Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 459
Filter
2.
Front Immunol ; 15: 1393852, 2024.
Article in English | MEDLINE | ID: mdl-38711526

ABSTRACT

Different eukaryotic cell organelles (e.g., mitochondria, endoplasmic reticulum, lysosome) are involved in various cancer processes, by dominating specific cellular activities. Organelles cooperate, such as through contact points, in complex biological activities that help the cell regulate energy metabolism, signal transduction, and membrane dynamics, which influence survival process. Herein, we review the current studies of mechanisms by which mitochondria, endoplasmic reticulum, and lysosome are related to the three major malignant gynecological cancers, and their possible therapeutic interventions and drug targets. We also discuss the similarities and differences of independent organelle and organelle-organelle interactions, and their applications to the respective gynecological cancers; mitochondrial dynamics and energy metabolism, endoplasmic reticulum dysfunction, lysosomal regulation and autophagy, organelle interactions, and organelle regulatory mechanisms of cell death play crucial roles in cancer tumorigenesis, progression, and response to therapy. Finally, we discuss the value of organelle research, its current problems, and its future directions.


Subject(s)
Genital Neoplasms, Female , Mitochondria , Organelles , Humans , Female , Genital Neoplasms, Female/pathology , Genital Neoplasms, Female/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Organelles/metabolism , Cell Survival , Animals , Lysosomes/metabolism , Endoplasmic Reticulum/metabolism , Autophagy , Energy Metabolism , Signal Transduction
3.
Br J Cancer ; 130(11): 1875-1884, 2024 May.
Article in English | MEDLINE | ID: mdl-38582811

ABSTRACT

BACKGROUND: Other than for breast cancer, endocrine therapy has not been highly effective for gynecologic cancers. Endocrine therapy resistance in estrogen receptor positive gynecologic cancers is still poorly understood. In this retrospective study, we examined the estrogen receptor (ER) signaling pathway activities of breast, ovarian, endometrial, and cervical cancers to identify those that may predict endocrine therapy responsiveness. METHODS: Clinical and genomic data of women with breast and gynecological cancers were downloaded from cBioPortal for Cancer Genomics. Estrogen receptor alpha (ESR1) expression level and sample-level pathway enrichment scores (EERES) were calculated to classify patients into four groups (low/high ESR1 and low/high EERES). Correlation between ESR1/EERES score and survival was further validated with RNAseq data from low-grade serous ovarian cancer. Pathway analyses were performed among different ESR1/EERES groups to identify genes that correlate with endocrine resistance, which are validated using Cancer Cell Line Encyclopedia gene expression and Genomics of Drug Sensitivity in Cancer data. RESULTS: We identified a novel combined prognostic value of ESR1 expression and the corresponding estrogen response signaling (EERES score) for breast cancer. The combined prognostic value (ESR1/EERES) may be applicable to other gynecologic cancers. More importantly, we discovered that ER signaling can cross-regulate MEK pathway activation. We identified downstream genes in the MEK pathway (EPHA2, INAVA, MALL, MPZL2, PCDH1, and TNFRSF21) that are potential endocrine therapy response biomarkers. CONCLUSION: This study demonstrated that targeting both the ER and the ER signaling activity related MEK pathway may aid the development of endocrine therapy strategies for personalized medicine.


Subject(s)
Estrogen Receptor alpha , Humans , Female , Prognosis , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Retrospective Studies , MAP Kinase Signaling System/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Drug Resistance, Neoplasm/genetics , Signal Transduction , Gene Expression Regulation, Neoplastic , Antineoplastic Agents, Hormonal/therapeutic use , Cell Line, Tumor , Genital Neoplasms, Female/genetics , Genital Neoplasms, Female/drug therapy , Genital Neoplasms, Female/metabolism , Genital Neoplasms, Female/pathology , Genital Neoplasms, Female/mortality , Ovarian Neoplasms/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/mortality
4.
Curr Pharm Des ; 30(5): 352-361, 2024.
Article in English | MEDLINE | ID: mdl-38303530

