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










Publication year range
1.
Int J Mol Sci ; 25(4)2024 Feb 17.
Article in English | MEDLINE | ID: mdl-38397067

ABSTRACT

Endometriosis is a common disease among women of reproductive age in which endometrial tissue grows in ectopic localizations, primarily within the pelvic cavity. These ectopic "lesions" grow as well as migrate and invade underlying tissues. Despite the prevalence of the disease, an understanding of factors that contribute to these cellular attributes remains poorly understood. Prefoldin-5 (PFDN5) has been associated with both aberrant cell proliferation and migration, but a potential role in endometriosis is unknown. As such, the purpose of this study was to examine PFDN5 expression in endometriotic tissue. PFDN5 mRNA and protein were examined in ectopic (lesion) and eutopic endometrial tissue from women with endometriosis and in eutopic endometrium from those without endometriosis using qRT-PCR and immunohistochemistry, respectively, while function of PFDN5 in vitro was evaluated using cell count and migration assays. PFDN5 mRNA and protein were expressed in eutopic and ectopic endometrial tissue, predominantly in the glandular epithelium, but not in endometrium from control subjects. Expression of both mRNA and protein was variable among endometriotic eutopic and ectopic endometrial tissue but showed an overall net increase. Knockdown of PFDN5 by siRNA transfection of endometriotic epithelial 12Z cells was associated with reduced cell proliferation/survival and migration. PFDN5 is expressed in eutopic and ectopic glandular epithelium and may play a role in proliferation and migration of these cells contributing to disease pathophysiology.


Subject(s)
Endometriosis , Molecular Chaperones , Repressor Proteins , Female , Humans , Cell Proliferation , Endometriosis/metabolism , Endometriosis/pathology , Endometrium/metabolism , Epithelium/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism
2.
Front Cell Dev Biol ; 11: 1225055, 2023.
Article in English | MEDLINE | ID: mdl-38078012

ABSTRACT

Endometriosis is a benign gynecological disease in which eutopic endometrial tissue composed of glands and stroma grow within the pelvic cavity. The disease affects females of reproductive age and is characterized by pelvic pain, infertility and reduced quality of life. The majority of pharmacologic treatment modalities for endometriosis focus on suppression of estradiol production and/or action; an approach associated with adverse side effects. c-MYC is elevated in eutopic endometrium and endometriotic lesion tissue in patients with endometriosis and the disease shares many similar pathological characteristics with that of endometrial carcinoma. While targeting of c-MYC with Omomyc has recently gained substantial interest in the field of cancer research, there has been no recent attempt to evaluate the potential utility in targeting c-MYC for endometriosis treatment. The following perspective article compares the similarities between endometriosis and endometrial cancer and presents preliminary data suggesting that targeting c-MYC with Omomyc reduces endometriotic cell proliferation and viability in vitro. Future application of targeting c-MYC in endometriosis treatment and potential pros and cons are then discussed.

3.
Clin Transl Med ; 13(5): e1223, 2023 05.
Article in English | MEDLINE | ID: mdl-37165578

ABSTRACT

BACKGROUND: Mutations in the receptor tyrosine kinase gene fibroblast growth factor receptor 2 (FGFR2) occur at a high frequency in endometrial cancer (EC) and have been linked to advanced and recurrent disease. However, little is known about how these mutations drive carcinogenesis. METHODS: Differential transcriptomic analysis and two-step quantitative real-time PCR (qRT-PCR) assays were applied to identify genes differentially expressed in two cohorts of EC patients carrying mutations in the FGFR2 gene as well as in EC cells harbouring mutations in the FGFR2. Candidate genes and target signalling pathways were investigated by qRT-PCR assays, immunohistochemistry and bioinformatics analysis. The functional roles of differently regulated genes were analysed using in vitro and in vivo experiments, including 3D-orthotypic co-culture systems, cell proliferation and migration protocols, as well as colony and focus formation assays together with murine xenograft tumour models. The molecular mechanisms were examined using CRISPR/Cas9-based loss-of-function and pharmacological approaches as well as luciferase reporter techniques, cell-based ectodomain shedding assays and bioinformatics analysis. RESULTS: We show that common FGFR2 mutations significantly enhance the sensitivity to FGF7-mediated activation of a disintegrin and metalloprotease (ADAM)17 and subsequent transactivation of the epidermal growth factor receptor (EGFR). We further show that FGFR2 mutants trigger the activation of ADAM10-mediated Notch signalling in an ADAM17-dependent manner, highlighting for the first time an intimate cooperation between EGFR and Notch pathways in EC. Differential transcriptomic analysis in EC cells in a cohort of patients carrying mutations in the FGFR2 gene identified a strong association between FGFR2 mutations and increased expression of members of the Notch pathway and ErbB receptor family. Notably, FGFR2 mutants are not constitutively active but require FGF7 stimulation to reprogram Notch and EGFR pathway components, resulting in ADAM17-dependent oncogenic growth. CONCLUSIONS: These findings highlight a pivotal role of ADAM17 in the pathogenesis of EC and provide a compelling rationale for targeting ADAM17 protease activity in FGFR2-driven cancers.


