1
|
Pandey V, Jain P, Chatterjee S, Rani A, Tripathi A, Dubey PK. Variants in exon 2 of MED12 gene causes uterine leiomyoma's through over-expression of MMP-9 of ECM pathway. Mutat Res 2024; 828:111839. [PMID: 38041927 DOI: 10.1016/j.mrfmmm.2023.111839] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 10/09/2023] [Accepted: 10/31/2023] [Indexed: 12/04/2023]
Abstract
AIMS To study the impact of Mediator complex subunit 12 (MED12) gene variants on the encoded protein's function and pathogenic relevance for genesis of uterine leiomyoma's (ULs). METHODS Mutational analysis in exon-2 of MED12 gene was performed by PCR amplification and DNA sequencing in 89 clinically diagnosed ULs tissues. Pathogenicity prediction of variation was performed by computational analysis. The functional effects of missense variation were done by quantity RT-PCR and western blot analysis. RESULT(S) Out of 89 samples, 40 (44.94%) had missense variation in 14 different CDS position of exon-2 of MED12 gene. Out of 40 missense variation, codon 44 had 25 (62.5%) looking as a hotspot region for mutation for ULs, because CDS position c130 and c131present at codon 44 that have necleotide change G>A, T, C at c130 and c131 have necleotide change G>A and C. We also find somenovel somatic mutations oncodon 36 (T > C), 38 (G>T) of exon-2 and 88 (G>C) of intron-2. No mutations were detected in uterine myometrium samples. Our computational analysis suggests that change in Med12c .131 G>A leads to single substitution of amino acid [Glycine (G) to Aspartate (D)] which has a pathogenic and lethal impact and may cause instability of MED12 protein. Further, analysis of extracellular matrix (ECM) component (MMP-2 & 9, COL4A2 and α-SMA) mRNA and protein expression levels in the set of ULs having MED12 mutation showed significantly higher expression of MMP-9 and α-SMA. CONCLUSION(S) The findings of present study suggest that missense variation in codon 44 of MED12 gene lead to the genesis of leiomyoma's through over-expression of MMP-9 of ECM pathway which could be therapeutically targeted for non-surgical management of ULs.
Collapse
Affiliation(s)
- Vivek Pandey
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Priyanka Jain
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida 201313, Uttar Pradesh, India
| | - Souradip Chatterjee
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Anjali Rani
- Department of Obstetrics and Gynecology, Institute of Medical Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Anima Tripathi
- MMV, Zoology Section, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Pawan K Dubey
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| |
Collapse
|
2
|
Tsikouras P, Gkaitatzi F, Gerede A, Anthoulaki X, Bothou A, Chalkidou A, Michalopoulos S, Tsirkas I, Gyroglou S, Peitsidis P, Nikolettos K, Alexiou A, Dragoutsos G, Sachnova N, Chloropoulou P, Zervoudis S, Iatrakis G, Rath W, Trypsiannis G, Nikolettos N, Souftas V. Life Quality in Premenopausal Women after Embolization of Uterine Myomas. J Pers Med 2022; 12:jpm12121990. [PMID: 36556210 PMCID: PMC9786225 DOI: 10.3390/jpm12121990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/01/2022] [Accepted: 11/10/2022] [Indexed: 12/02/2022] Open
Abstract
Objectives: Fibroids cause significant morbidity and are the most common indication for hysterectomies worldwide, delimiting a major public health problem. Uterine artery embolization (UAE) is an alternative therapy to surgical treatment of symptomatic fibroids; it has satisfactory long-time results and is no longer considered investigational for the treatment of symptomatic fibroids. This study was undertaken to evaluate changes in fibroid specific symptom severity and health-related quality of life (HRQOL) after UAE and to optimize the assessment of safety and outcomes measures for participants who receive UAE to objective compare UAE and surgical alternatives for therapy of symptomatic fibroids. Study design: The analysis was based on questionnaires completed by 270 pre-menopausal females with a mean age of 42 years (range, 38-50 years) who underwent UAE for uterine leiomyomas and/or adenomyosis from November 2013 through December 2019. Only symptomatic women were selected whose symptoms were not improving with medication and who did not wish to have children. The primary outcome measure was a change in fibroid symptoms and HRQOL (health related quality of life) after UAE. Secondary outcomes included the decrease in uterine volume after UAE. Results: Questionnaires were completed by 270 women (100%) at a mean of 12.1 months from UAE. The median follow-up period was two years. Uterine fibroid embolization led to a shrinkage at three months for the 90% of the participants. A reduction of bleeding symptoms, pain and bulk-related symptoms was observed in 89.7%, 88.9%, and 89.5% of the patients, respectively. In the long term, there was no significant difference in parameters assessed compared with the midterm follow-up findings. A total of 6 patients (2.3%) underwent fractional curettage an average of 32.1 months after intervention due to necrotic changes in submucosal fibroids. All participants continued to be satisfied with the intervention, and 240 patients (88.9%) answered that they would recommend uterine fibroid embolization to other patients. Conclusions: Women who undergo UAE have a significant decrease in symptom severity and increase in HRQOL which is associated with high levels of satisfaction with the procedure (even when subsequent therapies are pursued).
