1
|
Rizo JA, Ahmad V, Pru JM, Winuthayanon S, Challa S, Kim TH, Jeong JW, Spencer TE, Kelleher AM. Uterine organoids reveal insights into epithelial specification and plasticity in development and disease. Proc Natl Acad Sci U S A 2025; 122:e2422694122. [PMID: 39883834 PMCID: PMC11804710 DOI: 10.1073/pnas.2422694122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/26/2024] [Indexed: 02/01/2025] Open
Abstract
Understanding how epithelial cells in the female reproductive tract (FRT) differentiate is crucial for reproductive health, yet the underlying mechanisms remain poorly defined. At birth, FRT epithelium is highly malleable, allowing differentiation into various epithelial types, but the regulatory pathways guiding these early cell fate decisions are unclear. Here, we use neonatal mouse endometrial organoids and assembloid coculture models to investigate how innate cellular plasticity and external mesenchymal signals influence epithelial differentiation. Our findings demonstrate that uterine epithelium undergoes marked age-dependent changes, transitioning from a highly plastic state capable of forming both monolayered and multilayered structures to a more restricted fate as development progresses. Interestingly, parallels emerge between the developmental plasticity of neonatal uterine epithelium and pathological conditions such as endometrial cancer, where similar regulatory mechanisms may reactivate, driving abnormal epithelial differentiation and tumorigenesis. These results not only deepen our understanding of early uterine development but also offer a valuable model for studying the progression of reproductive diseases and cancers.
Collapse
Affiliation(s)
- Jason A. Rizo
- Division of Animal Sciences, University of Missouri, Columbia, MO65211
| | - Vakil Ahmad
- Division of Animal Sciences, University of Missouri, Columbia, MO65211
| | - Jacob M. Pru
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO65211
| | - Sarayut Winuthayanon
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO65211
| | - Sridevi Challa
- The University of Chicago Comprehensive Cancer Center, The University of Chicago, Chicago, IL60637
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, IL60637
| | - Tae Hoon Kim
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO65211
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO65211
| | - Thomas E. Spencer
- Division of Animal Sciences, University of Missouri, Columbia, MO65211
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO65211
| | - Andrew M. Kelleher
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO65211
| |
Collapse
|
2
|
Jia S, Zhao F. Decoding Müllerian Duct Epithelial Regionalization. Mol Reprod Dev 2025; 92:e70018. [PMID: 39994938 PMCID: PMC11850963 DOI: 10.1002/mrd.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025]
Abstract
Müllerian ducts (MD), also known as paramesonephric ducts, are the primordial anlage of the female reproductive tract organs including the oviduct, uterus, cervix and upper vagina along the craniocaudal axis. Although the general architecture of MD-derived organs is conserved, each organ possesses their unique epithelial structures and cell types to confer their region-specific functions, which collectively coordinate successful fertilization and pregnancy. MD epithelial fate decisions and differentiation along the craniocaudal axis is dependent on spatiotemporal regulation of intrinsic transcription factors and extrinsic signals derived from the mesenchyme. Findings from genetic mouse models, single-cell sequencing studies, and organoid cultures have significantly advanced our understanding of the cellular and molecular mechanisms of MD regionalization. In this review, we first discuss the diversity of epithelial morphologies and cell types in the female reproductive tract organs. Then, we discuss the roles of key transcription factors (Hox, transcriptional cascade driving multiciliogenesis, Foxa2, and P63), signaling pathways (estrogen/ESR1, Wnt/β-catenin, hedgehog, and retinoic acid), and epigenetic factors (microRNAs, chromatin remodeling factors, and histone modification enzymes) in region-specific MD differentiation. Further deciphering molecular mechanisms of MD craniocaudal patterning will open new avenues to improve our strategies for prevention, diagnosis, and treatment of Müllerian anomalies and female reproductive tract disorders.
Collapse
Affiliation(s)
- Shuai Jia
- Department of Comparative Biosciences, School of Veterinary MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Fei Zhao
- Department of Comparative Biosciences, School of Veterinary MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| |
Collapse
|
3
|
Wu X, Tian Y, Zhu H, Xu P, Zhang J, Hu Y, Ji X, Yan R, Yue H, Sang N. Invisible Hand behind Female Reproductive Disorders: Bisphenols, Recent Evidence and Future Perspectives. TOXICS 2023; 11:1000. [PMID: 38133401 PMCID: PMC10748066 DOI: 10.3390/toxics11121000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023]
Abstract
Reproductive disorders are considered a global health problem influenced by physiological, genetic, environmental, and lifestyle factors. The increased exposure to bisphenols, a chemical used in large quantities for the production of polycarbonate plastics, has raised concerns regarding health risks in humans, particularly their endocrine-disrupting effects on female reproductive health. To provide a basis for future research on environmental interference and reproductive health, we reviewed relevant studies on the exposure patterns and levels of bisphenols in environmental matrices and humans (including susceptible populations such as pregnant women and children). In addition, we focused on in vivo, in vitro, and epidemiological studies evaluating the effects of bisphenols on the female reproductive system (the uterus, ovaries, fallopian tubes, and vagina). The results indicate that bisphenols cause structural and functional damage to the female reproductive system by interfering with hormones; activating receptors; inducing oxidative stress, DNA damage, and carcinogenesis; and triggering epigenetic changes, with the damaging effects being intergenerational. Epidemiological studies support the association between bisphenols and diseases such as cancer of the female reproductive system, reproductive dysfunction, and miscarriage, which may negatively affect the establishment and maintenance of pregnancy. Altogether, this review provides a reference for assessing the adverse effects of bisphenols on female reproductive health.
Collapse
Affiliation(s)
- Xiaoyun Wu
- Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan 030006, China; (X.W.); (Y.T.); (H.Z.); (P.X.); (J.Z.); (Y.H.); (N.S.)
| | - Yuchai Tian
- Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan 030006, China; (X.W.); (Y.T.); (H.Z.); (P.X.); (J.Z.); (Y.H.); (N.S.)
| | - Huizhen Zhu
- Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan 030006, China; (X.W.); (Y.T.); (H.Z.); (P.X.); (J.Z.); (Y.H.); (N.S.)
| | - Pengchong Xu
- Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan 030006, China; (X.W.); (Y.T.); (H.Z.); (P.X.); (J.Z.); (Y.H.); (N.S.)
| | - Jiyue Zhang
- Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan 030006, China; (X.W.); (Y.T.); (H.Z.); (P.X.); (J.Z.); (Y.H.); (N.S.)
| | - Yangcheng Hu
- Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan 030006, China; (X.W.); (Y.T.); (H.Z.); (P.X.); (J.Z.); (Y.H.); (N.S.)
| | - Xiaotong Ji
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China;
| | - Ruifeng Yan
- Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan 030006, China; (X.W.); (Y.T.); (H.Z.); (P.X.); (J.Z.); (Y.H.); (N.S.)
| | - Huifeng Yue
- Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan 030006, China; (X.W.); (Y.T.); (H.Z.); (P.X.); (J.Z.); (Y.H.); (N.S.)
| | - Nan Sang
- Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan 030006, China; (X.W.); (Y.T.); (H.Z.); (P.X.); (J.Z.); (Y.H.); (N.S.)
| |
Collapse
|
4
|
Rizo JA, Davenport KM, Winuthayanon W, Spencer TE, Kelleher AM. Estrogen receptor alpha regulates uterine epithelial lineage specification and homeostasis. iScience 2023; 26:107568. [PMID: 37622003 PMCID: PMC10445454 DOI: 10.1016/j.isci.2023.107568] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/11/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023] Open
Abstract
Postnatal development of the uterus involves specification of undifferentiated epithelium into uterine-type epithelium. That specification is regulated by stromal-epithelial interactions as well as intrinsic cell-specific transcription factors and gene regulatory networks. This study utilized mouse genetic models of Esr1 deletion, endometrial epithelial organoids (EEO), and organoid-stromal co-cultures to decipher the role of Esr1 in uterine epithelial development. Organoids derived from wild-type (WT) mice developed a normal single layer of columnar epithelium. In contrast, EEO from Esr1 null mice developed a multilayered stratified squamous type of epithelium with basal cells. Co-culturing Esr1 null epithelium with WT uterine stromal fibroblasts inhibited basal cell development. Of note, estrogen treatment of EEO-stromal co-cultures and Esr1 conditional knockout mice increased basal epithelial cell markers. Collectively, these findings suggest that Esr1 regulates uterine epithelium lineage plasticity and homeostasis and loss of ESR1 promotes altered luminal-to-basal differentiation driven by ESR1-mediated paracrine factors from the stroma.
Collapse
Affiliation(s)
- Jason A. Rizo
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | | | - Wipawee Winuthayanon
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO 65211, USA
| | - Thomas E. Spencer
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO 65211, USA
| | - Andrew M. Kelleher
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
5
|
Kimura E, Mongan M, Xiao B, Wang J, Carreira VS, Bolon B, Zhang X, Burns KA, Biesiada J, Medvedovic M, Puga A, Xia Y. The Role of MAP3K1 in the Development of the Female Reproductive Tract. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023. [PMID: 37131749 PMCID: PMC10153227 DOI: 10.1101/2023.04.20.537715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Mitogen-Activated Protein 3 Kinase 1 (MAP3K1) is a dynamic signaling molecule with a plethora of cell-type specific functions, most of which are yet to be understood. Here we describe a role for MAP3K1 in the development of female reproductive tract (FRT). MAP3K1 kinase domain-deficient ( Map3k1 ΔKD ) females exhibit imperforate vagina, labor failure, and infertility. These defects correspond to a shunted Müllerian duct (MD), the principle precursor of the FRT, in embryos, while they manifest as a contorted caudal vagina with abrogated vaginal-urogenital sinus fusion in neonates. In epithelial cells, MAP3K1 acts through JNK and ERK to activate WNT, yet in vivo MAP3K1 is crucial for WNT activity in mesenchyme associated with the caudal MD. Expression of Wnt7b is high in wild type, but low in Map3k1 knockout MD epithelium and MAP3K1-deficient keratinocytes. Correspondingly, conditioned media derived from MAP3K1-competent epithelial cells activate TCF/Lef-luciferase reporter in fibroblasts, suggesting that MAP3K1-induced factors released from epithelial cells trans-activate WNT signaling in fibroblasts. Our results reveal a temporal-spatial and paracrine MAP3K1-WNT crosstalk contributing to MD caudal elongation and FRT development. Highlights MAP3K1 deficient female mice exhibit imperforate vagina and infertilityLoss of MAP3K1 kinase activity impedes Müllerian duct (MD) caudal elongation and fusion with urogenital sinus (UGS) in embryogenesisThe MAP3K1-MAPK pathway up-regulates WNT signaling in epithelial cellsMAP3K1 deficiency down-regulates Wnt7b expression in the MD epithelium and prevents WNT activity in mesenchyme of the caudal MD.
