1
|
Yaoxing X, Fangfang Z, Wenzhi L, Xianrong Z, Xin L, Xiang T. Morphological Diversity of the Endometrium in Choriocarcinoma Specimens and its Role in Differential Diagnosis. Int J Surg Pathol 2024; 32:935-942. [PMID: 37993989 DOI: 10.1177/10668969231211336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Introduction: The morphological characteristics of the endometrium in patients with choriocarcinoma have not been well described. We described the endometrial morphology patterns in 46 choriocarcinomas and analyzed their relationship with the clinicopathological characteristics of these patients. Methods: Forty-six patients diagnosed with choriocarcinoma that had sufficient endometrial tissues for histopathological diagnosis were selected. Diagnoses of choriocarcinoma and secretory status of endometrium were reviewed. LHCGR expression of endometrium was evaluated by immunostaining. Results: Endometrial morphology was classified as secretory or nonsecretory. The 15 secretory specimens included 2 highly secretory and 13 common secretory specimens. The 31 nonsecretory patterns included 1 hyperplasia without atypia, 7 disordered proliferations, 13 typical proliferations, and 10 resting endometria. Among these, 11 specimens with overall nonsecretory patterns showed focally weak secretory changes surrounding the choriocarcinoma lesion. Secretory patterns were observed in classic choriocarcinomas (8/17) and monomorphic choriocarcinomas (7/21) but not in scanty-trophoblast choriocarcinomas (0/8). Secretory changes appeared significantly less frequently in patients who received multi-agent chemotherapy (4/25) than in those who did not (7/14) or received single-agent chemotherapy (4/7) (P = 0.030). The differences in age, months since the last pregnancy, pregnancy type, recurrence, specimen type, gross diameter, human chorionic gonadotropin (hCG) levels, and expression of hCG receptors were not statistically significant. Conclusions: The endometrial morphologies in choriocarcinoma were diverse, including various proliferative and secretory changes, but rarely hypersecretory changes, compared to the prevailing hypersecretory endometrium in hydatidiform moles. The variety in endometrial morphology was the consequence of ovarian hormonal disturbances of the hypothalamic-pituitary-gonadal axis by hCG from choriocarcinoma. Therefore, the endometrium may serve as a clue for histopathological diagnosis of choriocarcinoma. Our study presents the largest cohort reported to date to describe the diverse spectrum of endometrial changes in choriocarcinoma patients.
Collapse
Affiliation(s)
- Xiao Yaoxing
- Department of Pathology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Zhong Fangfang
- Department of Pathology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Li Wenzhi
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Zhou Xianrong
- Department of Pathology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Lu Xin
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Tao Xiang
- Department of Pathology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
2
|
Wadhwa L, Rani A. Impact of Intrauterine Administration of Human Chorionic Gonadotropin before Intrauterine Insemination in Infertile Women: A Randomized Controlled Trial. J Hum Reprod Sci 2021; 14:156-161. [PMID: 34316231 PMCID: PMC8279052 DOI: 10.4103/jhrs.jhrs_196_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 05/08/2021] [Accepted: 05/09/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Implantation is the rate-limiting step in the success of both intrauterine Insemination (IUI) and in vitro fertilization cycles. Numerous interventions that target various local signals have been tried to improve the implantation and clinical pregnancy rate (CPR). The most significant of these signals is human chorionic gonadotropin (hCG) which acts as immunomodulator and improves implantation by decidualization of the endometrial stromal cells, trophoblast invasion, proliferation of uterine natural killer cells, stimulation of endometrial angiogenesis, and maintenance of progesterone secretion by the corpus luteum. Aim: The aim of the study is to evaluate the effect of intrauterine hCG administration before IUI on CPR. Settings and Design: A prospective parallel randomized control study was done from September 2017 to February 2019. Materials and Methods: A total of 200 eligible women planned for IUI were randomly divided just before IUI into 2 groups. A computer-generated randomization list with block size of 10 with 1:1 allocation was used to randomize the patients. Experimental group received 0.5 ml containing 500 IU hCG, on the other hand control group received 0.5 ml of normal saline 2–3 min before IUI in single sitting. The main outcomes were CPR, miscarriage rate, and ongoing pregnancy rate. Statistical Analysis: It was performed using statistical software version SPSS 17.0. Results: Patient's demographic and baseline characteristics were comparable in both the groups. CPR in experimental group was significantly high compared to control group (26% vs. 9%, P = 0.002). Ongoing pregnancy rate was also significantly higher in experimental group (23%) compared to control group (7%) (P = 0.003). No significant difference in miscarriage rate was seen between the two groups. No cases of ectopic pregnancy, ovarian hyperstimulation syndrome, or multiple pregnancy were reported. Conclusion: Intrauterine hCG administration is a simple procedure that can be used to improve pregnancy outcome in IUI cycles.
