collapse
Affiliation(s)
- Adele Chimento
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Arianna De Luca
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Paola Avena
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Ivan Casaburi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Rosa Sirianni
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Vincenzo Pezzi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| |
Collapse
|
15
|
Namvarsigaroudi N, Fard ZT. Endometrial Expression of Insulin Signaling Pathway Genes in Pregnancy Leading to Abortion under 20 Weeks in Infertile Women: A Case-Control Study. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2022; 16:286-291. [PMID: 36273315 PMCID: PMC9627012 DOI: 10.22074/ijfs.2021.534736.1163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND Impaired expression of genes which act on hormone signaling pathways is one of the factors affecting miscarriage. In this study, the expression levels of insulin receptor (INSR) and insulin receptor substrates-1 (IRS-1) genes in endometrial tissue of infertile women and fertile women with miscarriage in less than twenty weeks gestation for unknown reasons were evaluated. MATERIALS AND METHODS In this case-control study, forty-two fertile women with children and 42 infertile women, who underwent in vitro fertilization (IVF), were selected. Both groups had abortions under twenty weeks gestation for unknown reasons. The endometrial tissue of all patients was prepared to evaluate the expression of INSR and IRS-1 genes by quantitative real-time polymerase chain reaction (PCR) method after RNA extraction. RESULTS There was a statistically significant relationship between the expressions of INSR and IRS-1 genes in the endometrial tissue of the infertile women compared with the fertile women (P=0.002 and P=0.008, respectively). The expression level of genes was decreased in both groups by age and increasing body mass index (BMI). Comparison of genes expression levels in healthy and diabetic participants in each group showed a significant difference (P<0.0001), but no meaningful difference was indicated between diabetic infertile and fertile groups in terms of gene expression. INSR gene expression levels showed an increase in the fertile group in the second 10 weeks and a decrease in IRS-1 gene expression. But in the infertile group, both genes showed a slight increase in expression. CONCLUSION It seems a decreased expression of insulin signaling pathway genes in the endometrial tissue of infertile women can be one of reasons for unspecified abortion. These genes may be strong molecular markers for infertility.
Collapse
Affiliation(s)
| | - Zahra Tahmasebi Fard
- P.O.BOX: 3973188981Department of BiologyRoudehen
BranchIslamic Azad UniversityRoudehenIran
| |
Collapse
|
16
|
Cirillo F, Pellegrino M, Talia M, Perrotta ID, Rigiracciolo DC, Spinelli A, Scordamaglia D, Muglia L, Guzzi R, Miglietta AM, De Francesco EM, Belfiore A, Maggiolini M, Lappano R. Estrogen receptor variant ERα46 and insulin receptor drive in primary breast cancer cells growth effects and interleukin 11 induction prompting the motility of cancer-associated fibroblasts. Clin Transl Med 2021; 11:e516. [PMID: 34841688 PMCID: PMC8567034 DOI: 10.1002/ctm2.516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/21/2021] [Accepted: 07/20/2021] [Indexed: 02/06/2023] Open
Abstract
Among the prognostic and predictive biomarkers of breast cancer (BC), the role of estrogen receptor (ER)α wild-type has been acknowledged, although the action of certain ERα splice variants has not been elucidated. Insulin/insulin receptor (IR) axis has also been involved in the progression and metastasis of BC. For instance, hyperinsulinemia, which is often associated with obesity and type 2 diabetes, may be a risk factor for BC. Similarly, an aberrant expression of IR or its hyperactivation may correlate with aggressive BC phenotypes. In the present study, we have shown that a novel naturally immortalized BC cell line (named BCAHC-1) is characterized by a unique expression of 46 kDa ERα splice variant (ERα46) along with IR. Moreover, we have shown that a multifaceted crosstalk between ERα46 and IR occurs in BCAHC-1 cells upon estrogen and insulin exposure for growth and pulmonary metastasis. Through high-throughput RNA sequencing analysis, we have also found that the cytokine interleukin-11 (IL11) is the main factor linking BCAHC-1 cells to breast cancer-associated fibroblasts (CAFs). In particular, we have found that IL11 induced by estrogens and insulin in BCAHC-1 cells regulates pro-tumorigenic genes of the "extracellular matrix organization" signaling pathway, such as ICAM-1 and ITGA5, and promotes both migratory and invasive features in breast CAFs. Overall, our results may open a new scientific avenue to identify additional prognostic and therapeutic targets in BC.