ABSTRACT

Gynecologic cancers are among the most common malignancies with aggressive features and poor prognosis. Tumorigenesis in gynecologic cancers is a complicated process that is influenced by multiple factors, including genetic mutations that activate various oncogenic signaling pathways, including the TGF-ß pathway. Aberrant activation of TGF-ß signaling is correlated with tumor recurrence and metastasis. It has been shown that non-coding RNAs (ncRNAs) have crucial effects on cancer cell proliferation, migration, and metastasis. Upregulation of various ncRNAs, including long non-coding RNAs (lncRNA) and microRNAs (miRNAs), has been reported in several tumors, like cervical, ovarian, and endometrial cancers, but their cellular mechanisms remain to be investigated. Thus, recognizing the role of ncRNAs in regulating the TGF-ß pathway may provide novel strategies for better treatment of cancer patients. The present study summarizes recent findings on the role of ncRNAs in regulating the TGF-ß signaling involved in tumor progression and metastasis in gynecologic cancers.


Subject(s)
Genital Neoplasms, Female , MicroRNAs , RNA, Long Noncoding , Signal Transduction , Transforming Growth Factor beta , Humans , Female , Transforming Growth Factor beta/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Genital Neoplasms, Female/genetics , Genital Neoplasms, Female/pathology , Genital Neoplasms, Female/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Carcinogenesis/genetics , Carcinogenesis/pathology , Animals
6.
Cell Transplant ; 32: 9636897231195240, 2023.
Article in English | MEDLINE | ID: mdl-37632354

ABSTRACT

Exosomes are phospholipid bilayer vesicles that are released by all types of cells, containing proteins, lipids, and nucleic acids such as DNAs and RNAs. Exosomes can be transferred between cells and play a variety of physiological and pathological regulatory functions. Noncoding RNAs, including micro RNAs, long noncoding RNAs, and circular RNAs, are the most studied biomolecules from exosomes and more and more studies found that noncoding RNAs play an important role in the diagnosis, prognosis, and treatment of diseases, including various types of cancer. Gynecological malignancies such as ovarian, endometrial, and cervical cancer seriously threaten women's life. Therefore, this article reviews the roles and applications of exosomes in gynecological malignancies, including the promotion or inhibition of tumor progression and regulation of tumor microenvironments, and as potential therapeutic targets for treating gynecological cancers.


Subject(s)
Exosomes , Genital Neoplasms, Female , MicroRNAs , Neoplasms , RNA, Long Noncoding , Humans , Female , Exosomes/metabolism , Genital Neoplasms, Female/metabolism , Genital Neoplasms, Female/pathology , Neoplasms/metabolism , MicroRNAs/metabolism , RNA, Untranslated/metabolism , RNA, Long Noncoding/metabolism , Tumor Microenvironment
7.
Int J Oncol ; 62(5)2023 May.
Article in English | MEDLINE | ID: mdl-37052244

ABSTRACT

Gynecological malignancies are a leading cause of mortality among females worldwide, and difficulties in early diagnosis and acquired drug resistance constitute obstacles to effective therapies. Ovarian cancer causes more deaths than any other cancer of the female reproductive system. Specifically, in females aged 20 to 39 years, cervical cancer is the third leading cause of cancer­related mortality, and the incidence rates of cervical adenocarcinoma are increasing. Endometrial carcinoma is the most common gynecological cancer in developed countries, such as the United States. Vulvar cancer and uterine sarcomas are considered rare, and therefore require further investigation. Notably, the development of novel treatment options is critical. Previous research has revealed metabolic reprogramming as a distinct feature of tumor cells, which includes aerobic glycolysis. In this instance, cells produce adenosine triphosphate and various precursor molecules through glycolysis, despite oxygen levels being sufficient. This is to meet the energy required for rapid DNA replication. This phenomenon is also known as the Warburg effect. The Warburg effect results in an increased glucose uptake, lactate production and reduced pH values in tumor cells. The results of previous studies have demonstrated that microRNAs (miRNAs/miRs) regulate glycolysis, and participate in tumorigenesis and tumor progression via interactions with glucose transporters, essential enzymes, tumor suppressor genes, transcription factors and multiple cellular signaling pathways that play critical roles in glycolysis. Notably, miRNAs affect the levels of glycolysis in ovarian, cervical and endometrial cancers. The present review article provides a comprehensive overview of the literature surrounding miRNAs in the glycolysis of gynecological malignant cells. The present review also aimed to determine the role of miRNAs as potential therapeutic options rather than diagnostic markers.