Collapse
Affiliation(s)
- Leena Wadhwa
- Department of Obstetrics and Gynaecology, IVF and Fertility Research Centre, ESI-PGIMSR, New Delhi, India
| | - Anupama Rani
- Department of Obstetrics and Gynaecology, IVF and Fertility Research Centre, ESI-PGIMSR, New Delhi, India
| |
Collapse
|
3
|
Jonas KC, Rivero Müller A, Oduwole O, Peltoketo H, Huhtaniemi I. The Luteinizing Hormone Receptor Knockout Mouse as a Tool to Probe the In Vivo Actions of Gonadotropic Hormones/Receptors in Females. Endocrinology 2021; 162:6144965. [PMID: 33605422 PMCID: PMC8171189 DOI: 10.1210/endocr/bqab035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Indexed: 11/25/2022]
Abstract
Mouse models with altered gonadotropin functions have provided invaluable insight into the functions of these hormones/receptors. Here we describe the repurposing of the infertile and hypogonadal luteinizing hormone receptor (LHR) knockout mouse model (LuRKO), to address outstanding questions in reproductive physiology. Using crossbreeding strategies and physiological and histological analyses, we first addressed the physiological relevance of forced LHR homomerization in female mice using BAC expression of 2 ligand-binding and signaling deficient mutant LHR, respectively, that have previously shown to undergo functional complementation and rescue the hypogonadal phenotype of male LuRKO mice. In female LuRKO mice, coexpression of signaling and binding deficient LHR mutants failed to rescue the hypogonadal and anovulatory phenotype. This was apparently due to the low-level expression of the 2 mutant LHR and potential lack of luteinizing hormone (LH)/LHR-dependent pleiotropic signaling that has previously been shown at high receptor densities to be essential for ovulation. Next, we utilized a mouse model overexpressing human chorionic gonadotropin (hCG) with increased circulating "LH/hCG"-like bioactivity to ~40 fold higher than WT females, to determine if high circulating hCG in the LuRKO background could reveal putative LHR-independent actions. No effects were found, thus, suggesting that LH/hCG mediate their gonadal and non-gonadal effects solely via LHR. Finally, targeted expression of a constitutively active follicle stimulating hormone receptor (FSHR) progressed antral follicles to preovulatory follicles and displayed phenotypic markers of enhanced estrogenic activity but failed to induce ovulation in LuRKO mice. This study highlights the critical importance and precise control of functional LHR and FSHR for mediating ovarian functions and of the potential repurposing of existing genetically modified mouse models in answering outstanding questions in reproductive physiology.
Collapse
Affiliation(s)
- Kim Carol Jonas
- Department of Women and Children’s Health, King’s College London,
London SE1 1UL, UK
- Institute of Reproductive and Developmental Biology, Department of Metabolism,
Digestion and Reproduction, Imperial College London, London W12
0NN, UK
- Correspondence: Dr Kim Jonas, Department of Women and Children’s Health, King’s College London,
London SE1 1UL, UK; Institute of Reproductive and Developmental Biology, Department of
Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK.