Collapse
Affiliation(s)
- Panagiotis Tsikouras
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandropoulis, Greece
- Correspondence:
| | - Foteini Gkaitatzi
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandropoulis, Greece
| | - Aggeliki Gerede
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandropoulis, Greece
| | - Xanthoula Anthoulaki
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandropoulis, Greece
| | - Anastasia Bothou
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandropoulis, Greece
| | - Anna Chalkidou
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandropoulis, Greece
| | - Spyridon Michalopoulos
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandropoulis, Greece
| | - Ioannis Tsirkas
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandropoulis, Greece
| | - Selma Gyroglou
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandropoulis, Greece
| | | | - Konstantinos Nikolettos
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandropoulis, Greece
| | - Alexios Alexiou
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandropoulis, Greece
| | - George Dragoutsos
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandropoulis, Greece
| | - Natalia Sachnova
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandropoulis, Greece
| | - Pelagia Chloropoulou
- Department of Anaesthesiology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | | | - George Iatrakis
- Rea Maternity Hospital, University of West Attica, 17564 Athens, Greece
| | - Werner Rath
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandropoulis, Greece
| | - Grigorios Trypsiannis
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandropoulis, Greece
| | - Nikolaos Nikolettos
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandropoulis, Greece
| | - Vasileios Souftas
- Department of Interventional Radiology, Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
3
|
Shen M, Duan H, Chang Y, Wang S. Growth of surgically confirmed leiomyomas in postmenopausal women: analysis of the influencing factors. Menopause 2021; 28:1209-1213. [PMID: 34469931 DOI: 10.1097/gme.0000000000001846] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES The aim of this study was to examine the growth of uterine leiomyomas in postmenopausal women and evaluate the influencing factors associated with fibroid growth. METHODS We retrospectively analyzed the medical records of postmenopausal women with fibroids between 2015 and 2020. All women received at least 2 transvaginal ultrasound examinations within a 6-month interval. All fibroids were verified via surgery. The postoperative pathology of all tumors was uterine fibroid. We calculated the fibroid volume using the ellipsoid volume formula and evaluated the growth rate of fibroids within 6 months simultaneously. Univariable analysis and a linear mixed-effects model were used to assess the factors influencing fibroid growth. RESULTS A total of 102 postmenopausal women with a total of 132 fibroids were assessed. The median growth rate of surgically confirmed fibroids in postmenopausal women was 12.9% every 6 months (from -61.4% to 184.1%). Obesity was associated with fibroid growth (P < 0.05). Notably, the estimated growth rates of fibroids in obese and overweight women were 26.6% (95% confidence interval [CI]: 2.3-50.9) and 15.9% (95% CI: 0.4-31.4) higher than those in women of normal weight, respectively. The growth of fibroids varied by the initial tumor size (P < 0.05). The estimated growth rate of larger fibroids (≥5.0 cm diameter) was reduced 30.0% (95% CI: -52.4 to -7.5) compared with that of small fibroids (<3.0 cm diameter). CONCLUSIONS Uterine fibroids continually grow in some postmenopausal women. Obesity and small fibroids (<3.0 cm diameter) may contribute to higher growth rates of fibroids.
Collapse
Affiliation(s)
- Minghong Shen
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
- Department of Gynecology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian Province, China
| | - Hua Duan
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Yanan Chang
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Sha Wang
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
El Sabeh M, Saha SK, Afrin S, Islam MS, Borahay MA. Wnt/β-catenin signaling pathway in uterine leiomyoma: role in tumor biology and targeting opportunities. Mol Cell Biochem 2021; 476:3513-3536. [PMID: 33999334 DOI: 10.1007/s11010-021-04174-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023]
Abstract
Uterine leiomyoma is the most common tumor of the female reproductive system and originates from a single transformed myometrial smooth muscle cell. Despite the immense medical, psychosocial, and financial impact, the exact underlying mechanisms of leiomyoma pathobiology are poorly understood. Alterations of signaling pathways are thought to be instrumental in leiomyoma biology. Wnt/β-catenin pathway appears to be involved in several aspects of the genesis of leiomyomas. For example, Wnt5b is overexpressed in leiomyoma, and the Wnt/β-catenin pathway appears to mediate the role of MED12 mutations, the most common mutations in leiomyoma, in tumorigenesis. Moreover, Wnt/β-catenin pathway plays a paracrine role where estrogen/progesterone treatment of mature myometrial or leiomyoma cells leads to increased expression of Wnt11 and Wnt16, which induces proliferation of leiomyoma stem cells and tumor growth. Constitutive activation of β-catenin leads to myometrial hyperplasia and leiomyoma-like lesions in animal models. Wnt/β-catenin signaling is also closely involved in mechanotransduction and extracellular matrix regulation and relevant alterations in leiomyoma, and crosstalk is noted between Wnt/β-catenin signaling and other pathways known to regulate leiomyoma development and growth such as estrogen, progesterone, TGFβ, PI3K/Akt/mTOR, Ras/Raf/MEK/ERK, IGF, Hippo, and Notch signaling. Finally, evidence suggests that inhibition of the canonical Wnt pathway using β-catenin inhibitors inhibits leiomyoma cell proliferation. Understanding the molecular mechanisms of leiomyoma development is essential for effective treatment. The specific Wnt/β-catenin pathway molecules discussed in this review constitute compelling candidates for therapeutic targeting.