Collapse
|
6
|
McKey J, Anbarci DN, Bunce C, Ontiveros AE, Behringer RR, Capel B. Integration of mouse ovary morphogenesis with developmental dynamics of the oviduct, ovarian ligaments, and rete ovarii. eLife 2022; 11:e81088. [PMID: 36165446 PMCID: PMC9621696 DOI: 10.7554/elife.81088] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/26/2022] [Indexed: 01/29/2023] Open
Abstract
Morphogenetic events during the development of the fetal ovary are crucial to the establishment of female fertility. However, the effects of structural rearrangements of the ovary and surrounding reproductive tissues on ovary morphogenesis remain largely uncharacterized. Using tissue clearing and lightsheet microscopy, we found that ovary folding correlated with regionalization into cortex and medulla. Relocation of the oviduct to the ventral aspect of the ovary led to ovary encapsulation, and mutual attachment of the ovary and oviduct to the cranial suspensory ligament likely triggered ovary folding. During this process, the rete ovarii (RO) elaborated into a convoluted tubular structure extending from the ovary into the ovarian capsule. Using genetic mouse models in which the oviduct and RO are perturbed, we found the oviduct is required for ovary encapsulation. This study reveals novel relationships among the ovary and surrounding tissues and paves the way for functional investigation of the relationship between architecture and differentiation of the mammalian ovary.
Collapse
Affiliation(s)
- Jennifer McKey
- Department of Cell Biology, Duke University Medical CenterDurhamUnited States
| | - Dilara N Anbarci
- Department of Cell Biology, Duke University Medical CenterDurhamUnited States
| | - Corey Bunce
- Department of Cell Biology, Duke University Medical CenterDurhamUnited States
| | - Alejandra E Ontiveros
- Department of Genetics, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | - Richard R Behringer
- Department of Genetics, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | - Blanche Capel
- Department of Cell Biology, Duke University Medical CenterDurhamUnited States
| |
Collapse
|
7
|
Zhang M, Kiyono T, Aoki K, Goshima N, Kobayashi S, Hiranuma K, Shiraishi K, Saya H, Nakahara T. Development of an in vitro carcinogenesis model of human papillomavirus-induced cervical adenocarcinoma. Cancer Sci 2021; 113:904-915. [PMID: 34932848 PMCID: PMC8898731 DOI: 10.1111/cas.15246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 11/30/2022] Open
Abstract
Cervical adenocarcinoma (ADC) is the second most common pathological subtype of cervical cancer after squamous cell carcinoma. It accounts for approximately 20% of cervical cancers, and the incidence has increased in the past few decades, particularly among young patients. The persistent infection of high‐risk human papillomavirus (HPV) is responsible for most cervical ADC. However, almost all available in vitro models are designed to study the carcinogenesis of cervical squamous cell carcinoma. To gain better insights into molecular background of ADC, we aimed to establish an in vitro carcinogenesis model of ADC. We previously reported the establishment of an in vitro model for cervical squamous cell carcinoma by introducing defined viral and cellular oncogenes, HPV16 E6 and E7, c‐MYC, and activated RAS to human cervical keratinocytes. In this study, the expression of potential lineage‐specifying factors and/or SMAD4 reduction was introduced in addition to the defined four oncogenes to direct carcinogenesis toward ADC. The cell properties associated with the cell lineage were analyzed in monolayer and organoid cultures and the tumors in mouse xenografts. In the cells expressing Forkhead box A2 (FOXA2), apparent changes in cell properties were observed, such as elevated expression of columnar cell markers and decreased expression of squamous cell markers. Strikingly, the histopathology of tumors expressing FOXA2 resembled cervical ADC, proposing that FOXA2 plays a vital role in dictating the histopathology of cervical cancers.
Collapse
Affiliation(s)
- Mengzhu Zhang
- Department of Immune Medicine, National Cancer Center Research Institute, Tokyo, Japan.,Division of Gene Regulation, Institute for Advanced Medical Research, Graduate School of Medicine, Keio University, Tokyo, Japan
| | - Tohru Kiyono
- Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Kazunori Aoki
- Department of Immune Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Naoki Goshima
- Department of Human Sciences, Faculty of Human Sciences, Musashino University, Tokyo, Japan
| | - Shin Kobayashi
- Dynamic Pharmaco-Modality Research Group, Cellular and Molecular Biotechnology and Research Institute, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | - Kengo Hiranuma
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Kouya Shiraishi
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, Graduate School of Medicine, Keio University, Tokyo, Japan
| | - Tomomi Nakahara
- Department of Immune Medicine, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
8
|
Kwon DH, Malpica A, Zaleski M, Euscher ED, Ramalingam P. Immunohistochemical Loss of DPC4 in Tumors With Mucinous Differentiation Arising in or Involving the Gynecologic Tract. Int J Gynecol Pathol 2021; 40:523-532. [PMID: 33405429 DOI: 10.1097/pgp.0000000000000754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
DPC4 immunohistochemistry (IHC) is usually part of the work-up of mucinous neoplasms in the ovary where the distinction between an ovarian primary and metastatic pancreaticobiliary adenocarcinoma (PanACa) must be made. Although DPC4 IHC is lost in about 55% (46%-61%) of PanACas and typically retained in most primary ovarian mucinous neoplasms, no study has evaluated the expression of this marker in a large cohort of neoplasms arising in or involving gynecologic (GYN) organs. In this study, we retrospectively analyzed the expression of DPC4 IHC in a total of 251 tumors and lesions related to the GYN tract in which DPC4 IHC stain was performed during the initial pathology evaluation. Of these, 138 were primary GYN tumors and lesions, 31 were metastatic GYN tumors involving non-GYN sites, and 83 were metastatic non-GYN tumors involving the GYN tract. We identified 27 cases with loss of DPC4 IHC expression of which 20 cases met the inclusion criteria (i.e. clinical information was available to determine the site of tumor origin). We observed that loss of DPC4 nuclear expression was most commonly seen in tumors of endocervical origin (n=7), of which 5 were gastric-type cervical adenocarcinomas (GCxACa) and 2 were usual-type cervical adenocarcinomas, either primary or metastatic. This was followed by tumors of the pancreaticobiliary tract (n=5), ovary (n=2), and appendix (n=1). In addition, 1 gastric-type vaginal adenocarcinoma (GVaACa) also showed loss of DPC4. Our findings indicate that in female patients with mucinous neoplasms involving the ovary or other sites, with loss of DPC4 by IHC, and negative pancreaticobiliary imaging, the possibility of an occult GCx/GVaACa, and rarely an ovarian primary must be considered.
Collapse
Affiliation(s)
- Dong Hyang Kwon
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | | |
Collapse
|
9
|
Major AT, Estermann MA, Roly ZY, Smith CA. An evo-devo perspective of the female reproductive tract. Biol Reprod 2021; 106:9-23. [PMID: 34494091 DOI: 10.1093/biolre/ioab166] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 01/22/2023] Open
Abstract
The vertebrate female reproductive tract has undergone considerable diversification over evolution, having become physiologically adapted to different reproductive strategies. This review considers the female reproductive tract from the perspective of evolutionary developmental biology (evo-devo). Very little is known about how the evolution of this organ system has been driven at the molecular level. In most vertebrates, the female reproductive tract develops from paired embryonic tubes, the Müllerian ducts. We propose that formation of the Müllerian duct is a conserved process that has involved co-option of genes and molecular pathways involved in tubulogenesis in the adjacent mesonephric kidney and Wolffian duct. Downstream of this conservation, genetic regulatory divergence has occurred, generating diversity in duct structure. Plasticity of the Hox gene code and wnt signaling, in particular, may underlie morphological variation of the uterus in mammals, and evolution of the vagina. This developmental plasticity in Hox and Wnt activity may also apply to other vertebrates, generating the morphological diversity of female reproductive tracts evident today.
Collapse
Affiliation(s)
- Andrew T Major
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800. Australia
| | - Martin A Estermann
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800. Australia
| | - Zahida Y Roly
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800. Australia
| | - Craig A Smith
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800. Australia
| |
Collapse
|
10
|
Karasu S, Cilengir AH, Ocal I, Aydogmus S. Transperineal Ultrasonographic and Magnetic Resonance Imaging Findings of Vaginal Adenosis. Curr Med Imaging 2021; 18:82-85. [PMID: 34102982 DOI: 10.2174/1573405617666210608153631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/16/2021] [Accepted: 03/26/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Vaginal adenosis is a rare clinicopathological entity which refers to the presence of glandular endometrial tissue in the vaginal wall. CASE REPORT A gravida 46-year-old woman was presented to the gynecology department with symptoms of chronic pelvic pain. Maternal Diethylstilbestrol (DES) usage has been proven effective in the development of vaginal adenosis. It can also be idiopathic. Its sonographic or magnetic resonance imaging (MRI) findings have not been described in English literature previously. CONCLUSION Here, we report the clinical, transperineal ultrasonographic (US) and MRI findings of a pathologically proven case of vaginal adenosis.