; or Prof. Ilpo Huhtaniemi, Institute of Reproductive and
Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial
College London, London, W12 0NN, UK; Institute for Biomedicine, Department of Physiology,
University of Turku, 20520 Turku, Finland.
| | - Adolfo Rivero Müller
- Institute for Biomedicine, Department of Physiology, University of
Turku, 20520 Turku, Finland
- Department of Biochemistry and Molecular Biology, Medical University of
Lublin, 20-093 Lublin, Poland
| | - Olayiwola Oduwole
- Institute of Reproductive and Developmental Biology, Department of Metabolism,
Digestion and Reproduction, Imperial College London, London W12
0NN, UK
| | - Hellevi Peltoketo
- Institute of Reproductive and Developmental Biology, Department of Metabolism,
Digestion and Reproduction, Imperial College London, London W12
0NN, UK
- Laboratory of Cancer Genetics and Tumour Biology, Cancer and Translational
Medicine Research Unit, Biocenter Oulu and University of Oulu,
90220 Oulu, Finland
| | - Ilpo Huhtaniemi
- Institute of Reproductive and Developmental Biology, Department of Metabolism,
Digestion and Reproduction, Imperial College London, London W12
0NN, UK
- Institute for Biomedicine, Department of Physiology, University of
Turku, 20520 Turku, Finland
- Correspondence: Dr Kim Jonas, Department of Women and Children’s Health, King’s College London,
London SE1 1UL, UK; Institute of Reproductive and Developmental Biology, Department of
Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK.
; or Prof. Ilpo Huhtaniemi, Institute of Reproductive and
Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial
College London, London, W12 0NN, UK; Institute for Biomedicine, Department of Physiology,
University of Turku, 20520 Turku, Finland.
| |
Collapse
|
4
|
Jonusiene V, Sasnauskiene A. Notch and Endometrial Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1287:47-57. [PMID: 33034025 DOI: 10.1007/978-3-030-55031-8_4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The human endometrium is a unique, highly dynamic tissue that undergoes cyclic changes of cell proliferation, differentiation, and death. Endometrial cancer is the most common malignancy among women in developed countries. Importantly, the incidence of endometrial cancer is rising in high-income countries. Currently histological classification is used for subtyping of endometrial cancer, while ongoing research is evaluating markers for more accurate molecular classification. Evolutionary conserved Notch signaling pathway regulates diverse cellular processes such as proliferation, differentiation, and cell invasion. Accumulating evidence links aberrant Notch signaling with diseases such as hyperplasia and endometrial cancer. This chapter summarizes the current state of Notch signaling investigations in the endometrium, endometriosis, and endometrial cancer.
Collapse
Affiliation(s)
- Violeta Jonusiene
- Vilnius University, Life Sciences Center, Institute of Biosciences, Vilnius, Lithuania.
| | - Ausra Sasnauskiene
- Vilnius University, Life Sciences Center, Institute of Biosciences, Vilnius, Lithuania
| |
Collapse
|
5
|
Ott TL. Immunological detection of pregnancy: Evidence for systemic immune modulation during early pregnancy in ruminants. Theriogenology 2020; 150:498-503. [PMID: 32331860 DOI: 10.1016/j.theriogenology.2020.04.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 04/08/2020] [Indexed: 12/28/2022]
Abstract
Mammalian pregnancy creates unique challenges for immune systems highly evolved to detect and eliminate invading pathogens. Recognition of the challenges created by gestating a semi-allogeneic fetus evolved from the discipline of transplantation biology and were informed by studies on the unique natural parabiosis that occurs when female calves are gestated with twin male fetuses. These pregnancies typically result in an intersex female termed a freemartin, which revealed insights into development of the male and female reproductive tracts. However, they also uncovered important clues on immune tolerance with wide-ranging implications to reproductive biology, transplantation biology and autoimmune disease. Many studies focused on identifying mechanisms through which the fetus evades maternal immune detection and elimination. These included studies characterizing immune interactions between the fetus and mother at the nourishing interface of the placenta and uterine endometrium. This immunological forbearance only occurs under high concentrations of circulating progesterone. Beyond the requirement for progesterone, there has been considerable progress towards understanding the effects of conceptus signals on maternal immune function. One common theme is that pregnancy induces a T helper 2 immune bias as shown in several mammalian species, including domestic ruminants. However, a growing body of evidence shows that the fetus not only evades, but also provokes immune responses locally in the uterus and in peripheral tissues. This is perhaps most dramatically illustrated by domestic ruminants where the conceptus secretes a unique interferon in the opening salvo of hormonal communication with the maternal immune system. The role of interferon tau in regulating expression of genes of the innate immune system in the uterus has been extensively studied. More recently, it was determined that these same genes are also induced in peripheral immune cells and other tissues throughout the body. In addition to interferon tau and progesterone, pregnancy associate glycoproteins and chaperonin 10 (aka Early Pregnancy Factor) are implicated in altering immune function both locally and systemically during pregnancy. While it is tempting to speculate that this activation of innate immunity is designed to counteract selective immunosuppression, knowledge of the importance of local and systemic immune activation to the success of pregnancy remains incomplete. This area remains fertile ground for developing better approaches to diagnose and treat infertility in domestic farm species and humans alike.
