1
|
Bariani MV, Cui YH, Ali M, Bai T, Grimm SL, Coarfa C, Walker CL, He YY, Yang Q, Al-Hendy A. TGFβ signaling links early life endocrine-disrupting chemicals exposure to suppression of nucleotide excision repair in rat myometrial stem cells. Cell Mol Life Sci 2023; 80:288. [PMID: 37689587 PMCID: PMC10492698 DOI: 10.1007/s00018-023-04928-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/25/2023] [Accepted: 08/18/2023] [Indexed: 09/11/2023]
Abstract
Environmental exposure to endocrine-disrupting chemicals (EDCs) is linked to the development of uterine fibroids (UFs) in women. UFs, non-cancerous tumors, are thought to originate from abnormal myometrial stem cells (MMSCs). Defective DNA repair capacity may contribute to the emergence of mutations that promote tumor growth. The multifunctional cytokine TGFβ1 is associated with UF progression and DNA damage repair pathways. To investigate the impact of EDC exposure on TGFβ1 and nucleotide excision repair (NER) pathways, we isolated MMSCs from 5-month-old Eker rats exposed neonatally to diethylstilbestrol (DES), an EDC, or to vehicle (VEH). EDC-MMSCs exhibited overactivated TGFβ1 signaling and reduced mRNA and protein levels of NER pathway components compared to VEH-MMSCs. EDC-MMSCs also demonstrated impaired NER capacity. Exposing VEH-MMSCs to TGFβ1 decreased NER capacity while inhibiting TGFβ signaling in EDC-MMSCs restored it. RNA-seq analysis and further validation revealed decreased expression of Uvrag, a tumor suppressor gene involved in DNA damage recognition, in VEH-MMSCs treated with TGFβ1, but increased expression in EDC-MMSCs after TGFβ signaling inhibition. Overall, we demonstrated that the overactivation of the TGFβ pathway links early life exposure to EDCs with impaired NER capacity, which would lead to increased genetic instability, arise of mutations, and fibroid tumorigenesis. We demonstrated that the overactivation of the TGFβ pathway links early life exposure to EDCs with impaired NER capacity, which would lead to increased fibroid incidence.
Collapse
Affiliation(s)
| | - Yan-Hong Cui
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Mohamed Ali
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA
| | - Tao Bai
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sandra L Grimm
- Molecular and Cellular Biology Department, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Precision and Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Cristian Coarfa
- Molecular and Cellular Biology Department, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Precision and Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Cheryl L Walker
- Center for Precision and Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Qiwei Yang
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
2
|
Bariani MV, Cui YH, Ali M, Bai T, Grimm SL, Coarfa C, Walker CL, He YY, Yang Q, Al-Hendy A. TGFβ signaling links early-life endocrine-disrupting chemicals exposure to suppression of nucleotide excision repair in rat myometrial stem cells. RESEARCH SQUARE 2023:rs.3.rs-3001855. [PMID: 37333266 PMCID: PMC10274956 DOI: 10.21203/rs.3.rs-3001855/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Environmental exposure to endocrine-disrupting chemicals (EDCs) is linked to the development of uterine fibroids (UFs) in women. UFs, non-cancerous tumors, are thought to originate from abnormal myometrial stem cells (MMSCs). Defective DNA repair capacity may contribute to the emergence of mutations that promote tumor growth. The multifunctional cytokine TGFβ1 is associated with UF progression and DNA damage repair pathways. To investigate the impact of EDC exposure on TGFβ1 and nucleotide excision repair (NER) pathways, we isolated MMSCs from 5-months old Eker rats exposed neonatally to Diethylstilbestrol (DES), an EDC, or to vehicle (VEH). EDC-MMSCs exhibited overactivated TGFβ1 signaling and reduced mRNA and protein levels of NER pathway components compared to VEH-MMSCs. EDC-MMSCs also demonstrated impaired NER capacity. Exposing VEH-MMSCs to TGFβ1 decreased NER capacity while inhibiting TGFβ signaling in EDC-MMSCs restored it. RNA-seq analysis and further validation revealed decreased expression of Uvrag, a tumor suppressor gene involved in DNA damage recognition, in VEH-MMSCs treated with TGFβ1, but increased expression in EDC-MMSCs after TGFβ signaling inhibition. Overall, we demonstrated that the overactivation of the TGFβ pathway links early-life exposure to EDCs with impaired NER capacity, which would lead to increased genetic instability, arise of mutations, and fibroid tumorigenesis. We demonstrated that the overactivation of the TGFβ pathway links early-life exposure to EDCs with impaired NER capacity, which would lead to increased fibroid incidence.
Collapse
Affiliation(s)
| | | | - Mohamed Ali
- University of Chicago Department of Obstetrics and Gynecology
| | - Tao Bai
- University of Chicago Department of Obstetrics and Gynecology
| | | | | | | | - Yu-Ying He
- University of Chicago Department of Medicine
| | - Qiwei Yang
- University of Chicago Department of Obstetrics and Gynecology
| | - Ayman Al-Hendy
- University of Chicago Department of Obstetrics and Gynecology
| |
Collapse
|
3
|
Takao T, Ono M, Yoshimasa Y, Masuda H, Maruyama T. A mediator complex subunit 12 gain-of-function mutation induces partial leiomyoma cell properties in human uterine smooth muscle cells. F&S SCIENCE 2022; 3:288-298. [PMID: 35643626 DOI: 10.1016/j.xfss.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/18/2022] [Accepted: 04/12/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To clarify whether a mediator complex subunit 12 (MED12) gain-of-function mutation induces leiomyoma cell properties in human uterine smooth muscle cells (USMCs). DESIGN Experimental study. SETTING Academic research laboratory. PATIENT(S) Women undergoing hysterectomy for leiomyoma. INTERVENTION(S) CRISPR/Cas9-mediated genome editing to introduce an MED12 gain-of-function mutation (G44D) into human USMCs. MAIN OUTCOME MEASURE(S) Cell proliferation, collagen production, and in vivo tumorigenicity of USMCs with vs. without the MED12 mutation. RESULT(S) Uterine smooth muscle cells isolated from the uterine myometrium of a 44-year-old patient were subjected to lentiviral vector-mediated gene transduction of the fluorescent protein Venus, followed by long-term passage. Uterine smooth muscle cells with a normal female karyotype, high cell proliferative activity, and Venus expression, but without stem/progenitor cell populations, were obtained and designated as USMC44. Using CRISPR/Cas9-mediated genome editing, mtUSMC44 (MED12, 131G>A, p.G44D) and mock USMC44 without MED12 mutation (wtUSMC44) were established from USMC44. wtUSMC44 and mtUSMC44 showed similar cell proliferation activity, even in the presence of estradiol and progesterone (EP) together with transforming growth factor-beta 3 (TGFB3). In addition, wtUSMC44 and mtUSMC44 generated similar tiny smooth muscle-like tissue constructs when xenotransplanted beneath the kidney capsule in immunodeficient mice treated with EP alone or TGFB3. In contrast, mtUSMC44 produced more collagen type I than wtUSMC in vitro, and this production was likely enhanced by EP and TGFB3. CONCLUSION(S) The results suggest that the MED12 gain-of-function mutation is involved in collagen production. Although approximately 70% of leiomyomas have MED12 mutations, additional factors and/or events other than MED12 and/or myometrial stem/progenitor cells may be required for fully inducing leiomyoma cell properties, including transformation, in USMCs.