Collapse
Affiliation(s)
- Sebnem Karasu
- Kâtip Celebi University, Faculty of Medicine, Ataturk Training and Research Hospital, Department of Radiology, Turkey
| | | | - Irfan Ocal
- Kâtip Celebi University, Faculty of Medicine, Ataturk Training and Research Hospital, Department of Pathology, Turkey
| | - Serpil Aydogmus
- Kâtip Celebi University, Faculty of Medicine, Ataturk Training and Research Hospital, Department of Obstetrics and Gynecology, Turkey
| |
Collapse
|
11
|
Pontecorvi P, Megiorni F, Camero S, Ceccarelli S, Bernardini L, Capalbo A, Anastasiadou E, Gerini G, Messina E, Perniola G, Benedetti Panici P, Grammatico P, Pizzuti A, Marchese C. Altered Expression of Candidate Genes in Mayer-Rokitansky-Küster-Hauser Syndrome May Influence Vaginal Keratinocytes Biology: A Focus on Protein Kinase X. BIOLOGY 2021; 10:biology10060450. [PMID: 34063745 PMCID: PMC8223793 DOI: 10.3390/biology10060450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/13/2021] [Accepted: 05/20/2021] [Indexed: 11/16/2022]
Abstract
Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome is a rare and complex disease defined by congenital aplasia of the vagina and uterus in 46,XX women, often associated with kidney and urinary tract anomalies. The aetiopathogenesis of MRKH syndrome is still largely unknown. Herein, we investigated the role of selected candidate genes in the aetiopathogenesis of MRKH syndrome, with a focus on PRKX, which encodes for protein kinase X. Through RT-qPCR analyses performed on vaginal dimple samples from patients, and principal component analysis (PCA), we highlighted a phenotype-related expression pattern of PRKX, MUC1, HOXC8 and GREB1L in MRKH patients. By using an in vitro approach, we proved that PRKX ectopic overexpression in a cell model of vaginal keratinocytes promotes cell motility through epithelial-to-mesenchymal transition (EMT) activation, a fundamental process in urogenital tract morphogenesis. Moreover, our findings showed that PRKX upregulation in vaginal keratinocytes is able to affect transcriptional levels of HOX genes, implicated in urinary and genital tract development. Our study identified the dysregulation of PRKX expression as a possible molecular cause for MRKH syndrome. Moreover, we propose the specific role of PRKX in vaginal keratinocyte biology as one of the possible mechanisms underlying this complex disease.
Collapse
Affiliation(s)
- Paola Pontecorvi
- Department of Experimental Medicine, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (F.M.); (S.C.); (E.A.); (G.G.); (E.M.); (A.P.)
| | - Francesca Megiorni
- Department of Experimental Medicine, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (F.M.); (S.C.); (E.A.); (G.G.); (E.M.); (A.P.)
| | - Simona Camero
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (S.C.); (G.P.); (P.B.P.)
| | - Simona Ceccarelli
- Department of Experimental Medicine, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (F.M.); (S.C.); (E.A.); (G.G.); (E.M.); (A.P.)
| | - Laura Bernardini
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza Foundation-Viale Cappuccini, 1, 71013 San Giovanni Rotondo (FG), Italy; (L.B.); (A.C.)
| | - Anna Capalbo
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza Foundation-Viale Cappuccini, 1, 71013 San Giovanni Rotondo (FG), Italy; (L.B.); (A.C.)
| | - Eleni Anastasiadou
- Department of Experimental Medicine, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (F.M.); (S.C.); (E.A.); (G.G.); (E.M.); (A.P.)
| | - Giulia Gerini
- Department of Experimental Medicine, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (F.M.); (S.C.); (E.A.); (G.G.); (E.M.); (A.P.)
| | - Elena Messina
- Department of Experimental Medicine, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (F.M.); (S.C.); (E.A.); (G.G.); (E.M.); (A.P.)
| | - Giorgia Perniola
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (S.C.); (G.P.); (P.B.P.)
| | - Pierluigi Benedetti Panici
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (S.C.); (G.P.); (P.B.P.)
| | - Paola Grammatico
- Division of Medical Genetics, Department of Molecular Medicine, Sapienza University of Rome-San Camillo-Forlanini Hospital, Circonvallazione Gianicolense, 87, 00152 Rome, Italy;
| | - Antonio Pizzuti
- Department of Experimental Medicine, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (F.M.); (S.C.); (E.A.); (G.G.); (E.M.); (A.P.)
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza Foundation-Viale Cappuccini, 1, 71013 San Giovanni Rotondo (FG), Italy; (L.B.); (A.C.)
| | - Cinzia Marchese
- Department of Experimental Medicine, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (F.M.); (S.C.); (E.A.); (G.G.); (E.M.); (A.P.)
- Correspondence: ; Tel.: +39-06-4997-2872
| |
Collapse
|
12
|
Vlachos DE, Protopapas A, Vlachos G, Sarris K, Sotiropoulou M, Terzi M, Loutradis D. Menstural function preservation treatment of a primary vaginal clear cell carcinoma with ovarian transposition and vaginal brachytherapy. Gynecol Oncol Rep 2021; 36:100764. [PMID: 33912644 PMCID: PMC8063755 DOI: 10.1016/j.gore.2021.100764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/22/2021] [Accepted: 03/27/2021] [Indexed: 11/27/2022] Open
Abstract
Early stage vaginal carcinomas are typically treated with radical surgical procedures or radiation therapy. Both modalities impair the reproductive ability of the patients. We hereby report a case of menstrual function preservation in a 24-year-old patient with an early-stage primary vaginal clear cell carcinoma. We treated the patient with intravaginal brachytherapy after appropriate laparoscopic surgical staging and separate transposition of the ovaries and tubes. The patient is now 6 years without any evidence of disease. She reports minor. complaints during sexual intercourse, while her menstruation and hormonic profile are normal.
Collapse
Affiliation(s)
| | | | - Georgios Vlachos
- 1st Department of Obstetrics and Gynecology, University of Athens, Greece
| | - Kyrillos Sarris
- Department of Radiotherapy, General Hospital "Alexandra", Athens, Greece
| | | | - Maria Terzi
- Department of Pathology, 'Hygeia' Diagnostic and Therapeutic Centre of Athens - Hygeia, Greece
| | - Dimitris Loutradis
- 1st Department of Obstetrics and Gynecology, University of Athens, Greece
| |
Collapse
|
13
|
Pokorná Z, Vysloužil J, Hrabal V, Vojtěšek B, Coates PJ. The foggy world(s) of p63 isoform regulation in normal cells and cancer. J Pathol 2021; 254:454-473. [PMID: 33638205 DOI: 10.1002/path.5656] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/10/2021] [Accepted: 02/24/2021] [Indexed: 12/19/2022]
Abstract
The p53 family member p63 exists as two major protein variants (TAp63 and ΔNp63) with distinct expression patterns and functional properties. Whilst downstream target genes of p63 have been studied intensively, how p63 variants are themselves controlled has been relatively neglected. Here, we review advances in understanding ΔNp63 and TAp63 regulation, highlighting their distinct pathways. TAp63 has roles in senescence and metabolism, and in germ cell genome maintenance, where it is activated post-transcriptionally by phosphorylation cascades after DNA damage. The function and regulation of TAp63 in mesenchymal and haematopoietic cells is less clear but may involve epigenetic control through DNA methylation. ΔNp63 functions to maintain stem/progenitor cells in various epithelia and is overexpressed in squamous and certain other cancers. ΔNp63 is transcriptionally regulated through multiple enhancers in concert with chromatin modifying proteins. Many signalling pathways including growth factors, morphogens, inflammation, and the extracellular matrix influence ΔNp63 levels, with inconsistent results reported. There is also evidence for reciprocal regulation, including ΔNp63 activating its own transcription. ΔNp63 is downregulated during cell differentiation through transcriptional regulation, while post-transcriptional events cause proteasomal degradation. Throughout the review, we identify knowledge gaps and highlight discordances, providing potential explanations including cell-context and cell-matrix interactions. Identifying individual p63 variants has roles in differential diagnosis and prognosis, and understanding their regulation suggests clinically approved agents for targeting p63 that may be useful combination therapies for selected cancer patients. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Zuzana Pokorná
- Research Centre of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Jan Vysloužil
- Research Centre of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Václav Hrabal
- Research Centre of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Borˇivoj Vojtěšek
- Research Centre of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Philip J Coates
- Research Centre of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Brno, Czech Republic
| |
Collapse
|
14
|
Santana Gonzalez L, Rota IA, Artibani M, Morotti M, Hu Z, Wietek N, Alsaadi A, Albukhari A, Sauka-Spengler T, Ahmed AA. Mechanistic Drivers of Müllerian Duct Development and Differentiation Into the Oviduct. Front Cell Dev Biol 2021; 9:605301. [PMID: 33763415 PMCID: PMC7982813 DOI: 10.3389/fcell.2021.605301] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/28/2021] [Indexed: 12/15/2022] Open
Abstract
The conduits of life; the animal oviducts and human fallopian tubes are of paramount importance for reproduction in amniotes. They connect the ovary with the uterus and are essential for fertility. They provide the appropriate environment for gamete maintenance, fertilization and preimplantation embryonic development. However, serious pathologies, such as ectopic pregnancy, malignancy and severe infections, occur in the oviducts. They can have drastic effects on fertility, and some are life-threatening. Despite the crucial importance of the oviducts in life, relatively little is known about the molecular drivers underpinning the embryonic development of their precursor structures, the Müllerian ducts, and their successive differentiation and maturation. The Müllerian ducts are simple rudimentary tubes comprised of an epithelial lumen surrounded by a mesenchymal layer. They differentiate into most of the adult female reproductive tract (FRT). The earliest sign of Müllerian duct formation is the thickening of the anterior mesonephric coelomic epithelium to form a placode of two distinct progenitor cells. It is proposed that one subset of progenitor cells undergoes partial epithelial-mesenchymal transition (pEMT), differentiating into immature Müllerian luminal cells, and another subset undergoes complete EMT to become Müllerian mesenchymal cells. These cells invaginate and proliferate forming the Müllerian ducts. Subsequently, pEMT would be reversed to generate differentiated epithelial cells lining the fully formed Müllerian lumen. The anterior Müllerian epithelial cells further specialize into the oviduct epithelial subtypes. This review highlights the key established molecular and genetic determinants of the processes involved in Müllerian duct development and the differentiation of its upper segment into oviducts. Furthermore, an extensive genome-wide survey of mouse knockout lines displaying Müllerian or oviduct phenotypes was undertaken. In addition to widely established genetic determinants of Müllerian duct development, our search has identified surprising associations between loss-of-function of several genes and high-penetrance abnormalities in the Müllerian duct and/or oviducts. Remarkably, these associations have not been investigated in any detail. Finally, we discuss future directions for research on Müllerian duct development and oviducts.