Collapse
Affiliation(s)
- Malak El Sabeh
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD, 21205, USA
| | - Subbroto Kumar Saha
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD, 21205, USA
| | - Sadia Afrin
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD, 21205, USA
| | - Md Soriful Islam
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD, 21205, USA
| | - Mostafa A Borahay
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD, 21205, USA.
| |
Collapse
|
5
|
Hereditary leiomyomatosis and renal cell cancer syndrome: An update and review. J Am Acad Dermatol 2017; 77:149-158. [PMID: 28314682 DOI: 10.1016/j.jaad.2017.01.023] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 01/07/2017] [Accepted: 01/11/2017] [Indexed: 02/06/2023]
Abstract
Hereditary leiomyomatosis and renal cell cancer (HLRCC) syndrome is a rare genetic disorder that predisposes individuals to multiple cutaneous leiomyomas, renal cell carcinomas, and in women, uterine leiomyomas. Also known as Reed syndrome, it is caused by a germline heterozygous mutation of the fumarate hydratase tumor suppressor gene. HLRCC is associated with significant morbidity because of pain from cutaneous and uterine leiomyomas, the cutaneous pain often of unique character. Although genetic testing is currently considered the criterion standard to diagnose HLRCC, newer immunohistochemistry markers may provide rapid and cost effective alternatives to genetic testing. Because of the potentially aggressive nature of renal cell carcinomas that develop as early as in childhood, close annual cancer surveillance is desirable in individuals with HLRCC. In this review, we offer an update and an approach to the diagnosis, management, and renal cancer surveillance in HLRCC.
Collapse
|
6
|
Leiomyoma and leiomyoma cellulare of the fallopian tube: review of the literature and case reports. PRZEGLAD MENOPAUZALNY = MENOPAUSE REVIEW 2016; 15:143-147. [PMID: 27980525 PMCID: PMC5137476 DOI: 10.5114/pm.2016.63053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/16/2016] [Indexed: 11/23/2022]
Abstract
Introduction Leiomyoma of the fallopian tube is extremely rare, and its version – leiomyoma cellulare (LC) of the fallopian tube is absolutely unique. Aim of the study was to review literature reports on leiomyomas of the fallopian tubes, and to present cases of leiomyoma and LC of the fallopian tubes in the patients operated on in our ward. Material and methods There were fewer than 100 cases of leiomyomas of the fallopian tubes discussed in the literature up to 1993. Case 1. Leiomyoma of the left fallopian tube was detected postoperatively in a 68-year-old patient, G.K., on histopathological examination after laparoscopic total hysterectomy with bilateral adnexa. Case 2. A 56-year-old patient, K.T., with LC of the fallopian tube was qualified for laparoscopy. At operation, the procedure was converted to microlaparotomy due to the tumor size. The adnexa on the right side with the tumor of the fallopian tube were excised, and the left fallopian tube was excised, too. Histopathological microscopy found leiomyoma cellulare partim epithelioides. Results In the presented cases, the extent of operation was connected with the clinical picture, and in the case of LC of the right fallopian tube, with intraoperative histopathological findings. In both cases the postoperative course was uneventful. Conclusions Diagnosis of leiomyoma and LC of the fallopian tube, like in the other organs of the female genital tract, is possible only due to results of histopathological microscopy.