Subject(s)
Genital Neoplasms, Female , MicroRNAs , Ovarian Neoplasms , Uterine Cervical Neoplasms , Humans , Female , MicroRNAs/metabolism , Genital Neoplasms, Female/genetics , Genital Neoplasms, Female/metabolism , Genital Neoplasms, Female/pathology , Glycolysis/genetics
8.
Appl Biochem Biotechnol ; 194(1): 323-338, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34822059

ABSTRACT

Different metabolic and hormonal disorders like type 2 diabetes mellitus (T2DM), obesity, and polycystic ovary syndrome (PCOS) have tangible socio-economic impact. Prevalence of these metabolic and hormonal disorders is steadily increasing among women. There are clinical evidences that these physiological conditions are related to the manifestation of different gynecological cancers and their poor prognosis. The relationship between metabolic and hormonal disorders with gynecological cancers is quite complex. The need for gene level association study is extremely important to find markers and predicting risk factors. In the current work, we have selected metabolic disorders like T2DM and obesity, hormonal disorder PCOS, and 4 different gynecological cancers like endometrial, uterine, cervical, and triple negative breast cancer (TNBC). The gene list was downloaded from DisGeNET database (v 6.0). The protein interaction network was constructed using HIPPIE (v 2.2) and shared proteins were identified. Molecular comorbidity index and Jaccard coefficient (degree of similarity) between the diseases were determined. Pathway enrichment analysis was done using ReactomePA and significant modules (clusters in a network) of the constructed network was analyzed by MCODE plugin of Cytoscape. The comorbid conditions like PCOS-obesity found to increase the risk factor of ovarian and triple negative breast cancers whereas PCOS alone has highest contribution to the endometrial cancer. Different gynecological cancers were found to be differentially related to the metabolic/hormonal disorders and comorbid condition.


Subject(s)
Breast Neoplasms/metabolism , Diabetes Mellitus, Type 2/metabolism , Genital Neoplasms, Female/metabolism , Models, Biological , Polycystic Ovary Syndrome/metabolism , Breast Neoplasms/pathology , Diabetes Mellitus, Type 2/pathology , Female , Genital Neoplasms, Female/pathology , Humans , Polycystic Ovary Syndrome/pathology
9.
Front Endocrinol (Lausanne) ; 12: 747810, 2021.
Article in English | MEDLINE | ID: mdl-34745013

ABSTRACT

Prolactin (PRL) is a hormone produced by the pituitary gland and multiple non-pituitary sites, vital in several physiological processes such as lactation, pregnancy, cell growth, and differentiation. However, PRL is nowadays known to have a strong implication in oncogenic processes, making it essential to delve into the mechanisms governing these actions. PRL and its receptor (PRLR) activate a series of effects such as survival, cellular proliferation, migration, invasion, metastasis, and resistance to treatment, being highly relevant in developing certain types of cancer. Because women produce high levels of PRL, its influence in gynecological cancers is herein reviewed. It is interesting that, other than the 23 kDa PRL, whose mechanism of action is endocrine, other variants of PRL have been observed to be produced by tumoral tissue, acting in a paracrine/autocrine manner. Because many components, including PRL, surround the microenvironment, it is interesting to understand the hormone's modulation in cancer cells. This work aims to review the most important findings regarding the PRL/PRLR axis in cervical, ovarian, and endometrial cancers and its molecular mechanisms to support carcinogenesis.