Collapse
Affiliation(s)
- Laura Santana Gonzalez
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Ioanna A Rota
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Developmental Immunology Research Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Mara Artibani
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom.,Gene Regulatory Networks in Development and Disease Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Matteo Morotti
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Zhiyuan Hu
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Nina Wietek
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Abdulkhaliq Alsaadi
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Ashwag Albukhari
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tatjana Sauka-Spengler
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Gene Regulatory Networks in Development and Disease Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ahmed A Ahmed
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
15
|
Nakajima T, Kozuma M, Hirasawa T, Matsunaga YT, Tomooka Y. Extracellular matrix components and elasticity regulate mouse vaginal epithelial differentiation induced by mesenchymal cells†. Biol Reprod 2021; 104:1239-1248. [PMID: 33693507 DOI: 10.1093/biolre/ioab041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/20/2021] [Accepted: 03/01/2021] [Indexed: 11/12/2022] Open
Abstract
Oviduct, uterus, and vagina are derived from Müllerian ducts. But only in the vagina, the epithelium differentiates into stratified layers. Organ-specific secreted factors derived from the stroma of a neonatal mouse induce epithelial differentiation in the female reproductive tracts. However, the effects of the components and mechanical property of extracellular matrix (ECM) on the regulation of gene expression in the mesenchymal cells of neonatal stroma and differentiation of epithelium in the female reproductive tracts have been overlooked. In the present study, we have developed a simple 3D neonatal vaginal model using clonal cell lines to study the effect of ECM's components and stiffness on the epithelial stratification. Transcriptome analysis was performed by DNA-microarray to identify the components of ECM involved in the differentiation of vaginal epithelial stratification. The knockdown experiment of the candidate genes relating to vaginal epithelial stratification was focused on fibromodulin (Fmod), a collagen cross-linking protein. FMOD was essential for the expression of Bmp4, which encodes secreted factors to induce the epithelial stratification of vaginal mesenchymal cells. Furthermore, stiffer ECM as a scaffold for epithelial cells is necessary for vaginal epithelial stratification. Therefore, the components and stiffness of ECM are both crucial for the epithelial stratification in the neonatal vagina.
Collapse
Affiliation(s)
- Tadaaki Nakajima
- Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan.,Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
| | - Miyabi Kozuma
- Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - Tomoko Hirasawa
- Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan
| | | | - Yasuhiro Tomooka
- Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan
| |
Collapse
|
16
|
New frontiers of developmental endocrinology opened by researchers connecting irreversible effects of sex hormones on developing organs. Differentiation 2020; 118:4-23. [PMID: 33189416 DOI: 10.1016/j.diff.2020.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/12/2020] [Accepted: 10/25/2020] [Indexed: 01/17/2023]
Abstract
In the early 1960's, at Professor Bern's laboratory, University of California, Berkeley) in the US, Takasugi discovered ovary-independent, persistent vaginal changes in mice exposed neonatally to estrogen, which resulted in vaginal cancer later in life. Reproductive abnormalities in rodents were reported as a result of perinatal exposure to various estrogenic chemicals. Ten years later, vaginal cancers were reported in young women exposed in utero to the synthetic estrogen diethylstilbestrol (DES) and this has been called the "DES syndrome". The developing organism is particularly sensitive to developmental exposure to estrogens inducing long-term changes in various organs including the reproductive organs. The molecular mechanism underlying the persistent vaginal changes induced by perinatal estrogen exposure was partly demonstrated. Persistent phosphorylation and sustained expression of EGF-like growth factors, lead to estrogen receptor α (ESR1) activation, and then persistent vaginal epithelial cell proliferation. Agents which are weakly estrogenic by postnatal criteria may have major developmental effects, especially during a critical perinatal period. The present review outlines various studies conducted by four generations of investigators all under the influence of Prof. Bern. The studies include reports of persistent changes induced by neonatal androgen exposure, analyses of estrogen responsive genes, factors determining epithelial differentiation in the Müllerian duct, ESR and growth factor signaling, and polyovular follicles in mammals. This review is then expanded to the studies on the effects of environmental estrogens on wildlife and endocrine disruption in Daphnids.
Collapse
|
17
|
Roly ZY, Godini R, Estermann MA, Major AT, Pocock R, Smith CA. Transcriptional landscape of the embryonic chicken Müllerian duct. BMC Genomics 2020; 21:688. [PMID: 33008304 PMCID: PMC7532620 DOI: 10.1186/s12864-020-07106-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
Background Müllerian ducts are paired embryonic tubes that give rise to the female reproductive tract in vertebrates. Many disorders of female reproduction can be attributed to anomalies of Müllerian duct development. However, the molecular genetics of Müllerian duct formation is poorly understood and most disorders of duct development have unknown etiology. In this study, we describe for the first time the transcriptional landscape of the embryonic Müllerian duct, using the chicken embryo as a model system. RNA sequencing was conducted at 1 day intervals during duct formation to identify developmentally-regulated genes, validated by in situ hybridization. Results This analysis detected hundreds of genes specifically up-regulated during duct morphogenesis. Gene ontology and pathway analysis revealed enrichment for developmental pathways associated with cell adhesion, cell migration and proliferation, ERK and WNT signaling, and, interestingly, axonal guidance. The latter included factors linked to neuronal cell migration or axonal outgrowth, such as Ephrin B2, netrin receptor, SLIT1 and class A semaphorins. A number of transcriptional modules were identified that centred around key hub genes specifying matrix-associated signaling factors; SPOCK1, HTRA3 and ADGRD1. Several novel regulators of the WNT and TFG-β signaling pathway were identified in Müllerian ducts, including APCDD1 and DKK1, BMP3 and TGFBI. A number of novel transcription factors were also identified, including OSR1, FOXE1, PRICKLE1, TSHZ3 and SMARCA2. In addition, over 100 long non-coding RNAs (lncRNAs) were expressed during duct formation. Conclusions This study provides a rich resource of new candidate genes for Müllerian duct development and its disorders. It also sheds light on the molecular pathways engaged during tubulogenesis, a fundamental process in embryonic development.
Collapse
Affiliation(s)
- Zahida Yesmin Roly
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Rasoul Godini
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Martin A Estermann
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Andrew T Major
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Roger Pocock
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Craig A Smith
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia.
| |
Collapse
|
18
|
SIX1 cooperates with RUNX1 and SMAD4 in cell fate commitment of Müllerian duct epithelium. Cell Death Differ 2020; 27:3307-3320. [PMID: 32572167 PMCID: PMC7852590 DOI: 10.1038/s41418-020-0579-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 12/18/2022] Open
Abstract
During female mammal reproductive tract development, epithelial cells of the lower Müllerian duct are committed to become stratified squamous epithelium of the vagina and ectocervix, when the expression of ΔNp63 transcription factor is induced by mesenchymal cells. The absence of ΔNp63 expression leads to adenosis, the putative precursor of vaginal adenocarcinoma. Our previous studies with genetically engineered mouse models have established that fibroblast growth factor (FGF)/mitogen-activated protein kinase (MAPK), bone morphogenetic protein (BMP)/SMAD, and activin A/runt-related transcription factor 1 (RUNX1) signaling pathways are independently required for ΔNp63 expression in Müllerian duct epithelium (MDE). Here, we report that sine oculis homeobox homolog 1 (SIX1) plays a critical role in the activation of ΔNp63 locus in MDE as a downstream transcription factor of mesenchymal signals. In the developing mouse reproductive tract, SIX1 expression was restricted to MDE within the future cervix and vagina. SIX1 expression was totally absent in SMAD4 null MDE and was reduced in RUNX1 null and FGFR2 null MDE, indicating that SIX1 is under the control of vaginal mesenchymal factors: BMP4, activin A and FGF7/10. Furthermore, Six1, Runx1, and Smad4 gene-dose-dependently activated ΔNp63 expression in MDE within the vaginal fornix. Using a mouse model of diethylstilbestrol (DES)-associated vaginal adenosis, we found DES action through epithelial estrogen receptor α (ESR1) inhibits activation of ΔNp63 locus in MDE by transcriptionally repressing SIX1 and RUNX1 in the vaginal fornix.