Collapse
Affiliation(s)
- Troy L Ott
- Department of Animal Science, Center for Reproductive Biology and Health, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
6
|
Hsueh AJ, He J. Gonadotropins and their receptors: coevolution, genetic variants, receptor imaging, and functional antagonists. Biol Reprod 2019; 99:3-12. [PMID: 29462242 DOI: 10.1093/biolre/ioy012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/06/2018] [Indexed: 12/29/2022] Open
Abstract
Gonadotropins belong to the family of dimeric glycoprotein hormones and regulate gonadal physiology mediated by G protein-coupled, seven-transmembrane receptors. These glycoprotein hormones are widely used in the clinic to promote ovarian follicle development and for treating some cases of male infertility. We traced the coevolution of dimeric gonadotropin hormones and their receptors, together with thyrotropin and its receptor. We updated recent findings on human genetic variants of these genes and their association with dizygotic twining, polycystic ovarian syndrome, primary ovarian insufficiency, male-limited precocious puberty, and infertility. In addition to the known physiological roles of gonadotropin-receptor signaling in gonadal tissues, we also discussed emerging understanding of extragonadal functions of gonadotropins in bones and adipose tissues, together with recent advances in in vivo imaging of gonadotropin receptors in live animals. Recent development of gonadotropin receptor agonists and antagonists were summarized with an emphasis on the development of functional antagonists for FSH receptors to alleviate osteoporosis and obesity associated with menopause.
Collapse
Affiliation(s)
- Aaron J Hsueh
- Program of Reproductive and Stem Cell Biology, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California, USA
| | - Jiahuan He
- Program of Reproductive and Stem Cell Biology, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
7
|
Doroszko M, Chrusciel M, Stelmaszewska J, Slezak T, Anisimowicz S, Plöckinger U, Quinkler M, Bonomi M, Wolczynski S, Huhtaniemi I, Toppari J, Rahman NA. GnRH antagonist treatment of malignant adrenocortical tumors. Endocr Relat Cancer 2019; 26:103-117. [PMID: 30400009 PMCID: PMC6215908 DOI: 10.1530/erc-17-0399] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 08/21/2018] [Indexed: 12/11/2022]
Abstract
Aberrantly expressed G protein-coupled receptors in tumors are considered as potential therapeutic targets. We analyzed the expressions of receptors of gonadotropin-releasing hormone (GNRHR), luteinizing hormone/chorionic gonadotropin (LHCGR) and follicle-stimulating hormone (FSHR) in human adrenocortical carcinomas and assessed their response to GnRH antagonist therapy. We further studied the effects of the GnRH antagonist cetrorelix acetate (CTX) on cultured adrenocortical tumor (ACT) cells (mouse Cα1 and Y-1, and human H295R), and in vivo in transgenic mice (SV40 T-antigen expression under inhibin α promoter) bearing Lhcgr and Gnrhr in ACT. Both models were treated with control (CT), CTX, human chorionic gonadotropin (hCG) or CTX+hCG, and their growth and transcriptional changes were analyzed. In situ hybridization and qPCR analysis of human adrenocortical carcinomas (n = 11-13) showed expression of GNRHR in 54/73%, LHCGR in 77/100% and FSHR in 0%, respectively. CTX treatment in vitro decreased cell viability and proliferation, and increased caspase 3/7 activity in all treated cells. In vivo, CTX and CTX+hCG (but not hCG alone) decreased ACT weights and serum LH and progesterone concentrations. CTX treatment downregulated the tumor markers Lhcgr and Gata4. Upregulated genes included Grb10, Rerg, Nfatc and Gnas, all recently found to be abundantly expressed in healthy adrenal vs ACT. Our data suggest that CTX treatment may improve the therapy of human adrenocortical carcinomas by direct action on GNRHR-positive cancer cells inducing apoptosis and/or reducing gonadotropin release, directing tumor cells towards a healthy adrenal gene expression profile.