Collapse
Affiliation(s)
- Francesca Cirillo
- Department of PhysicsUniversity of CalabriaRendeItaly
- Department of Pharmacy, Health and Nutritional SciencesUniversity of CalabriaRendeItaly
| | - Michele Pellegrino
- Department of Pharmacy, Health and Nutritional SciencesUniversity of CalabriaRendeItaly
| | - Marianna Talia
- Department of Pharmacy, Health and Nutritional SciencesUniversity of CalabriaRendeItaly
| | - Ida Daniela Perrotta
- Centre for Microscopy and Microanalysis, Transmission Electron Microscopy Laboratory, and Department of Biology, Ecology and Earth SciencesUniversity of CalabriaRendeItaly
| | | | - Asia Spinelli
- Department of Pharmacy, Health and Nutritional SciencesUniversity of CalabriaRendeItaly
| | - Domenica Scordamaglia
- Department of Pharmacy, Health and Nutritional SciencesUniversity of CalabriaRendeItaly
| | - Lucia Muglia
- Department of Pharmacy, Health and Nutritional SciencesUniversity of CalabriaRendeItaly
| | - Rita Guzzi
- Department of PhysicsUniversity of CalabriaRendeItaly
| | | | | | - Antonino Belfiore
- Department of Clinical and Experimental Medicine, University of CataniaGaribaldi‐Nesima HospitalCataniaItaly
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional SciencesUniversity of CalabriaRendeItaly
| | - Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional SciencesUniversity of CalabriaRendeItaly
| |
Collapse
|
17
|
Katopodis P, Kerslake R, Zikopoulos A, Beri N, Anikin V. p38β - MAPK11 and its role in female cancers. J Ovarian Res 2021; 14:84. [PMID: 34174910 PMCID: PMC8236201 DOI: 10.1186/s13048-021-00834-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 06/07/2021] [Indexed: 12/24/2022] Open
Abstract
Background The p38MAPK family of Mitogen Activated Protein Kinases are a group of signalling molecules involved in cell growth, survival, proliferation and differentiation. The widely studied p38α isoform is ubiquitously expressed and is implicated in a number of cancer pathologies, as are p38γ and p38δ. However, the mechanistic role of the isoform, p38β, remains fairly elusive. Recent studies suggest a possible role of p38β in both breast and endometrial cancer with research suggesting involvement in bone metastasis and cancer cell survival. Female tissue specific cancers such as breast, endometrial, uterine and ovary account for over 3,000,000 cancer related incidents annually; advancements in therapeutics and treatment however require a deeper understanding of the molecular aetiology associated with these diseases. This study provides an overview of the MAPK signalling molecule p38β (MAPK11) in female cancers using an in-silico approach. Methods A detailed gene expression and methylation analysis was performed using datasets from cBioportal, CanSar and MEXPRESS. Breast, Uterine Endometrial, Cervical, Ovarian and Uterine Carcinosarcoma TCGA cancer datasets were used and analysed. Results Data using cBioportal and CanSAR suggest that expression of p38β is lower in cancers: BRCA, UCEC, UCS, CESC and OV compared to normal tissue. Methylation data from SMART and MEXPRESS indicate significant probe level variation of CpG island methylation status of the gene MAPK11. Analysis of the genes’ two CpG islands shows that the gene was hypermethylated in the CpG1 with increased methylation seen in BRCA, CESC and UCEC cancer data sets with a slight increase of expression recorded in cancer samples. CpG2 exhibited hypomethylation with no significant difference between samples and high levels of expression. Further analysis from MEXPRESS revealed no significance between probe methylation and altered levels of expression. In addition, no difference in the expression of BRCA oestrogen/progesterone/HER2 status was seen. Conclusion This data provides an overview of the expression of p38β in female tissue specific cancers, showing a decrease in expression of the gene in BRCA, UCEC, CESC, UCS and OV, increasing the understanding of p38β MAPK expression and offering insight for future in-vitro investigation and therapeutic application. Supplementary Information The online version contains supplementary material available at 10.1186/s13048-021-00834-9.