Collapse
|
19
|
Causal Evidence and Dispositions in Medicine and Public Health. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17061813. [PMID: 32168791 PMCID: PMC7142708 DOI: 10.3390/ijerph17061813] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 02/02/2023]
Abstract
Since the introduction of evidence-based medicine, there have been discussions about the epistemic primacy of randomised controlled trials (RCTs) for establishing causality in medicine and public health. A growing movement within philosophy of science calls instead for evidential pluralism: that we need more than one single method to investigate health outcomes. How should such evidential pluralism look in practice? How useful are the various methods available for causal inquiry? Further, how should different types of causal evidence be evaluated? This paper proposes a constructive answer and introduces a framework aimed at supporting scientists in developing appropriate methodological approaches for exploring causality. We start from the philosophical tradition that highlights intrinsic properties (dispositions, causal powers or capacities) as essential features of causality. This abstract idea has wide methodological implications. The paper explains how different methods, such as lab experiments, case studies, N-of-1 trials, case control studies, cohort studies, RCTs and patient narratives, all have some strengths and some limitations for picking out intrinsic causal properties. We explain why considering philosophy of causality is crucial for evaluating causality in the health sciences. In our proposal, we combine the various methods in a temporal process, which could then take us from an observed phenomenon (e.g., a correlation) to a causal hypothesis and, finally, to improved theoretical knowledge.
Collapse
|
20
|
Henning NF, LeDuc RD, Even KA, Laronda MM. Proteomic analyses of decellularized porcine ovaries identified new matrisome proteins and spatial differences across and within ovarian compartments. Sci Rep 2019; 9:20001. [PMID: 31882863 PMCID: PMC6934504 DOI: 10.1038/s41598-019-56454-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 12/12/2019] [Indexed: 12/14/2022] Open
Abstract
Premature ovarian insufficiency (POI) affects approximately 1% of women. We aim to understand the ovarian microenvironment, including the extracellular matrix (ECM) and associated proteins (matrisome), and its role in controlling folliculogenesis. We mapped the composition of the matrisome of porcine ovaries through the cortical compartment, where quiescent follicles reside and the medullary compartment, where the larger follicles grow and mature. To do this we sliced the ovaries, uniformly in two anatomical planes, enriched for matrisome proteins and performed bottom-up shotgun proteomic analyses. We identified 42 matrisome proteins that were significantly differentially expressed across depths, and 11 matrisome proteins that have not been identified in previous ovarian protein analyses. We validated these data for nine proteins and confirmed compartmental differences with a second processing method. Here we describe a processing and proteomic analysis pipeline that revealed spatial differences and matrisome protein candidates that may influence folliculogenesis.
Collapse
Affiliation(s)
- Nathaniel F Henning
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, USA
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, USA
| | - Richard D LeDuc
- Proteomics Center of Excellence, Northwestern University, Evanston, USA
| | - Kelly A Even
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, USA
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, USA
| | - Monica M Laronda
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, USA.
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, USA.
| |
Collapse
|
21
|
Liu N, Yin Y, Wang H, Zhou Z, Sheng X, Fu H, Guo R, Wang H, Yang J, Gong P, Ning W, Ju Z, Liu Y, Liu L. Telomere dysfunction impairs epidermal stem cell specification and differentiation by disrupting BMP/pSmad/P63 signaling. PLoS Genet 2019; 15:e1008368. [PMID: 31518356 PMCID: PMC6760834 DOI: 10.1371/journal.pgen.1008368] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 09/25/2019] [Accepted: 08/12/2019] [Indexed: 11/19/2022] Open
Abstract
Telomere shortening is associated with aging and age-associated diseases. Additionally, telomere dysfunction resulting from telomerase gene mutation can lead to premature aging, such as apparent skin atrophy and hair loss. However, the molecular signaling linking telomere dysfunction to skin atrophy remains elusive. Here we show that dysfunctional telomere disrupts BMP/pSmad/P63 signaling, impairing epidermal stem cell specification and differentiation of skin and hair follicles. We find that telomere shortening mediated by Terc loss up-regulates Follistatin (Fst), inhibiting pSmad signaling and down-regulating P63 and epidermal keratins in an ESC differentiation model as well as in adult development of telomere-shortened mice. Mechanistically, short telomeres disrupt PRC2/H3K27me3-mediated repression of Fst. Our findings reveal that skin atrophy due to telomere dysfunction is caused by a previously unappreciated link with Fst and BMP signaling that could be explored in the development of therapies.
Collapse
Affiliation(s)
- Na Liu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
- Key Laboratory of Bioactive Materials, Ministry of Education, Department of Cell Biology and Genetics, College of Life Sciences, Nankai University, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| | - Yu Yin
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
- Yunnan Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Haiying Wang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Zhongcheng Zhou
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Xiaoyan Sheng
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Haifeng Fu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Renpeng Guo
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Hua Wang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Jiao Yang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Peng Gong
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Wen Ning
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Yifei Liu
- Yale Fertility Center and Department of OB/GYN, Yale University School of Medicine, New Haven, CT, United States of America
- * E-mail: (YL); (LL)
| | - Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
- Key Laboratory of Bioactive Materials, Ministry of Education, Department of Cell Biology and Genetics, College of Life Sciences, Nankai University, Tianjin, China
- * E-mail: (YL); (LL)
| |
Collapse
|
22
|
Nakajima T, Sato T, Iguchi T, Takasugi N. Retinoic acid signaling determines the fate of the uterus from the mouse Müllerian duct. Reprod Toxicol 2019; 86:56-61. [DOI: 10.1016/j.reprotox.2019.03.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/20/2019] [Accepted: 03/24/2019] [Indexed: 10/27/2022]
|
23
|
Piazza MJ, Urbanetz AA. Environmental toxins and the impact of other endocrine disrupting chemicals in women's reproductive health. JBRA Assist Reprod 2019; 23:154-164. [PMID: 30875185 PMCID: PMC6501744 DOI: 10.5935/1518-0557.20190016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 12/27/2018] [Indexed: 12/14/2022] Open
Abstract
This review aimed to look into agents and mechanisms characterized as endocrine disrupting chemicals (EDCs). These agents are known to cause several harmful effects to the reproductive system of women and wildlife. There is a wide range of chemicals, developed for commercial use mainly in agriculture, which may cause endocrine disruption. Numerous studies show evidence of environmental contamination. However, no one is being held liable for the damages. The most important potentially harmful agents are identified and described, along with the different effects they have on the female genital area. Brazil is a large consumer of pesticides and others chemicals that may interfere with a normal women's life. We analyzed and described the mode of action and the impacts of different EDCs (bisphenols, phthalates, atrazine, polychlorinated and polybrominated biphenyls, DDT-dichlorodiphenyltrichloroethane; DDE-dichlorodiphenyldichloroethylene; DDD-dichlorodiphenyldichloroethane; and DES-diethylstilbestrol) on the genital area, ovarian steroidogenesis, polycystic ovary syndrome, endometriosis, the structure of the uterus and the vagina, and on the formation of leiomyomas.
Collapse
Affiliation(s)
- Mauri José Piazza
- Tocogynecology Department, Universidade Federal do
Paraná – UFPR – Curitiba (PR), Brazil
| | - Almir Antônio Urbanetz
- Tocogynecology Department, Universidade Federal do
Paraná – UFPR – Curitiba (PR), Brazil
| |
Collapse
|
24
|
Terakawa J, Serna VA, Taketo MM, Daikoku T, Suarez AA, Kurita T. Ovarian insufficiency and CTNNB1 mutations drive malignant transformation of endometrial hyperplasia with altered PTEN/PI3K activities. Proc Natl Acad Sci U S A 2019; 116:4528-4537. [PMID: 30782821 PMCID: PMC6410785 DOI: 10.1073/pnas.1814506116] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Endometrioid endometrial carcinomas (EECs) carry multiple driver mutations even when they are low grade. However, the biological significance of these concurrent mutations is unknown. We explored the interactions among three signature EEC mutations: loss-of-function (LOF) mutations in PTEN, gain-of-function (GOF) mutations of phosphoinositide 3-kinase (PI3K), and CTNNB1 exon 3 mutations, utilizing in vivo mutagenesis of the mouse uterine epithelium. While epithelial cells with a monoallelic mutation in any one of three genes failed to propagate in the endometrium, any combination of two or more mutant alleles promoted the growth of epithelium, causing simple hyperplasia, in a dose-dependent manner. Notably, Ctnnb1 exon 3 deletion significantly increased the size of hyperplastic lesions by promoting the growth of PTEN LOF and/or PI3K GOF mutant cells through the activation of neoadenogenesis pathways. Although these three mutations were insufficient to cause EEC in intact female mice, castration triggered malignant transformation, leading to myometrial invasion and serosal metastasis. Treatment of castrated mice with progesterone or estradiol attenuated the neoplastic transformation. This study demonstrates that multiple driver mutations are required for premalignant cells to break the growth-repressing field effect of normal endometrium maintained by ovarian steroids and that CTNNB1 exon 3 mutations play critical roles in the growth of preneoplastic cells within the endometrium of premenopausal women and in the myometrial invasion of EECs in menopausal women.