Collapse
Affiliation(s)
- Tomoka Takao
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan; Department of Regenerative Science, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masanori Ono
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo, Japan
| | - Yushi Yoshimasa
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Hirotaka Masuda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Tetsuo Maruyama
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
4
|
Rooda I, Kaselt B, Liivrand M, Smolander OP, Salumets A, Velthut-Meikas A. Hsa-mir-548 family expression in human reproductive tissues. BMC Genom Data 2021; 22:40. [PMID: 34625017 PMCID: PMC8501715 DOI: 10.1186/s12863-021-00997-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Hsa-miR-548ba expressed in ovarian granulosa cells targets PTEN and LIFR, which are essential for ovarian follicle activation and growth. The expression pattern of hsa-miR-548ba correlates with its host gene follicle-stimulating hormone receptor (FSHR), and FSH has a positive influence on hsa-miR-548ba expression. However, hsa-miR-548ba is a member of a large hsa-mir-548 family with potentially overlapping targets. The current study aims to investigate the co-expression of hsa-mir-548 family members in FSHR-positive reproductive tissues and to explore the potential co-regulation of pathways. RESULTS For the above-described analysis, small RNA sequencing data from public data repositories were used. Sequencing results revealed that hsa-miR-548ba was expressed at the highest level in the ovarian granulosa cells and uterine myometrial samples together with another twelve and one hsa-miR-548 family members, respectively. Pathway enrichment analysis of microRNA targets in the ovarian samples revealed the hsa-miR-548ba and hsa-miR-548b-5p co-regulation of RAB geranylgeranylation in mural granulosa cells. Moreover, other hsa-mir-548 family members co-regulate pathways essential for ovarian functions (PIP3 activates AKT signalling and signalling by ERBB4). In addition to hsa-miR-548ba, hsa-miR-548o-3p is expressed in the myometrium, which separately targets the peroxisome proliferator-activated receptor alpha (PPARA) pathway. CONCLUSION This study reveals that hsa-mir-548 family members are expressed in variable combinations in the reproductive tract, where they potentially fulfil different regulatory roles. The results provide a reference for further studies of the hsa-mir-548 family role in the reproductive tract.
Collapse
Affiliation(s)
- Ilmatar Rooda
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia.
- Competence Centre on Health Technologies, Teaduspargi 13, 50411, Tartu, Estonia.
| | - Birgitta Kaselt
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| | - Maria Liivrand
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| | - Olli-Pekka Smolander
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| | - Andres Salumets
- Competence Centre on Health Technologies, Teaduspargi 13, 50411, Tartu, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 14186, Stockholm, Sweden
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, L. Puusepa St. 8, 50406, Tartu, Estonia
- Institute of Genomics, University of Tartu, Riia 23b, 51010, Tartu, Estonia
| | - Agne Velthut-Meikas
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| |
Collapse
|
5
|
Governini L, Marrocco C, Semplici B, Pavone V, Belmonte G, Luisi S, Petraglia F, Luddi A, Piomboni P. Extracellular matrix remodeling and inflammatory pathway in human endometrium: insights from uterine leiomyomas. Fertil Steril 2021; 116:1404-1414. [PMID: 34256950 DOI: 10.1016/j.fertnstert.2021.06.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 06/10/2021] [Accepted: 06/10/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To compare the expression profiles of matrix metalloproteinases (MMPs) and their inhibitors (tissue inhibitors of matrix metalloproteinases, TIMPs) in the endometrium of women with and without type 3 leiomyomas and to understand their relationship with inflammatory status. DESIGN Molecular and in silico studies. SETTING University hospital. PATIENT(S) Patients with type 3 leiomyomas ranging from 3 to 10 cm in diameter (n = 18) and control age-matched women undergoing surgery for ovarian cysts (n = 18) who underwent endometrial biopsies. INTERVENTION(S) Endometrial biopsies. MAIN OUTCOME MEASURE(S) To evaluate the expression levels of MMPs and TIMPs in the endometrium, quantitative reverse transcriptase polymerase chain reaction and Western blot were performed. With the use of immunofluorescence analysis, the investigated proteins were localized in the tissues. The expression levels of inflammatory mediators such as IL-1β, IL-6, IL-10, TGF, COX1, COX2, STAT3, and VEGF were evaluated by quantitative reverse transcriptase polymerase chain reaction, and their relationships were detected by the STRING approach. RESULT(S) The endometrium of women with type 3 leiomyomas exhibited differential expression of MMPs and TIMPs, particularly MMP2, MMP11, and MMP14, as well as different topographic distribution, suggesting that leiomyomas may influence the endometrial molecular profile. Significant decreases in IL-1β, IL-6, and IL-10 expression, along with increases in COX1 and COX2, as well as VEGF, were highlighted. The STRING approach suggests that this altered gene expression profile may affect the Th17 cell differentiation pathway. CONCLUSION(S) The differential expression and localization of MMPs and TIMPs observed in women with type 3 leiomyomas, along with the reported derangement in the expression of key molecules involved in the inflammatory pathway, may contribute to changes in endometrial receptivity in these patients.
Collapse
Affiliation(s)
- Laura Governini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Camilla Marrocco
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Bianca Semplici
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Valentina Pavone
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Giuseppe Belmonte
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Stefano Luisi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Felice Petraglia
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Alice Luddi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy.
| | - Paola Piomboni
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
6
|
Nakayama M, Ono M, Iizuka T, Kagami K, Fujiwara T, Sekizuka-Kagami N, Maida Y, Obata T, Yamazaki R, Daikoku T, Fujiwara H. Hypertensive disorders of pregnancy are associated with dysmenorrhea in early adulthood: A cohort study. J Obstet Gynaecol Res 2020; 46:2292-2297. [PMID: 32808405 DOI: 10.1111/jog.14431] [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: 01/31/2020] [Revised: 05/30/2020] [Accepted: 07/25/2020] [Indexed: 01/05/2023]
Abstract
AIM Hypertensive disorders of pregnancy (HDP) are serious conditions that occur in 5-10% of pregnancies. Maternal factors, such as maternal age, obesity, and renal disease, have been described as risk factors. In order to extract the background lifestyle and gynecological characteristics for HDP, we conducted a prospective cohort study. METHODS Pregnant participants were administered a questionnaire on characteristics, menstrual abnormalities and lifestyle factors. The women were followed individually until 1-month postpartum. We used medical records to examine the relationship between menstrual abnormalities and the onset of HDP. RESULTS We collected data from 193 pregnant women, and excluding 3 who had miscarriage, examined the records of 190. A total of 26 patients developed HDP, of which 10 had early-onset HDP and 16 had late-onset HDP. Although there was no significant association between HDP and dysmenorrhea just prior to pregnancy, there was a significant increase in the incidence of HDP in patients who experienced dysmenorrhea around the age of 20 years (odds ratio 4.362 [95% CI 1.61-11.81]). CONCLUSION We found that patients with a history of dysmenorrhea around the age of 20 years have a significantly higher risk of developing HDP. Although dysmenorrhea in young adulthood is ameliorated, it may become apparent as a perinatal disease when a physical load such as pregnancy is applied.
Collapse
Affiliation(s)
- Midori Nakayama
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Masanori Ono
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Takashi Iizuka
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Kyosuke Kagami
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Tomoko Fujiwara
- Department of Social Work and Life Design, Kyoto Notre Dame University, Kyoto, Japan
| | - Naomi Sekizuka-Kagami
- Department of Nursing, College of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan
| | - Yoshiko Maida
- Department of Nursing, College of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan
| | - Takeshi Obata
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Rena Yamazaki
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Takiko Daikoku
- Advanced Science Research Center, Kanazawa University, Ishikawa, Japan
| | - Hiroshi Fujiwara
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| |
Collapse
|
7
|
Sakai K, Tanikawa C, Hirasawa A, Chiyoda T, Yamagami W, Kataoka F, Susumu N, Terao C, Kamatani Y, Takahashi A, Momozawa Y, Hirata M, Kubo M, Fuse N, Takai-Igarashi T, Shimizu A, Fukushima A, Kadota A, Arisawa K, Ikezaki H, Wakai K, Yamaji T, Sawada N, Iwasaki M, Tsugane S, Aoki D, Matsuda K. Identification of a novel uterine leiomyoma GWAS locus in a Japanese population. Sci Rep 2020; 10:1197. [PMID: 31988393 PMCID: PMC6985131 DOI: 10.1038/s41598-020-58066-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/12/2019] [Indexed: 12/26/2022] Open
Abstract
Uterine leiomyoma is one of the most common gynaecologic benign tumours, but its genetic basis remains largely unknown. Six previous GWAS identified 33 genetic factors in total. Here, we performed a two-staged GWAS using 13,746 cases and 70,316 controls from the Japanese population, followed by a replication analysis using 3,483 cases and 4,795 controls. The analysis identified 9 significant loci, including a novel locus on 12q23.2 (rs17033114, P = 6.12 × 10-25 with an OR of 1.177 (1.141-1.213), LINC00485). Subgroup analysis indicated that 5 loci (3q26.2, 5p15.33, 10q24.33, 11p15.5, 13q14.11) exhibited a statistically significant effect among multiple leiomyomas, and 2 loci (3q26.2, 10q24.33) exhibited a significant effect among submucous leiomyomas. Pleiotropic analysis indicated that all 9 loci were associated with at least one proliferative disease, suggesting the role of these loci in the common neoplastic pathway. Furthermore, the risk T allele of rs2251795 (3q26.2) was associated with longer telomere length in both normal and tumour tissues. Our findings elucidated the significance of genetic factors in the pathogenesis of leiomyoma.