Collapse
Affiliation(s)
- Periklis Katopodis
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UK. .,Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, London, UB9 6JH, UK.
| | - Rachel Kerslake
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UK
| | - Athanasios Zikopoulos
- Obstetrics and Gynaecology Department, Royal Cornwall Hospitals NHS Foundation Trust, Royal Cornwall Hospital, Truro, TR1 3LJ, UK
| | - Nefeli Beri
- Department of Medicine, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Vladimir Anikin
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, London, UB9 6JH, UK.,Department of Oncology and Reconstructive Surgery, Sechenov First Moscow State Medical University, Moscow, Russian Federation, 119146
| |
Collapse
|
18
|
Khojasteh Poor F, Keivan M, Ramazii M, Ghaedrahmati F, Anbiyaiee A, Panahandeh S, Khoshnam SE, Farzaneh M. Mini review: The FDA-approved prescription drugs that target the MAPK signaling pathway in women with breast cancer. Breast Dis 2021; 40:51-62. [PMID: 33896802 DOI: 10.3233/bd-201063] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Breast cancer (BC) is the most common cancer and the prevalent type of malignancy among women. Multiple risk factors, including genetic changes, biological age, dense breast tissue, and obesity are associated with BC. The mitogen-activated protein kinases (MAPK) signaling pathway has a pivotal role in regulating biological functions such as cell proliferation, differentiation, apoptosis, and survival. It has become evident that the MAPK pathway is associated with tumorigenesis and may promote breast cancer development. The MAPK/RAS/RAF cascade is closely associated with breast cancer. RAS signaling can enhance BC cell growth and progression. B-Raf is an important kinase and a potent RAF isoform involved in breast tumor initiation and differentiation. Depending on the reasons for cancer, there are different strategies for treatment of women with BC. Till now, several FDA-approved treatments have been investigated that inhibit the MAPK pathway and reduce metastatic progression in breast cancer. The most common breast cancer drugs that regulate or inhibit the MAPK pathway may include Farnesyltransferase inhibitors (FTIs), Sorafenib, Vemurafenib, PLX8394, Dabrafenib, Ulixertinib, Simvastatin, Alisertib, and Teriflunomide. In this review, we will discuss the roles of the MAPK/RAS/RAF/MEK/ERK pathway in BC and summarize the FDA-approved prescription drugs that target the MAPK signaling pathway in women with BC.
Collapse
Affiliation(s)
- Fatemeh Khojasteh Poor
- Department of Obstetrics and Gynecology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mona Keivan
- Fertility and Infertility Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Ramazii
- Kerman University of Medical Sciences, University of Kerman, Kerman, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Anbiyaiee
- Department of Surgery, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Samira Panahandeh
- School of Health, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
19
|
Mei S, Ge S, Wang J, Li H, Jing X, Liang K, Zhang X, Xue C, Zhang C, Zhang T. PRMT5 promotes progression of endometrioid adenocarcinoma via ERα and cell cycle signaling pathways. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2021; 7:154-164. [PMID: 33416213 PMCID: PMC7869932 DOI: 10.1002/cjp2.194] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 11/15/2020] [Accepted: 11/18/2020] [Indexed: 12/31/2022]
Abstract
Protein arginine methyltransferase 5 (PRMT5) has previously been reported to be upregulated in many malignant tumors. This study investigated the significance of PRMT5 in endometrial carcinoma (EC) and explored its function in tumorigenesis. Immunohistochemistry was performed to evaluate PRMT5 expression in 62 EC and 66 endometrial hyperplasia samples. The functions of PRMT5 were investigated by cell counting kit‐8, plate colony formation, wound healing, and transwell and flow cytometry assays. Quantitative reverse transcription‐polymerase chain reaction and western blotting were used to measure the expression of PRMT5, changes in estrogen receptor α (ERα), and related functional proteins. Coimmunoprecipitation was performed to examine the interaction of PRMT5 with ERα and its coactivator steroid receptor coactivator‐1 (SRC1). Compared to endometrial hyperplasia tissue, PRMT5 was overexpressed in endometrioid adenocarcinoma (EAC) but not overexpressed in mucinous EC. The main expression pattern of PRMT5 in EAC was cytoplasmic. However, the positive cases of endometrial hyperplasia showed both cytoplasmic and nuclear positivity in the endometrial glands or were mainly positive in stromal cells. Knockdown of PRMT5 significantly inhibited the growth and migration ability of EAC cells and promoted their apoptosis by regulating cyclin D1, c‐myc, p53, and Bcl2 proteins. Furthermore, PRMT5 could form a complex with ERα and SRC1 to promote the expression of ERα. In conclusion, PRMT5 plays a significant role in the progression of EAC by interacting with ERα and impacting the cell cycle signaling pathways.