Collapse
Affiliation(s)
- Jumpei Terakawa
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210
- The Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
| | - Vanida Ann Serna
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210
- The Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
| | - Makoto Mark Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, 606-8506 Kyoto, Japan
| | - Takiko Daikoku
- Division of Transgenic Animal Science, Advanced Science Research Center, Kanazawa University, 920-8640 Kanazawa, Japan
| | - Adrian A Suarez
- The Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
- Department of Pathology, Ohio State University, Columbus, OH 43210
| | - Takeshi Kurita
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210;
- The Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
| |
Collapse
|
25
|
Cunha GR, Robboy SJ, Kurita T, Isaacson D, Shen J, Cao M, Baskin LS. Development of the human female reproductive tract. Differentiation 2018; 103:46-65. [PMID: 30236463 PMCID: PMC6234064 DOI: 10.1016/j.diff.2018.09.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/31/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022]
Abstract
Development of the human female reproductive tract is reviewed from the ambisexual stage to advanced development of the uterine tube, uterine corpus, uterine cervix and vagina at 22 weeks. Historically this topic has been under-represented in the literature, and for the most part is based upon hematoxylin and eosin stained sections. Recent immunohistochemical studies for PAX2 (reactive with Müllerian epithelium) and FOXA1 (reactive with urogenital sinus epithelium and its known pelvic derivatives) shed light on an age-old debate on the derivation of vaginal epithelium supporting the idea that human vaginal epithelium derives solely from urogenital sinus epithelium. Aside for the vagina, most of the female reproductive tract is derived from the Müllerian ducts, which fuse in the midline to form the uterovaginal canal, the precursor of uterine corpus and uterine cervix an important player in vaginal development as well. Epithelial and mesenchymal differentiation markers are described during human female reproductive tract development (keratins, homeobox proteins (HOXA11 and ISL1), steroid receptors (estrogen receptor alpha and progesterone receptor), transcription factors and signaling molecules (TP63 and RUNX1), which are expressed in a temporally and spatially dynamic fashion. The utility of xenografts and epithelial-mesenchymal tissue recombination studies are reviewed.
Collapse
Affiliation(s)
- Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, USA.
| | - Stanley J Robboy
- Department of Pathology, Duke University Medical Center, DUMC 3712, Durham, NC 27710, USA
| | - Takeshi Kurita
- Department of Cancer Biology and Genetics, College of Medicine, Comprehensive Cancer Center, Ohio State University, 812 Biomedical Research Tower, 460 W. 12th Avenue, Columbus, OH 43210, USA
| | - Dylan Isaacson
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Joel Shen
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Mei Cao
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Laurence S Baskin
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, USA
| |
Collapse
|
26
|
Cunha GR, Kurita T, Cao M, Shen J, Cooke PS, Robboy SJ, Baskin LS. Tissue interactions and estrogenic response during human female fetal reproductive tract development. Differentiation 2018; 101:39-45. [PMID: 29684808 DOI: 10.1016/j.diff.2018.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/11/2018] [Accepted: 04/16/2018] [Indexed: 12/17/2022]
Abstract
The role of tissue interactions was explored to determine whether epithelial differentiation within the developing human reproductive tract is induced and specified by mesenchyme in tissue recombinants composed of mouse vaginal mesenchyme + human uterine tubal epithelium (mVgM+hTubE). The tissue recombinants were grown in DES-treated ovariectomized athymic mice. After 2-4 weeks of in vivo growth, several vaginal specific features were expressed in the human tubal epithelium. The mesenchyme-induced effects included morphological change as well as expression of several immunohistochemical markers. Although the mesenchyme-induced shift in vaginal differentiation in the human tubal epithelium was not complete, the partial induction of vaginal markers in human tubal epithelium verifies the importance of mesenchymal-epithelial interactions in development of the human female reproductive tract. In a separate experiment, DES-induction of uterine epithelial progesterone receptor (PGR) and estrogen receptor 1 (ESR1) was explored in tissue recombinants composed of wild-type or Esr1KO mouse uterine mesenchyme + human fetal uterine epithelium (wt UtM+hUtE and Esr1KO UtM+hUtE). The rationale of this experiment was to determine whether DES-induction of PGR and ESR1 is mediated directly via epithelial ESR1 or indirectly (paracrine mechanism) via mesenchymal ESR1. DES-induction of uterine epithelial ESR1 and PGR in Esr1KO UtM+hUtE tissue recombinants (devoid of mesenchymal ESR1) formally eliminates the paracrine mechanism and demonstrates that DES induction of human uterine epithelial ESR1 and PGR is directly mediated via epithelial ESR1.
Collapse
Affiliation(s)
- Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States.
| | - Takeshi Kurita
- Department of Cancer Biology and Genetics, College of Medicine, Comprehensive Cancer Center, Ohio State University, 812 Biomedical Research Tower, 460 W. 12th Avenue, Columbus, OH 43210, United States
| | - Mei Cao
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States
| | - Joel Shen
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States
| | - Paul S Cooke
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, 1333 Center Drive, Gainesville, FL 32610, United States
| | - Stanley J Robboy
- Department of Pathology, Duke University Medical Center, DUMC 3712, Durham, NC 27710, United States
| | - Laurence S Baskin
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States
| |
Collapse
|
27
|
Roly ZY, Backhouse B, Cutting A, Tan TY, Sinclair AH, Ayers KL, Major AT, Smith CA. The cell biology and molecular genetics of Müllerian duct development. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2018; 7:e310. [DOI: 10.1002/wdev.310] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 11/06/2017] [Accepted: 11/22/2017] [Indexed: 12/29/2022]
Affiliation(s)
- Zahida Yesmin Roly
- Monash Biomedicine Discovery Institute, Department of Anatomy and Development BiologyMonash UniversityClaytonVictoriaAustralia
| | - Brendan Backhouse
- Murdoch Children's Research Institute and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalMelbourneVictoriaAustralia
| | - Andrew Cutting
- Biology Laboratory, Faculty of ScienceThe University of MelbourneMelbourneVictoriaAustralia
| | - Tiong Yang Tan
- Murdoch Children's Research Institute and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalMelbourneVictoriaAustralia
| | - Andrew H. Sinclair
- Murdoch Children's Research Institute and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalMelbourneVictoriaAustralia
| | - Katie L. Ayers
- Murdoch Children's Research Institute and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalMelbourneVictoriaAustralia
| | - Andrew T. Major
- Monash Biomedicine Discovery Institute, Department of Anatomy and Development BiologyMonash UniversityClaytonVictoriaAustralia
| | - Craig A. Smith
- Monash Biomedicine Discovery Institute, Department of Anatomy and Development BiologyMonash UniversityClaytonVictoriaAustralia
| |
Collapse
|
28
|
Cunha GR, Kurita T, Cao M, Shen J, Robboy SJ, Baskin L. Response of xenografts of developing human female reproductive tracts to the synthetic estrogen, diethylstilbestrol. Differentiation 2017; 98:35-54. [PMID: 29102757 DOI: 10.1016/j.diff.2017.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 09/15/2017] [Accepted: 10/03/2017] [Indexed: 12/16/2022]
Abstract
Human female fetal reproductive tracts 9.5-22 weeks of gestation were grown for 1 month in ovariectomized athymic adult female mouse hosts that were either untreated or treated continuously with diethylstilbestrol (DES) via subcutaneous pellet. Normal morphogenesis and normal patterns of differentiation marker expression (KRT6, KRT7, KRT8, KRT10, KRT14, KRT19, ESR1, PGR, TP63, RUNX1, ISL1, HOXA11 and α-ACT2) were observed in xenografts grown in untreated hosts and mimicked observations of previously reported (Cunha et al., 2017) non-grafted specimens of comparable age. DES elicited several notable morphological affects: (a) induction of endometrial/cervical glands, (b) increased plication (folding) of tubal epithelium, (c) stratified squamous maturation of vaginal epithelium and (d) vaginal adenosis. DES also induced ESR1 in epithelia of the uterine corpus, cervix and globally induced PGR in most cells of the developing human female reproductive tract. Keratin expression (KRT6, KRT7, KRT8, KRT14 and KRT19) was minimally affected by DES. Simple columnar adenotic epithelium was devoid of TP63 and RUNX1, while DES-induced mature vaginal epithelium was positive for both transcription factors. Another striking effect of DES was observed in grafts of human uterine tube, in which DES perturbed smooth muscle patterning. These results define for the first time IHC protein markers of DES action on the developing human reproductive tract, which provide bio-endpoints of estrogen-induced teratogenesis in the developing human female reproductive tract for future testing of estrogenic endocrine disruptors.
Collapse
Affiliation(s)
- Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States.
| | - Takeshi Kurita
- Department of Cancer Biology and Genetics, College of Medicine, Comprehensive Cancer Center, 812 Biomedical Research Tower, 460 West 12th Avenue, Columbus, OH 43210, United States
| | - Mei Cao
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States
| | - Joel Shen
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States
| | - Stanley J Robboy
- Departments of Pathology and Obstetrics and Gynecology, Duke University Medical Center, Durham, NC 27710, United States
| | - Laurence Baskin
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States
| |
Collapse
|
29
|
Robboy SJ, Kurita T, Baskin L, Cunha GR. New insights into human female reproductive tract development. Differentiation 2017; 97:9-22. [PMID: 28918284 DOI: 10.1016/j.diff.2017.08.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/31/2017] [Accepted: 08/01/2017] [Indexed: 01/01/2023]
Abstract
We present a detailed review of the embryonic and fetal development of the human female reproductive tract utilizing specimens from the 5th through the 22nd gestational week. Hematoxylin and eosin (H&E) as well as immunohistochemical stains were used to study the development of the human uterine tube, endometrium, myometrium, uterine cervix and vagina. Our study revisits and updates the classical reports of Koff (1933) and Bulmer (1957) and presents new data on development of human vaginal epithelium. Koff proposed that the upper 4/5ths of the vagina is derived from Müllerian epithelium and the lower 1/5th derived from urogenital sinus epithelium, while Bulmer proposed that vaginal epithelium derives solely from urogenital sinus epithelium. These conclusions were based entirely upon H&E stained sections. A central player in human vaginal epithelial development is the solid vaginal plate, which arises from the uterovaginal canal (fused Müllerian ducts) cranially and squamous epithelium of urogenital sinus caudally. Since Müllerian and urogenital sinus epithelium cannot be unequivocally identified in H&E stained sections, we used immunostaining for PAX2 (reactive with Müllerian epithelium) and FOXA1 (reactive with urogenital sinus epithelium). By this technique, the PAX2/FOXA1 boundary was located at the extreme caudal aspect of the vaginal plate at 12 weeks. During the ensuing weeks, the PAX2/FOXA1 boundary progressively extended cranially such that by 21 weeks the entire vaginal epithelium was FOXA1-reactive and PAX2-negative. This observation supports Bulmer's proposal that human vaginal epithelium derives solely from urogenital sinus epithelium. Clearly, the development of the human vagina is far more complex than previously envisioned and appears to be distinctly different in many respects from mouse vaginal development.
