Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Gynecol Obstet Invest ; 89(2): 73-86, 2024.
Article in English | MEDLINE | ID: mdl-38382486

ABSTRACT

BACKGROUND: Uterine fibroids are benign monoclonal tumors originating from the smooth muscle cells of the myometrium, constituting the most prevalent pathology within the female genital tract. Uterine sarcomas, although rare, still represent a diagnostic challenge and should be managed in centers with adequate expertise in gynecological oncology. OBJECTIVES: This article is aimed to summarize and discuss cutting-edge elements about the diagnosis and management of uterine fibroids and sarcomas. METHODS: This paper is a report of the lectures presented in an expert meeting about uterine fibroids and sarcomas held in Palermo in February 2023. OUTCOME: Overall, the combination of novel molecular pathways may help combine biomarkers and expert ultrasound for the differential diagnosis of uterine fibroids and sarcomas. On the one hand, molecular and cellular maps of uterine fibroids and matched myometrium may enhance our understanding of tumor development compared to histologic analysis and whole tissue transcriptomics, and support the development of minimally invasive treatment strategies; on the other hand, ultrasound imaging allows in most of the cases a proper mapping the fibroids and to differentiate between benign and malignant lesions, which need appropriate management. CONCLUSIONS AND OUTLOOK: The choice of uterine fibroid management, including pharmacological approaches, surgical treatment, or other strategies, such as high-intensity focused ultrasound (HIFU), should be carefully considered, taking into account the characteristics of the patient and reproductive prognosis.


Subject(s)
High-Intensity Focused Ultrasound Ablation , Leiomyoma , Sarcoma , Uterine Myomectomy , Uterine Neoplasms , Female , Humans , Treatment Outcome , Leiomyoma/diagnosis , Leiomyoma/therapy , Leiomyoma/pathology , Uterine Neoplasms/diagnosis , Uterine Neoplasms/therapy , Uterine Neoplasms/pathology , Prognosis , Sarcoma/diagnosis , Sarcoma/therapy , High-Intensity Focused Ultrasound Ablation/methods
2.
Nat Commun ; 15(1): 945, 2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38296945

ABSTRACT

Age-associated myometrial dysfunction can prompt complications during pregnancy and labor, which is one of the factors contributing to the 7.8-fold increase in maternal mortality in women over 40. Using single-cell/single-nucleus RNA sequencing and spatial transcriptomics, we have constructed a cellular atlas of the aging myometrium from 186,120 cells across twenty perimenopausal and postmenopausal women. We identify 23 myometrial cell subpopulations, including contractile and venous capillary cells as well as immune-modulated fibroblasts. Myometrial aging leads to fewer contractile capillary cells, a reduced level of ion channel expression in smooth muscle cells, and impaired gene expression in endothelial, smooth muscle, fibroblast, perivascular, and immune cells. We observe altered myometrial cell-to-cell communication as an aging hallmark, which associated with the loss of 25 signaling pathways, including those related to angiogenesis, tissue repair, contractility, immunity, and nervous system regulation. These insights may contribute to a better understanding of the complications faced by older individuals during pregnancy and labor.


Subject(s)
Labor, Obstetric , Myometrium , Pregnancy , Humans , Female , Myometrium/metabolism , Labor, Obstetric/genetics , Labor, Obstetric/metabolism , Muscle, Smooth , Aging/genetics , Muscle Contraction
3.
Cell Mol Life Sci ; 80(9): 274, 2023 Aug 31.
Article in English | MEDLINE | ID: mdl-37650943

ABSTRACT

BACKGROUND: The stage, when tissues and organs are growing, is very vulnerable to environmental influences, but it's not clear how exposure during this time causes changes to the epigenome and increases the risk of hormone-related illnesses like uterine fibroids (UFs). METHODS: Developmental reprogramming of myometrial stem cells (MMSCs), the putative origin from which UFs originate, was investigated in vitro and in the Eker rat model by RNA-seq, ChIP-seq, RRBS, gain/loss of function analysis, and luciferase activity assays. RESULTS: When exposed to the endocrine-disrupting chemical (EDC) diethylstilbestrol during Eker rat development, MMSCs undergo a reprogramming of their estrogen-responsive transcriptome. The reprogrammed genes in MMSCs are known as estrogen-responsive genes (ERGs) and are activated by mixed lineage leukemia protein-1 (MLL1) and DNA hypo-methylation mechanisms. Additionally, we observed a notable elevation in the expression of ERGs in MMSCs from Eker rats exposed to natural steroids after developmental exposure to EDC, thereby augmenting estrogen activity. CONCLUSION: Our studies identify epigenetic mechanisms of MLL1/DNA hypo-methylation-mediated MMSC reprogramming. EDC exposure epigenetically targets MMSCs and leads to persistent changes in the expression of a subset of ERGs, imparting a hormonal imprint on the ERGs, resulting in a "hyper-estrogenic" phenotype, and increasing the hormone-dependent risk of UFs.