Collapse
Affiliation(s)
| | | | - Joanna Stelmaszewska
- Department of Reproduction and Gynecological EndocrinologyMedical University of Bialystok, Bialystok, Poland
| | - Tomasz Slezak
- Department of Biochemistry and Molecular BiologyUniversity of Chicago, Chicago, Illinois, USA
| | | | - Ursula Plöckinger
- Interdisciplinary Center of Metabolism: EndocrinologyDiabetes and Metabolism, Charité University Medicine Berlin, Berlin, Germany
| | - Marcus Quinkler
- Endocrinology in CharlottenburgBerlin, Germany
- Department of Clinical EndocrinologyCharité Campus Mitte, Charité University Medicine Berlin, Berlin, Germany
| | - Marco Bonomi
- Department of Clinical Sciences & Community HealthUniversity of Milan, Milan, Italy
| | - Slawomir Wolczynski
- Department of Reproduction and Gynecological EndocrinologyMedical University of Bialystok, Bialystok, Poland
| | - Ilpo Huhtaniemi
- Institute of BiomedicineUniversity of Turku, Turku, Finland
- Department of Surgery and CancerFaculty of Medicine, Imperial College London, London, U.K.
| | - Jorma Toppari
- Institute of BiomedicineUniversity of Turku, Turku, Finland
- Department of PediatricsTurku University Hospital, Turku, Finland
| | - Nafis A Rahman
- Institute of BiomedicineUniversity of Turku, Turku, Finland
- Department of Reproduction and Gynecological EndocrinologyMedical University of Bialystok, Bialystok, Poland
- Correspondence should be addressed to N Rahman:
| |
Collapse
|
8
|
Bazer FW, Burghardt RC, Johnson GA, Spencer TE, Wu G. Mechanisms for the establishment and maintenance of pregnancy: synergies from scientific collaborations. Biol Reprod 2018; 99:225-241. [PMID: 29462279 PMCID: PMC6044348 DOI: 10.1093/biolre/ioy047] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 02/13/2018] [Accepted: 02/15/2018] [Indexed: 11/14/2022] Open
Abstract
Research on the functions of interferon tau (IFNT) led to the theory of pregnancy recognition signaling in ruminant species. But IFNT does much more as it induces expression of interferon regulatory factor 2 (IRF2) in uterine luminal (LE), superficial glandular (sGE), but not glandular (GE) epithelia. First, IRF2 silences transcription of the estrogen receptor alpha gene and, indirectly, transcription of the oxytocin receptor gene to abrogate development of the luteolytic mechanism to prevent regression of the corpus luteum and its production of progesterone for establishing and maintaining pregnancy. Second, IRF2 silences expression of classical interferon-stimulated genes in uterine LE and sGE; however, uterine LE and sGE respond to progesterone (P4) and IFNT to increase expression of genes for transport of nutrients into the uterine lumen such as amino acids and glucose. Other genes expressed by uterine LE and sGE encode for adhesion molecules such as galectin 15, cathepsins, and cystatins for tissue remodeling, and hypoxia-inducible factor relevant to angiogenesis and survival of blastocysts in a hypoxic environment. IFNT is also key to a servomechanism that allows uterine epithelia, particularly GE, to proliferate and to express genes in response to placental lactogen and placental growth hormone in sheep. The roles of secreted phosphoprotein 1 are also discussed regarding its role in implantation in sheep and pigs, as well as its stimulation of expression of mechanistic target of rapamycin mRNA and protein which is central to proliferation, migration, and gene expression in the trophectoderm cells.