Collapse
Affiliation(s)
- Shuyu Mei
- Institute of Pathology and Pathophysiology, Shandong University School of Medicine, Jinan, PR China.,Department of Pathology, Bao Di Hospital, Bao Di Clinical College of Tianjin Medical University, Tianjin, PR China
| | - Shuang Ge
- Institute of Pathology and Pathophysiology, Shandong University School of Medicine, Jinan, PR China
| | - Jun Wang
- Institute of Pathology and Pathophysiology, Shandong University School of Medicine, Jinan, PR China
| | - Hailing Li
- Institute of Pathology and Pathophysiology, Shandong University School of Medicine, Jinan, PR China
| | - Xiaotong Jing
- Institute of Pathology and Pathophysiology, Shandong University School of Medicine, Jinan, PR China
| | - Ke Liang
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, PR China
| | - Xiaoying Zhang
- Institute of Pathology and Pathophysiology, Shandong University School of Medicine, Jinan, PR China
| | - Chaoshuai Xue
- Institute of Pathology and Pathophysiology, Shandong University School of Medicine, Jinan, PR China
| | - Cuijuan Zhang
- Institute of Pathology and Pathophysiology, Shandong University School of Medicine, Jinan, PR China.,Department of Pathology, Qilu Hospital of Shandong University, Jinan, PR China
| | - Tingguo Zhang
- Institute of Pathology and Pathophysiology, Shandong University School of Medicine, Jinan, PR China.,Department of Pathology, Qilu Hospital of Shandong University, Jinan, PR China
| |
Collapse
|
20
|
Chang Y, Hao M, Jia R, Zhao Y, Cai Y, Liu Y. Metapristone (RU486-derivative) inhibits endometrial cancer cell progress through regulating miR-492/Klf5/Nrf1 axis. Cancer Cell Int 2021; 21:29. [PMID: 33413440 PMCID: PMC7792070 DOI: 10.1186/s12935-020-01682-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 11/27/2020] [Indexed: 12/14/2022] Open
Abstract
Background Endometrial cancer is an invasive gynecological cancer prevalent in the world. The pathogenesis of endometrial cancer is related to multiple levels of regulation, referring to oestrogen, tumor-suppressor gene (e.g. PTEN) or microRNAs (e.g. miR-23a and miR-29b). Metapristone is a hormone-related drug, which is widely used in clinical treatment of endometrial cancer. However, the underlying regulatory mechanism of metapristone on endometrial cancer is still unclear, especially the regulatory effect on microRNAs. The aim of this study is to investigate the specific molecular mechanism of metapristone regulating microRNAs in the treatment of endometrial cancer. Methods RL95-2 cells and Ishikawa cells were used as the endometrial cancer models. MiR-492 or si-miR-492 was transfected into RL95-2 cells and Ishikawa cells to explore the role of miR-492 in endometrial cancer. The cell cancer model and mice cancer model were used to confirm the function and mechanism of metapristone affected on endometrial cancer in vitro and in vivo. Mechanically, cell proliferation was monitored using MTT assay, cell colony formation assay and EdU assay. Luciferase reporter assay was used to identify the downstream target gene of miR-492. The protein expression and RNA expression were respectively measured by western blot and qRT-PCR for cell signaling pathway research, subsequently, were verified in the mice tumor model via immunohistochemistry. Results Metapristone as a kind of hormone-related drug significantly inhibited the endometrial cancer cell growth through regulating cell apoptosis-related gene expression. Mechanically, miR-492 and its target genes Klf5 and Nrf1 were highly expressed in the endometrial cancer cell lines, which promoted cell proliferation and inhibited cell apoptosis. Metapristone decreased the expression of miR-492 and its target genes Klf5 and Nrf1, leading to endometrial cancer cell growth inhibition in vitro and in vivo. Conclusion Metapristone inhibited the endometrial cancer cell growth through regulating the cell apoptosis-related signaling pathway and decreasing the expression of miR-492 and its downstream target genes (Klf5 and Nrf1), which provided the theoretical basis in clinical treatment of endometrial cancer.