Subject(s)
Endocrine Disruptors , Leiomyoma , Animals , Rats , Endocrine Disruptors/toxicity , Estrogens , Biological Assay , Leiomyoma/chemically induced , Leiomyoma/genetics , Myeloid-Lymphoid Leukemia Protein , DNA
4.
Int J Mol Sci ; 24(14)2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37511399

ABSTRACT

The period during which tissue and organ development occurs is particularly vulnerable to the influence of environmental exposures. However, the specific mechanisms through which biological pathways are disrupted in response to developmental insults, consequently elevating the risk of hormone-dependent diseases, such as uterine fibroids (UFs), remain poorly understood. Here, we show that developmental exposure to the endocrine-disrupting chemical (EDC), diethylstilbestrol (DES), activates the inflammatory pathways in myometrial stem cells (MMSCs), which are the origin of UFs. Significantly, the secretome of reprogrammed MMSCs enhances the expression of critical inflammation-related genes in differentiated myometrial cells through the paracrine mechanism, which amplifies pro-inflammatory and immune suppression signaling in the myometrium. The expression of reprogrammed inflammatory responsive genes (IRGs) is driven by activated mixed-lineage leukemia protein-1 (MLL1) in MMSCs. The deactivation of MLL reverses the reprogramming of IRG expression. In addition, the inhibition of histone deacetylases (HDACs) also reversed the reprogrammed IRG expression induced by EDC exposure. This work identifies the epigenetic mechanisms of MLL1/HDAC-mediated MMSC reprogramming, and EDC exposure epigenetically targets MMSCs and imparts an IRG expression pattern, which may result in a "hyper-inflammatory phenotype" and an increased hormone-dependent risk of UFs later in life.


Subject(s)
Leiomyoma , Uterine Neoplasms , Female , Humans , Myometrium/metabolism , Leiomyoma/genetics , Leiomyoma/metabolism , Stem Cells/metabolism , Hormones/metabolism , Epigenesis, Genetic , Uterine Neoplasms/genetics
5.
Virchows Arch ; 2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37466765

ABSTRACT

Three main uterine leiomyoma molecular subtypes include tumors with MED12 mutation, molecular aberrations leading to HMGA2 overexpression, and biallelic loss of FH. These aberrations are mutually exclusive and can be found in approximately 80-90% of uterine leiomyoma, in which they seem to be a driver event. Approximately 10% of uterine leiomyoma, however, does not belong to any of these categories. Uterine leiomyoma with HMGA2 overexpression is the most common subtype in cellular and second most common category of usual leiomyoma. In some of these tumors, rearrangement of HMGA2 gene is present. The most common fusion partner of HMGA2 gene is RAD51B. Limited data suggests that RAD51B fusions with other genes may be present in uterine leiomyoma. In our study, we described two cases of uterine leiomyoma with RAD51B::NUDT3 fusion, which occur in one case of usual and one case of highly cellular leiomyoma. In both cases, no other driver molecular aberrations were found. The results of our study showed that RAD51::NUDT3 fusion can occur in both usual and cellular leiomyoma. RAD51B may be a fusion partner of multiple genes other than HMGA2 and HMGA1. In these cases, RAD51B fusion seems to be mutually exclusive with other driver aberrations defining molecular leiomyoma subtypes. RAD51B::NUDT3 fusion should be added to the spectrum of fusions which may occur in uterine leiomyoma, which can be of value especially in cellular leiomyoma in the context of differential diagnosis against endometrial stromal tumors.