Collapse
Affiliation(s)
- Stanley J Robboy
- Department of Pathology, Duke University, Davison Building, Box 3712, Durham, NC 27710, United States.
| | - Takeshi Kurita
- Department of Cancer Biology and Genetics, The Comprehensive Cancer Center, Ohio State University, 460 W. 12th Avenue, 812 Biomedical Research Tower, Columbus, OH 43210, United States
| | - Laurence Baskin
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States
| | - Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States
| |
Collapse
|
30
|
Molecular mechanisms of development of the human fetal female reproductive tract. Differentiation 2017; 97:54-72. [PMID: 29053991 DOI: 10.1016/j.diff.2017.07.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/21/2017] [Accepted: 07/24/2017] [Indexed: 01/30/2023]
Abstract
Human female reproductive tract development rests mostly upon hematoxilyn and eosin stained sections despite recent advances on molecular mechanisms in mouse studies. We report application of immunohistochemical methods to explore the ontogeny of epithelial and mesenchymal differentiation markers (keratins, homobox proteins, steroid receptors), transcription factors and signaling molecules (TP63 and RUNX1) during human female reproductive tract development. Keratins 6, 7, 8, 10, 14 and 19 (KRT6, KRT7, KRT8, KRT10, KRT14, KRT19) were expressed in a temporally and spatially dynamic fashion. The undifferentiated Müllerian duct and uterovaginal canal, lined by simple columnar epithelia, expressed KRT7, KRT8 and KRT19. Glandular derivatives of the Müllerian duct (uterine tube, uterine corpus and endocervix) maintained expression of these keratins, while tissues that undergo stratified squamous differentiation (exocervix and vagina) expressed KRT6, KRT14 and KRT10 during development in an age-dependent fashion. TP63 and RUNX1 were expressed prior to KRT14, as these two transcription factors are known to be upstream from KRT14 in developing Müllerian epithelium. In the vagina, KRT10, a marker of terminal differentiation, appeared after endogenous estrogens transformed the epithelium to a thick glycogenated squamous epithelium. Uroplakin, a protein unique to urothelium, was expressed only in the bladder, urethra and vaginal introitus, but not in the female reproductive tract itself. Mesenchymal differentiation was examined through immunostaining for HOXA11 (expressed in uterine mesenchyme) and ISL1 (expressed in vaginal mesenchyme). A detailed ontogeny of estrogen receptor alpha (ESR1), progesterone receptor (PGR) and the androgen receptor (AR) provides the mechanistic underpinning for the teratogenicity of estrogens, progestins and androgens on female reproductive tract development. Immunohistochemical analysis of differentiation markers and signaling molecules advance our understanding of normal development of the human female reproductive tract. These observations demonstrate remarkable similarities in mouse and human female reproductive tract development, but also highlight some key differences.
Collapse
|
31
|
Wagamitsu S, Takase D, Aoki F, Suzuki MG. Identification of the Doublesex protein binding sites that activate expression of lozenge in the female genital disc in Drosophila melanogaster. Mech Dev 2017; 143:26-31. [PMID: 28087460 DOI: 10.1016/j.mod.2017.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 12/28/2016] [Accepted: 01/08/2017] [Indexed: 11/29/2022]
Abstract
Normal sexual differentiation in the genital organs is essential for the animal species that use sexual reproduction. Although it is known that doublesex (dsx) is required for the sexual development of the genitalia in various insect species, the direct target genes responsible for the sexual differentiation of the genitalia have not been identified. The lozenge (lz) gene is expressed in the female genital disc and is essential for developments of spermathecae and accessory glands in Drosophila melanogaster. The female-specific isoform of DSX (DSXF) is required for activating lz expression in the female genital disc. However, it still remains unclear whether the DSXF directly activates the transcription of lz in the female genital disc. In this study, we found two sequences (lz-DBS1 and lz-DBS2) within lz locus that showed high homoloty to the DSX binding motif identified previously. Competition assays using recombinant DSX DNA-binding domain (DSX-DBD) protein verified that the DSX-DBD protein bound to lz-DBS1 and lz-DBS2 in a sequence-specific manner with lower affinity than to the known DSX binding site in the bric-à-brac 1 (bab1) gene. Reporter gene analyses revealed that a 2.5-kbp lz genomic fragment containing lz-DBS1 and lz-DBS2 drove reporter gene (EGFP) expression in a manner similar to endogenous lz expression in the female genital disc. Mutations in lz-DBS1 alone significantly reduced the area of EGFP-expressing region, while EGFP expression in the female genital disc was abolished when both sites were mutated. These results demonstrated that DSX directly activates female-specific lz expression in the genital disc through lz-DBS1 and lz-DBS2.
Collapse
Affiliation(s)
- Shunsuke Wagamitsu
- Division of Biological Sciences, Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 302 Bioscience-Bldg, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Dan Takase
- Division of Biological Sciences, Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 302 Bioscience-Bldg, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Fugaku Aoki
- Division of Biological Sciences, Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 302 Bioscience-Bldg, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Masataka G Suzuki
- Division of Biological Sciences, Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 302 Bioscience-Bldg, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan.
| |
Collapse
|
32
|
|
33
|
Retinoic acid signaling determines the fate of uterine stroma in the mouse Müllerian duct. Proc Natl Acad Sci U S A 2016; 113:14354-14359. [PMID: 27911779 DOI: 10.1073/pnas.1608808113] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The Müllerian duct develops into the oviduct, uterus, and vagina, all of which are quite distinct in their morphology and function. The epithelial fate of these female reproductive organs in developing mice is determined by factors secreted from the stroma; however, how stromal differentiation occurs in the female reproductive organs derived from the Müllerian duct is still unclear. In the present study, roles of retinoic acid (RA) signaling in developing female reproductive tracts were investigated. Retinol dehydrogenase 10 (RDH10) and aldehyde dehydrogenase family 1 subfamily A2 (ALDH1A2) mRNAs and proteins and transactivation activity of endogenous RA were found in the stroma of proximal Müllerian ducts and gradually decreased from the proximal to caudal regions in fetal mice. In organ-cultured Müllerian ducts, retinaldehyde or RA treatment induced uterine epithelial differentiation, defined as a layer of columnar epithelial cells negative for oviductal and vaginal epithelial markers. In contrast, inhibition of RA receptor (RAR) signaling induced vaginal epithelial differentiation, characterized as vaginal epithelial marker genes-positive stratified epithelium. Grafting experiments of the organ-cultured Müllerian duct revealed irreversible epithelial fate determination. Although RAR did not directly bind to the homeobox A10 (Hoxa10) promoter region, RA-RAR signaling stimulated Hoxa10 expression. Thus, RA-RAR signaling in the Müllerian duct determines the fate of stroma to form the future uterus and vagina.
Collapse
|
34
|
Harimenshi JM, Jean-Jacques B, Michels JJ. [Vaginal adenosis: A case report and literature review]. Ann Pathol 2016; 36:282-5. [PMID: 27475006 DOI: 10.1016/j.annpat.2016.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 06/12/2016] [Accepted: 06/15/2016] [Indexed: 10/21/2022]
Abstract
We report a case of vaginal adenosis in a woman of 42years. This is a rare congenital disorder since cessation of use of diethylstilbestrol (DES), usually of benign course, not to ignore in its tubo-endometrial histological form which may progress to atypical adenosis precursor of vaginal clear cell adenocarcinoma in patients exposed in utero to DES.
Collapse
Affiliation(s)
- Jean-Marie Harimenshi
- Service d'anatomie pathologique, groupe hospitalier Sud-Réunion, CHU de la Réunion, avenue du Président-Mitterrand, BP 350, 97448 Saint-Pierre cedex, France
| | - Bastien Jean-Jacques
- Service d'anatomie pathologique, CHU de Caen, avenue Côte-de-Nacre, 14033 Caen, France.
| | - Jean-Jacques Michels
- Service d'anatomie pathologique, centre François-Baclesse, 3, avenue du Général-Harris, 14000 Caen, France; Service d'anatomie pathologique, centre hospitalier Public du Cotentin, 46, rue du Val-de-Saire, 50100 Cherbourg-Octeville, France
| |
Collapse
|
35
|
Terakawa J, Rocchi A, Serna VA, Bottinger EP, Graff JM, Kurita T. FGFR2IIIb-MAPK Activity Is Required for Epithelial Cell Fate Decision in the Lower Müllerian Duct. Mol Endocrinol 2016; 30:783-95. [PMID: 27164167 DOI: 10.1210/me.2016-1027] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Cell fate of lower Müllerian duct epithelium (MDE), to become uterine or vaginal epithelium, is determined by the absence or presence of ΔNp63 expression, respectively. Previously, we showed that SMAD4 and runt-related transcription factor 1 (RUNX1) were independently required for MDE to express ΔNp63. Here, we report that vaginal mesenchyme directs vaginal epithelial cell fate in MDE through paracrine activation of fibroblast growth factor (FGF) receptor-MAPK pathway. In the developing reproductive tract, FGF7 and FGF10 were enriched in vaginal mesenchyme, whereas FGF receptor 2IIIb was expressed in epithelia of both the uterus and vagina. When Fgfr2 was inactivated, vaginal MDE underwent uterine cell fate, and this differentiation defect was corrected by activation of MEK-ERK pathway. In vitro, FGF10 in combination with bone morphogenetic protein 4 and activin A (ActA) was sufficient to induce ΔNp63 in MDE, and ActA was essential for induction of RUNX1 through SMAD-independent pathways. Accordingly, inhibition of type 1 receptors for activin in neonatal mice induced uterine differentiation in vaginal epithelium by down-regulating RUNX1, whereas conditional deletion of Smad2 and Smad3 had no effect on vaginal epithelial differentiation. In conclusion, vaginal epithelial cell fate in MDE is induced by FGF7/10-MAPK, bone morphogenetic protein 4-SMAD, and ActA-RUNX1 pathway activities, and the disruption in any one of these pathways results in conversion from vaginal to uterine epithelial cell fate.