Collapse
Affiliation(s)
- Yue Chang
- Department of Obstetrics and Gynecology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China
| | - Min Hao
- Department of Obstetrics and Gynecology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China
| | - Ru Jia
- Department of Obstetrics and Gynecology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yihui Zhao
- Department of Obstetrics and Gynecology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yixuan Cai
- Department of Obstetrics and Gynecology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yun Liu
- Department of Obstetrics and Gynecology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China.
| |
Collapse
|
21
|
Karadayi FZ, Yaman M, Kisla MM, Konu O, Ates-Alagoz Z. Design, synthesis, anticancer activity, molecular docking and ADME studies of novel methylsulfonyl indole-benzimidazoles in comparison with ethylsulfonyl counterparts. NEW J CHEM 2021. [DOI: 10.1039/d1nj01019k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Specific sidechain modifications on the indole-benzimidazole scaffold play fundamental roles for determining molecule's affinity against ERα and its anti-cancer activity.
Collapse
Affiliation(s)
| | - Murat Yaman
- Interdisciplinary Program in Neuroscience
- Bilkent University
- Ankara
- Turkey
| | - Mehmet Murat Kisla
- Department of Pharmaceutical Chemistry
- Faculty of Pharmacy
- Ankara University
- Ankara
- Turkey
| | - Ozlen Konu
- Interdisciplinary Program in Neuroscience
- Bilkent University
- Ankara
- Turkey
- Department of Molecular Biology and Genetics
| | - Zeynep Ates-Alagoz
- Department of Pharmaceutical Chemistry
- Faculty of Pharmacy
- Ankara University
- Ankara
- Turkey
| |
Collapse
|
22
|
Wang JY, Chen LJ, Qiang P. The Potential Role of N6-Methyladenosine (m6A) Demethylase Fat Mass and Obesity-Associated Gene (FTO) in Human Cancers. Onco Targets Ther 2020; 13:12845-12856. [PMID: 33364780 PMCID: PMC7751723 DOI: 10.2147/ott.s283417] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/27/2020] [Indexed: 12/12/2022] Open
Abstract
N6-methyladenosine (m6A) demethylase fat mass and obesity-associated gene(FTO), previously recognized to be related with obesity and diabetes, was gradually discovered to be dysregulated in multiple cancers and plays an oncogenic or tumor-suppressive role. However, the specific expression and pro- or anti-cancer role of FTO in various cancers remained controversial. In this review, through summarizing the available literature, we found that FTO single nucleotide polymorphisms (SNPs) were closely related with cancer risk. Additionally, the dysregulation of FTO was implicated in multiple biological processes, such as cancer cell apoptosis, proliferation, migration, invasion, metastasis, cell-cycle, differentiation, stem cell self-renewal and so on. These modulations mostly relied on the communications between FTO and specific signaling pathways, including PI3K/AKT, MAPK and mTOR signaling pathways. Furthermore, FTO had great potential for clinical application by serving as a prognostic biomarker.