6.
Physiol Rev ; 103(3): 1965-2038, 2023 07 01.
Article in English | MEDLINE | ID: mdl-36796099

ABSTRACT

Pregnancy is established during the periconceptional period as a continuum beginning with blastocyst attachment to the endometrial epithelial surface followed by embryo invasion and placenta formation. This period sets the foundation for the child and mother's health during pregnancy. Emerging evidence indicates that prevention of downstream pathologies in both the embryo/newborn and pregnant mother may be possible at this stage. In this review, we discuss current advances in the periconceptional space, including the preimplantation human embryo and maternal endometrium. We also discuss the role of the maternal decidua, the periconceptional maternal-embryonic interface, the dialogue between these elements, and the importance of the endometrial microbiome in the implantation process and pregnancy. Finally, we discuss the myometrium in the periconceptional space and review its role in determining pregnancy health.


Subject(s)
Embryo Implantation , Endometrium , Pregnancy , Female , Child , Infant, Newborn , Humans , Blastocyst , Placenta
7.
Int J Mol Sci ; 23(22)2022 Nov 09.
Article in English | MEDLINE | ID: mdl-36430255

ABSTRACT

High-grade serous ovarian carcinoma (HGSOC) represents the most common form of epithelial ovarian carcinoma. The absence of specific symptoms leads to late-stage diagnosis, making HGSOC one of the gynecological cancers with the worst prognosis. The cellular origin of HGSOC and the role of reproductive hormones, genetic traits (such as alterations in P53 and DNA-repair mechanisms), chromosomal instability, or dysregulation of crucial signaling pathways have been considered when evaluating prognosis and response to therapy in HGSOC patients. However, the detection of HGSOC is still based on traditional methods such as carbohydrate antigen 125 (CA125) detection and ultrasound, and the combined use of these methods has yet to support significant reductions in overall mortality rates. The current paradigm for HGSOC management has moved towards early diagnosis via the non-invasive detection of molecular markers through liquid biopsies. This review presents an integrated view of the relevant cellular and molecular aspects involved in the etiopathogenesis of HGSOC and brings together studies that consider new horizons for the possible early detection of this gynecological cancer.


Subject(s)
Ovarian Neoplasms , Female , Humans , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/therapy , Carcinoma, Ovarian Epithelial
8.
Int J Mol Sci ; 23(4)2022 Feb 16.
Article in English | MEDLINE | ID: mdl-35216305

ABSTRACT

The absence of standardized molecular profiling to differentiate uterine leiomyosarcomas versus leiomyomas represents a current diagnostic challenge. In this study, we aimed to search for a differential molecular signature for these myometrial tumors based on artificial intelligence. For this purpose, differential exome and transcriptome-wide research was performed on histologically confirmed leiomyomas (n = 52) and leiomyosarcomas (n = 44) to elucidate differences between and within these two entities. We identified a significantly higher tumor mutation burden in leiomyosarcomas vs. leiomyomas in terms of somatic single-nucleotide variants (171,863 vs. 81,152), indels (9491 vs. 4098), and copy number variants (8390 vs. 5376). Further, we discovered alterations in specific copy number variant regions that affect the expression of some tumor suppressor genes. A transcriptomic analysis revealed 489 differentially expressed genes between these two conditions, as well as structural rearrangements targeting ATRX and RAD51B. These results allowed us to develop a machine learning approach based on 19 differentially expressed genes that differentiate both tumor types with high sensitivity and specificity. Our findings provide a novel molecular signature for the diagnosis of leiomyoma and leiomyosarcoma, which could be helpful to complement the current morphological and immunohistochemical diagnosis and may lay the foundation for the future evaluation of malignancy risk.


Subject(s)
Leiomyoma , Leiomyosarcoma , Uterine Neoplasms , Artificial Intelligence , Diagnosis, Differential , Female , Humans , Leiomyoma/diagnosis , Leiomyoma/genetics , Leiomyoma/metabolism , Leiomyosarcoma/diagnosis , Leiomyosarcoma/genetics , Leiomyosarcoma/metabolism , Transcriptome , Uterine Neoplasms/diagnosis , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism
9.
Int J Mol Sci ; 22(16)2021 Aug 06.
Article in English | MEDLINE | ID: mdl-34445194

ABSTRACT

Uterine leiomyomas represent the most common benign gynecologic tumor. These hormone-dependent smooth-muscle formations occur with an estimated prevalence of ~70% among women of reproductive age and cause symptoms including pain, abnormal uterine bleeding, infertility, and recurrent abortion. Despite the prevalence and public health impact of uterine leiomyomas, available treatments remain limited. Among the potential causes of leiomyomas, early hormonal exposure during periods of development may result in developmental reprogramming via epigenetic changes that persist in adulthood, leading to disease onset or progression. Recent developments in unbiased high-throughput sequencing technology enable powerful approaches to detect driver mutations, yielding new insights into the genomic instability of leiomyomas. Current data also suggest that each leiomyoma originates from the clonal expansion of a single transformed somatic stem cell of the myometrium. In this review, we propose an integrated cellular and molecular view of the origins of leiomyomas, as well as paradigm-shifting studies that will lead to better understanding and the future development of non-surgical treatments for these highly frequent tumors.


