Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters











Publication year range
1.
Biomedicines ; 11(10)2023 Sep 23.
Article in English | MEDLINE | ID: mdl-37892992

ABSTRACT

We investigated whether anogenital distance (AGD) is associated with adenomyosis, endometriosis and uterine leiomyomas (UL, also called uterine fibroids). We recruited 81 women with UL, 105 with ovarian endometrioma (OE), 116 with adenomyosis, 28 with both adenomyosis and UL, and 100 control subjects with other acquired gynecological conditions but not endometriosis, adenomyosis, UL, or polycystic ovarian syndrome. Measurements from the anterior clitoral surface to the center of the anus (AGDAC), from the tip of the clitoris to the center of the anus (AGDACt), and from the posterior fourchette to the center of the anus (AGDAF) were made in all subjects. Multiple regression was performed to estimate the association between AGDs and presence of OE, adenomyosis, and UL while controlling for possible confounding factors. We found that, compared with controls, women with OE and adenomyosis, but not UL, had significantly shorter AGDAF, but not AGDAC. However, the amount of variance that could be explained by the disease status is rather moderate, suggesting that factors other than disease status, bodyweight and height were also responsible for AGD. Thus, prenatal exposure to reduced levels of androgen may increase the risk of developing endometriosis and adenomyosis. However, other factors may also contribute to the pathogenesis of endometriosis and adenomyosis.

3.
Biotechnol Genet Eng Rev ; : 1-11, 2023 Jan 04.
Article in English | MEDLINE | ID: mdl-36600526

ABSTRACT

With the continuous innovation of digital technology in my country at this stage and the design of medical imaging technology systems, the depth and breadth of the development of digital medical imaging technology have been greatly expanded. This paper focus on application of medical images in breast milk smearing of the umbilical nursing for full-term newborns. OBJECTIVE: To explore the effect of breast milk application in umbilical nursing of full-term newborns. METHODS: 596 full-term newborns were divided into three groups: Experimental Group, Control Group A and Control Group B, Experimental Group A treated with breast milk, control group a treated with 75% alcohol, and control group B treated with 37 ~ 42°C warm boiled water, the time of umbilical cord abscission, infection and other complications were compared among the three groups. The process was recorded by images. RESULTS: According to the images, compared with the Control Group A and B, the experimental group significantly shortened the time of umbilical cord shedding, the difference was statistically significant (p < 0.001). CONCLUSION: The application of breast milk in umbilical region of full-term neonates can reduce the time of umbilical cord abscission without increasing the incidence of Omphalitis.

4.
Trends Cancer ; 9(1): 69-82, 2023 01.
Article in English | MEDLINE | ID: mdl-36184492

ABSTRACT

Development of cancer therapeutics has traditionally focused on targeting driver oncogenes. Such an approach is limited by toxicity to normal tissues and treatment resistance. A class of 'cancer fitness genes' with crucial roles in metastasis have been identified. Elevated or altered activities of these genes do not directly cause cancer; instead, they relieve the stresses that tumor cells encounter and help them adapt to a changing microenvironment, thus facilitating tumor progression and metastasis. Importantly, as normal cells do not experience high levels of stress under physiological conditions, targeting cancer fitness genes is less likely to cause toxicity to noncancerous tissues. Here, we summarize the key features and function of cancer fitness genes and discuss their therapeutic potential.


Subject(s)
Neoplasms , Oncogenes , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Tumor Microenvironment/genetics
5.
J Med Chem ; 65(18): 12188-12199, 2022 09 22.
Article in English | MEDLINE | ID: mdl-36044768

ABSTRACT

Blocking the interaction of MTDH/SND1 complex is an attractive strategy for cancer therapeutics. In this work, we designed and obtained a novel class of potent stabilized peptide inhibitors derived from MTDH sequence to disrupt MTDH/SND1 interaction. Through structure-based optimization and biological evaluation, stabilized peptides were obtained with tight binding affinity, improved cell penetration, and antitumor effects in the triple-negative breast cancer (TNBC) cells without nonspecific toxicity. To date, our study was the first report to demonstrate that stabilized peptides truncated from MTDH could serve as promising candidates to disrupt the MTDH/SND1 interaction for potential breast cancer treatment.