Collapse
Affiliation(s)
- Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| | - Robert C Burghardt
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Gregory A Johnson
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Thomas E Spencer
- Division of Animal Science, University of Missouri, Columbia, Missouri, USA
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
9
|
Screening chromosomal anomalies in early pregnancy: When and why. INDIAN JOURNAL OF MEDICAL SPECIALITIES 2018. [DOI: 10.1016/j.injms.2018.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
10
|
Humaidan P, Chin W, Rogoff D, D'Hooghe T, Longobardi S, Hubbard J, Schertz J. Efficacy and safety of follitropin alfa/lutropin alfa in ART: a randomized controlled trial in poor ovarian responders. Hum Reprod 2018; 32:544-555. [PMID: 28137754 PMCID: PMC5850777 DOI: 10.1093/humrep/dew360] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 01/10/2017] [Indexed: 01/15/2023] Open
Abstract
STUDY QUESTION How does the efficacy and safety of a fixed-ratio combination of recombinant human FSH plus recombinant human LH (follitropin alfa plus lutropin alfa; r-hFSH/r-hLH) compare with that of r-hFSH monotherapy for controlled ovarian stimulation (COS) in patients with poor ovarian response (POR)? SUMMARY ANSWER The primary and secondary efficacy endpoints were comparable between treatment groups and the safety profile of both treatment regimens was favourable. WHAT IS KNOWN ALREADY Although meta-analyses of clinical trials have suggested some beneficial effect on reproductive outcomes with r-hLH supplementation in patients with POR, the definitions of POR were heterogeneous and limit the comparability across studies. STUDY DESIGN, SIZE, DURATION Phase III, single-blind, active-comparator, randomized, parallel-group clinical trial. Patients were followed for a single ART cycle. A total of 939 women were randomized (1:1) to receive either r-hFSH/r-hLH or r-hFSH. Randomization, stratified by study site and participant age, was conducted via an interactive voice response system. PARTICIPANTS/MATERIALS, SETTING, METHODS Women classified as having POR, based on criteria incorporating the ESHRE Bologna criteria, were down-regulated with a long GnRH agonist protocol and following successful down-regulation were randomized (1:1) to COS with r-hFSH/r-hLH or r-hFSH alone. The primary efficacy endpoint was the number of oocytes retrieved following COS. Safety endpoints included the incidence of adverse events, including ovarian hyperstimulation syndrome (OHSS). Post hoc analyses investigated safety outcomes and correlations between live birth and baseline characteristics (age and number of oocytes retrieved in previous ART treatment cycles or serum anti-Müllerian hormone (AMH)). The significance of the treatment effect was tested by generalized linear models (Poisson regression for counts and logistic regression for binary endpoints) adjusting for age and country. MAIN RESULTS AND THE ROLE OF CHANCE Of 949 subjects achieving down-regulation, 939 were randomized to r-hFSH/r-hLH (n = 477) or r-hFSH (n = 462) and received treatment. Efficacy assessment: In the intention-to-treat (ITT) population, the mean (SD) number of oocytes retrieved (primary endpoint) was 3.3 (2.71) in the r-hFSH/r-hLH group compared with 3.6 (2.82) in the r-hFSH group (between-group difference not statistically significant). The observed difference between treatment groups (r-hFSH/r-hLH and r-hFSH, respectively) for efficacy outcomes decreased over the course of pregnancy (biochemical pregnancy rate: 17.3% versus 23.9%; clinical pregnancy rate: 14.1% versus 16.8%; ongoing pregnancy rate: 11.0% versus 12.4%; and live birth rate: 10.6% versus 11.7%). An interaction (identified post hoc) between baseline characteristics related to POR and treatment effect was noted for live birth, with r-hFSH/r-hLH associated with a higher live birth rate for patients with moderate or severe POR, whereas r-hFSH was associated with a higher live birth rate for those with mild POR. A post hoc logistic regression analysis indicated that the incidence of total pregnancy outcome failure was lower in the r-hFSH/r-hLH group (6.7%) compared with the r-hFSH group (12.4%) with an odds ratio of 0.52 (95% CI 0.33, 0.82; P = 0.005). Safety assessment: The overall proportion of patients with treatment-emergent adverse events (TEAEs) occurring during or after r-hFSH/r-hLH or r-hFSH use (stimulation or post-stimulation phase) was 19.9% and 26.8%, respectively. There was no consistent pattern of TEAEs associated with either treatment. LIMITATIONS, REASONS FOR CAUTION Despite using inclusion criteria for POR incorporating the ESHRE Bologna criteria, further investigation is needed to determine