Subject(s)
Leiomyoma/pathology , Uterine Neoplasms/pathology , Uterus/pathology , Animals , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Leiomyoma/etiology , Leiomyoma/genetics , Mutation , Uterine Neoplasms/etiology , Uterine Neoplasms/genetics , Uterus/metabolism
10.
Am J Obstet Gynecol ; 221(4): 320.e1-320.e23, 2019 10.
Article in English | MEDLINE | ID: mdl-31121144

ABSTRACT

BACKGROUND: Although uterine leiomyomas and leiomyosarcomas are considered biologically unrelated tumors, they share morphologic and histologic characteristics that complicate their differential diagnosis. The long-term therapeutic option for leiomyoma is laparoscopic myomectomy with morcellation, particularly for patients who wish to preserve their fertility. However, because of the potential dissemination of undiagnosed or hidden leiomyosarcoma from morcellation, there is a need to develop a preoperative assessment of malignancy risk. OBJECTIVE: Through an integrated comparative genomic and transcriptomic analysis, we aim to identify differential genetic targets in leiomyomas vs leiomyosarcomas using next-generation sequencing as the first step toward preoperative differential diagnosis. STUDY DESIGN: Targeted sequencing of DNA and RNA coding regions for solid tumor-associated genes was performed on formalin-fixed paraffin-embedded samples from 13 leiomyomas and 13 leiomyosarcoma cases. DNA sequencing was used to identify copy number variations, single-nucleotide variants, and small insertions/deletions. RNA sequencing was used to identify gene fusions, splice variants, and/or differential gene expression profiles. RESULTS: In leiomyosarcomas, tumor mutation burden was higher in terms of copy number variations, single nucleotide variants, small insertions/deletions, and gene fusions compared with leiomyomas. For copy number variations, 20 genes were affected by deletions in leiomyosarcomas, compared with 6 observed losses in leiomyomas. Gains (duplications) were identified in 19 genes in leiomyosarcomas, but only 3 genes in leiomyomas. The most common mutations (single-nucleotide variants and insertions/deletions) for leiomyosarcomas were identified in 105 genes of all analyzed leiomyosarcomas; 82 genes were affected in leiomyomas. Of note, 1 tumor previously diagnosed as leiomyosarcoma was established as inflammatory myofibroblastic tumor along this study with a novel ALK-TNS1 fusion. Finally, a differential transcriptomic profile was observed for 11 of 55 genes analyzed in leiomyosarcomas; 8.5% of initially diagnosed leiomyosarcomas showed high-confidence, novel gene fusions that were associated with these tumors. CONCLUSION: Through integrated comparative genomic and transcriptomic analyses, we identified novel differential genetic targets that potentially differentiate leiomyosarcomas and leiomyomas. This provides a new insight into the differential diagnosis of these myometrial tumors.


Subject(s)
Leiomyoma/diagnosis , Leiomyosarcoma/diagnosis , Uterine Neoplasms/diagnosis , Adult , Aged , DNA Copy Number Variations , Diagnosis, Differential , Female , Gene Deletion , Gene Duplication , Gene Expression Profiling , Gene Fusion , Genomics , High-Throughput Nucleotide Sequencing , Humans , Leiomyoma/genetics , Leiomyosarcoma/genetics , Middle Aged , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Sequence Analysis, RNA , Uterine Neoplasms/genetics
12.
Biol Reprod ; 101(6): 1115-1123, 2019 12 24.
Article in English | MEDLINE | ID: mdl-30184111

ABSTRACT

Uterine leiomyomas (LM) and leiomyosarcomas (LMS) are considered biologically unrelated tumors due to their cytogenetic and molecular disparity. Yet, these tumors share morphological and molecular characteristics that cannot be differentiated through current clinical diagnostic tests, and thus cannot be definitively classified as benign or malignant until surgery. Newer approaches are needed for the identification of these tumors, as has been done for other tissues. The application of next generation sequencing enables the detection of new mutations that, when coupled to machine learning bioinformatic tools, advances our understanding of chromosomal instability. These approaches in the context of LM and LMS could allow the discovery of genetic variants and possible genomic markers. Additionally, the potential clinical utility of circulating cell-free tumor DNA could revolutionize the noninvasive detection and monitoring of these tumors. Here, we seek to provide a perspective on the molecular background of LM and LMS, recognizing their distinct molecular features that may lead to improved diagnosis and personalized treatments, which would have a measurable impact on women's reproductive health.


Subject(s)
Leiomyoma/diagnosis , Leiomyoma/genetics , Leiomyosarcoma/diagnosis , Leiomyosarcoma/genetics , Uterine Neoplasms/diagnosis , Uterine Neoplasms/genetics , Circulating Tumor DNA/blood , Circulating Tumor DNA/genetics , Diagnosis, Differential , Female , High-Throughput Nucleotide Sequencing , Humans , Liquid Biopsy , Molecular Diagnostic Techniques , Mutation , Precision Medicine
13.
Stem Cell Res Ther ; 9(1): 333, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30486855

ABSTRACT

BACKGROUND: Myometrium, the muscular wall of the uterus, is an active organ markedly remodeled during a woman's reproductive life, especially during pregnancy. Different studies using the 5-bromo-2'-deoxyuridine and side population methods in murine and human myometrium have suggested the presence of somatic stem cells in this tissue because of its remarkable regenerative capacity. Recently, our group has developed a surface-marker (Stro1/CD44)-specific approach to isolate and characterize myometrial somatic stem cells (SSCs) from humans and rats. OBJECTIVE: In this study, we aimed to identify and localize the putative myometrial stem cell population in the murine uterus by using the specific surface markers, Nanog/CD44. METHODS: Uteri from OCT4-GFP transgenic mice at different early-life time points were analyzed via single and double immunohistochemistry to co-localize myometrial stem cell marker CD44 with other general stemmness markers, e.g., Nanog and Oct-4. Finally, we correlated the frequency of myometrial stem cells in vivo with the expression of sex steroid hormone receptors, estrogen receptor α (ERα), and progesterone receptors A and B (PR A&B). RESULTS: Nanog+/CD44+ stem cells were present in murine myometrium. Both stem cell markers were shown to co-localize with Oct-4 expression. Time-course experiments demonstrated that their percentages were significantly lower at the pre-sexual age of 1 week than at the sexually mature ages of 3 to 24 weeks. Importantly, both ERα and PR A&B were abundantly expressed in the myometrium at ages 1, 3 and 4 weeks. CONCLUSIONS: We demonstrated that murine CD44+ myometrial cells have features of somatic stem cells with the expression of typical undifferentiated markers. Furthermore, our results suggest that myometrial stem cells are sex steroid hormone dependent, likely via paracrine pathway, and increase in numbers with reproductive maturity and rise in serum estrogen and progesterone levels around 3 weeks of age in mice. The abundance and early onset expression of ER/PR emphasize the vulnerability of neonatal myometrium to environmental endocrine disruptors which can potentially lead to permanent reprograming and adult onset of myometrial disorders such as uterine fibroids.


Subject(s)
Green Fluorescent Proteins/metabolism , Myometrium/cytology , Octamer Transcription Factor-3/metabolism , Stem Cells/cytology , Animals , Female , Hyaluronan Receptors/metabolism , Mice, Transgenic , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism
14.
Hum Reprod Update ; 24(6): 673-693, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30239705

ABSTRACT

BACKGROUND: Stem cell research in the endometrium and myometrium from animal models and humans has led to the identification of endometrial/myometrial stem cells and their niches. This basic knowledge is beginning to be translated to clinical use for incurable uterine pathologies. Additionally, the implication of bone marrow-derived stem cells (BMDSCs) in uterine physiology has opened the field for the exploration of an exogenous and autologous source of stem cells. OBJECTIVE AND RATIONALE: In this review, we outline the progress of endometrial and myometrial stem/progenitor cells in both human and mouse models from their characterization to their clinical application, indicating roles in Asherman syndrome, atrophic endometrium and tissue engineering, among others. SEARCH METHODS: A comprehensive search of PubMed and Google Scholar up to December 2017 was conducted to identify peer-reviewed literature related to the contribution of bone marrow, endometrial and myometrial stem cells to potential physiological regeneration as well as their implications in pathologies of the human uterus. OUTCOMES: The discovery and main characteristics of stem cells in the murine and human endometrium and myometrium are presented together with the relevance of their niches and cross-regulation. The current state of advanced stem cell therapy using BMDSCs in the treatment of Asherman syndrome and atrophic endometrium is analyzed. In the myometrium, the understanding of genetic and epigenetic defects that result in the development of tumor-initiating cells in the myometrial stem niche and thus contribute to the growth of uterine leiomyoma is also presented. Finally, recent advances in tissue engineering based on the creation of novel three-dimensional scaffolds or decellularisation open up new perspectives for the field of uterine transplantation. WIDER IMPLICATIONS: More than a decade after their discovery, the knowledge of uterine stem cells and their niches is crystalising into novel therapeutic approaches aiming to treat with cells those conditions that cannot be cured with drugs, particularly the currently incurable uterine pathologies. Additional work and improvements are needed, but the basis has been formed for this therapeutic application of uterine cells.


Subject(s)
Endometrium/cytology , Myometrium/cytology , Stem Cell Transplantation/methods , Stem Cells/physiology , Animals , Endometrium/physiology , Female , Humans , Leiomyoma/pathology , Mice , Models, Animal , Myometrium/physiology , Translational Research, Biomedical/methods , Uterine Diseases/pathology , Uterine Diseases/therapy
15.
Am J Pathol ; 188(10): 2293-2306, 2018 10.
Article in English | MEDLINE | ID: mdl-30075150

ABSTRACT

Uterine fibroids (UFs) are clonal, hormonally regulated, benign smooth-muscle myometrial tumors that severely affect female reproductive health, although their unknown etiology limits effective care. UFs occur fourfold more commonly in African American women than in Caucasian women, and African American women generally have earlier disease onset and greater UF tumor burden, although the mechanism of this ethnic disparity has not been identified. Recent findings have linked cancer (ie, tumor) risk to increased tissue-specific stem cell division and self-renewal and suggest that somatic mutations in myometrial stem cells (MyoSCs) convert them into tumor-initiating cells, leading to UF. Specifically, preliminary results in paraffin-embedded myometrial tissues have shown increased STRO-1+/CD44+ MyoSCs in African American versus Caucasian women. Using specific methods of flow cytometry and automated quantitative pathology imaging, a large cohort of myometrial samples were investigated to determine how the STRO-1+/CD44+ MyoSCs change with regard to a patient's race, age, parity, fibroid and hormone statuses, and the location of UFs within the uterus. We confirmed that the STRO-1+/CD44+ MyoSC population is expanded in African American women, is correlated with parity and fibroid number, and fluctuates with cyclic menstrual cycle hormone changes and age. Our data suggest that an expanded MyoSC population increases the formation of tumor-initiating cells, ultimately contributing to increased UF prevalence and burden in African American women.


Subject(s)
Leiomyoma/ethnology , Myometrium/pathology , Precancerous Conditions/ethnology , Uterine Neoplasms/ethnology , Adult , Black or African American/ethnology , Age Factors , Antigens, Surface/metabolism , Cell Proliferation/physiology , Female , Hormones/physiology , Humans , Hyaluronan Receptors/metabolism , Leiomyoma/pathology , Middle Aged , Parity , Precancerous Conditions/pathology , Pregnancy , Stem Cells/pathology , Stem Cells/physiology , Uterine Neoplasms/pathology , White People/ethnology
16.
Biol Reprod ; 99(4): 735-748, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29688260

ABSTRACT

Despite the major negative impact uterine fibroids (UFs) have on female reproductive health, little is known about early events that initiate development of these tumors. Somatic fibroid-causing mutations in mediator complex subunit 12 (MED12), the most frequent genetic alterations in UFs (up to 85% of tumors), are implicated in transforming normal myometrial stem cells (MSCs) into tumor-forming cells, though the underlying mechanism(s) leading to these mutations remains unknown. It is well accepted that defective DNA repair increases the risk of acquiring tumor-driving mutations, though defects in DNA repair have not been explored in UF tumorigenesis. In the Eker rat UF model, a germline mutation in the Tsc2 tumor suppressor gene predisposes to UFs, which arise due to "second hits" in the normal allele of this gene. Risk for developing these tumors is significantly increased by early-life exposure to endocrine-disrupting chemicals (EDCs), suggesting increased UF penetrance is modulated by early drivers for these tumors. We analyzed DNA repair capacity using analyses of related gene and protein expression and DNA repair function in MSCs from adult rats exposed during uterine development to the model EDC diethylstilbestrol. Adult MSCs isolated from developmentally exposed rats demonstrated decreased DNA end-joining ability, higher levels of DNA damage, and impaired ability to repair DNA double-strand breaks relative to MSCs from age-matched, vehicle-exposed rats. These data suggest that early-life developmental EDC exposure alters these MSCs' ability to repair and reverse DNA damage, providing a driver for acquisition of mutations that may promote the development of these tumors in adult life.


Subject(s)
DNA Repair/drug effects , Endocrine Disruptors/toxicity , Leiomyoma/etiology , Myometrium/drug effects , Myometrium/metabolism , Uterine Neoplasms/etiology , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Animals , Cellular Reprogramming/drug effects , Cellular Reprogramming/genetics , DNA Damage , DNA Repair/genetics , Diethylstilbestrol/toxicity , Disease Models, Animal , Female , Germ-Line Mutation , Humans , Leiomyoma/genetics , Leiomyoma/metabolism , Mediator Complex/genetics , Myometrium/growth & development , Rats , Rats, Mutant Strains , Tuberous Sclerosis Complex 2 Protein/genetics , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism
17.
Int J Womens Health ; 9: 607-617, 2017.
Article in English | MEDLINE | ID: mdl-28919823

ABSTRACT

Uterine anatomy and uterine fibroids (UFs) characteristics have been classically considered as almost a unique issue in gynecology and reproductive medicine. Nowadays, the management of UF pathology is undergoing an important evolution, with the patient's quality of life being the most important aspect to consider. Accordingly, surgical techniques and aggressive treatments are reserved for only those cases with heavy symptomatology, while the clinical diagnostic based on size and number of UFs remains in a second plane in these situations. Moreover, the development of several noninvasive surgical techniques, especially the appearance of ulipristal acetate as a medical etiological treatment, has substantially changed the clinical indications. As a consequence, after almost 2 decades without relevant updates, it has been necessary to update the protocols for the management of UFs in the Spanish Society of Gynecology and Obstetrics twice. Accordingly, we believe that it is necessary to translate our experience to protocolize the medical care for patients with UFs, incorporating these new therapeutic options, and selecting the best treatment for them. We highlight the importance of achieving the patient's goals and decisions by improving the clinical diagnosis for these type of pathologies, allowing enhanced personalized treatments, as well as the reduction of potential risks and unnecessary surgeries.

18.
Biol Reprod ; 96(1): 70-80, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28395335

ABSTRACT

Regulation of myometrial functions during pregnancy has been considered the result of the integration of endocrine and mechanical signals. Nevertheless, uterine regeneration is poorly understood, and the cellular source within the gravid uterus is largely unexplored.In this study, we isolated and quantified the myometrial stem cells (MSC) population from pregnant female Eker rat uteri, by using Stro1/CD44 surface markers. We demonstrated that prior parity significantly increased the percentage of Stro1+/CD44+ MSC because of injured tissue response. Interestingly, we established that Stro1+/CD44+ MSC respond efficiently to physiological cues when they were treated in vitro under different dose-dependent pregnant rat serum.Previous studies reveal strong regulatory links between O2 availability and stem cell function. Based on these premises, cell proliferation assays showed that isolated Stro1+/CD44+ MSC possess a higher proliferative rate under hypoxic versus normoxic conditions. We also detected a total of 37 upregulated and 44 downregulated hypoxia-related genes, which were differentially expressed in Stro1+/CD44+ MSC, providing an alternative approach to infer into complex molecular mechanisms such as energy metabolism, inflammatory response, uterine expansion, and/or remodeling.Since these cells preferentially grow under low oxygen conditions, we propose that the increase of the rat uterus during pregnancy involves myometrial oxygen consumption, thereby enhancing MSC proliferation. Moreover, pregnancy-induced mechanical stretching results in hypoxic conditions, ultimately creating an environment that promotes stem cell proliferation and further uterine enlargement, which is essential for a successful pregnancy. In summary, all of these data support that rat Stro1+/CD44+ MSC contribute to uterine enlargement during pregnancy.


Subject(s)
Myometrium/physiology , Pregnancy, Animal/physiology , Stem Cells/physiology , Animals , Antigens, Surface/metabolism , Cell Proliferation , Female , Hyaluronan Receptors/metabolism , Hypoxia/metabolism , Pregnancy , Rats
19.
Stem Cells ; 35(3): 666-678, 2017 03.
Article in English | MEDLINE | ID: mdl-27739139

ABSTRACT

Despite the high prevalence and major negative impact of uterine fibroids (UFs) on women's health, their pathogenesis remains largely unknown. While tumor-initiating cells have been previously isolated from UFs, the cell of origin for these tumors in normal myometrium has not been identified. We isolated cells with Stro1/CD44 surface markers from normal myometrium expressing stem cell markers Oct-4/c-kit/nanog that exhibited the properties of myometrial stem/progenitor-like cells (MSCs). Using a murine model for UFs, we showed that the cervix was a hypoxic "niche" and primary site (96%) for fibroid development in these animals. The pool size of these MSCs also responded to environmental cues, contracting with age and expanding in response to developmental environmental exposures that promote fibroid development. Translating these findings to women, the number of MSCs in unaffected human myometrium correlated with risk for developing UFs. Caucasian (CC) women with fibroids had increased numbers of MSCs relative to CC women without fibroids, and African-American (AA) women at highest risk for these tumors had the highest number of MSCs: AA-with fibroids > CC-with fibroids > AA-without fibroids > CC-without fibroids. These data identify Stro1+ /CD44+ MSCs as MSC/progenitor cell for UFs, and a target for ethnic and environmental factors that increase UF risk. Stem Cells 2017;35:666-678.


Subject(s)
Carcinogenesis/pathology , Cell Compartmentation , Endocrine Disruptors/toxicity , Leiomyoma/pathology , Myometrium/pathology , Aging , Animals , Antigens, Surface/metabolism , Biomarkers, Tumor/metabolism , Carcinogenesis/drug effects , Cell Count , Environmental Exposure , Female , Hormones/pharmacology , Humans , Hyaluronan Receptors/metabolism , Mice, Inbred NOD , Mice, SCID , Oxygen/pharmacology , Rats , Risk Factors , Steroids/pharmacology , Uterine Neoplasms/pathology
20.
Reprod Sci ; 24(9): 1235-1244, 2017 09.
Article in English | MEDLINE | ID: mdl-27872195

ABSTRACT

Uterine fibroids are the most common gynecologic tumors with a significant medical and financial burden. Several genetic, hormonal, and biological factors have been shown to contribute to the development and growth of fibroid tumors. Of these factors, estrogen is particularly critical since fibroids are considered estrogen dependent because no prepubertal cases have been described in the literature and tumors tend to regress after menopause. Understanding the role of estrogen in fibroids is not only important for understanding the pathobiology of fibroids but also for the development of successful therapeutics. In this review, we discuss the types and structure of estrogen receptors (nuclear and membrane bound, including α and ß receptors and G protein-coupled estrogen receptor 1 GPER1). Estrogen-signaling pathways in fibroids include genomic (direct and indirect) and nongenomic including Ras-Raf-MEK (MAPK/Erk Kinase)-mitogen-activated protein kinase (MAPK) and phosphatidylinositide 3-kinase (PI3K)-phosphatidylinositol-3,4,5-trisphosphate (PIP3)-Akt (Protein kinase B)-mammalian target of rapamycin (mTOR) pathways; shortly Ras-Raf-MEK-MAPK and PI3K-PIP3-Akt-mTOR pathways. Several aberrations in estrogen receptors and signaling pathways are implicated in fibroid pathobiology. Current therapeutic and research agents targeting ERs/signaling include gonadotropin-releasing hormone (GnRH) agonists, GnRH antagonists, aromatase inhibitors, selective ER modulators, gene therapy, and others. Future research can identify potential targets for the development of novel treatments. In particular, epigenomics of estrogen activity and individualized (precision) medicine appear to be attractive areas for future research.


Subject(s)
Leiomyoma/metabolism , Receptors, Estrogen/metabolism , Signal Transduction/physiology , Uterine Neoplasms/metabolism , Animals , Female , Humans , Leiomyoma/pathology , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Uterine Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...