Subject(s)
Endometrial Neoplasms , Receptor, Fibroblast Growth Factor, Type 2 , Female , Humans , Mice , Animals , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Signal Transduction/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Mutation/genetics
4.
Int J Mol Sci ; 24(3)2023 Jan 31.
Article in English | MEDLINE | ID: mdl-36769010

ABSTRACT

Adenomyosis is defined as the development of endometrial epithelial glands and stroma within the myometrial layer of the uterus. These "ectopic" lesions share many cellular characteristics with endometriotic epithelial cells as well as endometrial adenocarcinoma cells, including enhanced proliferation, migration, invasion and progesterone resistance. We recently reported that the 60S acidic ribosomal protein P1, RPLP1, is up-regulated in endometriotic epithelial cells and lesion tissue where it plays a role in cell survival. To evaluate if a similar pattern of expression and function for RPLP1 exists in adenomyosis and endometrial cancer, we examined RPLP1 expression in adenomyosis and endometrial cancer tissue specimens and assessed its function in vitro using well-characterized cell lines. A total of 12 control endometrial biopsies and 20 eutopic endometrial and matched adenomyosis biopsies as well as 103 endometrial adenocarcinoma biopsies were evaluated for RPLP1 localization by immunohistochemistry. Endometrial adenocarcinoma cell lines, Ishikawa, HEC1A, HEC1B and AN3 were evaluated for RPLP1 protein and transcript expression, while in vitro function was evaluated by knocking down RPLP1 expression and assessing cell survival and migration. RPLP1 protein was up-regulated in eutopic epithelia as well as in adenomyosis lesions compared to eutopic endometria from control subjects. RPLP1 was also significantly up-regulated in endometrial adenocarcinoma tissue. Knockdown of RPLP1 in endometrial adenocarcinoma cell lines was associated with reduced cell survival and migration. RPLP1 expression is up-regulated in eutopic and ectopic adenomyotic epithelia as well as in the epithelia of endometrial cancer specimens. In vitro studies support an essential role for RPLP1 in mediating cell survival and migration, processes which are all involved in pathophysiology associated with both diseases.


Subject(s)
Adenocarcinoma , Adenomyosis , Endometrial Neoplasms , Endometriosis , Uterine Neoplasms , Female , Humans , Adenocarcinoma/pathology , Adenomyosis/pathology , Cell Survival/genetics , Endometrial Neoplasms/metabolism , Endometriosis/pathology , Endometrium/metabolism , Epithelial Cells/metabolism , Uterine Neoplasms/pathology
5.
Proc Natl Acad Sci U S A ; 119(44): e2205524119, 2022 11.
Article in English | MEDLINE | ID: mdl-36282915