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW The review analyzes how fibroids may influence pregnancy and how myomas may be modified by pregnancy. The most important clinical aspect concerns the impact of myoma on pregnancy and the possibility of a well tolerated surgical treatment for the mother and her fetus, preserving maternal reproductive capacity. RECENT FINDINGS Fibroids significantly increase in size during early pregnancy and then decrease in the third trimester. Although most women with uterine fibroids have a regular pregnancy, data from the literature suggest that they may have a higher risk of fertility problems and pregnancy complications. SUMMARY Myomectomy can increase the rate of pregnancy in women with infertility, attempting to restore a normal anatomy and reduce uterine contractility and local inflammation associated with the presence of fibroids, improving the blood supply. Current evidence does not suggest routine myomectomy during pregnancy or at the cesarean birth, as fibroids-related complications are rare and may be overcome by the risks of surgery. However, in selected cases, myomectomy is a feasible and safe technique and associated to a good outcome.The diagnosis of myomas in pregnancy may require attention for the adequate management to preserve maternal and fetal well-being.
Collapse
|
8
|
Prevalence of uterine myomas in women in Germany: data of an epidemiological study. Arch Gynecol Obstet 2015; 293:1243-53. [DOI: 10.1007/s00404-015-3930-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 10/16/2015] [Indexed: 11/25/2022]
|
9
|
Fujisawa C, Castellot JJ. Matrix production and remodeling as therapeutic targets for uterine leiomyoma. J Cell Commun Signal 2014; 8:179-94. [PMID: 25012731 DOI: 10.1007/s12079-014-0234-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 05/22/2014] [Indexed: 01/06/2023] Open
Abstract
Uterine leiomyoma, commonly known as fibroids, is a benign neoplasm of smooth muscle in women. The incidence of clinically symptomatic fibroids in reproductive-age women is approximately 20 %, with nearly 80 % of black women suffering from this condition. Symptoms include severe pain and hemorrhage; fibroids are also a major cause of infertility or sub-fertility in women. Uterine leiomyoma consist of hyperplastic smooth muscle cells and an excess deposition of extracellular matrix, specifically collagen, fibronectin, and sulfated proteoglycans. Extracellular matrix components interact and signal through integrin-β1 on the surface of uterine leiomyoma smooth muscle cells, provide growth factor storage, and act as co-receptors for growth factor-receptor binding. ECM and growth factor signaling through integrin-β1 and growth factor receptors significantly increases cell proliferation and ECM deposition in uterine leiomyoma. Growth factors TGF-β, IGF, PDGF, FGF and EGF are all shown to promote uterine leiomyoma progression and signal through multiple pathways to increase the expression of genes encoding matrix or matrix-modifying proteins. Decreasing integrin expression, reducing growth factor action and inhibiting ECM action on uterine leiomyoma smooth muscle cells are important opportunities to treat uterine leiomyoma without use of the current surgical procedures. Both natural compounds and chemicals are shown to decrease fibrosis and uterine leiomyoma progression, but further analysis is needed to make inroads in treating this common women's health issue.
Collapse
Affiliation(s)
- Caitlin Fujisawa
- Public Heath and Professional Degrees Program, Tufts University School of Medicine, Boston, MA, 02111, USA
| | | |
Collapse
|
10
|
Uterine fibroids: pathogenesis and interactions with endometrium and endomyometrial junction. Obstet Gynecol Int 2013; 2013:173184. [PMID: 24163697 PMCID: PMC3791844 DOI: 10.1155/2013/173184] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 06/10/2013] [Accepted: 08/13/2013] [Indexed: 01/30/2023] Open
Abstract
Uterine leiomyomas (fibroids or myomas) are benign tumors of uterus and clinically apparent in a large part of reproductive aged women. Clinically, they present with a variety of symptoms: excessive menstrual bleeding, dysmenorrhoea and intermenstrual bleeding, chronic pelvic pain, and pressure symptoms such as a sensation of bloatedness, increased urinary frequency, and bowel disturbance. In addition, they may compromise reproductive functions, possibly contributing to subfertility, early pregnancy loss, and later pregnancy complications. Despite the prevalence of this condition, myoma research is underfunded compared to other nonmalignant diseases. To date, several pathogenetic factors such as genetics, microRNA, steroids, growth factors, cytokines, chemokines, and extracellular matrix components have been implicated in the development and growth of leiomyoma. This paper summarizes the available literature regarding the ultimate relative knowledge on pathogenesis of uterine fibroids and their interactions with endometrium and subendometrial myometrium.
Collapse
|
11
|
Review: Human uterine stem/progenitor cells: Implications for uterine physiology and pathology. Placenta 2013; 34 Suppl:S68-72. [PMID: 23332213 DOI: 10.1016/j.placenta.2012.12.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 12/14/2012] [Accepted: 12/17/2012] [Indexed: 01/17/2023]
Abstract
The human uterus is composed of the endometrial lining and the myometrium. The endometrium, in particular the functionalis layer, regenerates and regresses with each menstrual cycle under hormonal control. A mouse xenograft model has been developed in which the functional changes of the endometrium are reproduced. The myometrium possesses similar plasticity, critical to permit the changes connected with uterine expansion and involution associated with pregnancy. Regeneration and remodeling in the uterus are likely achieved through endometrial and myometrial stem cell systems. Putative stem/progenitor cells in humans and rodents recently have been identified, isolated and characterized. Their roles in endometrial physiology and pathophysiology are presently under study. These stem/progenitor cells ultimately may provide a novel means by which to produce tissues and organs in vitro and in vivo.