Collapse
Affiliation(s)
- Jumpei Terakawa
- Department of Molecular Virology Immunology and Medical Genetics (J.T., V.A.S., T.K.), The Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210; Department of Cell and Molecular Biology (A.R.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611; The Charles Bronfman Institute for Personalized Medicine (E.P.B.), Icahn School of Medicine at Mt Sinai, New York, New York 10029; and Developmental Biology (J.M.G.), Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Altea Rocchi
- Department of Molecular Virology Immunology and Medical Genetics (J.T., V.A.S., T.K.), The Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210; Department of Cell and Molecular Biology (A.R.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611; The Charles Bronfman Institute for Personalized Medicine (E.P.B.), Icahn School of Medicine at Mt Sinai, New York, New York 10029; and Developmental Biology (J.M.G.), Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Vanida A Serna
- Department of Molecular Virology Immunology and Medical Genetics (J.T., V.A.S., T.K.), The Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210; Department of Cell and Molecular Biology (A.R.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611; The Charles Bronfman Institute for Personalized Medicine (E.P.B.), Icahn School of Medicine at Mt Sinai, New York, New York 10029; and Developmental Biology (J.M.G.), Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Erwin P Bottinger
- Department of Molecular Virology Immunology and Medical Genetics (J.T., V.A.S., T.K.), The Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210; Department of Cell and Molecular Biology (A.R.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611; The Charles Bronfman Institute for Personalized Medicine (E.P.B.), Icahn School of Medicine at Mt Sinai, New York, New York 10029; and Developmental Biology (J.M.G.), Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Jonathan M Graff
- Department of Molecular Virology Immunology and Medical Genetics (J.T., V.A.S., T.K.), The Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210; Department of Cell and Molecular Biology (A.R.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611; The Charles Bronfman Institute for Personalized Medicine (E.P.B.), Icahn School of Medicine at Mt Sinai, New York, New York 10029; and Developmental Biology (J.M.G.), Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Takeshi Kurita
- Department of Molecular Virology Immunology and Medical Genetics (J.T., V.A.S., T.K.), The Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210; Department of Cell and Molecular Biology (A.R.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611; The Charles Bronfman Institute for Personalized Medicine (E.P.B.), Icahn School of Medicine at Mt Sinai, New York, New York 10029; and Developmental Biology (J.M.G.), Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
36
|
Kim SY, Ebbert K, Cordeiro MH, Romero MM, Whelan KA, Suarez AA, Woodruff TK, Kurita T. Constitutive Activation of PI3K in Oocyte Induces Ovarian Granulosa Cell Tumors. Cancer Res 2016; 76:3851-61. [PMID: 27197196 DOI: 10.1158/0008-5472.can-15-3358] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 04/19/2016] [Indexed: 11/16/2022]
Abstract
Cell-cell interactions play crucial roles in the maintenance of tissue homeostasis, a loss of which often leads to varying diseases, including cancer. Here, we report that uncontrolled PI3K activity within oocytes irreversibly transforms granulosa cells (GC), causing GC tumors (GCT) through perturbed local cell communication. Previously, we reported reproductive phenotypes of transgenic mice, in which expression of constitutively active mutant PI3K was induced in primordial oocytes by Gdf9-iCre. The transgenic mice (Cre(+)) demonstrated severe ovarian phenotypes, including the overgrowth of excess ovarian follicles and anovulation. Surprisingly, the Cre(+) mice became cachectic by postnatal day 80 due to bilateral GCT. Although GCT cells proliferated independently of oocytes, local interactions with mutant PI3K-positive oocytes during early folliculogenesis were essential for the GC transformation. Growing GCT cells expressed high levels of inhibin βA and nuclear SMAD3, and the proliferation rate was positively correlated with a high activin A to inhibin A ratio. These results suggested that the tumor cells stimulated their growth through an activin A autocrine signaling pathway, a hypothesis confirmed by activin A secretion in cultured GCT cells, which proliferated in response. Although communication between the oocyte and surrounding somatic cells is critical for the normal development of ovarian follicles, perturbations in oocyte-GC communication during early folliculogenesis can induce GCT by activating an autocrine growth circuit program in GC. Cancer Res; 76(13); 3851-61. ©2016 AACR.
Collapse
Affiliation(s)
- So-Youn Kim
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| | - Katherine Ebbert
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Marilia H Cordeiro
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Megan M Romero
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Kelly A Whelan
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Adrian A Suarez
- Department of Pathology, The Comprehensive Cancer Center, Ohio State University, Columbus, Ohio
| | - Teresa K Woodruff
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Takeshi Kurita
- Department of Molecular Virology, Immunology and Medical Genetics, The Comprehensive Cancer Center, Ohio State University, Columbus, Ohio.
| |
Collapse
|
37
|
Investigation of sexual dimorphisms through mouse models and hormone/hormone-disruptor treatments. Differentiation 2016; 91:78-89. [DOI: 10.1016/j.diff.2015.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 11/11/2015] [Indexed: 01/23/2023]
|
38
|
Gore AC, Chappell VA, Fenton SE, Flaws JA, Nadal A, Prins GS, Toppari J, Zoeller RT. EDC-2: The Endocrine Society's Second Scientific Statement on Endocrine-Disrupting Chemicals. Endocr Rev 2015; 36:E1-E150. [PMID: 26544531 PMCID: PMC4702494 DOI: 10.1210/er.2015-1010] [Citation(s) in RCA: 1417] [Impact Index Per Article: 141.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 09/01/2015] [Indexed: 02/06/2023]
Abstract
The Endocrine Society's first Scientific Statement in 2009 provided a wake-up call to the scientific community about how environmental endocrine-disrupting chemicals (EDCs) affect health and disease. Five years later, a substantially larger body of literature has solidified our understanding of plausible mechanisms underlying EDC actions and how exposures in animals and humans-especially during development-may lay the foundations for disease later in life. At this point in history, we have much stronger knowledge about how EDCs alter gene-environment interactions via physiological, cellular, molecular, and epigenetic changes, thereby producing effects in exposed individuals as well as their descendants. Causal links between exposure and manifestation of disease are substantiated by experimental animal models and are consistent with correlative epidemiological data in humans. There are several caveats because differences in how experimental animal work is conducted can lead to difficulties in drawing broad conclusions, and we must continue to be cautious about inferring causality in humans. In this second Scientific Statement, we reviewed the literature on a subset of topics for which the translational evidence is strongest: 1) obesity and diabetes; 2) female reproduction; 3) male reproduction; 4) hormone-sensitive cancers in females; 5) prostate; 6) thyroid; and 7) neurodevelopment and neuroendocrine systems. Our inclusion criteria for studies were those conducted predominantly in the past 5 years deemed to be of high quality based on appropriate negative and positive control groups or populations, adequate sample size and experimental design, and mammalian animal studies with exposure levels in a range that was relevant to humans. We also focused on studies using the developmental origins of health and disease model. No report was excluded based on a positive or negative effect of the EDC exposure. The bulk of the results across the board strengthen the evidence for endocrine health-related actions of EDCs. Based on this much more complete understanding of the endocrine principles by which EDCs act, including nonmonotonic dose-responses, low-dose effects, and developmental vulnerability, these findings can be much better translated to human health. Armed with this information, researchers, physicians, and other healthcare providers can guide regulators and policymakers as they make responsible decisions.
Collapse
Affiliation(s)
- A C Gore
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - V A Chappell
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - S E Fenton
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - J A Flaws
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - A Nadal
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - G S Prins
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - J Toppari
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - R T Zoeller
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| |
Collapse
|
39
|
Yin Y, Lin C, Zhang I, Fisher AV, Dhandha M, Ma L. Homeodomain Transcription Factor Msx-2 Regulates Uterine Progenitor Cell Response to Diethylstilbestrol. ACTA ACUST UNITED AC 2015; 1. [PMID: 26457333 DOI: 10.19104/jstb.2015.105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
The fate of mouse uterine epithelial progenitor cells is determined between postnatal days 5 to 7. Around this critical time window, exposure to an endocrine disruptor, diethylstilbestrol (DES), can profoundly alter uterine cytodifferentiation. We have shown previously that a homeo domain transcription factor MSX-2 plays an important role in DES-responsiveness in the female reproductive tract (FRT). Mutant FRTs exhibited a much more severe phenotype when treated with DES, accompanied by gene expression changes that are dependent on Msx2. To better understand the role that MSX-2 plays in uterine response to DES, we performed global gene expression profiling experiment in mice lacking Msx2 By comparing this result to our previously published microarray data performed on wild-type mice, we extracted common and differentially regulated genes in the two genotypes. In so doing, we identified potential downstream targets of MSX-2, as well as genes whose regulation by DES is modulated through MSX-2. Discovery of these genes will lead to a better understanding of how DES, and possibly other endocrine disruptors, affects reproductive organ development.
Collapse
Affiliation(s)
- Yan Yin
- Division of Dermatology, Washington University School of Medicine, St. Louis, MO
| | - Congxing Lin
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Ivy Zhang
- Department of Dermatology, Saint Louis University School of Medicine, St. Louis, MO
| | | | | | | |
Collapse
|