Subject(s)
Cell Transformation, Neoplastic , Genital Neoplasms, Female/pathology , Prolactin/physiology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Female , Genital Neoplasms, Female/metabolism , Humans , Prolactin/metabolism , Receptors, Prolactin/metabolism , Receptors, Prolactin/physiology , Signal Transduction/physiology , Tumor Microenvironment/physiology
10.
Gynecol Oncol ; 162(3): 619-625, 2021 09.
Article in English | MEDLINE | ID: mdl-34272090

ABSTRACT

OBJECTIVES: To estimate the maximally tolerated dose (MTD) and describe toxicities associated with lenvatinib and weekly paclitaxel in patients with recurrent endometrial and platinum resistant epithelial ovarian cancer. METHODS: Using a 3 + 3 design patients were given weekly paclitaxel 80 mg/m2 IV day 1, 8, 15 and oral levantinib daily on a 28-day cycle. Lenvatinib dose levels were 8 mg, 12 mg, 16 mg, 20 mg. Toxicities were recorded using CTCAE v4.03 and response was determined with imaging after cycle 2, then every 3rd cycle, using RECIST 1.1 criteria. RESULTS: 26 patients were enrolled; 19 with ovarian cancer (14 high grade serous, 1 low grade serous, 2 clear cell, 1 endometrioid, and 1 carcinosarcoma), and 7 with endometrial cancer (3 serous, and 4 endometrioid). The MTD was established at lenvatinib 16 mg and weekly paclitaxel 80 mg/m2. Toxicities (all grades) occurring in ≥25% of patients included anemia, neutropenia, lymphopenia, mucositis, nausea, diarrhea, anorexia, hypertension, fatigue, proteinuria, epistaxis, hoarseness. Twenty-three patients were evaluable for response and PFS; 15 (65%) had a partial response, 7 (30%) stable, 1 (4%) progressive disease with an objective response rate of 65%; 71% in ovarian and 50% in endometrial cancer. Median progression free survival (PFS) is 12.4 months; 14.0 months in endometrial cancer, 7.2 months in ovarian cancer; 54% had a PFS > 6 months. The median duration of response for PR patients (n = 15) was 10.9 months. CONCLUSIONS: The regimen was tolerable with manageable side effects. Encouraging activity was observed in endometrial and ovarian cancer, and warrants further development.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Genital Neoplasms, Female/drug therapy , Peritoneal Neoplasms/drug therapy , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Carcinoma, Ovarian Epithelial/drug therapy , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Resistance, Neoplasm , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/metabolism , Fallopian Tube Neoplasms/drug therapy , Fallopian Tube Neoplasms/metabolism , Female , Genital Neoplasms, Female/metabolism , Humans , Middle Aged , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Paclitaxel/pharmacokinetics , Peritoneal Neoplasms/metabolism , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/adverse effects , Phenylurea Compounds/pharmacokinetics , Quinolines/administration & dosage , Quinolines/adverse effects , Quinolines/pharmacokinetics
11.
Curr Oncol Rep ; 23(8): 92, 2021 06 14.
Article in English | MEDLINE | ID: mdl-34125319

ABSTRACT

PURPOSE OF REVIEW: We review the emerging evidence regarding the relationship between the microbiota of the gastrointestinal and female reproductive tracts and gynecologic cancer. RECENT FINDINGS: The microbiome has essential roles in maintaining health. In recent years, the microbiota of the gastrointestinal and female reproductive tracts have been linked to many diseases, including gynecologic cancer. Alterations to the bacterial populations in a microbiota, or dysbiosis, have been shown to favor a pro-carcinogenic state through altered immune responses, dysregulated hormone metabolism, and modulation of the cell cycle. Pre-clinical and clinical studies have emerged, demonstrating that specific bacteria or microbial communities may be associated with increased risk for uterine, ovarian, and cervical cancers. Notably, numerous studies have linked a non-Lactobacillus-dominant vaginal microbiota, composed of anaerobic bacteria, with HPV infection, persistence, and development of invasive cervical cancer. Similarly, next-generation high-throughput sequencing techniques have enabled the characterization of unique microbiotas in patients with malignant and benign gynecologic conditions, shedding light on new associations between bacterial species and gynecologic cancers. Harnessing the power of the microbiome for early diagnosis, therapeutic intervention and modulation creates tremendous potential to optimize gynecologic cancer outcomes in the future.