Collapse
|
12
|
Drosch M, Bullerdiek J, Zollner TM, Prinz F, Koch M, Schmidt N. A novel mouse model that closely mimics human uterine leiomyomas. Fertil Steril 2012; 99:927-935.e6. [PMID: 23260859 DOI: 10.1016/j.fertnstert.2012.11.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 11/15/2012] [Accepted: 11/16/2012] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To develop a predictive mouse model for uterine fibroids. DESIGN Human fibroid cells xenografted to immunodeficient mice. SETTING University and industrial research center. ANIMAL(S) Immunodeficient scid/beige mice. INTERVENTION(S) Subcutaneous and intrauterine injection of fibroid-derived cells, SV40 transformation of primary cells by lentiviral transduction, proliferation determined by immunohistochemistry, FISH. MAIN OUTCOME MEASURE(S) Characterization of primary and immortalized cells by Western blot and soft agar assay, determination of in vivo tumorigenicity, comparative histology and immunohistochemistry, fluorescence in situ hybridization. RESULT(S) Tumorigenicity of primary myoma cells disappears upon in vitro culture. Transformation and immortalization does not restore or conserve the in vivo growth potential of cultured cells. Injection of primary cells into myometrium of mice leads to xenografts with a leiomyoma-like histology. CONCLUSION(S) Primary myoma cells are suited to generate fibroid-like xenografts for studying pathogenesis without genetic modifications. In contrast, in vitro culture abolishes transplantability, and neither transformation nor immortalization is sufficient to restore tumorigenic capacity.
Collapse
Affiliation(s)
- Michael Drosch
- Center of Human Genetics, University of Bremen, Bremen, Germany
| | | | | | | | | | | |
Collapse
|
13
|
Roeder H, Jayes F, Feng L, Leppert PC. CDB-4124 does not cause apoptosis in cultured fibroid cells. Reprod Sci 2011; 18:850-7. [PMID: 21422050 DOI: 10.1177/1933719111399929] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Selective progesterone receptor modulators (SPRMs), such as asoprisnil (J867) and ulipristal (CDB-2914), have been shown to reduce fibroid volume in vivo and to induce apoptosis in vitro. CDB-4124 (telapristone), a SPRM with different side groups, also reduced fibroid volume in vivo, and we hypothesized that this SPRM would also cause apoptosis in cultured fibroid cells. METHODS Immortalized, progesterone receptor-positive fibroid cells, known to be capable of apoptosis, were grown to 80% confluence in serum-containing media. Cells were then treated for 48 hours in serum-free media with 0, 10, 100, or 1000 nmol/L CDB-4124. Actinomycin-D and staurosporine were used as positive controls to induce apoptosis. Apoptosis was quantified using a TUNEL-fluorescein kit. Images were captured with a widefield-fluorescence microscope and analyzed using MetaMorph image analysis software. To validate results, Western blots of total cell lysates were probed for cleaved caspase-3 (c-CASP3). Experiments were repeated 3 times using independent cell batches. RESULTS Analysis of 19 712 nuclei indicated 14.8% ± 10.9% (mean ± SEM), 8.4% ± 4.6%, 8.2% ± 4.7%, and 9.3% ± 6.3% apoptosis in 0, 10, 100, and 1000 nmol/L CDB-4124-treated cells, respectively. There was no evidence of elevated c-CASP3 over vehicle control after treatment with CDB-4124. CONCLUSION CDB-4124 did not significantly induce apoptosis in cultured fibroid cells under the conditions described suggesting apoptosis may not be the main pathway responsible for CDB-4124-induced fibroid shrinkage. Variations in SPRM biological effects may be due to differences in fibroid source cells, binding kinetics, or extracellular matrix characteristics, and can be exploited in further investigations of the mechanisms of action of SPRMs in fibroid biology.