Subject(s)
Breast Neoplasms , Triple Negative Breast Neoplasms , Breast Neoplasms/pathology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Endonucleases/metabolism , Female , Humans , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Peptides/metabolism , Peptides/pharmacology , RNA-Binding Proteins , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism
6.
Front Med (Lausanne) ; 9: 869010, 2022.
Article in English | MEDLINE | ID: mdl-35402443

ABSTRACT

Colorectal cancer (CRC) is one of the most devastating diseases that accounts for numerous deaths worldwide. Tumor cell-autonomous pathways, such as the oncogenic signaling activation, significantly contribute to CRC progression and metastasis. Recent accumulating evidence suggests that the CRC microenvironment also profoundly promotes or represses this process. As the roles of the tumor microenvironment (TME) in CRC progression and metastasis is gradually uncovered, the importance of these non-cell-autonomous signaling pathways is appreciated. However, we are still at the beginning of this TME function exploring process. In this review, we summarize the current understanding of the TME in CRC progression and metastasis by focusing on the gut microbiota and host cellular and non-cellular components. We also briefly discuss TME-remodeling therapies in CRC.

7.
Nat Cancer ; 3(3): 355-370, 2022 03.
Article in English | MEDLINE | ID: mdl-35301507

ABSTRACT

Ligand-dependent corepressor (LCOR) mediates normal and malignant breast stem cell differentiation. Cancer stem cells (CSCs) generate phenotypic heterogeneity and drive therapy resistance, yet their role in immunotherapy is poorly understood. Here we show that immune-checkpoint blockade (ICB) therapy selects for LCORlow CSCs with reduced antigen processing/presentation machinery (APM) driving immune escape and ICB resistance in triple-negative breast cancer (TNBC). We unveil an unexpected function of LCOR as a master transcriptional activator of APM genes binding to IFN-stimulated response elements (ISREs) in an IFN signaling-independent manner. Through genetic modification of LCOR expression, we demonstrate its central role in modulation of tumor immunogenicity and ICB responsiveness. In TNBC, LCOR associates with ICB clinical response. Importantly, extracellular vesicle (EV) Lcor-messenger RNA therapy in combination with anti-PD-L1 overcame resistance and eradicated breast cancer metastasis in preclinical models. Collectively, these data support LCOR as a promising target for enhancement of ICB efficacy in TNBC, by boosting of tumor APM independently of IFN.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy , Interferons/pharmacology , Melanoma , Repressor Proteins/therapeutic use , Skin Neoplasms , Triple Negative Breast Neoplasms/drug therapy , Melanoma, Cutaneous Malignant
8.
Nat Cancer ; 3(1): 60-74, 2022 01.
Article in English | MEDLINE | ID: mdl-35121988

ABSTRACT

Despite increased overall survival rates, curative options for metastatic breast cancer remain limited. We have previously shown that metadherin (MTDH) is frequently overexpressed in poor prognosis breast cancer, where it promotes metastasis and therapy resistance through its interaction with staphylococcal nuclease domain-containing 1 (SND1). Through genetic and pharmacological targeting of the MTDH-SND1 interaction, we reveal a key role for this complex in suppressing antitumor T cell responses in breast cancer. The MTDH-SND1 complex reduces tumor antigen presentation and inhibits T cell infiltration and activation by binding to and destabilizing Tap1/2 messenger RNAs, which encode key components of the antigen-presentation machinery. Following small-molecule compound C26-A6 treatment to disrupt the MTDH-SND1 complex, we showed enhanced immune surveillance and sensitivity to anti-programmed cell death protein 1 therapy in preclinical models of metastatic breast cancer, in support of this combination therapy as a viable approach to increase immune-checkpoint blockade therapy responses in metastatic breast cancer.