Collapse
Affiliation(s)
- Jin-Yan Wang
- Department of Obstetrics and Gynecology, Zhangjiagang First People's Hospital, Zhangjiagang Jiangsu 215600, People's Republic of China.,Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Li-Juan Chen
- Department of Obstetrics and Gynecology, Zhangjiagang First People's Hospital, Zhangjiagang Jiangsu 215600, People's Republic of China
| | - Ping Qiang
- Department of Gynecology, Zhangjiagang First People's Hospital, Zhangjiagang Affiliated Hospital of Soochow University, Zhangjiagang, Jiangsu 215600, People's Republic of China
| |
Collapse
|
23
|
Rotenberg O, Doulaveris G, Fridman D, Renz M, Kaplan J, Xie X, Goldberg GL, Dar P. Long-term outcome of postmenopausal women with proliferative endometrium on endometrial sampling. Am J Obstet Gynecol 2020; 223:896.e1-896.e7. [PMID: 32640199 DOI: 10.1016/j.ajog.2020.06.045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 06/17/2020] [Accepted: 06/22/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Proliferative endometrium has been reported in 15% of endometrial biopsies of women aged 50 years and older. Contrary to endometrial hyperplasia, proliferative endometrium has not been associated with the risk of endometrial cancer. OBJECTIVE This study aimed to report on the long-term outcome of postmenopausal women who received a diagnosis of proliferative endometrium. STUDY DESIGN This is a retrospective cohort study of 1808 women aged 55 years and older who underwent endometrial sampling between January 1997 and December 2008. Outcome data were available through February 2018. Women with a proliferative endometrium were compared with those with an atrophic endometrium for future development of endometrial hyperplasia or cancer. A subanalysis was performed for those who presented with postmenopausal bleeding. Uni- and multivariable logistic regression analyses were used to assess for confounders. RESULTS In this study, 297 women (16.4%) received a diagnosis of proliferative endometrium. Furthermore, 962 women met the inclusion criteria. Among those women, 278 had a proliferative endometrium, and 684 had an atrophic endometrium. Women with a proliferative endometrium were younger (61.2 vs 64.5 years; P<.0001) and had a higher body mass index (33.9 vs 30.6 kg/m2; P<.0001). More African American women had a proliferative endometrium. Both groups had a similar length of surveillance (11.9 vs 11.5 years; P=.27). Women with a proliferative endometrium had a higher risk of developing endometrial hyperplasia or cancer (11.9% vs 2.9%; P<.0001), any endometrial cancer (5.8% vs 1.8%; P=.002), atypical endometrial hyperplasia (2.2% vs 0.4%; P=.02), and nonatypical endometrial hyperplasia (2.0% vs 0.7%; P=.001). The risk of developing endometrial cancer and endometrial hyperplasia remained similar after excluding cases on hormonal replacement therapy (12.2% vs 3%; P=.001). On logistic regression analysis, proliferative endometrium histology (odds ratio, 3.89; 95% confidence interval, 2.03-7.49; P<.0001), age >60 years (odds ratio, 1.98; 95% confidence interval, 1.03-3.82; P=.04), and body mass index >35 kg/m2 (odds ratio, 2.3; 95% confidence interval, 1.09-4.83; P<.0001) remained significant risk factors for progression to cancer. CONCLUSION One of the 6 postmenopausal women who underwent endometrial sampling had a proliferative endometrium. Furthermore, 11.9% of women developed endometrial hyperplasia or cancer, a 4-fold greater incidence than women with an atrophic endometrium. The findings of this study suggest that long-term monitoring is warranted for women with postmenopausal bleeding and a proliferative endometrium histology. Further studies are needed to examine if a treatment is required to negate the risk of unopposed estrogen.
Collapse
|
24
|
Digging deeper through glucose metabolism and its regulators in cancer and metastasis. Life Sci 2020; 264:118603. [PMID: 33091446 DOI: 10.1016/j.lfs.2020.118603] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/04/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022]
Abstract
Glucose metabolism enzymes and transporters play major role in cancer development and metastasis. In this study, we discuss glucose metabolism, transporters, receptors, hormones, oncogenes and tumor suppressors which interact with glucose metabolism and we try to discuss their major role in cancer development and cancer metabolism. We try to highlight the. Metabolic changes in cancer and metastasis upregulation of glycolysis is observed in many primary and metastatic cancers and aerobic glycolysis is the most favorable mechanism for glucose metabolism in cancer cells, and it is a kind of evolutionary change. The question that is posed at this juncture is: Can we use aerobic glycolysis phenotype and enzymes beyond this mechanism in estimating cancer prognosis and metastasis? Lactate is a metabolite of glucose metabolism and it is a key player in cancer and metastasis in both normoxic and hypoxic condition so lactate dehydrogenase can be a good prognostic biomarker. Furthermore, monocarboxylic transporter which is the main lactate transporter can be good target in therapeutic studies. Glycolysis enzymes are valuable enzymes in cancer and metastasis diagnosis and can be used as therapeutic targets in cancer treatment. Designing a diagnostic and prognostic profile for cancer metastasis seems to be possible base on glycolysis enzymes and glucose transporters. Also, glucose metabolism enzymes and agents can give us a clear vision in estimating cancer metastasis. We can promote a panel of genes that detect genetic changes in glucose metabolism agents to diagnose cancer metastasis.