Subject(s)
Gastrointestinal Microbiome , Genital Neoplasms, Female/microbiology , Genitalia, Female/microbiology , Female , Gastrointestinal Diseases/microbiology , Gastrointestinal Tract/microbiology , Genital Neoplasms, Female/metabolism , Genital Neoplasms, Female/therapy , Genitalia, Female/metabolism , Humans
12.
Br J Cancer ; 125(6): 789-797, 2021 09.
Article in English | MEDLINE | ID: mdl-34099896

ABSTRACT

BAG3, a member of the BAG family of co-chaperones, is a multidomain protein with a role in several cellular processes, including the control of apoptosis, autophagy and cytoskeletal dynamics. The expression of bag3 is negligible in most cells but can be induced by stress stimuli or malignant transformation. In some tumours, BAG3 has been reported to promote cell survival and resistance to therapy. The expression of BAG3 has been documented in ovarian, endometrial and cervical cancers, and studies have revealed biochemical and functional connections of BAG3 with proteins involved in the survival, invasion and resistance to therapy of these malignancies. BAG3 expression has also been shown to correlate with the grade of dysplasia in squamous intraepithelial lesions of the uterine cervix. Some aspects of BAG3 activity, such as its biochemical and functional interaction with the human papillomavirus proteins, could help in our understanding of the mechanisms of oncogenesis induced by the virus. This review aims to highlight the potential value of BAG3 studies in the field of gynaecological tumours.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Drug Resistance, Neoplasm , Genital Neoplasms, Female/metabolism , Female , Gene Expression Regulation, Neoplastic , Genital Neoplasms, Female/drug therapy , Genital Neoplasms, Female/pathology , Humans , Neoplasm Grading , Neoplasm Invasiveness , Survival Analysis
13.
Nat Commun ; 12(1): 2592, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33972543

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is the most commonly cited mechanism for cancer metastasis, but it is difficult to distinguish from profiles of normal stromal cells in the tumour microenvironment. In this study we use published single cell RNA-seq data to directly compare mesenchymal signatures from cancer and stromal cells. Informed by these comparisons, we developed a computational framework to decouple these two sources of mesenchymal expression profiles using bulk RNA-seq datasets. This deconvolution offers the opportunity to characterise EMT across hundreds of tumours and examine its association with metastasis and other clinical features. With this approach, we find three distinct patterns of EMT, associated with squamous, gynaecological and gastrointestinal cancer types. Surprisingly, in most cancer types, EMT patterns are not associated with increased chance of metastasis, suggesting that other steps in the metastatic cascade may represent the main bottleneck. This work provides a comprehensive evaluation of EMT profiles and their functional significance across hundreds of tumours while circumventing the confounding effect of stromal cells.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Lymphatic Metastasis/genetics , Neoplasms/metabolism , Neoplasms/pathology , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Genital Neoplasms, Female/genetics , Genital Neoplasms, Female/metabolism , Genital Neoplasms, Female/pathology , Humans , Neoplasm Grading , Neoplasms/genetics , Neoplasms, Squamous Cell/genetics , Neoplasms, Squamous Cell/metabolism , Neoplasms, Squamous Cell/pathology , RNA-Seq , Single-Cell Analysis , Stromal Cells/metabolism , Stromal Cells/pathology
14.
Mol Cell Biochem ; 476(8): 3177-3190, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33864572

ABSTRACT

Melatonin (N-acetyl-5-methoxytryptamine), the main product of pineal gland in vertebrates, is well known for its multifunctional role which has great influences on the reproductive system. Recent studies documented that melatonin is a powerful free radical scavenger that affects the reproductive system function and female infertility by MT1 and MT2 receptors. Furthermore, cancer researches indicate the influence of melatonin on the modulation of tumor cell signaling pathways resulting in growth inhibitor of the both in vivo/in vitro models. Cancer adjuvant therapy can also benefit from melatonin through therapeutic impact and decreasing the side effects of radiation and chemotherapy. This article reviews the scientific evidence about the influence of melatonin and its mechanism of action on the fertility potential, physiological alteration, and anticancer efficacy, during experimental and clinical studies.