Subject(s)
Breast Neoplasms , Antigen Presentation , Breast Neoplasms/drug therapy , Endonucleases/metabolism , Female , Humans , Membrane Proteins/metabolism , Micrococcal Nuclease/metabolism , Nuclear Proteins/genetics , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism
9.
Nat Cancer ; 3(1): 43-59, 2022 01.
Article in English | MEDLINE | ID: mdl-35121987

ABSTRACT

Metastatic breast cancer is a leading health burden worldwide. Previous studies have shown that metadherin (MTDH) promotes breast cancer initiation, metastasis and therapy resistance; however, the therapeutic potential of targeting MTDH remains largely unexplored. Here, we used genetically modified mice and demonstrate that genetic ablation of Mtdh inhibits breast cancer development through disrupting the interaction with staphylococcal nuclease domain-containing 1 (SND1), which is required to sustain breast cancer progression in established tumors. We performed a small-molecule compound screening to identify a class of specific inhibitors that disrupts the protein-protein interaction (PPI) between MTDH and SND1 and show that our lead candidate compounds C26-A2 and C26-A6 suppressed tumor growth and metastasis and enhanced chemotherapy sensitivity in preclinical models of triple-negative breast cancer (TNBC). Our results demonstrate a significant therapeutic potential in targeting the MTDH-SND1 complex and identify a new class of therapeutic agents for metastatic breast cancer.


Subject(s)
Endonucleases/metabolism , Membrane Proteins/metabolism , Micrococcal Nuclease , RNA-Binding Proteins/metabolism , Triple Negative Breast Neoplasms , Animals , Cell Adhesion Molecules/genetics , Humans , Membrane Proteins/genetics , Mice , RNA-Binding Proteins/genetics , Transcription Factors
10.
Pregnancy Hypertens ; 26: 133-140, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34794010

ABSTRACT

OBJECTIVES: Preeclampsia is a common complication of pregnancy that causes health problems for both the mother and her fetus. This study aimed to develop and externally validate a model to predict adverse outcomes in preeclampsia in a trans-regional two-center retrospective cohort of Chinese women. STUDY DESIGN: To generate a model for the risk of women with adverse outcomes, we incorporated candidate variables in the development set in univariate, least absolute shrinkage and selection operator analysis and multivariable logistic regression. The performance of the model was evaluated for the receiver operating characteristic (ROC) curve, calibration and decision curve analysis. Further, we externally validated the model in an independent dataset. MAIN OUTCOME MEASURES: Composite adverse outcomes within 48 h of admission. RESULTS: There were 1 783 and 116 preeclampsia women in the development and validation set, respectively. The model included 10 predictors: gestational age at admission, irregular prenatal care, number of symptoms, mean arterial pressure, hematocrit, platelet count, fibrinogen, albumin, total bilirubin, and serum urea. The area under the ROC curve of the model was 0.867 in the development set and 0.841 in the external validation set. The calibration plots for the probability of adverse outcomes demonstrated a good correlation. Decision curve analysis further showed that our model had clinical application value. The nomogram and a software-based calculator (https://sdfyyfck.shinyapps.io/preeclampsia/) were constructed for convenient clinical use. CONCLUSIONS: Such a model could be used as a useful tool for the assessment of hypertensive-related complications in Chinese preeclampsia patients.


Subject(s)
Pre-Eclampsia/diagnosis , Adult , China , Female , Gestational Age , Humans , Nomograms , Pregnancy , ROC Curve , Retrospective Studies , Risk Assessment/methods
11.
Cancer Res ; 81(4): 1014-1025, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33239430

ABSTRACT

Colorectal and lung cancers account for one-third of all cancer-related deaths worldwide. Previous studies suggested that metadherin (MTDH) is involved in the development of colorectal and lung cancers. However, how MTDH regulates the pathogenesis of these cancers remains largely unknown. Using genetically modified mouse models of spontaneous colorectal and lung cancers, we found that MTDH promotes cancer progression by facilitating Wnt activation and by inducing cytotoxic T-cell exhaustion, respectively. Moreover, we developed locked nucleic acid-modified (LNA) MTDH antisense oligonucleotides (ASO) that effectively and specifically suppress MTDH expression in vitro and in vivo. Treatments with MTDH ASOs in mouse models significantly attenuated progression and metastasis of colorectal, lung, and breast cancers. Our study opens a new avenue for developing therapies against colorectal and lung cancers by targeting MTDH using LNA-modified ASO. SIGNIFICANCE: This study provides new insights into the mechanism of MTDH in promoting colorectal and lung cancers, as well as genetic and pharmacologic evidence supporting the development of MTDH-targeting therapeutics.