Collapse
Affiliation(s)
- Hilary Roeder
- Department of Obstetrics & Gynecology, Duke University, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
14
|
Jiang Y, Suo G, Sadarangani A, Cowan B, Wang JYJ. Expression profiling of protein tyrosine kinases and their ligand activators in leiomyoma uteri. Syst Biol Reprod Med 2010; 56:318-26. [PMID: 20462350 DOI: 10.3109/19396361003646109] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The aim of this study is to compare the expression patterns of tyrosine kinases and their ligand activators between matched myometrium and leiomyoma tissues. Total RNA extracted from 42 pairs of matched leiomyomal and adjacent myometrial tissues were hybridized to a set of 840 customized oligonucleotide microarrays to compare the expression profiles of 244 selected human genes including 90 tyrosine kinases and 103 ligands. Among the 244 genes surveyed, 38 were found to exhibit differential expression between pairs of myometrium and leiomyoma tissues. Clustering analysis of the expression ratios of these 38 genes from 42 pairs of samples led to the subdivision of fibroid samples into three groups, based in principle on the expression ratios of two peptide ligands, CYR61 and EFNA4. Real-time quantitative RT-PCR measurements of an additional 32 pairs of samples further confirmed the three subgroups. The altered expression of ligand activators between myometrium and leiomyoma suggest that tyrosine kinases regulated by CYR61 and EFNA4 may be exploited as therapeutic targets to develop non-surgical treatments of symptomatic leiomyomas.
Collapse
Affiliation(s)
- Yong Jiang
- Department of Medicine and Moores, UCSD Cancer Center, La Jolla, CA 92093-0820, USA
| | | | | | | | | |
Collapse
|
15
|
Markowski DN, Bartnitzke S, Belge G, Drieschner N, Helmke BM, Bullerdiek J. Cell culture and senescence in uterine fibroids. ACTA ACUST UNITED AC 2010; 202:53-7. [PMID: 20804922 DOI: 10.1016/j.cancergencyto.2010.06.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Revised: 06/07/2010] [Accepted: 06/14/2010] [Indexed: 11/27/2022]
Abstract
The in vitro growth of cells from uterine fibroids is characterized by an early onset of senescence. Often, an even lower growth potential than that of matching myometrial cells is noted. Also, the tremendous differences in the expression of the high mobility group protein HMGA2 seen when comparing fibroids of different genetic subtypes are surprisingly not reflected by significant differences in their growth potential in vitro. We aimed to evaluate possible changes of the HMGA2 expression level between the native tissue and cell cultures, so we performed quantitative real-time polymerase chain reaction studies that revealed a marked decrease of the HMGA2 mRNA in culture in those cases with overexpression of HMGA2. In the two cases initially showing the highest expression, it decreased by approximately 97%. Associated with the decrease of HMGA2 was a clearly increased expression of the senescence-associated p19(Arf). Together, these findings explain the similar behavior of cell cultures from fibroids of different genetic subgroups and may also offer an explanation for the early onset of in vitro senescence in these cell cultures.
Collapse
|
16
|
Chegini N. Proinflammatory and profibrotic mediators: principal effectors of leiomyoma development as a fibrotic disorder. Semin Reprod Med 2010; 28:180-203. [PMID: 20414842 DOI: 10.1055/s-0030-1251476] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Leiomyomas are believed to derive from the transformation of myometrial smooth muscle cells/connective tissue fibroblasts. Although the identity of the molecule(s) that initiate such cellular transformation and orchestrate subsequent growth is still unknown, conventional evidence indicates that ovarian steroids are essential for leiomyoma growth. Ovarian steroid action in their target cell/tissue is mediated in part through local expression of various growth factors, cytokines, and chemokines. These autocrine/paracrine molecules with proinflammatory and profibrotic activities serve as major contributing factors in regulating cellular transformation, cell growth and apoptosis, angiogenesis, cellular hypertrophy, and excess tissue turnover, events central to leiomyoma growth. This review addresses the key regulatory functions of proinflammatory and profibrotic mediators and their molecular mechanisms, downstream signaling that regulates cellular events that result in transformation, and commitments of specific cells into forming a cellular environment with a possible role in development and subsequent growth of leiomyomas.
Collapse
Affiliation(s)
- Nasser Chegini
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Florida, Gainesville, Florida 32610, USA.