the impact of the heterogeneity of POR in the Bologna patient population. The observed correlation between baseline clinical characteristics related to POR and live birth rate, as well as the observed differences between groups regarding total pregnancy outcome failure were from post hoc analyses, and the study was not powered for these endpoints. In addition, the attrition rate for pregnancy outcomes in this trial may not reflect general medical practice. Furthermore, as the patient population was predominantly White these results might not be applicable to other ethnicities. WIDER IMPLICATIONS OF THE FINDINGS In the population of women with POR investigated in this study, although the number of oocytes retrieved was similar following stimulation with either a fixed-ratio combination of r-hFSH/r-hLH or r-hFSH monotherapy, post hoc analyses showed that there was a lower rate of total pregnancy outcome failure in patients receiving r-hFSH/r-hLH, in addition to a higher live birth rate in patients with moderate and severe POR. These findings are clinically relevant and require additional investigation. The benefit:risk balance of treatment with either r-hFSH/r-hLH or r-hFSH remains positive. STUDY FUNDING/COMPETING INTEREST(S) This study was funded by Merck KGaA, Darmstadt, Germany. P.H. has received honoraria for lectures and unrestricted research grants from Ferring, Merck KGaA and MSD. D.R. is a former employee of EMD Serono, a business of Merck KGaA, Darmstadt, Germany. J.S., J.H. and W.C. are employees of EMD Serono Research and Development Institute, a business of Merck KGaA, Darmstadt, Germany. T.D.’H. and S.L. are employees of Merck KGaA, Darmstadt, Germany. TRIAL REGISTRATION NUMBER ClinicalTrials.gov identifier: NCT02047227; EudraCT Number: 2013-003817-16. TRIAL REGISTRATION DATE ClinicalTrials.gov: 24 January 2014; EudraCT: 19 December 2013. DATE OF FIRST PATIENT'S ENROLMENT 30 January 2014.
Collapse
Affiliation(s)
- P Humaidan
- The Fertility Clinic, Skive Regional Hospital, Skive, Denmark.,Faculty of Health, Aarhus University, Aarhus, Denmark
| | - W Chin
- Global Biostatistics and Epidemiology, EMD Serono, Billerica, MA, USA, a business of Merck KGaA, Darmstadt, Germany
| | - D Rogoff
- Global Clinical Development, EMD Serono Research and Development Institute, Billerica, MA, USA, a business of Merck KGaA, Darmstadt, Germany
| | - T D'Hooghe
- Global Medical Affairs Fertility, Merck KGaA, Darmstadt, Germany
| | - S Longobardi
- Global Medical Affairs Fertility, Merck KGaA, Darmstadt, Germany
| | - J Hubbard
- Global Clinical Development, EMD Serono Research and Development Institute, Billerica, MA, USA, a business of Merck KGaA, Darmstadt, Germany
| | - J Schertz
- Global Clinical Development, EMD Serono Research and Development Institute, Billerica, MA, USA, a business of Merck KGaA, Darmstadt, Germany
| | | |
Collapse
|
11
|
Stouffer RL, Woodruff TK. Nonhuman Primates: A Vital Model for Basic and Applied Research on Female Reproduction, Prenatal Development, and Women's Health. ILAR J 2017; 58:281-294. [PMID: 28985318 PMCID: PMC5886348 DOI: 10.1093/ilar/ilx027] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 06/30/2017] [Indexed: 12/20/2022] Open
Abstract
The comparative biology of reproduction and development in mammalian species is remarkable. Hence, because of similarities in environmental and neuroendocrine control of the reproductive axis, the cyclic function of the ovary and reproductive tract, establishment and control of the maternal-fetal-placental unit during pregnancy, and reproductive aging from puberty through menopause, nonhuman primates (NHPs) are valuable models for research related to women's reproductive health and its disorders. This chapter provides examples of research over the past 10+ years using Old World monkeys (notably macaque species), baboons, and to a lesser extent New World monkeys (especially marmosets) that contributed to our understanding of the etiology and therapies or prevention of: (1) ovarian disorders, e.g., polycystic ovary syndrome, mitochondrial DNA-based diseases from the oocyte; (2) uterine disorders, for example, endometriosis and uterine transplantation; and (3) pregnancy disorders, for example, preterm labor and delivery, environmental factors. Also, emerging opportunities such as viral (e.g., Zika) induced fetal defects and germline genomic editing to generate valuable primate models of human diseases (e.g., Huntington and muscular dystrophy) are addressed. Although the high costs, specialized resources, and ethical debate challenge the use of primates in biomedical research, their inclusion in fertility and infertility research is vital for continued improvements in women's reproductive health.