Subject(s)
Adenocarcinoma/therapy , Colorectal Neoplasms/therapy , Lung Neoplasms/therapy , Membrane Proteins/antagonists & inhibitors , Oligonucleotides, Antisense/therapeutic use , RNA-Binding Proteins/antagonists & inhibitors , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Genetic Therapy/methods , HEK293 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy/methods , Neoplasm Metastasis , Oligonucleotides/pharmacology , Oligonucleotides/therapeutic use , Oligonucleotides, Antisense/pharmacology , RNA-Binding Proteins/genetics , Xenograft Model Antitumor Assays
12.
Genes Dev ; 34(23-24): 1577-1598, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33262145

ABSTRACT

Metastasis is the ultimate "survival of the fittest" test for cancer cells, as only a small fraction of disseminated tumor cells can overcome the numerous hurdles they encounter during the transition from the site of origin to a distinctly different distant organ in the face of immune and therapeutic attacks and various other stresses. During cancer progression, tumor cells develop a variety of mechanisms to cope with the stresses they encounter, and acquire the ability to form metastases. Restraining these stress-releasing pathways could serve as potentially effective strategies to prevent or reduce metastasis and improve the survival of cancer patients. Here, we provide an overview of the tumor-intrinsic, microenvironment- and treatment-induced stresses that tumor cells encounter in the metastatic cascade and the molecular pathways they develop to relieve these stresses. We also summarize the preclinical and clinical studies that evaluate the potential therapeutic benefit of targeting these stress-relieving pathways.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasm Metastasis/physiopathology , Neoplasms/physiopathology , Neoplasms/therapy , Stress, Physiological/physiology , Tumor Microenvironment/physiology , Gene Expression Regulation, Neoplastic , Humans
13.
Genes Dev ; 34(19-20): 1310-1315, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32943575

ABSTRACT

SNAI2/SLUG, a metastasis-promoting transcription factor, is a labile protein that is degraded through the ubiquitin proteasome degradation system. Here, we conducted comprehensive gain- and loss-of-function screens using a human DUB cDNA library of 65 genes and an siRNA library of 98 genes, and identified USP20 as a deubiquitinase (DUB) that regulates SNAI2 ubiquitination and stability. Further investigation of USP20 demonstrated its function in promoting migration, invasion, and metastasis of breast cancer. USP20 positively correlates with SNAI2 protein level in breast tumor samples, and higher USP20 expression is associated with poor prognosis in ER- breast cancer patients.


Subject(s)
Breast Neoplasms/physiopathology , Neoplasm Metastasis/genetics , Snail Family Transcription Factors/metabolism , Ubiquitin Thiolesterase/metabolism , Breast Neoplasms/genetics , Cell Movement/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Library , Humans , Neoplasm Invasiveness/genetics , Protein Stability , Proteolysis , RNA, Small Interfering/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitination
14.
Genes Dev ; 34(19-20): 1359-1372, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32943576

ABSTRACT

Transcription factor SNAI2 plays key roles during development and has also been known to promote metastasis by inducing invasive phenotype and tumor-initiating activity of cancer cells. However, the post-translational regulation of SNAI2 is less well studied. We performed a dual-luciferase-based, genome-wide E3 ligase siRNA library screen and identified ASB13 as an E3 ubiquitin ligase that targets SNAI2 for ubiquitination and degradation. ASB13 knockout in breast cancer cells promoted cell migration and decreased F-actin polymerization, while overexpression of ASB13 suppressed lung metastasis. Furthermore, ASB13 knockout decreased YAP expression, and such regulation is dependent on an increased protein level of SNAI2, which in turn represses YAP transcription. YAP suppresses tumor progression in breast cancer, as YAP knockout increases tumorsphere formation, anchorage-independent colony formation, cell migration in vitro, and lung metastasis in vivo. Clinical data analysis reveals that ASB13 expression is positively correlated with improved overall survival in breast cancer patients. These findings establish ASB13 as a suppressor of breast cancer metastasis by promoting degradation of SNAI2 and relieving its transcriptional repression of YAP.