| |
Collapse
|
17
|
Crabtree JS, Jelinsky SA, Harris HA, Choe SE, Cotreau MM, Kimberland ML, Wilson E, Saraf KA, Liu W, McCampbell AS, Dave B, Broaddus RR, Brown EL, Kao W, Skotnicki JS, Abou-Gharbia M, Winneker RC, Walker CL. Comparison of human and rat uterine leiomyomata: identification of a dysregulated mammalian target of rapamycin pathway. Cancer Res 2009; 69:6171-8. [PMID: 19622772 DOI: 10.1158/0008-5472.can-08-4471] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Uterine leiomyomata, or fibroids, are benign tumors of the uterine myometrium that significantly affect up to 30% of reproductive-age women. Despite being the primary cause of hysterectomy in the United States, accounting for up to 200,000 procedures annually, the etiology of leiomyoma remains largely unknown. As a basis for understanding leiomyoma pathogenesis and identifying targets for pharmacotherapy, we conducted transcriptional profiling of leiomyoma and unaffected myometrium from humans and Eker rats, the best characterized preclinical model of leiomyomata. A global comparison of mRNA from leiomyoma versus myometrium in human and rat identified a highly significant overlap of dysregulated gene expression in leiomyomata. An unbiased pathway analysis using a method of gene-set enrichment based on the sigPathway algorithm detected the mammalian target of rapamycin (mTOR) pathway as one of the most highly up-regulated pathways in both human and rat tumors. To validate this pathway as a therapeutic target for uterine leiomyomata, preclinical studies were conducted in Eker rats. These rats develop uterine leiomyomata as a consequence of loss of Tsc2 function and up-regulation of mTOR signaling. Inhibition of mTOR in female Eker rats with the rapamycin analogue WAY-129327 for 2 weeks decreased mTOR signaling and cell proliferation in tumors, and treatment for 4 months significantly decreased tumor incidence, multiplicity, and size. These results identify dysregulated mTOR signaling as a component of leiomyoma etiology across species and directly show the dependence of uterine leiomyomata with activated mTOR on this signaling pathway for growth.
Collapse
Affiliation(s)
- Judy S Crabtree
- Endocrinology & Reproductive Disorders Division, Women's Health and Musculoskeletal Biology, Wyeth Research, Collegeville, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Check JH. 30 years of IVF: what does the future hold? WOMENS HEALTH 2008; 4:307-10. [PMID: 19072493 DOI: 10.2217/17455057.4.4.307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
19
|
Growth of uterine leiomyomata among premenopausal black and white women. Proc Natl Acad Sci U S A 2008; 105:19887-92. [PMID: 19047643 DOI: 10.1073/pnas.0808188105] [Citation(s) in RCA: 245] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Uterine leiomyomata (fibroids) are the leading cause of hysterectomy in the United States. Black women have a greater fibroid burden than whites, yet no study has systematically evaluated the growth of fibroids in blacks and whites. We prospectively tracked growth for 262 fibroids (size range: 1-13 cm in diameter) from 72 premenopausal participants (38 blacks and 34 whites). Fibroid volume was measured by computerized analysis of up to four MRI scans over 12 months. We used mixed effects models to identify factors that are associated with growth, and results were converted to percent change per 6 months for clinical relevance. The median growth rate was 9% (range: -89% to +138%). Seven percent of fibroids regressed (>20% shrinkage). Tumors from the same woman grew at different rates (within-woman component of variation was twice the component among women; both were significant, P < 0.001). Black and white women less than 35 years of age had similar fibroid growth rates. However, growth rates declined with age for whites but not for blacks (P = 0.05). The odds of a tumor growing more than 20% in 6 months also decreased with age for whites but not for blacks (P < 0.01). Growth rates were not influenced by tumor size, location, body mass index, or parity. We conclude that (i) spontaneous regression of fibroids occurs; (ii) fibroids from the same woman grow at different rates, despite a uniform hormonal milieu; (iii) fibroid size does not predict growth rate; and (iv) age-related differences in fibroid growth between blacks and whites may contribute to the higher symptom burden for black women.
Collapse
|
20
|
Luo X, Chegini N. The expression and potential regulatory function of microRNAs in the pathogenesis of leiomyoma. Semin Reprod Med 2008; 26:500-14. [PMID: 18951332 PMCID: PMC2710997 DOI: 10.1055/s-0028-1096130] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Leiomyomas are benign uterine tumors considered to arise from transformation of myometrial cells. What initiates the conversion of myometrial cells into leiomyoma is unknown, however cytogenetic analysis often shows occurrence of nonrandom chromosomal abnormalities that may account for their establishment. It is clear that ovarian steroids are essential for leiomyoma growth, and local expression of many autocrine/paracrine mediators serving as key regulators of cell-cycle progression, cellular hypertrophy, extracellular matrix accumulation, and apoptosis appear to play central roles in this capacity. However, the stability of the expression of these genes represents the hallmarks of leiomyoma establishment, growth, and regression. With the emergence of microRNA (miRNA) as a key regulator of gene expression stability, in this review we present evidence for the expression and potential regulatory functions on miRNAs in leiomyoma with particular emphasis on the expression of their selective target genes whose products influence various cellular activities critical to pathogenesis of leiomyomas.