Collapse
Affiliation(s)
- Kensuke Sakai
- Laboratory of Genome Technology, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Keio University School of Medicine, Department of Obstetrics and Gynecology, Tokyo, Japan
| | - Chizu Tanikawa
- Laboratory of Genome Technology, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Akira Hirasawa
- Keio University School of Medicine, Department of Obstetrics and Gynecology, Tokyo, Japan.,Department of Clinical Genomic Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Tatsuyuki Chiyoda
- Keio University School of Medicine, Department of Obstetrics and Gynecology, Tokyo, Japan
| | - Wataru Yamagami
- Keio University School of Medicine, Department of Obstetrics and Gynecology, Tokyo, Japan
| | - Fumio Kataoka
- Keio University School of Medicine, Department of Obstetrics and Gynecology, Tokyo, Japan
| | - Nobuyuki Susumu
- International University of Health and Welfare School of Medicine, Department of Obstetrics and Gynecology, Chiba, Japan
| | - Chikashi Terao
- RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | | | - Atsushi Takahashi
- RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan.,Department of Genomic Medicine, Research Institute, National Cerebral and Cardiovascular Center, Osaka, Japan
| | | | - Makoto Hirata
- Laboratory of Genome Technology, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Nobuo Fuse
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | | | - Atsushi Shimizu
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, Japan
| | - Akimune Fukushima
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, Japan
| | - Aya Kadota
- Department of Health Science, Shiga University of Medical Science, Shiga, Japan
| | - Kokichi Arisawa
- Department of Preventive Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Hiroaki Ikezaki
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Kenji Wakai
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Taiki Yamaji
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Norie Sawada
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Motoki Iwasaki
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Shoichiro Tsugane
- Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Daisuke Aoki
- Keio University School of Medicine, Department of Obstetrics and Gynecology, Tokyo, Japan
| | - Koichi Matsuda
- Laboratory of Clinical Genome Sequencing, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan.
| |
Collapse
|
8
|
Patterson AL, George JW, Chatterjee A, Carpenter TJ, Wolfrum E, Chesla DW, Teixeira JM. Putative human myometrial and fibroid stem-like cells have mesenchymal stem cell and endometrial stromal cell properties. Hum Reprod 2020; 35:44-57. [PMID: 31913469 PMCID: PMC6993861 DOI: 10.1093/humrep/dez247] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/08/2019] [Indexed: 12/16/2022] Open
Abstract
STUDY QUESTION Can endometrial stromal stem/progenitor cell markers, SUSD2 and CD146/CD140b, enrich for human myometrial and fibroid stem/progenitor cells? SUMMARY ANSWER SUSD2 enriches for myometrial and fibroid cells that have mesenchymal stem cell (MSC) characteristics and can also be induced to decidualise. WHAT IS KNOWN ALREADY Mesenchymal stem-like cells have been separately characterised in the endometrial stroma and myometrium and may contribute to diseases in their respective tissues. STUDY DESIGN, SIZE, DURATION Normal myometrium, fibroids and endometrium were collected from hysterectomies with informed consent. Primary cells or tissues were used from at least three patient samples for each experiment. PARTICIPANTS/MATERIALS, SETTING, METHODS Flow cytometry, immunohistochemistry and immunofluorescence were used to characterise tissues. In vitro colony formation in normoxic and hypoxic conditions, MSC lineage differentiation (osteogenic and adipogenic) and decidualisation were used to assess stem cell activity. Xenotransplantation into immunocompromised mice was used to determine in vivo stem-like activity. Endpoint measures included quantitative PCR, colony formation, trichrome, Oil Red O and alkaline phosphatase activity staining. MAIN RESULTS AND THE ROLE OF CHANCE CD146+CD140b+ and/or SUSD2+ myometrial and fibroid cells were located in the perivascular region and formed more colonies in vitro compared to control cells and differentiated down adipogenic and osteogenic mesenchymal lineages in vitro. SUSD2+ myometrial cells had greater in vitro decidualisation potential, and SUSD2+ fibroid cells formed larger tumours in vivo compared to control cells. LARGE-SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Markers used in this study enrich for cells with stem/progenitor cell activity; however, they do not distinguish stem from progenitor cells. SUSD2+ myometrial cells express markers of decidualisation when treated in vitro, but in vivo assays are needed to fully demonstration their ability to decidualise. WIDER IMPLICATIONS OF THE FINDINGS These results suggest a possible common MSC for the endometrial stroma and myometrium, which could be the tumour-initiating cell for uterine fibroids. STUDY FUNDING/COMPETING INTEREST(S) These studies were supported by NIH grants to JMT (R01OD012206) and to ALP (F32HD081856). The authors certify that we have no conflicts of interest to disclose.
Collapse
Affiliation(s)
- Amanda L Patterson
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Division of Animal Sciences and Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO 65203, USA
| | - Jitu W George
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Anindita Chatterjee
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Tyler J Carpenter
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Emily Wolfrum
- Bioinformatics and Biostatistics Core, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - David W Chesla
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Office of Research, Spectrum Health, Grand Rapids, MI 49503, USA
| | - Jose M Teixeira
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| |
Collapse
|
9
|
Baranov VS, Osinovskaya NS, Yarmolinskaya MI. Pathogenomics of Uterine Fibroids Development. Int J Mol Sci 2019; 20:E6151. [PMID: 31817606 PMCID: PMC6940759 DOI: 10.3390/ijms20246151] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/12/2022] Open
Abstract
We review recent studies dealing with the molecular genetics and basic results of omics analysis of uterine leiomyoma (LM)-a common benign muscle tumor of the uterus. Whole genome studies of LM resulted in the discovery of many new gene nets and biological pathways, including its origin, transcriptomic, and epigenetic profiles, as well as the impact of the inter-cell matrix in LM growth and involvement of microRNA in its regulation. New data on somatic cell mutations ultimately involved in the origin, distribution and growth of LM are reviewed. Putative identification of LM progenitor SC (stem cells) giving rise to maternal fibroid nodes and junctional zones provide a new clue for hypotheses on the pathogenomics of LM. The reviewed data are consistent with at least two different but probably intimately interacted molecular mechanisms of LM. One of them (the genetic hypothesis) is focused primarily on the MED12 gene mutations and suggests its onset in the side population of embryonic myoblasts of the female reproductive system, which later gave rise to multiple small and medium fibroids. The single and usually large-size fibroids are induced by predominantly epigenetic disorders in LM SC, provoked by enhanced expression of the HMGA2 gene caused by its hypomethylation and epigenetic deregulation enhanced by hypoxia, muscle tension, or chromosome instability/aberrations. The pathogenomics of both genetic and epigenetic programs of LM with many peculiarities at the beginning later became rather similar and partly overlapped due to the proximity of their gene nets and epigenetic landscape. Pathogenomic studies of LM open ways for elaboration of novel strategies of prevention and treatment of this common disease.
Collapse
Affiliation(s)
- Vladislav S. Baranov
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia; (N.S.O.); (M.I.Y.)
| | | | | |
Collapse
|
10
|
Brakta S, Mas A, Al-Hendy A. The ontogeny of myometrial stem cells in OCT4-GFP transgenic mouse model. Stem Cell Res Ther 2018; 9:333. [PMID: 30486855 PMCID: PMC6264618 DOI: 10.1186/s13287-018-1079-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 11/12/2018] [Accepted: 11/19/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Myometrium, the muscular wall of the uterus, is an active organ markedly remodeled during a woman's reproductive life, especially during pregnancy. Different studies using the 5-bromo-2'-deoxyuridine and side population methods in murine and human myometrium have suggested the presence of somatic stem cells in this tissue because of its remarkable regenerative capacity. Recently, our group has developed a surface-marker (Stro1/CD44)-specific approach to isolate and characterize myometrial somatic stem cells (SSCs) from humans and rats. OBJECTIVE In this study, we aimed to identify and localize the putative myometrial stem cell population in the murine uterus by using the specific surface markers, Nanog/CD44. METHODS Uteri from OCT4-GFP transgenic mice at different early-life time points were analyzed via single and double immunohistochemistry to co-localize myometrial stem cell marker CD44 with other general stemmness markers, e.g., Nanog and Oct-4. Finally, we correlated the frequency of myometrial stem cells in vivo with the expression of sex steroid hormone receptors, estrogen receptor α (ERα), and progesterone receptors A and B (PR A&B). RESULTS Nanog+/CD44+ stem cells were present in murine myometrium. Both stem cell markers were shown to co-localize with Oct-4 expression. Time-course experiments demonstrated that their percentages were significantly lower at the pre-sexual age of 1 week than at the sexually mature ages of 3 to 24 weeks. Importantly, both ERα and PR A&B were abundantly expressed in the myometrium at ages 1, 3 and 4 weeks. CONCLUSIONS We demonstrated that murine CD44+ myometrial cells have features of somatic stem cells with the expression of typical undifferentiated markers. Furthermore, our results suggest that myometrial stem cells are sex steroid hormone dependent, likely via paracrine pathway, and increase in numbers with reproductive maturity and rise in serum estrogen and progesterone levels around 3 weeks of age in mice. The abundance and early onset expression of ER/PR emphasize the vulnerability of neonatal myometrium to environmental endocrine disruptors which can potentially lead to permanent reprograming and adult onset of myometrial disorders such as uterine fibroids.
Collapse
Affiliation(s)
- Soumia Brakta
- Department of Obstetrics and Gynecology, University of Augusta, Augusta, GA, 30912, USA
| | - Aymara Mas
- Reproductive Medicine Research Group, La Fe Research Institute, Valencia, Spain
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Illinois at Chicago (UIC), Chicago, IL, 60612, USA.
| |
Collapse
|
11
|
Prusinski Fernung LE, Jones K, Mas A, Kleven D, Waller JL, Al-Hendy A. Expanding upon the Human Myometrial Stem Cell Hypothesis and the Role of Race, Hormones, Age, and Parity in a Profibroid Environment. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2293-2306. [PMID: 30075150 DOI: 10.1016/j.ajpath.2018.06.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/14/2018] [Accepted: 06/26/2018] [Indexed: 12/20/2022]
Abstract
Uterine fibroids (UFs) are clonal, hormonally regulated, benign smooth-muscle myometrial tumors that severely affect female reproductive health, although their unknown etiology limits effective care. UFs occur fourfold more commonly in African American women than in Caucasian women, and African American women generally have earlier disease onset and greater UF tumor burden, although the mechanism of this ethnic disparity has not been identified. Recent findings have linked cancer (ie, tumor) risk to increased tissue-specific stem cell division and self-renewal and suggest that somatic mutations in myometrial stem cells (MyoSCs) convert them into tumor-initiating cells, leading to UF. Specifically, preliminary results in paraffin-embedded myometrial tissues have shown increased STRO-1+/CD44+ MyoSCs in African American versus Caucasian women. Using specific methods of flow cytometry and automated quantitative pathology imaging, a large cohort of myometrial samples were investigated to determine how the STRO-1+/CD44+ MyoSCs change with regard to a patient's race, age, parity, fibroid and hormone statuses, and the location of UFs within the uterus. We confirmed that the STRO-1+/CD44+ MyoSC population is expanded in African American women, is correlated with parity and fibroid number, and fluctuates with cyclic menstrual cycle hormone changes and age. Our data suggest that an expanded MyoSC population increases the formation of tumor-initiating cells, ultimately contributing to increased UF prevalence and burden in African American women.
Collapse
Affiliation(s)
- Lauren E Prusinski Fernung
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Kimya Jones
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Aymara Mas
- Reproductive Medicine Research Group, La Fe Health Research Institute, Valencia, Spain
| | - Daniel Kleven
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Jennifer L Waller
- Department of Biostatistics and Data Science, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Illinois, Chicago, Illinois.
| |
Collapse
|
12
|
Torres-de la Roche LA, Becker S, Cezar C, Hermann A, Larbig A, Leicher L, Di Spiezio Sardo A, Tanos V, Wallwiener M, Verhoeven H, De Wilde RL. Pathobiology of myomatosis uteri: the underlying knowledge to support our clinical practice. Arch Gynecol Obstet 2017; 296:701-707. [PMID: 28864872 DOI: 10.1007/s00404-017-4494-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 08/16/2017] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Uterine myomatosis, a benign condition, is the most common indication for hysterectomies worldwide, affecting the reproductive goals and quality of health of women. However, gynecologists have been provided with interesting insights on its pathobiology, which are the basis for uterine-preserving therapies. The aim of this paper is to discuss the latest evidence on these mechanisms and its importance in the clinical practice. METHOD A comprehensive literature search was made in Pubmed, Medline, the Cochrane Library, Orbis plus and Google Scholar for articles related to the epidemiological, biological, and genetic bases. RESULTS There is extensive evidence that genetic, epigenetic, hormonal, environmental, proinflamatory, angiogenetic, and growing factors are involved in the biology of myomatosis. Such factors are capable of activating and promoting inhibitory signaling pathways leading to initiation, development, and regression of myomata through changes in myometrial cells and myomata fibers, such as cellular proliferation, differentiation, apoptosis, angiogenesis, and changes in the surrounding environment. CONCLUSION A good understanding of the pathobiologic mechanisms of myomatosis gives reasons to physicians to elect conservative or combined therapies, and allow affected women to receive an individualized management, according to age, reproduction desire, and clinical condition.
Collapse
Affiliation(s)
- Luz Angela Torres-de la Roche
- Clinic for Gynecology, Obstetrics and Gynecological Oncology, Pius Hospital, University Hospital for Gynecology, Carl von Ossietzky University Medical School, Georgstrasse 12, 26121, Oldenburg, Germany
| | - Sven Becker
- Clinic of Gynecology and Obstetrics, Universitäts Klinikum Frankfurt, Frankfurt, Germany
| | - Cristina Cezar
- Clinic for Gynecology and Obstetrics, Carl von Ossietzky University Medical School, Oldenburg, Germany
| | - Anja Hermann
- Clinic for Gynecology and Obstetrics, Carl von Ossietzky University Medical School, Oldenburg, Germany
| | - Angelika Larbig
- Clinic for Gynecology, Obstetrics and Gynecological Oncology, Pius Hospital, University Hospital for Gynecology, Carl von Ossietzky University Medical School, Georgstrasse 12, 26121, Oldenburg, Germany
| | - Lasse Leicher
- Clinic for Gynecology, Obstetrics and Gynecological Oncology, Pius Hospital, University Hospital for Gynecology, Carl von Ossietzky University Medical School, Georgstrasse 12, 26121, Oldenburg, Germany
| | | | - Vasilis Tanos
- Department of Surgery, Aretaeio Hospital, St. Georges Medical School, Nicosia University, Nicosia, Cyprus
| | - Markus Wallwiener
- Clinic of Gynecology and Obstetrics and Gynecological Oncology, University of Heidelberg, Heidelberg, Germany
| | - Hugo Verhoeven
- Clinic for Gynecology, Obstetrics and Gynecological Oncology, Pius Hospital, University Hospital for Gynecology, Carl von Ossietzky University Medical School, Georgstrasse 12, 26121, Oldenburg, Germany
| | - Rudy Leon De Wilde
- Clinic for Gynecology, Obstetrics and Gynecological Oncology, Pius Hospital, University Hospital for Gynecology, Carl von Ossietzky University Medical School, Georgstrasse 12, 26121, Oldenburg, Germany.
| |
Collapse
|
13
|
Delaney MA, Wan YW, Kim GE, Creighton CJ, Taylor MG, Masand R, Park A, Valdes C, Gibbons W, Liu Z, Anderson ML. A Role for Progesterone-Regulated sFRP4 Expression in Uterine Leiomyomas. J Clin Endocrinol Metab 2017; 102. [PMID: 28637297 PMCID: PMC5587057 DOI: 10.1210/jc.2016-4014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
CONTEXT Despite progesterone's key role in uterine smooth muscle tumorigenesis, the mechanisms by which it promotes the growth of uterine leiomyomas remain poorly understood. OBJECTIVE The aim of this study was to identify gene products mediating the effects of progesterone in uterine leiomyomas. DESIGN Gene expression profiling was used to identify putative progesterone-regulated genes differentially expressed in uterine leiomyomas, which were then studied in vitro. METHODS Gene expression was comprehensively profiled with the Illumina WG BeadChip (version 2.6) and analyzed with a bioinformatic algorithm that integrates known protein-protein interactions. Genomic binding sites for progesterone receptor (PR) were interrogated by chromatin immunoprecipitation-quantitative polymerase chain reaction (ChIP-qPCR). Small interfering RNA was used to study gene function in primary cell lines. RESULTS Our analyses identified secreted Frizzled-related protein 4 (sFRP4) as a key gene product functionally linked to PR activation whose expression was 2.6 times higher in leiomyomas than myometrium (n = 26, P < 0.01) and 2.5 times higher during the proliferative phase of the menstrual cycle (n = 26, P < 0.01). Direct binding between PR and sFRP4 promoter was observed by ChIP-qPCR. Robust overexpression of sFRP4 was also observed in primary cultures derived from leiomyoma. Progesterone preferentially inhibited sFRP4 expression and secretion in leiomyoma cultures in a dose-dependent manner sensitized by estradiol. Knockdown of sFRP4 inhibited proliferation and apoptosis in primary cultures of both myometrium and leiomyoma. CONCLUSIONS Overexpression of sFRP4 is a robust, progesterone-regulated feature of leiomyomas that increases smooth muscle proliferation. More work is needed to elucidate how progesterone's ability to modulate sFRP4 expression contributes to uterine smooth muscle tumorigenesis.
Collapse
Affiliation(s)
- Meaghan A. Delaney
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, Texas 77030
| | - Ying-Wooi Wan
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, Texas 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Gyoung-Eun Kim
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, Texas 77030
| | - Chad J. Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030
| | - Margaret G. Taylor
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, Texas 77030
| | - Ramya Masand
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030
| | - Andrew Park
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, Texas 77030
| | - Cecilia Valdes
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, Texas 77030
| | - William Gibbons
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, Texas 77030
| | - Zhandong Liu
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030
| | - Matthew L. Anderson
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, Texas 77030
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
14
|
Mas A, Prusinski L, Yang Q, Diaz-Gimeno P, Stone L, Diamond MP, Simón C, Al-Hendy A. Role of Stro1+/CD44+ stem cells in myometrial physiology and uterine remodeling during pregnancy. Biol Reprod 2017; 96:70-80. [PMID: 28395335 PMCID: PMC5803774 DOI: 10.1095/biolreprod.116.143461] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/31/2016] [Accepted: 11/22/2016] [Indexed: 12/17/2022] Open
Abstract
Regulation of myometrial functions during pregnancy has been considered the result of the integration of endocrine and mechanical signals. Nevertheless, uterine regeneration is poorly understood, and the cellular source within the gravid uterus is largely unexplored.In this study, we isolated and quantified the myometrial stem cells (MSC) population from pregnant female Eker rat uteri, by using Stro1/CD44 surface markers. We demonstrated that prior parity significantly increased the percentage of Stro1+/CD44+ MSC because of injured tissue response. Interestingly, we established that Stro1+/CD44+ MSC respond efficiently to physiological cues when they were treated in vitro under different dose-dependent pregnant rat serum.Previous studies reveal strong regulatory links between O2 availability and stem cell function. Based on these premises, cell proliferation assays showed that isolated Stro1+/CD44+ MSC possess a higher proliferative rate under hypoxic versus normoxic conditions. We also detected a total of 37 upregulated and 44 downregulated hypoxia-related genes, which were differentially expressed in Stro1+/CD44+ MSC, providing an alternative approach to infer into complex molecular mechanisms such as energy metabolism, inflammatory response, uterine expansion, and/or remodeling.Since these cells preferentially grow under low oxygen conditions, we propose that the increase of the rat uterus during pregnancy involves myometrial oxygen consumption, thereby enhancing MSC proliferation. Moreover, pregnancy-induced mechanical stretching results in hypoxic conditions, ultimately creating an environment that promotes stem cell proliferation and further uterine enlargement, which is essential for a successful pregnancy. In summary, all of these data support that rat Stro1+/CD44+ MSC contribute to uterine enlargement during pregnancy.
Collapse
Affiliation(s)
- Aymara Mas
- Department of Obstetrics and Gynecology, Augusta University, Augusta, Georgia, USA
| | - Lauren Prusinski
- Department of Obstetrics and Gynecology, Augusta University, Augusta, Georgia, USA
| | - Qiwei Yang
- Department of Obstetrics and Gynecology, Augusta University, Augusta, Georgia, USA
| | - Patricia Diaz-Gimeno
- Fundacion Instituto Valenciano de Infertilidad (FIVI), Department of Obstetrics & Gynecology, School of Medicine, Valencia University and Instituto Universitario IVI/INCLIVA, Valencia, Spain
| | - Lelyand Stone
- Department of Obstetrics and Gynecology, Augusta University, Augusta, Georgia, USA
| | - Michael P Diamond
- Department of Obstetrics and Gynecology, Augusta University, Augusta, Georgia, USA
| | - Carlos Simón
- Igenomix, Valencia, Spain; Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain; Department of Pediatrics, Obstetrics, and Gynecology, Universidad de Valencia, Instituto Universitario IVI, Valencia, Spain; Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Stanford, CA
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
15
|
Yang Q, Al-Hendy A. Developmental Environmental Exposure Alters the Epigenetic Features of Myometrial Stem Cells. ACTA ACUST UNITED AC 2016. [PMID: 28626795 DOI: 10.17140/goroj-3-e005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Qiwei Yang
- Division of Translation Research, Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ayman Al-Hendy
- Division of Translation Research, Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
16
|
Yatsenko SA, Mittal P, Wood-Trageser MA, Jones MW, Surti U, Edwards RP, Sood AK, Rajkovic A. Highly heterogeneous genomic landscape of uterine leiomyomas by whole exome sequencing and genome-wide arrays. Fertil Steril 2016; 107:457-466.e9. [PMID: 27889101 DOI: 10.1016/j.fertnstert.2016.10.035] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 10/26/2016] [Accepted: 10/26/2016] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To determine the genomic signatures of human uterine leiomyomas and prevalence of MED12 mutations in human uterine leiomyosarcomas. DESIGN Retrospective cohort study. SETTING Not applicable. PATIENT(S) This study included a set of 16 fresh frozen leiomyoma and corresponding unaffected myometrium specimens as well as 153 leiomyosarcomas collected from women diagnosed with uterine leiomyomas or leiomyosarcomas who underwent clinically indicated abdominal hysterectomy. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Whole exome sequencing and high-resolution X-chromosome and whole genome single nucleotide polymorphism microarray analyses were performed on leiomyoma samples negative for the known MED12 mutations and compared with their corresponding myometrium. Leiomyosarcoma specimens were examined for exon 2 MED12 mutations to evaluate the frequency of MED12 mutated leiomyosarcomas. RESULT(S) Our results indicate remarkable genomic heterogeneity of leiomyoma lesions. MED12-negative leiomyomas contain copy number alterations involving the Mediator complex subunits such as MED8, MED18, CDK8, and long intergenic nonprotein coding RNA340 (CASC15), which may affect the Mediator architecture and/or its transcriptional activity. We also identified mutations in a number of genes that were implicated in leiomyomagenesis such as COL4A6, DCN, and AHR, as well as novel genes: NRG1, ADAM18, HUWE1, FBXW4, FBXL13, and CAPRIN1. CONCLUSION(S) Mutations in genes implicated in cell-to-cell interactions and remodeling of the extracellular matrix and genomic aberrations involving genes coding for the Mediator complex subunits were identified in uterine leiomyomas. Additionally, we discovered that ∼4.6% of leiomyosarcomas harbored MED12 exon 2 mutations, but the relevance of this association with molecular pathogenesis of leiomyosarcoma remains unknown.
Collapse
Affiliation(s)
- Svetlana A Yatsenko
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Pathology, Magee-Women's Hospital of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Priya Mittal
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; Magee-Women's Research Institute, Pittsburgh, Pennsylvania; Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Michelle A Wood-Trageser
- Department of Pathology, Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mirka W Jones
- Department of Pathology, Magee-Women's Hospital of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Urvashi Surti
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Pathology, Magee-Women's Hospital of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; Magee-Women's Research Institute, Pittsburgh, Pennsylvania
| | - Robert P Edwards
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania; Magee-Women's Research Institute, Pittsburgh, Pennsylvania
| | - Anil K Sood
- Department of Gynecologic Oncology and Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aleksandar Rajkovic
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Pathology, Magee-Women's Hospital of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; Magee-Women's Research Institute, Pittsburgh, Pennsylvania.
| |
Collapse
|
17
|
Katz TA, Yang Q, Treviño LS, Walker CL, Al-Hendy A. Endocrine-disrupting chemicals and uterine fibroids. Fertil Steril 2016; 106:967-77. [PMID: 27553264 PMCID: PMC5051569 DOI: 10.1016/j.fertnstert.2016.08.023] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 08/09/2016] [Accepted: 08/09/2016] [Indexed: 12/17/2022]
Abstract
Uterine fibroids are the most frequent gynecologic tumor, affecting 70% to 80% of women over their lifetime. Although these tumors are benign, they can cause significant morbidity and may require invasive treatments such as myomectomy and hysterectomy. Many risk factors for these tumors have been identified, including environmental exposures to endocrine-disrupting chemicals (EDCs) such as genistein and diethylstilbestrol. Uterine development may be a particularly sensitive window to environmental exposures, as some perinatal EDC exposures have been shown to increase tumorigenesis in both rodent models and human epidemiologic studies. The mechanisms by which EDC exposures may increase tumorigenesis are still being elucidated, but epigenetic reprogramming of the developing uterus is an emerging hypothesis. Given the remarkably high incidence of uterine fibroids and their significant impact on women's health, understanding more about how prenatal exposures to EDCs (and other environmental agents) may increase fibroid risk could be key to developing prevention and treatment strategies in the future.
Collapse
Affiliation(s)
- Tiffany A Katz
- Health Science Center, Institute of Biotechnology, Center for Translational Cancer Research, Texas A&M University, Houston, Texas
| | - Qiwei Yang
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Lindsey S Treviño
- Health Science Center, Institute of Biotechnology, Center for Translational Cancer Research, Texas A&M University, Houston, Texas
| | - Cheryl Lyn Walker
- Health Science Center, Institute of Biotechnology, Center for Translational Cancer Research, Texas A&M University, Houston, Texas
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia.
| |
Collapse
|
18
|
Janjusevic M, Greco S, Islam MS, Castellucci C, Ciavattini A, Toti P, Petraglia F, Ciarmela P. Locostatin, a disrupter of Raf kinase inhibitor protein, inhibits extracellular matrix production, proliferation, and migration in human uterine leiomyoma and myometrial cells. Fertil Steril 2016; 106:1530-1538.e1. [PMID: 27565262 DOI: 10.1016/j.fertnstert.2016.08.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/30/2016] [Accepted: 08/01/2016] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To investigate the presence of Raf kinase inhibitor protein (RKIP) in human myometrium and leiomyoma as well as to determine the effect of locostatin (RKIP inhibitor) on extracellular matrix (ECM) production, proliferation, and migration in human myometrial and leiomyoma cells. DESIGN Laboratory study. SETTING Human myometrium and leiomyoma. PATIENT(S) Thirty premenopausal women who were admitted to the hospital for myomectomy or hysterectomy. INTERVENTION(S) Myometrial and leiomyoma tissues were used to investigate the localization and the expression level of RKIP through immunohistochemistry and Western blotting. Myometrial and leiomyoma cells were treated with locostatin (10 μM) to measure ECM expression by real-time polymerase chain reaction, GSK3β expression by Western blotting, cell migration by wound-healing assay, and cell proliferation by MTT assay and immunocytochemistry. MAIN OUTCOME MEASURE(S) The expression of RKIP in human myometrial and leiomyoma tissue; ECM components and GSK3β expression, migration, and proliferation in myometrial and leiomyoma cells. RESULT(S) RKIP is expressed in human myometrial and leiomyoma tissue. Locostatin treatment resulted in the activation of the mitogen-activated protein kinase (MAPK) signal pathway (ERK phosphorylation), providing a powerful validation of our targeting protocol. Further, RKIP inhibition by locostatin reduces ECM components. Moreover, the inhibition of RKIP by locostatin impaired cell proliferation and migration in both leiomyoma and myometrial cells. Finally, locostatin treatment reduced GSK3β expression. Therefore, even if the activation of MAPK pathway should increase proliferation and migration, the destabilization of GSK3β leads to the reduction of proliferation and migration of myometrial and leiomyoma cells. CONCLUSION(S) Our results indicate that RKIP may be involved in leiomyoma pathophysiology.
Collapse
Affiliation(s)
- Milijana Janjusevic
- Department of Experimental and Clinical Medicine, Faculty of Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Stefania Greco
- Department of Experimental and Clinical Medicine, Faculty of Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Md Soriful Islam
- Department of Experimental and Clinical Medicine, Faculty of Medicine, Università Politecnica delle Marche, Ancona, Italy; Biotechnology and Microbiology Laboratory, Department of Botany, University of Rajshahi, Rajshahi, Bangladesh
| | - Clara Castellucci
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Andrea Ciavattini
- Department of Clinical Science, Università Politecnica delle Marche, Ancona, Italy
| | - Paolo Toti
- Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Felice Petraglia
- Department of Molecular and Developmental Medicine, Obstetrics, and Gynecology, University of Siena, Siena, Italy
| | - Pasquapina Ciarmela
- Department of Experimental and Clinical Medicine, Faculty of Medicine, Università Politecnica delle Marche, Ancona, Italy; Department of Information Engineering, Università Politecnica delle Marche, Ancona, Italy.
| |
Collapse
|
19
|
Reis FM, Bloise E, Ortiga-Carvalho TM. Hormones and pathogenesis of uterine fibroids. Best Pract Res Clin Obstet Gynaecol 2016; 34:13-24. [DOI: 10.1016/j.bpobgyn.2015.11.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/18/2015] [Indexed: 12/30/2022]
|
20
|
Friedman N, Shushan A, Rojansky N, Shveiky D, Levitzki R, Chaouat M, Ben-Bassat H. Targeting leiomyomas with all-trans-retinoic acid at phosphoinositide 3-kinase pathway suppression: Effective roles of β-catenin and of signaling interactions. J Obstet Gynaecol Res 2016; 42:1343-1353. [PMID: 27354299 DOI: 10.1111/jog.13068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 05/01/2016] [Indexed: 12/31/2022]
Abstract
AIM Leiomyomas, monoclonal tumors developed by the transformation of myometrium somatic stem cells, are a major health concern that can severely impair quality of life. Pathological alterations of signaling pathways have been recognized as a key feature in a variety of human diseases. Our objective was to analyze treatment with all-trans-retinoic acid (ATRA) by suppression of the phosphoinositide 3-kinase (PI3K) pathway on growth, signaling pattern and interactions among PI3K/B-cell lymphoma 2 (Bcl2)/retinol leiomyoma proteins. METHODS Cultures of paired myometrium and leiomyoma cells from premenopausal women undergoing hysterectomy were collected. Western blot and analysis of variance were used for analysis. RESULTS Significant differences were detected between treatment with ATRA alone or with LY294002 (a PI3K growth suppressor) in response to treatment and among cell samples and cell numbers. Leiomyoma cells were less affected. Immunochemical analysis of signaling patterns demonstrated that treatments affected most of the examined protein levels differently. Significant differences between the cell type responses to treatment in pyruvate phosphate dikinase 1 (pPDK1), Bad and pβ-catenin levels were identified. The pβ-catenin level showed highly significant interaction between response to treatment and cell type. CONCLUSIONS ATRA treatment on PI3K pathway suppression significantly affected growth, signaling pattern and interactions among PI3K/Bcl2/retinol proteins involved in the growth, survival and apoptosis of leiomyomas. Interpretation of our results suggests that increasing knowledge of the role of signaling interplay in the pathogenesis of leiomyomas may present an opportunity to use specific signal transduction inhibitors for treating and preventing this disorder.
Collapse
Affiliation(s)
- Noa Friedman
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Jerusalem, Israel.,Laboratory of Experimental Surgery, Hadassah Medical Center, Jerusalem, Israel
| | - Asher Shushan
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Jerusalem, Israel
| | - Nathan Rojansky
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Jerusalem, Israel
| | - David Shveiky
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Jerusalem, Israel
| | - Rubina Levitzki
- Laboratory of Experimental Surgery, Hadassah Medical Center, Jerusalem, Israel
| | - Malka Chaouat
- Israel National Skin Bank, Hadassah Medical Center, Jerusalem, Israel.,Laboratory of Experimental Surgery, Hadassah Medical Center, Jerusalem, Israel
| | - Hannah Ben-Bassat
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Jerusalem, Israel. .,Israel National Skin Bank, Hadassah Medical Center, Jerusalem, Israel.
| |
Collapse
|
21
|
Hellström M, Moreno-Moya JM, Bandstein S, Bom E, Akouri RR, Miyazaki K, Maruyama T, Brännström M. Bioengineered uterine tissue supports pregnancy in a rat model. Fertil Steril 2016; 106:487-496.e1. [PMID: 27068301 DOI: 10.1016/j.fertnstert.2016.03.048] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/21/2016] [Accepted: 03/29/2016] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To create a bioengineered uterine patch for uterine repair of a partially defect uterus. DESIGN Three different decellularized uterine scaffolds were recellularized in vitro with primary uterine cells and green fluorescent protein- (GPF-) labeled bone marrow-derived mesenchymal stem cells (GFP-MSCs). The patches were transplanted in vivo to investigate their tissue adaptation and supporting capacity during pregnancy. SETTING Research laboratory. ANIMAL(S) Female Lewis rats (n = 9) as donors to generate whole-uterus scaffolds using three different protocols (n = 3 per protocol); Sprague Dawley rats (n = 40) for primary uterus cell isolation procedures (n = 10) and for transplantation/pregnancy studies (n = 30); and male Sprague Dawley rats (n = 12) for mating. INTERVENTION(S) Decellularization was achieved by whole-uterus perfusion with buffered or nonbuffered Triton-X100 and dimethyl sulfoxide (DMSO; group P1/P2) or with sodium deoxycholate (group P3). Primary uterine cells and GFP-MSCs were used to develop uterine tissue constructs, which were grafted to uteri with partial tissue defects. MAIN OUTCOME MEASURE(S) Recellularization efficiency and graft quality were analyzed morphologically, immunohistochemically, and by real-time quantitative polymerase chain reaction (PCR). The location and number of fetuses were documented during pregnancy days 16-20. RESULT(S) Pregnancy and fetal development were normal in groups P1 and P2, with fetal development over patched areas. Group P3 showed significant reduction of fetal numbers, and embryos were not seen in the grafted area. Quantitative PCR and immunohistochemistry revealed uterus-like tissue in the patches, which had been further reconstructed by infiltrating host cells after transplantation. CONCLUSION(S) Primary uterine cells and MSCs can be used to reconstruct decellularized uterine tissue. The bioengineered patches made from triton-X100+DMSO-generated scaffolds were supportive during pregnancy. These protocols should be explored further to develop suitable grafting material to repair partially defect uteri and possibly to create a complete bioengineered uterus.
Collapse
Affiliation(s)
- Mats Hellström
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Juan M Moreno-Moya
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sara Bandstein
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Eva Bom
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Surgery, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Randa R Akouri
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kaoru Miyazaki
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Tokyo, Japan
| | - Tetsuo Maruyama
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Tokyo, Japan
| | - Mats Brännström
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
22
|
Shalaby SM, Khater MK, Perucho AM, Mohamed SA, Helwa I, Laknaur A, Lebedyeva I, Liu Y, Diamond MP, Al-Hendy AA. Magnetic nanoparticles as a new approach to improve the efficacy of gene therapy against differentiated human uterine fibroid cells and tumor-initiating stem cells. Fertil Steril 2016; 105:1638-1648.e8. [PMID: 27020169 DOI: 10.1016/j.fertnstert.2016.03.001] [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] [Received: 09/18/2015] [Revised: 03/02/2016] [Accepted: 03/03/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To study whether efficient transduction and subsequent elimination of fibroid tumor-initiating stem cells during debulking of tumor cells will aid in completely eradicating the tumor as well as decreasing the likelihood of recurrence. DESIGN Case control study. SETTING Research laboratory. PATIENT(S) None. INTERVENTION(S) Magnetic nanoparticles (MNPs) complexed to adenovirus (Ad-GFP) or (Ad-LacZ) used to transfect differentiated human fibroid cells in vitro. MAIN OUTCOME MEASURE(S) Rate of transduction and tumor growth inhibition. RESULT(S) We have developed a localized nonsurgical adenovirus-based alternative for the treatment of uterine fibroids that combines viral-based gene delivery with nanotechnology for more efficient targeting. Magnetic nanoparticles complexed to adenovirus, in the presence of an external magnetic field, accelerate adenovirus transduction. We observed a statistically significant increase in transduction efficiency among differentiated human fibroid cells at two different multiplicities of infection (MOI), 1 and 10, respectively, with MNPs as compared with adenovirus alone. Human fibroid stem cells transfected with Ad-LacZ expressed β-galactosidaze at a MOI of 1, 10, and 50 at 19%, 62%, and 90%, respectively, which were statistically significantly enhanced with MNPs. CONCLUSION(S) When applied with adenovirus herpes simplex thymidine kinase, magnetofection statistically significantly suppressed proliferation and induced apoptosis in both cell types. Through the use of magnetofection, we will prove that a lower viral dose will effectively increase the overall safety profile of suicide gene therapy against fibroid tumors.
Collapse
Affiliation(s)
- Shahinaz Mahmood Shalaby
- Department of Obstetrics and Gynecology, Georgia Regents University, Augusta, Georgia; Department of Pharmacology, Tanta Faculty of Medicine, Tanta, Egypt
| | - Mostafa K Khater
- Department of Obstetrics and Gynecology, Georgia Regents University, Augusta, Georgia
| | - Aymara Mas Perucho
- Department of Obstetrics and Gynecology, Georgia Regents University, Augusta, Georgia
| | - Sara A Mohamed
- Department of Obstetrics and Gynecology, Georgia Regents University, Augusta, Georgia; Department of Obstetrics and Gynecology, Mansoura University Hospital, Mansoura Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Inas Helwa
- Department of Cell Biology and Anatomy, Georgia Regents University, Augusta, Georgia
| | - Archana Laknaur
- Department of Obstetrics and Gynecology, Georgia Regents University, Augusta, Georgia
| | - Iryna Lebedyeva
- Department of Chemistry and Physics, Georgia Regents University, Augusta, Georgia
| | - Yutao Liu
- Department of Cell Biology and Anatomy, Georgia Regents University, Augusta, Georgia
| | - Michael P Diamond
- Department of Obstetrics and Gynecology, Georgia Regents University, Augusta, Georgia
| | - Ayman A Al-Hendy
- Department of Obstetrics and Gynecology, Georgia Regents University, Augusta, Georgia.
| |
Collapse
|
23
|
Ono M, Kajitani T, Uchida H, Arase T, Oda H, Uchida S, Ota K, Nagashima T, Masuda H, Miyazaki K, Asada H, Hida N, Mabuchi Y, Morikawa S, Ito M, Bulun SE, Okano H, Matsuzaki Y, Yoshimura Y, Maruyama T. CD34 and CD49f Double-Positive and Lineage Marker-Negative Cells Isolated from Human Myometrium Exhibit Stem Cell-Like Properties Involved in Pregnancy-Induced Uterine Remodeling. Biol Reprod 2015; 93:37. [PMID: 26108791 PMCID: PMC6058739 DOI: 10.1095/biolreprod.114.127126] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 01/10/2015] [Accepted: 06/09/2015] [Indexed: 01/08/2023] Open
Abstract
Repeated and dramatic pregnancy-induced uterine enlargement and remodeling throughout reproductive life suggests the existence of uterine smooth muscle stem/progenitor cells. The aim of this study was to isolate and characterize stem/progenitor-like cells from human myometrium through identification of specific surface markers. We here identify CD49f and CD34 as markers to permit selection of the stem/progenitor cell-like population from human myometrium and show that human CD45(-) CD31(-) glycophorin A(-) and CD49f(+) CD34(+) myometrial cells exhibit stem cell-like properties. These include side population phenotypes, an undifferentiated status, high colony-forming ability, multilineage differentiation into smooth muscle cells, osteoblasts, adipocytes, and chondrocytes, and in vivo myometrial tissue reconstitution following xenotransplantation. Furthermore, CD45(-) CD31(-) glycophorin A(-) and CD49f(+) CD34(+) myometrial cells proliferate under hypoxic conditions in vitro and, compared with the untreated nonpregnant myometrium, show greater expansion in the estrogen-treated nonpregnant myometrium and further in the pregnant myometrium in mice upon xenotransplantation. These results suggest that the newly identified myometrial stem/progenitor-like cells influenced by hypoxia and sex steroids may participate in pregnancy-induced uterine enlargement and remodeling, providing novel insights into human myometrial physiology.
Collapse
Affiliation(s)
- Masanori Ono
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Takashi Kajitani
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Hiroshi Uchida
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Toru Arase
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Hideyuki Oda
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Sayaka Uchida
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Kuniaki Ota
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Takashi Nagashima
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Hirotaka Masuda
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Kaoru Miyazaki
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Hironori Asada
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Naoko Hida
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Yo Mabuchi
- Department of Physiology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Satoru Morikawa
- Department of Physiology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Mamoru Ito
- Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan
| | - Serdar E Bulun
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Hideyuki Okano
- Department of Physiology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Yumi Matsuzaki
- Department of Physiology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Yasunori Yoshimura
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Tetsuo Maruyama
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| |
Collapse
|
24
|
Taylor DK, Holthouser K, Segars JH, Leppert PC. Recent scientific advances in leiomyoma (uterine fibroids) research facilitates better understanding and management. F1000Res 2015; 4:183. [PMID: 26236472 PMCID: PMC4513689 DOI: 10.12688/f1000research.6189.1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/02/2015] [Indexed: 01/12/2023] Open
Abstract
Uterine leiomyomas (fibroids) are the most prevalent medical problem of the female reproductive tract, but there are few non-surgical treatment options. Although many advances in the understanding of the molecular components of these tumors have occurred over the past five years, an effective pharmaceutical approach remains elusive. Further, there is currently no clinical method to distinguish a benign uterine leiomyoma from a malignant leiomyosarcoma prior to treatment, a pressing need given concerns about the use of the power morcellator for minimally invasive surgery. This paper reviews current studies regarding the molecular biology of uterine fibroids, discusses non-surgical approaches and suggests new cutting-edge therapeutic and diagnostic approaches.
Collapse
Affiliation(s)
- Darlene K Taylor
- Department of Chemistry, North Carolina Central University, Durham, NC, 27707, USA
| | - Kristine Holthouser
- Department of Obstetrics and Gynecology, University of Louisville, Louisville, KY, USA
| | - James H Segars
- Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Phyllis C Leppert
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, 27710, USA
| |
Collapse
|
25
|
Simmen RCM, Heard ME, Simmen AM, Montales MTM, Marji M, Scanlon S, Pabona JMP. The Krüppel-like factors in female reproductive system pathologies. J Mol Endocrinol 2015; 54:R89-R101. [PMID: 25654975 PMCID: PMC4369192 DOI: 10.1530/jme-14-0310] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Female reproductive tract pathologies arise largely from dysregulation of estrogen and progesterone receptor signaling, leading to aberrant cell proliferation, survival, and differentiation. The signaling pathways orchestrated by these nuclear receptors are complex, require the participation of many nuclear proteins serving as key binding partners or targets, and involve a range of paracrine and autocrine regulatory circuits. The members of the Krüppel-like factor (KLF) family of transcription factors are ubiquitously expressed in reproductive tissues and have been increasingly implicated as critical co-regulators and integrators of steroid hormone actions. Herein, we explore the involvement of KLF family members in uterine pathology, describe their currently known molecular mechanisms, and discuss their potential as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Rosalia C M Simmen
- Department of Physiology and BiophysicsUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USADepartment of Obstetrics and GynecologyUniversity of Michigan Health System, Ann Arbor, Michigan 48109, USADepartment of Internal MedicineHarlem Hospital Center, Columbia University Medical Center, New York, New York 10037, USA
| | - Melissa E Heard
- Department of Physiology and BiophysicsUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USADepartment of Obstetrics and GynecologyUniversity of Michigan Health System, Ann Arbor, Michigan 48109, USADepartment of Internal MedicineHarlem Hospital Center, Columbia University Medical Center, New York, New York 10037, USA
| | - Angela M Simmen
- Department of Physiology and BiophysicsUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USADepartment of Obstetrics and GynecologyUniversity of Michigan Health System, Ann Arbor, Michigan 48109, USADepartment of Internal MedicineHarlem Hospital Center, Columbia University Medical Center, New York, New York 10037, USA
| | - Maria Theresa M Montales
- Department of Physiology and BiophysicsUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USADepartment of Obstetrics and GynecologyUniversity of Michigan Health System, Ann Arbor, Michigan 48109, USADepartment of Internal MedicineHarlem Hospital Center, Columbia University Medical Center, New York, New York 10037, USA
| | - Meera Marji
- Department of Physiology and BiophysicsUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USADepartment of Obstetrics and GynecologyUniversity of Michigan Health System, Ann Arbor, Michigan 48109, USADepartment of Internal MedicineHarlem Hospital Center, Columbia University Medical Center, New York, New York 10037, USA
| | - Samantha Scanlon
- Department of Physiology and BiophysicsUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USADepartment of Obstetrics and GynecologyUniversity of Michigan Health System, Ann Arbor, Michigan 48109, USADepartment of Internal MedicineHarlem Hospital Center, Columbia University Medical Center, New York, New York 10037, USA
| | - John Mark P Pabona
- Department of Physiology and BiophysicsUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USADepartment of Obstetrics and GynecologyUniversity of Michigan Health System, Ann Arbor, Michigan 48109, USADepartment of Internal MedicineHarlem Hospital Center, Columbia University Medical Center, New York, New York 10037, USA
| |
Collapse
|