Subject(s)
Breast Neoplasms/physiopathology , Gene Expression Regulation, Neoplastic/genetics , Neoplasm Metastasis/physiopathology , Proteolysis , Proto-Oncogene Proteins c-yes/genetics , Snail Family Transcription Factors/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Female , Genome-Wide Association Study , Humans , Neoplasm Metastasis/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/genetics
15.
Dev Cell ; 51(4): 417-419, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31743661

ABSTRACT

Carcinoma cells often acquire mobility and invasiveness by undergoing epithelial-to-mesenchymal transition, and yet most metastatic lesions remain epithelial in nature. Recently, in Nature, Padmanaban et al. (2019) demonstrated that the quintessential epithelial marker E-cadherin promotes metastasis of invasive ductal breast carcinoma by enhancing the survival of tumor cells.


Subject(s)
Breast Neoplasms , Cadherins , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Humans
16.
Cancer Cell ; 35(1): 64-80.e7, 2019 01 14.
Article in English | MEDLINE | ID: mdl-30612941

ABSTRACT

Triple-negative breast cancer (TNBC) patients have the worst prognosis and distant metastasis-free survival among all major subtypes of breast cancer. The poor clinical outlook is further exacerbated by a lack of effective targeted therapies for TNBC. Here we show that ectopic expression and therapeutic delivery of the secreted protein Tubulointerstitial nephritis antigen-like 1 (Tinagl1) suppresses TNBC progression and metastasis through direct binding to integrin α5ß1, αvß1, and epidermal growth factor receptor (EGFR), and subsequent simultaneous inhibition of focal adhesion kinase (FAK) and EGFR signaling pathways. Moreover, Tinagl1 protein level is associated with good prognosis and reversely correlates with FAK and EGFR activation status in TNBC. Our results suggest Tinagl1 as a candidate therapeutic agent for TNBC by dual inhibition of integrin/FAK and EGFR signaling pathways.


Subject(s)
Extracellular Matrix Proteins/genetics , Integrin alpha5beta1/metabolism , Lipocalins/genetics , Lung Neoplasms/therapy , Receptors, Vitronectin/metabolism , Triple Negative Breast Neoplasms/therapy , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , ErbB Receptors/metabolism , Extracellular Matrix Proteins/administration & dosage , Extracellular Matrix Proteins/metabolism , Female , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lipocalins/administration & dosage , Lipocalins/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Prognosis , Signal Transduction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
17.
Reprod Sci ; 26(3): 377-385, 2019 03.
Article in English | MEDLINE | ID: mdl-29871559

ABSTRACT

Epithelial-mesenchymal transition (EMT) has been reported to be involved in adenomyosis by promoting cell invasion and fibrogenesis. But few studies have identified critical factors that regulate EMT process during adenomyosis. The eukaryotic translation initiation factor 3 subunit e (eIF3e) protein is a component of the multisubunit eIF3 complex essential for cap-dependent translation initiation. The aim of this study was to investigate whether eIF3e is involved in EMT in adenomyosis. Ectopic endometrial tissue samples were collected from 40 premenopausal women with ultrasonographically diagnosed and histologically confirmed adenomyosis. As controls, endometrial samples were obtained from 40 cycling premenopausal women patients who underwent surgery for benign gynecologic disorders or cervical intraepithelial neoplasia but without endometriosis, adenomyosis, nor uterine fibroids. All tissue samples were subjected to immunohistochemistry analysis of eIF3e, transforming growth factor-ß1 (TGF-ß1), E-cadherin, vimentin, Snail, and proliferating cell nuclear antigen (PCNA). The epithelial component of ectopic endometrium showed significantly reduced immunoreactivity against eIF3e and E-cadherin but elevated immunoreactivity against TGF-ß1, Snail, vimentin, and PCNA as compared with that of control endometrium (all P values <.05), and the difference was not affected by age, parity, or menstrual phase. The eIF3e staining levels correlated negatively with those of TGF-ß1, vimentin, Snail, and PCNA (both P values <.05). These data suggest that decreased eIF3e expression may pave way for EMT in the development of adenomyosis through activating the TGF-ß1 signaling pathway. Our study provided novel insights into the development and treatments of adenomyosis.