Collapse
|
25
|
Teng F, Ma X, Yu X, Yan Y, Zhao J, Gao J, Gao C, Wang Y, Tian W, Xue F. High serum Androgen and Insulin concentrations increase the tendency of Endometrial Carcinoma. J Cancer 2020; 11:5656-5664. [PMID: 32913460 PMCID: PMC7477453 DOI: 10.7150/jca.46391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/16/2020] [Indexed: 12/22/2022] Open
Abstract
Purpose: The objective of the study was to evaluate the important role played by androgen and insulin in the development of endometrial carcinoma (EC), and their combined effect on EC risk. Methods: We enrolled 510 type I EC patients and 510 age-, time-, and nationality-matched subjects into this study. Metabolic and hormonal parameters of enrolled subjects were examined. Univariate and multivariate logistic regression analyses for EC and control subjects were performed. Type I EC risk was evaluated with respect to testosterone, androstenedione, and insulin levels based on odds ratios (ORs) using stratified data. Results: EC risk was positively associated with C-peptide, estrone, androgen (including testosterone and androstenedione) and insulin levels, BMI, WHR, family history of cancer, nulliparity, irregular menstruation, diabetes, and hypertension. In multivariate logistic regression models, high C-peptide and testosterone levels, diabetes, and hypertension were independent risk factors after adjustment for BMI, WHR, family history of cancer, high serum insulin, and estrone levels. Increased serum total testosterone and insulin levels were positively correlated with EC risk in total, premenopausal, and postmenopausal women. Androstenedione was correlated with EC in total and postmenopausal, but not in premenopausal subjects. Compared with higher testosterone and insulin, odds ratios (ORs) for higher testosterone with lower insulin and lower testosterone with higher insulin were decreased in total, premenopausal, and postmenopausal women. Similarly, compared to both higher FAI and insulin, ORs for higher FAI with lower insulin and lower FAI with higher insulin were decreased in all three groups. Coordinately, ORs for higher androstenedione with lower insulin and lower androstenedione with higher insulin were decreased in total and postmenopausal, but not premenopausal subjects. Conclusions: These findings suggested that androgen and insulin were risk factors of type I EC, and relatively high levels of both testosterone and insulin synergistically affected EC risk.
Collapse
Affiliation(s)
- Fei Teng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaotong Ma
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaopei Yu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Ye Yan
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing Zhao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinping Gao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Chao Gao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenyan Tian
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
26
|
Li XC, Dong YY, Cheng Y, Zhou JY, Yang X, Shen BQ, Wu XT, Li XP, Wang JL. Increased Serum Calcium Level Promotes the Risk of Lymph Node Metastasis in Endometrial Cancer. Cancer Manag Res 2020; 12:5023-5030. [PMID: 32612389 PMCID: PMC7323808 DOI: 10.2147/cmar.s253914] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 05/25/2020] [Indexed: 12/17/2022] Open
Abstract
Purpose The early predictive values of diagnostic markers for lymph node metastasis (LNM) in endometrial cancer (EC) are still unclear at present. The purpose of this study is to explore the relationship between serum calcium and LNM in EC. Methods We identified all patients with EC who underwent surgery between January 2012 and December 2016. Patient characteristics and various preoperative clinicopathologic data were obtained from medical records and were reviewed retrospectively. These patients were divided into two groups according to the pathology of their lymph node. Logistic regression models analyzed the relationship between the ionized calcium and LNM of EC patients, while adjusting for the potential confounders. Results A total of 448 patients were assessed. Univariate analysis showed that ionized calcium, CA125 level, tumor grade, peritoneal cytology, FIGO stage, histological type, LVSI, and myometrial invasion were positively correlated with LNM (all P<0.05). The risk of LNM increased with the promotion of serum ionized calcium (P for trend <0.01). Ionized calcium level was significant before and after the adjustment of cofounders (unadjusted: OR=11.9, 95% CI: 4.8-29.6, P< 0.01; model I: OR=11.3, 95% CI: 4.5-28.8, P< 0.01; model II: OR=5.2, 95% CI: 1.6-17.2, P< 0.05). Additionally, the risk of ionized calcium was especially evident in patients whose age was older than 60, BMI<28 kg/m2, grade 3, negative peritoneal cytology and endometrioid endometrial adenocarcinoma. Conclusion Ionized calcium level was highly associated with LNM in EC and acted as a potential biomarker in predicting the risk of LNM in EC.
Collapse
Affiliation(s)
- Xing-Chen Li
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, People's Republic of China
| | - Yang-Yang Dong
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, People's Republic of China
| | - Yuan Cheng
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, People's Republic of China
| | - Jing-Yi Zhou
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, People's Republic of China.,Beijing Key Laboratory of Female Pelvic Floor Disorders Diseases, Beijing 100044, People's Republic of China
| | - Xiao Yang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, People's Republic of China
| | - Bo-Qiang Shen
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, People's Republic of China
| | - Xiao-Tong Wu
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, People's Republic of China.,Beijing Key Laboratory of Female Pelvic Floor Disorders Diseases, Beijing 100044, People's Republic of China
| | - Xiao-Ping Li
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, People's Republic of China
| | - Jian-Liu Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, People's Republic of China.,Beijing Key Laboratory of Female Pelvic Floor Disorders Diseases, Beijing 100044, People's Republic of China
| |
Collapse
|
27
|
Dana PM, Sadoughi F, Mobini M, Shafabakhsh R, Chaichian S, Moazzami B, Chamani M, Asemi Z. Molecular and Biological Functions of Melatonin in Endometrial Cancer. Curr Drug Targets 2020; 21:519-526. [DOI: 10.2174/1389450120666190927123746] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/01/2019] [Accepted: 09/12/2019] [Indexed: 12/15/2022]
Abstract
Endometrial cancer is the fifth leading cancer among women. This rate is higher in developed countries and its incidence is increasing worldwide. Diabetes mellitus, obesity, hypertension and arteriosclerosis are major risk factors for endometrial cancer. Melatonin is a hormone synthesized in the pineal and extra-pineal organs such as the digestive tract, bone marrow, retina and more. Evidence shows the potential effects of melatonin in endometrial cancer inhibition. Therefore, the focus of this paper is to review this outstanding evidence and to summarize the molecular and biological mechanisms of melatonin for the inhibition of endometrial cancer.
Collapse
Affiliation(s)
- Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Sadoughi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Moein Mobini
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Rana Shafabakhsh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Shala Chaichian
- Pars Advanced and Minimally Invasive Medical Manners Research Center, Pars Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Bahram Moazzami
- Pars Advanced and Minimally Invasive Medical Manners Research Center, Pars Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Chamani
- Department of Gynecology and Obstetrics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
28
|
Liu Y, Ma H, Yao J. ERα, A Key Target for Cancer Therapy: A Review. Onco Targets Ther 2020; 13:2183-2191. [PMID: 32210584 PMCID: PMC7073439 DOI: 10.2147/ott.s236532] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/20/2020] [Indexed: 12/18/2022] Open
Abstract
Estrogen receptor α (ERα) is closely associated with both hormone-dependent and hormone-independent tumors, and it is also essential for the development of these cancers. The functions of ERα are bi-faceted; it can contribute to cancer progression as well as cancer inhibition. Therefore, understanding ERα is vital for the treatment of those cancers that are closely associated with its expression. Here, we will elaborate on ERα based on its structure, localization, activation, modification, and mutation. Also, we will look at co-activators of ERα, elucidate the signaling pathway activated by ERα, and identify cancers related to its activation. A comprehensive understanding of ERα could help us to find new ways to treat cancers.
Collapse
Affiliation(s)
- Yanfang Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Hong Ma
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Jing Yao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| |
Collapse
|