ABSTRACT

Uterine leiomyomas (UL) are benign tumors that arise in the myometrial layer of the uterus. The standard treatment option for UL is hysterectomy, although hormonal therapies, such as selective progesterone receptor modulators, are often used as temporary treatment options to reduce symptoms or to slow the growth of tumors. However, since the pathogenesis of UL is poorly understood and most hormonal therapies are not based on UL-specific, divergent hormone signaling pathways, hallmarks that predict long-term efficacy and safety of pharmacotherapies remain largely undefined. In a previous study, we reported that aberrant expression of repressor element 1 silencing transcription factor/neuron-restrictive silencing factor (REST/NRSF) target genes activate UL growth due to the near ubiquitous loss of REST. Here, we show that ablation of the Rest gene in mouse uterus leads to UL phenotype and gene-expression patterns analogous to UL, including altered estrogen and progesterone signaling pathways. We demonstrate that many of the genes dysregulated in UL harbor cis-regulatory elements bound by REST and progesterone receptor (PGR) adjacent to each other. Crucially, we identify an interaction between REST and PGR in healthy myometrium and present a putative mechanism for the dysregulation of progesterone-responsive genes in UL ensuing in the loss of REST. Using three Rest conditional knockout mouse lines, we provide a comprehensive picture of the impact loss of REST has in UL pathogenesis and in altering the response of UL to steroid hormones.


Subject(s)
Leiomyoma , Uterine Neoplasms , Animals , Female , Humans , Mice , Estrogens/metabolism , Leiomyoma/genetics , Leiomyoma/metabolism , Leiomyoma/pathology , Progesterone/metabolism , Receptors, Progesterone/genetics , Transcription Factors , Uterine Neoplasms/pathology
6.
Biomedicines ; 10(7)2022 Jul 14.
Article in English | MEDLINE | ID: mdl-35885004

ABSTRACT

Endometriosis is an enigmatic disease characterized by pain and infertility in which endometrial tissue grows in ectopic locations, predominantly the pelvic cavity. The pathogenesis and pathophysiology of endometriosis is complex and postulated to involve alterations in inflammatory, cell proliferation and post-transcriptional regulatory pathways among others. Our understanding on the pathogenesis and pathophysiology of endometriosis is further complicated by the fact that endometriosis can only be diagnosed by laparoscopy only after the disease has manifested. This makes it difficult to understand the true pathogenesis as a cause-and-effect relationship is difficult to ascertain. To aid in our understanding on endometriosis pathogenesis and pathophysiology, numerous rodent models have been developed. In this case, we discuss further assessment of a miR-451a-macrophage migration inhibitory factor (Mif) pathway which contributes to lesion survival. Specifically, we evaluate the temporal expression of lesion Mif receptors, Cd74 and Cxcr4 using host mice which express wild-type or miR-451a deficient lesions. Similar to that observed in humans and a non-human primate model of endometriosis, Cd74 expression is elevated in lesion tissue in a temporal fashion while that of Cxcr4 shows minimal increase during initial lesion establishment but is reduced later during the lifespan. Absence of miR-451a during initial lesion establishment is associated with an augmentation of Cd74, but no Cxcr4 expression. The data obtained in this study provide further support for a role of Mif receptors, Cd74 and Cxcr4 in the pathophysiology of endometriosis.

7.
Int J Mol Sci ; 23(11)2022 May 24.
Article in English | MEDLINE | ID: mdl-35682544

ABSTRACT

BACKGROUND: miR-451a can function as a tumor suppresser and has been shown to be elevated in both endometriotic lesion tissue and serum from women with endometriosis. To further explore the role of miR-451a in the pathophysiology of endometriosis, specifically, further evaluating its association with the tumor suppressor, phosphatase and tensin homolog (PTEN), we examined their expression in individual endometriotic lesion tissue to gain insight into their relationship and further explore if miR-451a regulates PTEN expression. METHODS: A total of 55 red, peritoneal endometriotic lesions and matched eutopic endometrial specimens were obtained from 46 patients with endometriosis. miR-451a, miR-25-3p and PTEN mRNA levels were assessed by qRT-PCR and reported for each matched eutopic and ectopic sample. To evaluate miR-451a and miR-25-3p expression of miR-25-3p and PTEN, respectively, 12Z cells (endometriotic epithelial cell line) were transfected and miR-25-3p expression was assessed by qRT-PCR, while PTEN protein expression was assessed by Western blotting. RESULTS: PTEN and miR-25-3p expression exhibited an inverse relationship, as did miR-25-3p and miR-451a in individual lesions. Over-expression of miR-451a in 12Z cells resulted in down-regulation of miR-25-3p, while up-regulation of miR-25-3p resulted in down-regulation of PTEN protein expression. CONCLUSIONS: By assessing individual endometriotic lesion expression, we discovered an inverse relationship between miR-451a, miR-25-3p and PTEN, while in vitro cell transfection studies suggest that miR-451a may regulate PTEN expression via modulating miR-25-3p.