Subject(s)
Adenomyosis/metabolism , Endometrium/metabolism , Epithelial-Mesenchymal Transition , Eukaryotic Initiation Factor-3/metabolism , Adult , Cell Proliferation , Female , Humans , Middle Aged , Signal Transduction , Transforming Growth Factor beta1/metabolism
18.
Front Med ; 12(4): 426-439, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30097962

ABSTRACT

Tumor microenvironment (TME) is comprised of cellular and non-cellular components that exist within and around the tumor mass. The TME is highly dynamic and its importance in different stages of cancer progression has been well recognized. A growing body of evidence suggests that TME also plays pivotal roles in cancer treatment responses. TME is significantly remodeled upon cancer therapies, and such change either enhances the responses or induces drug resistance. Given the importance of TME in tumor progression and therapy resistance, strategies that remodel TME to improve therapeutic responses are under developing. In this review, we provide an overview of the essential components in TME and the remodeling of TME in response to anti-cancer treatments. We also summarize the strategies that aim to enhance therapeutic efficacy by modulating TME.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms , Tumor Microenvironment , Drug Resistance , Humans , Neoplasm Staging , Neoplasms/drug therapy , Neoplasms/pathology , Treatment Outcome , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology
19.
Trends Mol Med ; 24(9): 733-735, 2018 09.
Article in English | MEDLINE | ID: mdl-30029985

ABSTRACT

The tumor microenvironment contains heterogeneous populations of stromal cells with important roles in cancer progression and metastasis. In a recent study published in Nature Medicine, pSTAT3+ reactive astrocytes were found to promote brain metastasis by altering the tumor microenvironment, and represent a promising target for the treatment of brain metastasis.


Subject(s)
Astrocytes , Brain Neoplasms , Brain , Humans , STAT3 Transcription Factor , Stromal Cells , Tumor Microenvironment
20.
Reprod Sci ; 25(1): 102-109, 2018 01.
Article in English | MEDLINE | ID: mdl-28438065

ABSTRACT

Epithelial-mesenchymal transition (EMT) is now well documented to be involved in the development of endometriosis through the promotion of invasion and fibrogenesis. To date, several factors have been reported to be involved in EMT in endometriosis. The eukaryotic translation initiation factor 3 subunit e (eIF3e) protein is a component of the multisubunit eIF3 complex essential for cap-dependent translation initiation. The aim of this study was to investigate whether eIF3e is involved in EMT in endometriosis. We recruited 40 premenopausal women (34.7 [6.8] years) with laparoscopically and histologically diagnosed ovarian endometriomas, and their ectopic endometrial tissue samples were collected after informed consent. As controls, endometrial tissue samples were obtained after informed consent from 40 premenopausal women, roughly age-matched (36.9 [6.4] years) and menstrual phase-matched with endometriosis group, who underwent surgery for benign gynecologic disorders or cervical intraepithelial neoplasia but without endometriosis, adenomyosis, or uterine fibroids. All tissue samples were subjected to immunohistochemistry analysis of eIF3e, transforming growth factor (TGF-ß1), Snail, E-cadherin, vimentin, and proliferating cell nuclear antigen (PCNA). We found significantly reduced immunoreactivity against eIF3e and E-cadherin but elevated immunoreactivity against TGF-ß1, Snail, vimentin, and PCNA in endometriotic epithelial cells when compared to that of control endometrium (all P values <.05). The eIF3e staining levels correlated negatively with that of TGF-ß1 and Snail but positively with that of E-cadherin (all P values <.05). These data suggest that eIF3e downregulation may be involved in EMT in endometriosis, possibly through preferential translation of Snail. Future studies are warranted to confirm whether this is the mechanism.


Subject(s)
Endometriosis/metabolism , Endometrium/metabolism , Epithelial-Mesenchymal Transition/physiology , Eukaryotic Initiation Factor-3/metabolism , Adult , Cadherins/metabolism , Female , Humans , Middle Aged , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta1/metabolism , Vimentin/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL