LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 32

Search options

  1. Article ; Online: Molecular and Cellular Insights into the Development of Uterine Fibroids.

    Machado-Lopez, Alba / Simón, Carlos / Mas, Aymara

    International journal of molecular sciences

    2021  Volume 22, Issue 16

    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 ... ...

    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.
    MeSH term(s) Animals ; Epigenesis, Genetic ; Female ; Gene Expression Regulation, Neoplastic ; Genetic Predisposition to Disease ; Humans ; Leiomyoma/etiology ; Leiomyoma/genetics ; Leiomyoma/pathology ; Mutation ; Uterine Neoplasms/etiology ; Uterine Neoplasms/genetics ; Uterine Neoplasms/pathology ; Uterus/metabolism ; Uterus/pathology
    Language English
    Publishing date 2021-08-06
    Publishing country Switzerland
    Document type Journal Article ; Review
    ZDB-ID 2019364-6
    ISSN 1422-0067 ; 1422-0067 ; 1661-6596
    ISSN (online) 1422-0067
    ISSN 1422-0067 ; 1661-6596
    DOI 10.3390/ijms22168483
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: Epigenetic Modulation of Inflammatory Pathways in Myometrial Stem Cells and Risk of Uterine Fibroids.

    Yang, Qiwei / Ali, Mohamed / Treviño, Lindsey S / Mas, Aymara / Ismail, Nahed / Al-Hendy, Ayman

    International journal of molecular sciences

    2023  Volume 24, Issue 14

    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, ... ...

    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.
    MeSH term(s) Female ; Humans ; Myometrium/metabolism ; Leiomyoma/genetics ; Leiomyoma/metabolism ; Stem Cells/metabolism ; Hormones/metabolism ; Epigenesis, Genetic ; Uterine Neoplasms/genetics
    Chemical Substances Hormones
    Language English
    Publishing date 2023-07-19
    Publishing country Switzerland
    Document type Journal Article
    ZDB-ID 2019364-6
    ISSN 1422-0067 ; 1422-0067 ; 1661-6596
    ISSN (online) 1422-0067
    ISSN 1422-0067 ; 1661-6596
    DOI 10.3390/ijms241411641
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Developmental reprogramming of myometrial stem cells by endocrine disruptor linking to risk of uterine fibroids.

    Yang, Qiwei / Ali, Mohamed / Treviño, Lindsey S / Mas, Aymara / Al-Hendy, Ayman

    Cellular and molecular life sciences : CMLS

    2023  Volume 80, Issue 9, Page(s) 274

    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 ...

    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.
    MeSH term(s) Animals ; Rats ; Endocrine Disruptors/toxicity ; Estrogens ; Biological Assay ; Leiomyoma/chemically induced ; Leiomyoma/genetics ; Myeloid-Lymphoid Leukemia Protein ; DNA
    Chemical Substances Endocrine Disruptors ; Estrogens ; Myeloid-Lymphoid Leukemia Protein (149025-06-9) ; DNA (9007-49-2)
    Language English
    Publishing date 2023-08-31
    Publishing country Switzerland
    Document type Journal Article
    ZDB-ID 1358415-7
    ISSN 1420-9071 ; 1420-682X
    ISSN (online) 1420-9071
    ISSN 1420-682X
    DOI 10.1007/s00018-023-04919-0
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Molecular differential diagnosis of uterine leiomyomas and leiomyosarcomas.

    Mas, Aymara / Simón, Carlos

    Biology of reproduction

    2018  Volume 101, Issue 6, Page(s) 1115–1123

    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 ... ...

    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.
    MeSH term(s) Circulating Tumor DNA/blood ; Circulating Tumor DNA/genetics ; Diagnosis, Differential ; Female ; High-Throughput Nucleotide Sequencing ; Humans ; Leiomyoma/diagnosis ; Leiomyoma/genetics ; Leiomyosarcoma/diagnosis ; Leiomyosarcoma/genetics ; Liquid Biopsy ; Molecular Diagnostic Techniques ; Mutation ; Precision Medicine ; Uterine Neoplasms/diagnosis ; Uterine Neoplasms/genetics
    Chemical Substances Circulating Tumor DNA
    Language English
    Publishing date 2018-09-05
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Review
    ZDB-ID 1118-6
    ISSN 1529-7268 ; 0006-3363
    ISSN (online) 1529-7268
    ISSN 0006-3363
    DOI 10.1093/biolre/ioy195
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Molecular Management of High-Grade Serous Ovarian Carcinoma.

    Punzón-Jiménez, Paula / Lago, Victor / Domingo, Santiago / Simón, Carlos / Mas, Aymara

    International journal of molecular sciences

    2022  Volume 23, Issue 22

    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 ... ...

    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.
    MeSH term(s) Female ; Humans ; Ovarian Neoplasms/diagnosis ; Ovarian Neoplasms/genetics ; Ovarian Neoplasms/therapy ; Carcinoma, Ovarian Epithelial
    Language English
    Publishing date 2022-11-09
    Publishing country Switzerland
    Document type Journal Article ; Review
    ZDB-ID 2019364-6
    ISSN 1422-0067 ; 1422-0067 ; 1661-6596
    ISSN (online) 1422-0067
    ISSN 1422-0067 ; 1661-6596
    DOI 10.3390/ijms232213777
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: The ontogeny of myometrial stem cells in OCT4-GFP transgenic mouse model.