Subject(s)
Genital Neoplasms, Female/drug therapy , Genitalia, Female/drug effects , Melatonin/pharmacology , Animals , Female , Genital Neoplasms, Female/metabolism , Genital Neoplasms, Female/pathology , Genitalia, Female/metabolism , Humans
15.
Int J Mol Med ; 47(6)2021 Jun.
Article in English | MEDLINE | ID: mdl-33907821

ABSTRACT

Cancers of the urinary tract, as well as those of the female and male reproductive systems, account for a large percentage of malignancies worldwide. Mortality is frequently affected by late diagnosis or therapeutic difficulties. The Sonic Hedgehog (SHH) pathway is an evolutionary conserved molecular cascade, which is mainly associated with the development of the central nervous system in fetal life. The present review aimed to provide an in­depth summary of the SHH signaling pathway, including the characterization of its major components, the mechanism of its upstream regulation and non­canonical activation, as well as its interactions with other cellular pathways. In addition, the three possible mechanisms of the cellular SHH cascade in cancer tissue are discussed. The aim of the present review was to summarize significant findings with regards to the expression of the SHH pathway components in kidney, bladder, ovarian, cervical and prostate cancer. Reports associated with common deficits and de­regulations of the SHH pathway were summarized, despite the differences in molecular and histological patterns among these malignancies. However, currently, neither are SHH pathway elements included in panels of prognostic/therapeutic molecular patterns in any of the discussed cancers, nor have the drugs targeting SMO or GLIs been approved for therapy. The findings of the present review may support future studies on the treatment of and/or molecular targets for gynecological and genitourinary cancers.


Subject(s)
Genital Neoplasms, Female/metabolism , Hedgehog Proteins/metabolism , Signal Transduction , Urogenital Neoplasms/metabolism , Female , Genital Neoplasms, Female/genetics , Hedgehog Proteins/genetics , Humans , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Signal Transduction/genetics , Tumor Microenvironment/genetics , Urogenital Neoplasms/genetics
16.
Biochim Biophys Acta Rev Cancer ; 1875(2): 188525, 2021 04.
Article in English | MEDLINE | ID: mdl-33600824

ABSTRACT

Gynecological cancers are characterized by a high mortality rate when chemoresistance develops. Autophagy collaborates with apoptosis and participates in homeostasis of chemoresistance. Recent findings supported that crosstalk of necrotic, apoptotic and autophagic factors, and chemotherapy-driven hypoxia, oxidative stress and ER stress play critical roles in chemoresistance in gynecological cancers. Meanwhile, current studies have shown that autophagy could be regulated by and cooperate with metabolic regulator, survival factors, stemness factors and specific post-translation modification in chemoresistant tumor cells. Meanwhile, non-coding RNA and autophagy crosstalk also contribute to the chemoresistance. Until now, analysis of individual autophagy factors towards the clinical significance and chemoresistance in gynecological cancer is still lacking. We suggest comprehensive integrated analysis of cellular homeostasis and tumor microenvironment to clarify the role of autophagy and the associated factors in cancer progression and chemoresistance. Panel screening of pan-autophagic factors will pioneer the development of risk models for predicting efficacy of chemotherapy and guidelines for systematic treatment and precision medicine.