Subject(s)
Endometriosis , MicroRNAs , Peritoneal Diseases , Endometriosis/pathology , Female , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Peritoneal Diseases/genetics , RNA, Messenger/metabolism
8.
F S Sci ; 3(3): 246-254, 2022 08.
Article in English | MEDLINE | ID: mdl-35654738

ABSTRACT

OBJECTIVE: To examine the expression of uterine natural killer (uNK) cells and cytotoxic T lymphocytes (CTLs) in endometrial biopsies from reproductive-age women with and without nonstructural abnormal uterine bleeding (AUB) and evaluate the expression of granulysin within these cell populations and potential modulation of matrix metalloproteinase (MMP) expression. DESIGN: Experimental study, retrospective design. SETTING: Academic research laboratory. PATIENT(S): Patients with nonstructural AUB with no other gynecological pathologies and control patients without AUB. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Immunohistochemical analysis of granulysin, CD56 (uNK cell marker), and CD8 (CTL marker) expression as well as granulysin messenger ribonucleic acid (mRNA) expression levels in endometrial biopsy samples. Assessment of granulysin regulation of human endometrial stromal cell MMP-1 and MMP-3 mRNA expression. RESULT(S): The numbers of uNK cells and CTLs were significantly greater in endometrial biopsy tissue from women with AUB than those from controls. In accord with the increased expression of uNK cells and CTLs, granulysin expression was significantly greater in endometrial biopsies from patients with AUB than in from controls and colocalized to both cell types but not endometrial stromal or epithelial cells. The increased granulysin protein expression was associated with the increased granulysin mRNA expression in adjacent serial sections from these same samples. The treatment of the human endometrial stromal cell line t-HESC with granulysin resulted in a significant increase in MMP-1 and MMP-3 mRNA expression. CONCLUSION(S): In the current study, immunohistochemistry showed an increased expression of uNK cells, CTLs, and granulysin among subjects with AUB compared with that of subjects without AUB, leading to conclusions that disturbances in the balance of immune cells and an increase in granulysin expression may have implications in the pathophysiology of AUB and include enhanced MMP-1 and MMP-3 expression.


Subject(s)
Matrix Metalloproteinase 1 , Matrix Metalloproteinase 3 , Endometrium , Female , Humans , Killer Cells, Natural , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/genetics , RNA, Messenger/genetics , Retrospective Studies , T-Lymphocytes, Cytotoxic/metabolism , Uterine Hemorrhage/metabolism
9.
Reprod Sci ; 29(8): 2089-2104, 2022 08.
Article in English | MEDLINE | ID: mdl-35476352

ABSTRACT

Emerging data indicates an association between endometriosis and subclinical atherosclerosis, with women with endometriosis at a higher risk for cardiovascular disease later in life. Inflammation is proposed to play a central role in the pathophysiology of both diseases and elevated levels of systemic pro-inflammatory cytokines including macrophage migration inhibitory factor (MIF), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) are well documented. However, a thorough understanding on the mediators and mechanisms which contribute to altered cytokine expression in both diseases remain poorly understood. MicroRNAs (miRNAs) are important post-transcriptional regulators of inflammatory pathways and numerous studies have reported altered circulating levels of miRNAs in both endometriosis and atherosclerosis. Potential contribution of miRNA-mediated inflammatory cascades common to the pathophysiology of both diseases has not been evaluated but could offer insight into common pathways and early manifestation relevant to both diseases which may help understand cause and effect. In this review, we discuss and summarize differentially expressed inflammatory circulating miRNAs in endometriosis subjects, compare this profile to that of circulating levels associated with atherosclerosis when possible, and then discuss mechanistic studies focusing on these miRNAs in relevant cell, tissue, and animal models. We conclude by discussing the potential utility of targeting the relevant miRNAs in the MIF-IL-6-TNF-α pathway as therapeutic options and offer insight into future studies which will help us better understand not only the role of these miRNAs in the pathophysiology of both endometriosis and atherosclerosis but also commonality between both diseases.


Subject(s)
Atherosclerosis , Endometriosis , MicroRNAs , Animals , Atherosclerosis/genetics , Endometriosis/metabolism , Female , Humans , Interleukin-6 , MicroRNAs/genetics , MicroRNAs/metabolism , Tumor Necrosis Factor-alpha
10.
Cells ; 11(7)2022 03 24.
Article in English | MEDLINE | ID: mdl-35406659

ABSTRACT

Endometriosis is a significant disease characterized by infertility and pelvic pain in which endometrial stromal and glandular tissue grow in ectopic locations. Altered responsiveness to progesterone is a contributing factor to endometriosis pathophysiology, but the precise mechanisms are poorly understood. Progesterone resistance influences both the eutopic and ectopic (endometriotic lesion) endometrium. An inability of the eutopic endometrium to properly respond to progesterone is believed to contribute to the infertility associated with the disease, while an altered responsiveness of endometriotic lesion tissue may contribute to the survival of the ectopic tissue and associated symptoms. Women with endometriosis express altered levels of several endometrial progesterone target genes which may be due to the abnormal expression and/or function of progesterone receptors and/or chaperone proteins, as well as inflammation, genetics, and epigenetics. MiRNAs are a class of epigenetic modulators proposed to play a role in endometriosis pathophysiology, including the modulation of progesterone signaling. In this paper, we summarize the role of progesterone receptors and progesterone signaling in endometriosis pathophysiology, review miRNAs, which are over-expressed in endometriosis tissues and fluids, and follow this with a discussion on the potential regulation of key progesterone signaling components by these miRNAs, concluding with suggestions for future research endeavors in this area.


Subject(s)
Endometriosis , Infertility , MicroRNAs , Endometriosis/metabolism , Female , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Progesterone , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism
12.
Mol Cell Endocrinol ; 525: 111190, 2021 04 05.
Article in English | MEDLINE | ID: mdl-33549604

ABSTRACT

The endometrium is an essential component of the female uterus which provides the environment for pregnancy establishment and maintenance. Abnormalities of the endometrium not only lead to difficulties in establishing and maintaining pregnancy but also play a causative role in diseases of endometrial origin including endometriosis and endometrial cancer. Non-coding RNAs are proposed to play a role in regulating the genome in both normal endometrial physiology and pathophysiology. In this review, we first provide a general overview of non-coding RNAs and reproductive physiology of the endometrium. We then discuss the role on non-coding RNAs in normal endometrial physiology and pathophysiology of endometrial infertility. We then conclude with non-coding RNAs in the pathophysiology of endometriosis and endometrial cancer.


Subject(s)
Endometrium/metabolism , Endometrium/physiopathology , RNA, Long Noncoding/metabolism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/physiopathology , Female , Humans , Menstruation , RNA, Long Noncoding/genetics , Reproduction , Uterine Diseases/genetics , Uterine Diseases/physiopathology
13.
Adv Anat Embryol Cell Biol ; 232: 79-97, 2020.
Article in English | MEDLINE | ID: mdl-33278008

ABSTRACT

Endometriosis is an enigmatic disease for which we still have a poor understanding on how and why the disease develops. In recent years, miRNAs, small noncoding RNAs which regulate gene expression posttranscriptionally, have been evaluated for their role in endometriosis pathophysiology. This review will provide a brief summary on the role of miRNAs in endometrial physiology and pathophysiology as related to endometriosis. We will then discuss mouse models used in endometriosis research and the incorporation of some of these models in studies which examined the role of miRNAs in endometriosis pathophysiology. We conclude with providing future prospective on the role of mouse models in dissecting the role of miRNAs in endometriosis pathophysiology.


Subject(s)
Endometriosis/metabolism , Endometrium/metabolism , MicroRNAs/metabolism , Animals , Disease Models, Animal , Endometriosis/genetics , Female , Humans , Mice , MicroRNAs/genetics
14.
Mol Hum Reprod ; 26(1): 53-64, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31899515

ABSTRACT

Endometriosis is a female disease which is defined as the presence of ectopic endometrial tissue and is dependent on estrogen for its survival in these ectopic locations. Expression of the ribosomal protein large P1 (RPLP1) is associated with cell proliferation and invasion in several pathologies, but a role in the pathophysiology of endometriosis has not been explored. In this study, we aimed to evaluate the expression and function of RPLP1 with respect to endometriosis pathophysiology. RPLP1 protein was localised by immunohistochemistry (IHC) in eutopic and ectopic tissue from 28 subjects with confirmed endometriosis and from 20 women without signs or symptoms of the disease, while transcript levels were evaluated by qRT-PCR in 77 endometriotic lesions and 55 matched eutopic endometrial biopsies, and protein expression was evaluated using western blotting in 20 of these matched samples. To evaluate the mechanism for enhanced lesion expression of RPLP1, an experimental murine model of endometriosis was used and RPLP1 expression was localized using IHC. In vitro studies using an endometriosis cell line coupled with shRNA knockdown was used to demonstrate its role in cell survival. Expression of RPLP1 mRNA and protein were significantly higher in ectopic lesion tissue compared to paired eutopic endometrium and immunohistochemical localisation revealed predominant localisation to epithelial cells. This pattern of lesion RPLP1 was recapitulated in mice with experimentally induced endometriosis. Stable knockdown of RPLP1 protein resulted in a significant decrease in cell survival in vitro. These studies reveal that RPLP1 is associated with cell proliferation and/or survival and may play a role in the pathophysiology of endometriosis.


Subject(s)
Apoptosis/genetics , Endometriosis/genetics , Epithelial Cells/metabolism , Phosphoproteins/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/genetics , Ribosomal Proteins/genetics , Adult , Animals , Case-Control Studies , Cell Line , Cell Proliferation , Disease Models, Animal , Endometriosis/metabolism , Endometriosis/pathology , Endometrium/metabolism , Endometrium/pathology , Epithelial Cells/pathology , Female , Gene Expression Regulation , Humans , Mice, Inbred C57BL , Middle Aged , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribosomal Proteins/antagonists & inhibitors , Ribosomal Proteins/metabolism , Severity of Illness Index , Signal Transduction
15.
F S Sci ; 1(1): 90-97, 2020 Aug.
Article in English | MEDLINE | ID: mdl-35559743

ABSTRACT

OBJECTIVE: To study C-X-C motif chemokine 12 (CXCL12) and CXCR4 expression in endometrial tissue from both women with and without abnormal uterine bleeding (AUB) of endometrial origin and evaluate their relationship with microRNA (miRNA). DESIGN: Retrospective and laboratory study. SETTING: University-based research laboratory. PATIENT(S): Nine women with and without abnormal uterine bleeding, all of whom were in the secretory stage of their menstrual cycle, who provided endometrial biopsy tissue. INTERVENTION(S): Immunohistochemical localization of CXCL12 and CXCR4 as well as quantitative reverse-transcription polymerase chain reaction (qRT-PCR) assessment of mRNA expression in archived endometrial biopsy tissue and in vitro cell culture using the immortalized endometrial stromal cell line, t-HESC. Endometrial stromal cell line, t-HESC transfection with nontargeting, negative control miRNA mimics or miRNA mimics for miR-23b-3p and mRNA assessment miR-23b-3p expression confirmed by qRT-PCR and evaluation of impact on CXCL12 expression at the protein level by enzyme-linked immunosorbent assay and mRNA levels by qRT-PCR. MAIN OUTCOME MEASURE(S): Expression of CXCL12 and CXCR4 protein via immunohistochemistry and mRNA and miRNA levels of CXCL12 and CXCR4 as well as miR-23b-3p, miR-24b-3p, and miR-27b-3p, respectively, via qRT-PCR. RESULT(S): CXCL12 and its receptor CXCR4 expression were up-regulated in the endometrial tissue of women with AUB at the protein level, but this up-regulation of expression was only associated with increased CXCR4 mRNA expression. To evaluate whether CXCL12 may be post-transcriptionally regulated, we assessed expression of miR-23b-3p, a bona fide post-transcriptional regulator of CXCL12 expression. The expression of miR-23b-3p was statistically significantly lower in AUB endometrial tissue, as were fellow cluster members miR-24-3p and miR-27-3p. Transfection of t-HESC cells with pre-miR-23b-3p mimics statistically significantly reduced the levels of CXCL12 secreted protein but not mRNA levels, suggesting that miR-23b-3p retards protein translation independent of transcript degradation. CONCLUSION(S): Reduced expression of the miR-23b-3p/24-3p/27b-3p cluster is associated with elevated expression of CXCL12, which may contribute to the pathophysiology of AUB.

16.
Methods Mol Biol ; 2080: 123-134, 2020.
Article in English | MEDLINE | ID: mdl-31745876

ABSTRACT

Archived, formalin-fixed, paraffin-embedded tissue provides a robust resource for assessing protein expression in a variety of complex tissue types. Immunohistochemical localization techniques allow one to identify proteins of interest in the different cell populations which compose these tissues, but quantitative comparison within and between samples is semiquantitative. In contrast, Western blot analysis provides a more quantitative assessment but without the ability to identify the cellular sources of expressed protein. Here we describe a dual approach using human endometrium to assess both the localization and quantitation of the macrophage migration inhibitory factor (MIF) receptor CD74 by immunohistochemical techniques and Western blotting.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , Blotting, Western , Histocompatibility Antigens Class II/metabolism , Immunohistochemistry , Receptors, Immunologic/metabolism , Biomarkers , Blotting, Western/methods , Formaldehyde , Immunohistochemistry/methods , Paraffin Embedding , Protein Binding , Protein Transport , Receptors, Immunologic/genetics , Tissue Fixation
17.
Sci Rep ; 9(1): 8823, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31217548

ABSTRACT

Endometriosis is an inflammatory condition in which endometrial tissue grows in ectopic locations. Survival and growth of these ectopic lesions is associated with pain and infertility. MicroRNAs (miRNAs) have been postulated to play a role in the pathophysiology of the disease and we have previously demonstrated expression of miR-451 in human endometriotic lesion tissue. Here we report elevated expression of the miR-144-3p/miR-451a cluster in human endometriotic lesion tissue. Use of an endometriotic epithelial cell line (12Z) in which the miRNA processing enzyme, DROSHA, was knocked down resulted in an enrichment in the primary (pri) form of miR-144-3p but not that of pri-miR-451a. Using an experimental mouse model of endometriosis in which ectopic endometriotic lesions were deficient for both of these miRNAs revealed that miR-451a, but not miR-144-3p may be derived from exogenous sources such as the circulation/erythrocytes. Together, these data suggest that the miR-144-3p/miR-451a cluster is expressed in human endometriotic lesion tissue, the level of expression correlates with survival status of the lesion tissue and that miR-451a, but not miR-144-3p may be derived from exogenous sources such as erythrocytes.


Subject(s)
Endometrium/metabolism , Endometrium/pathology , Gene Expression Regulation , MicroRNAs/metabolism , Adult , Cell Survival/genetics , Female , Humans , MicroRNAs/genetics , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonuclease III/metabolism , Young Adult
18.
Reprod Sci ; 25(11): 1557-1566, 2018 11.
Article in English | MEDLINE | ID: mdl-29592775

ABSTRACT

CD74 is the primary receptor for macrophage migration inhibitory factor (MIF). Although expression of MIF has been described in endometriotic lesions, the cellular localization and function of the MIF receptor, CD74, are poorly understood. To further explore the role of CD74 in the pathophysiology of endometriosis, we utilized specimens from women with diagnostically confirmed endometriosis, women with no signs or symptoms of endometriosis (controls), and 8 baboons with experimentally induced endometriosis. Compared to eutopic endometrium from women with endometriosis, CD74 transcript expression was significantly increased in endometriotic lesion tissue. Similarly, cellular expression of CD74 was significantly greater in ectopic lesion tissue compared to paired eutopic endometrium, which both expressed greater CD74 expression compared to eutopic endometrium from control patients. Localization of CD74 was predominant to epithelial cells of ectopic and matched eutopic endometrium and was not influenced by the stage of the menstrual cycle. Eutopic endometrium from control patients did not express detectable levels of CD74 protein by immunohistochemistry. This pattern of expression and CD74 protein localization could be recapitulated in endometriotic lesion tissue from baboons with experimentally induced disease. Transfection of the endometriotic epithelial cell lines, 12Z with CD74 short hairpin RNA (shRNA), resulted in a significant decrease in CD74 protein expression, which was associated with a significant reduction in cellular proliferation as well as the expression of the prosurvival cytokine interleukin 8. Together, these data support the hypothesis that CD74 is elevated in endometriotic lesion tissue and may contribute to the pathogenesis of endometriosis by promoting cell survival.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , Endometriosis/metabolism , Epithelial Cells/metabolism , Histocompatibility Antigens Class II/metabolism , Interleukin-8/metabolism , Papio anubis/metabolism , Receptors, Immunologic/metabolism , Adult , Animals , Cell Survival , Endometriosis/pathology , Endometrium/metabolism , Endometrium/pathology , Epithelial Cells/pathology , Female , Humans , Middle Aged , Species Specificity , Young Adult
19.
Curr Womens Health Rev ; 14(2): 189-194, 2018.
Article in English | MEDLINE | ID: mdl-31435203

ABSTRACT

BACKGROUND: Endometriosis is a disease common among women of reproductive age characterized by pain, anxiety and infertility. Defined as the growth of endometrial tissue in ectopic locations, endometriosis remains an enigmatic disease for which current treatments are less than ideal. Much of these shortcomings to current therapy stem from our incomplete understanding on the pathogenesis of the disease. It is generally accepted that endometriosis is an estrogen-dependent disease and, as such, the majority of treatment approaches aim at reducing estrogen action and/or production. Unfortunately, this approach is not effective in all women with endometriosis and in those women where success is achieved with their use, there is potential for health-comprising side effects. OBJECTIVE: The objective of this review is to summarize current approaches for treatment of endometriosis, discuss their limitations and potential reasons for lack of progress towards better therapeutics for this disease. RESULTS: In this review we summarize the current approaches for treatment of endometriosis, discuss their limitations and potential reasons for lack of progress towards better therapeutics for this disease. CONCLUSION: Based upon the current state of knowledge, there is a strong necessity for through assessment at the level of the genome, miRNAome and proteome as well as the importance of integrating clinically-relevant endpoints in future studies which evaluate potential endometriosis therapies in experimental models of endometriosis.

20.
Reprod Sci ; 24(9): 1264-1274, 2017 09.
Article in English | MEDLINE | ID: mdl-28256954

ABSTRACT

Decidualization is essential for successful embryo implantation and is regulated by concerted actions of growth factors and hormones. More recently, microRNAs, small RNA molecules that regulate posttranscriptional gene expression, have been implicated to play a role in the decidualization process. Of these microRNAs, miR-181b-5p has been associated with decidualization but its precise role and targets are not well established. To address this gap in our knowledge, we assessed the expression of miR-181b-5p, and its target tissue inhibitor of metalloproteinase 3 (TIMP-3), during in vitro decidualization using the well-characterized human endometrial stromal cell line, t-HESC. miR-181b-5p expression was highest prior to decidualization and significantly decreased in response to decidualization stimulus. In contrast, TIMP-3 expression was absent prior to in vitro decidualization and increased during decidualization. Regulation of TIMP-3 expression by miR-181b-5p was confirmed in vitro by quantitative reverse transcription polymerase chain reaction (qRT-PCR), Western blot analysis, and 3' untranslated region reporter constructs. To identify unforeseen targets of miR-181b-5p during in vitro decidualization, t-HESC cells were transfected with pre- miR-181b-5p, and protein profiles were determined by 2-dimensional differential in-gel electrophoresis followed by matrix-assisted laser desorption-ionization time-of-flight/time-of-flight (MALDI TOF/TOF) tandem mass spectrometry. Of these proteins, several downregulated proteins associated with cell migration were identified including annexin A2, which we subsequently confirmed by qRT-PCR and Western blot analysis to be regulated by miR-181b-5p. In conclusion, miR-181b-5p is downregulated during the process of in vitro decidualization and may regulate the expression of proteins associated with cell migration including TIMP-3 and annexin A2.


Subject(s)
Annexin A2/metabolism , Cell Movement/physiology , Endometrium/metabolism , MicroRNAs/metabolism , Stromal Cells/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , Annexin A2/genetics , Cell Line , Down-Regulation , Embryo Implantation/physiology , Endometrium/cytology , Female , Gene Expression Regulation , Humans , MicroRNAs/genetics , Stromal Cells/cytology , Tissue Inhibitor of Metalloproteinase-3/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...