    Brakta, Soumia / Mas, Aymara / Al-Hendy, Ayman

    Stem cell research & therapy

    2018  Volume 9, Issue 1, Page(s) 333

    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 ...

    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
    Conclusions: We demonstrated that murine CD44
    MeSH term(s) Animals ; Female ; Green Fluorescent Proteins/metabolism ; Hyaluronan Receptors/metabolism ; Mice, Transgenic ; Myometrium/cytology ; Octamer Transcription Factor-3/metabolism ; Receptors, Estrogen/metabolism ; Receptors, Progesterone/metabolism ; Stem Cells/cytology
    Chemical Substances Hyaluronan Receptors ; Octamer Transcription Factor-3 ; Receptors, Estrogen ; Receptors, Progesterone ; Green Fluorescent Proteins (147336-22-9)
    Language English
    Publishing date 2018-11-29
    Publishing country England
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ZDB-ID 2548671-8
    ISSN 1757-6512 ; 1757-6512
    ISSN (online) 1757-6512
    ISSN 1757-6512
    DOI 10.1186/s13287-018-1079-7
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: Effect of aging on the human myometrium at single-cell resolution.

    Punzon-Jimenez, Paula / Machado-Lopez, Alba / Perez-Moraga, Raul / Llera-Oyola, Jaime / Grases, Daniela / Galvez-Viedma, Marta / Sibai, Mustafa / Satorres-Perez, Elena / Lopez-Agullo, Susana / Badenes, Rafael / Ferrer-Gomez, Carolina / Porta-Pardo, Eduard / Roson, Beatriz / Simon, Carlos / Mas, Aymara

    Nature communications

    2024  Volume 15, Issue 1, Page(s) 945

    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 ... ...

    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.
    MeSH term(s) Pregnancy ; Humans ; Female ; Myometrium/metabolism ; Labor, Obstetric/genetics ; Labor, Obstetric/metabolism ; Muscle, Smooth ; Aging/genetics ; Muscle Contraction
    Language English
    Publishing date 2024-01-31
    Publishing country England
    Document type Journal Article
    ZDB-ID 2553671-0
    ISSN 2041-1723 ; 2041-1723
    ISSN (online) 2041-1723
    ISSN 2041-1723
    DOI 10.1038/s41467-024-45143-z
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: The human periconceptional maternal-embryonic space in health and disease.

    Moreno, Inmaculada / Capalbo, Antonio / Mas, Aymara / Garrido-Gomez, Tamara / Roson, Beatriz / Poli, Maurizio / Dimitriadis, Eva / Santamaria, Xavier / Vilella, Felipe / Simon, Carlos

    Physiological reviews

    2023  Volume 103, Issue 3, Page(s) 1965–2038

    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 ... ...

    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.
    MeSH term(s) Pregnancy ; Female ; Child ; Infant, Newborn ; Humans ; Embryo Implantation ; Endometrium ; Blastocyst ; Placenta
    Language English
    Publishing date 2023-02-16
    Publishing country United States
    Document type Journal Article ; Review ; Research Support, Non-U.S. Gov't
    ZDB-ID 209902-0
    ISSN 1522-1210 ; 0031-9333
    ISSN (online) 1522-1210
    ISSN 0031-9333
    DOI 10.1152/physrev.00050.2021
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: Uterine leiomyoma with RAD51B::NUDT3 fusion: a report of 2 cases.

    Dundr, Pavel / Machado-Lopez, Alba / Mas, Aymara / Věcková, Zuzana / Mára, Michal / Richtárová, Adéla / Matěj, Radoslav / Stružinská, Ivana / Kendall Bártů, Michaela / Němejcová, Kristýna / Dvořák, Jiří / Hojný, Jan

    Virchows Archiv : an international journal of pathology

    2023  

    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 ... ...

    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.
    Language English
    Publishing date 2023-07-19
    Publishing country Germany
    Document type Journal Article
    ZDB-ID 1184867-4
    ISSN 1432-2307 ; 0945-6317
    ISSN (online) 1432-2307
    ISSN 0945-6317
    DOI 10.1007/s00428-023-03603-9
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article ; Online: Uterine stem cells: from basic research to advanced cell therapies.

    Santamaria, Xavier / Mas, Aymara / Cervelló, Irene / Taylor, Hugh / Simon, Carlos

    Human reproduction update

    2018  Volume 24, Issue 6, Page(s) 673–693

    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 ... ...

    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.
    MeSH term(s) Animals ; Endometrium/cytology ; Endometrium/physiology ; Female ; Humans ; Leiomyoma/pathology ; Mice ; Models, Animal ; Myometrium/cytology ; Myometrium/physiology ; Stem Cell Transplantation/methods ; Stem Cells/physiology ; Translational Medical Research/methods ; Uterine Diseases/pathology ; Uterine Diseases/therapy
    Language English
    Publishing date 2018-11-12
    Publishing country England
    Document type Journal Article
    ZDB-ID 1286738-x
    ISSN 1460-2369 ; 1355-4786
    ISSN (online) 1460-2369
    ISSN 1355-4786
    DOI 10.1093/humupd/dmy028
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top