Subject(s)
Autophagy , Drug Resistance, Neoplasm , Genital Neoplasms, Female/metabolism , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Oxidative Stress , Signal Transduction , Tumor Microenvironment
17.
Reprod Sci ; 28(9): 2436-2447, 2021 09.
Article in English | MEDLINE | ID: mdl-33569749

ABSTRACT

Long non-coding RNAs (lncRNAs) are emerging regulators of cellular pathways, especially in cancer development. Among the lncRNAs, nuclear paraspeckle assembly transcript 1 (NEAT1) forms a scaffold for a nuclear body; the paraspeckle and aberrant expression of NEAT1 have been reported in breast and gynecologic cancers (ovarian, cervical, endometrial, and vulvar). Abundantly expressed NEAT1 in breast and gynecologic cancers generally contribute to tumor development by sponging its corresponding tumor-suppressive microRNAs or interacting with various regulatory proteins. The distinct expression of NEAT1 and its contribution to tumorigenic pathways make it a promising therapeutic target in breast and gynecologic cancers. Herein, we summarize the functions and molecular mechanisms of NEAT1 in human breast, ovarian, cervical, endometrial, and vulvar cancers. Furthermore, we emphasize its critical role in the formation of paraspeckle development and its functions. Conclusively, NEAT1 is a considerable biomarker with a bright prospect and can be therapeutically targeted to manage breast and gynecologic cancers.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Genital Neoplasms, Female/metabolism , RNA, Long Noncoding/metabolism , Animals , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Clinical Decision-Making , Female , Gene Expression Regulation, Neoplastic , Genital Neoplasms, Female/drug therapy , Genital Neoplasms, Female/genetics , Genital Neoplasms, Female/pathology , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Precision Medicine , Predictive Value of Tests , Prognosis , RNA, Long Noncoding/genetics , Signal Transduction , Uterine Neoplasms/drug therapy , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology , Vulvar Neoplasms/drug therapy , Vulvar Neoplasms/genetics , Vulvar Neoplasms/metabolism , Vulvar Neoplasms/pathology
18.
Gynecol Oncol ; 161(1): 314-327, 2021 04.
Article in English | MEDLINE | ID: mdl-33581845

ABSTRACT

Gynecologic cancer is a group of any malignancies affecting reproductive tissues and organs of women, including ovaries, uterine, cervix, vagina, vulva, and endometrium. Several types of molecular mechanisms are associated with the progression of gynecologic cancers. Among it can be referred to the most widely studied non-coding RNAs (ncRNAs), specifically microRNAs (miRNAs) and long ncRNAs (lncRNAs). As yet, lncRNAs are known to serve key biological roles via various mechanisms, such as splicing regulation, chromatin rearrangement, translation regulation, cell-cycle control, genetic imprinting and mRNA decay. Besides, miRNAs govern gene expression by modulation of mRNAs and lncRNAs degradation, suggestive of needing more research in this field. Generally, driving gynecological cancers pathways by miRNAs and lncRNAs lead to the current improvement in cancer-related technologies. Exosomes are extracellular microvesicles which can carry cargo molecules among cells. In recent years, more studies have been focused on exosomal non-coding RNAs (exo-ncRNAs) and exosomal microRNAs (exo-miRs) because of being natural carriers of lnc RNAs and microRNAs via programmed process. In this review we summarized recent reports concerning the function of exosomal microRNAs and exosomal long non-coding RNAs in gynecological cancers.


Subject(s)
Exosomes/genetics , Genital Neoplasms, Female/genetics , RNA, Long Noncoding/genetics , Exosomes/metabolism , Female , Genital Neoplasms, Female/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism
19.
Sci Rep ; 11(1): 2257, 2021 01 26.
Article in English | MEDLINE | ID: mdl-33500424