Collapse
Affiliation(s)
- Xiaoping Luo
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Florida, Gainesville, Florida 32610, USA.
| | | |
Collapse
|
21
|
Dimitrova IK, Richer JK, Rudolph MC, Spoelstra NS, Reno EM, Medina TM, Bradford AP. Gene expression profiling of multiple leiomyomata uteri and matched normal tissue from a single patient. Fertil Steril 2008; 91:2650-63. [PMID: 18672237 DOI: 10.1016/j.fertnstert.2008.03.071] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2008] [Revised: 03/24/2008] [Accepted: 03/24/2008] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To identify differentially expressed genes between fibroid and adjacent normal myometrium in an identical hormonal and genetic background. DESIGN Array analysis of three leiomyomata and matched adjacent normal myometrium in a single patient. SETTING University of Colorado Hospital. PATIENT(S) A single female undergoing medically indicated hysterectomy for symptomatic fibroids. INTERVENTIONS(S) mRNA isolation and microarray analysis, reverse-transcriptase polymerase chain reaction, Western blotting, and immunohistochemistry. MAIN OUTCOME MEASURE(S) Changes in mRNA and protein levels in leiomyomata and matched normal myometrium. RESULT(S) Expression of 197 genes was increased and 619 decreased significantly by at least twofold, in leiomyomata relative to normal myometrium. Expression profiles between tumors were similar and normal myometrial samples showed minimal variation. Changes in, and variation of, expression of selected genes were confirmed in additional normal and leiomyoma samples from multiple patients. CONCLUSION(S) Analysis of multiple tumors from a single patient confirmed changes in expression of genes described in previous, apparently disparate, studies, and identified novel targets. Gene expression profiles in leiomyomata are consistent with increased activation of mitogenic pathways and inhibition of apoptosis. Down-regulation of genes implicated in invasion and metastasis, of cancers, was observed in fibroids. This expression pattern may underlie the benign nature of uterine leiomyomata and may aid in the differential diagnosis of leiomyosarcoma.
Collapse
Affiliation(s)
- Irina K Dimitrova
- Department of Obstetrics and Gynecology, University of Colorado Health Sciences Center, Aurora, Colorado 80045, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Huyck KL, Panhuysen CIM, Cuenco KT, Zhang J, Goldhammer H, Jones ES, Somasundaram P, Lynch AM, Harlow BL, Lee H, Stewart EA, Morton CC. The impact of race as a risk factor for symptom severity and age at diagnosis of uterine leiomyomata among affected sisters. Am J Obstet Gynecol 2008; 198:168.e1-9. [PMID: 18226615 PMCID: PMC2265083 DOI: 10.1016/j.ajog.2007.05.038] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Revised: 05/23/2007] [Accepted: 05/23/2007] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The objective of the study was to identify risk factors for uterine leiomyomata (UL) in a racially diverse population of women with a family history of UL, and to evaluate their contribution to disease severity and age at diagnosis. STUDY DESIGN We collected and analyzed epidemiologic data from 285 sister pairs diagnosed with UL. Risk factors for UL-related outcomes were compared among black (n = 73) and white (n = 212) sister pairs using univariate and multivariate regression models. RESULTS Black women reported an average age at diagnosis of 5.3 years younger (SE, 1.1; P < .001) and were more likely to report severe disease (odds ratio, 5.22; 95% confidence interval, 1.99-13.7, P < .001) than white women of similar socioeconomic status. CONCLUSION Self-reported race is a significant factor in the severity of UL among women with a family history of UL. Differences in disease presentation between races likely reflect underlying genetic heterogeneity. The affected sister-pair study design can address both epidemiological and genetic hypotheses about UL.
Collapse
Affiliation(s)
- Karen L Huyck
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Hodge JC, Morton CC. Genetic heterogeneity among uterine leiomyomata: insights into malignant progression. Hum Mol Genet 2007; 16 Spec No 1:R7-13. [PMID: 17613550 DOI: 10.1093/hmg/ddm043] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Uterine leiomyomata (UL), also known as fibroids, are the most common pelvic tumors in women of reproductive age and are the primary indication for hysterectomy in the USA. Many lines of evidence indicate a strong genetic component to the development of these tumors. In fact, approximately 40% of UL have non-random, tumor-specific chromosome abnormalities which have allowed classification into well-defined subgroups (deletion of portions of 7q, trisomy 12 or rearrangements of 12q15, 6p21 or 10q22) as well as identification of candidate genes for UL predisposition. Although benign, UL have been linked to malignancy through two genomic regions on chromosome 1. Mutation of fumarate hydratase (FH) at 1q43 is known to cause the Mendelian syndromes of multiple cutaneous and uterine leiomyomata (MCL) and hereditary leiomyomatosis and renal cell cancer (HLRCC), and recently, FH mutations have been detected in some non-syndromic UL. In addition, transcriptional profiling suggests that loss of the short arm of chromosome 1 in cellular leiomyomata, an uncommon histological variant of UL, may account in part for the presumed yet rare malignant transformation of UL to uterine leiomyosarcoma.
Collapse
Affiliation(s)
- Jennelle C Hodge
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | |
Collapse
|