ABSTRACT

We investigated the prognostic significance and the underlying mechanism of increased bone marrow (BM) 2-(18F) fluoro-2-deoxy-D-glucose as a tracer (FDG)-uptake in patients with gynecological cancer. A list of patients diagnosed with cervical, endometrial, and ovarian cancer from January 2008 to December 2014 were identified. Then, through chart reviews, 559 patients who underwent staging by FDG-positron emission tomography (PET)/computed tomography (CT) and subsequent surgical resection were identified, and their clinical data were reviewed retrospectively. BM FDG-uptake was evaluated using maximum standardized uptake value (SUVmax) and BM-to-aorta uptake ratio (BAR). As a result, we have found that increased BAR was observed in 20 (8.7%), 21 (13.0%), 21 (12.6%) of cervical, endometrial, and ovarian cancer, respectively, and was associated with significantly shorter survival. Increased BAR was also closely associated with increased granulopoiesis. In vitro and in vivo experiments revealed that tumor-derived granulocyte colony-stimulating factor (G-CSF) was involved in the underlying causative mechanism of increased BM FDG-uptake, and that immune suppression mediated by G-CSF-induced myeloid-derived suppressor cells (MDSCs) is responsible for the poor prognosis of this type of cancer. In conclusion, increased BM FDG-uptake, as represented by increased BAR, is an indicator of poor prognosis in patients with gynecological cancer.


Subject(s)
Bone Marrow/metabolism , Fluorodeoxyglucose F18/pharmacokinetics , Genital Neoplasms, Female/metabolism , Adult , Aged , Aged, 80 and over , Animals , Aorta/metabolism , Cell Line, Tumor , Female , Genital Neoplasms, Female/diagnostic imaging , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Mice, Inbred C57BL , Middle Aged , Multivariate Analysis , Myeloid-Derived Suppressor Cells/metabolism , Positron Emission Tomography Computed Tomography , Prognosis , Progression-Free Survival , Rats
20.
Histopathology ; 79(2): 160-167, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33432679

ABSTRACT

AIMS: Genomic inactivation of ARID1B in ARID1A-inactivated tumour and genomic inactivation of SMARCB1 represent two recurrent mechanisms, core SWItch/sucrose non-fermentable (SWI/SNF) complex inactivation, that are associated with de-differentiation in endometrial carcinoma. Approximately one-third of dedifferentiated/undifferentiated endometrial carcinomas (DDEC/UEC) show loss of ARID1B expression with a minor subset showing loss of SMARCB1 expression, but little is known regarding the specificity of ARID1B or SMARCB1 loss in gynaecological tract tumours in general. The aim of this study was to examine the frequency of ARID1B and SMARCB1 loss by immunohistochemistry in a series of gynaecological tract epithelial/mesenchymal neoplasms. METHODS AND RESULTS: We evaluated 1849 tumours that included 748 endometrial carcinomas, 101 uterine carcinosarcomas/adenosarcomas, 64 uterine sarcomas, 221 cervical carcinomas and 715 ovarian carcinomas/borderline tumours by tissue microarrays (TMA). We observed ARID1B loss in 35 of 86 (41%) and SMARCB1 loss in three of 86 (3%) DDEC/UEC, but not in any other uterine tumour types examined. ARID1B-deficient DDEC/UEC also showed concurrent loss of ARID1A expression. All SMARCB1-deficient tumours showed loss of MLH1 and PMS2, while 29 of 35 ARID1B-deficient tumours showed loss of MLH1 and PMS2 or loss of MSH6. All ovarian carcinomas/borderline tumours and cervical carcinomas showed intact expression of ARID1B and SMARCB1. CONCLUSION: Our findings indicate that the loss of expression of ARID1B or SMARCB1 by immunohistochemistry is highly specific for undifferentiated carcinoma among tumours of the upper gynaecological tract and cervix, and therefore can be used to identify these highly aggressive malignant tumours.


Subject(s)
Carcinoma/diagnosis , Cell Dedifferentiation , DNA-Binding Proteins/deficiency , Genital Neoplasms, Female/diagnosis , SMARCB1 Protein/deficiency , Transcription Factors/deficiency , Biomarkers, Tumor/metabolism , Carcinoma/metabolism , Carcinoma/pathology , Cell Dedifferentiation/genetics , Cohort Studies , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Genital Neoplasms, Female/genetics , Genital Neoplasms, Female/metabolism , Genital Neoplasms, Female/pathology , Humans , Immunohistochemistry , SMARCB1 Protein/genetics , SMARCB1 Protein/metabolism , Sarcoma/diagnosis , Sarcoma/metabolism , Sarcoma/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...