Collapse
Affiliation(s)
- Richard L Stouffer
- Richard L. Stouffer, Ph.D., is Professor in the Division of Reproductive and Developmental Sciences at the Oregon National Primate Research Center in Beaverton, Oregon and Professor in the Department of Obstetrics and Gynecology at Oregon Health & Sciences University in Portland, Oregon. Teresa K. Woodruff, Ph.D., is Thomas J. Watkins Professor of Obstetrics and Gynecology, Vice Chair of Research (OB/GYN), and Chief of the Division of Reproductive Science in Medicine at the Feinberg School of Medicine, and Professor of Molecular Biosciences at Weinberg College of Arts and Sciences, Northwestern University in Chicago, Illinois.
| | - Teresa K Woodruff
- Richard L. Stouffer, Ph.D., is Professor in the Division of Reproductive and Developmental Sciences at the Oregon National Primate Research Center in Beaverton, Oregon and Professor in the Department of Obstetrics and Gynecology at Oregon Health & Sciences University in Portland, Oregon. Teresa K. Woodruff, Ph.D., is Thomas J. Watkins Professor of Obstetrics and Gynecology, Vice Chair of Research (OB/GYN), and Chief of the Division of Reproductive Science in Medicine at the Feinberg School of Medicine, and Professor of Molecular Biosciences at Weinberg College of Arts and Sciences, Northwestern University in Chicago, Illinois.
| |
Collapse
|
12
|
Hansen TR, Sinedino LDP, Spencer TE. Paracrine and endocrine actions of interferon tau (IFNT). Reproduction 2017; 154:F45-F59. [DOI: 10.1530/rep-17-0315] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/18/2017] [Accepted: 09/04/2017] [Indexed: 12/22/2022]
Abstract
This review focuses on the paracrine and endocrine actions of interferon tau (IFNT) during pregnancy recognition and establishment in ruminants. Pregnancy recognition involves the suppression of the endometrial luteolytic mechanism by the conceptus to maintain progesterone production by the corpus luteum (CL). The paracrine antiluteolytic effects of conceptus-derived IFNT inhibit upregulation of oxytocin receptors in the endometrial epithelia of the uterus, thereby preventing the production of luteolytic prostaglandin F2 alpha (PGF2α) pulses. In the endometrium, IFNT induces or upregulates a large number of classical IFN-stimulated genes (ISGs) and regulates expression of many other genes in a cell-specific manner that are likely important for conceptus elongation, implantation and establishment of pregnancy. Further, IFNT has endocrine effects on extrauterine cells and tissues. In sheep, IFNT induces luteal resistance to PGF2α, thereby ensuring survival of the CL for maintenance of pregnancy. The ISGs induced in circulating peripheral blood mononuclear cells by IFNT may also be useful as an indicator of pregnancy status in cattle. An increased knowledge of IFNT and ISGs is important to improve the reproductive efficiency in ruminants.
Collapse
|
13
|
Sun X, Liu X. Cancer metastasis: enactment of the script for human reproductive drama. Cancer Cell Int 2017; 17:51. [PMID: 28469531 PMCID: PMC5414196 DOI: 10.1186/s12935-017-0421-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 04/24/2017] [Indexed: 12/14/2022] Open
Abstract
Based on compelling evidence from many biological disciplines, we put forth a hypothesis for cancer metastasis. In the hypothesis, the metastatic cascade is depicted as human reproduction in miniature. Illustrated in a reproductive light, the staggering resemblance of cancer metastasis to human reproduction becomes evident despite some ostensible dis-similarities. In parallel to the appearance of primordial germ cells during early embryogenesis, the cancer reproductive saga starts with the separation of metastasis initiating cells (MICs) from cancer initiating cells when the primary cancer is still in its infancy. Prime MICs embark on a journey to the host bone marrow where they undergo further development and regulation. Migrating MICs are guided by the same CXCR4/CYCL12 axis as used in the migration of primordial germ cells to the genital ridge. Like the ovary, the host bone marrow features immune privileges, coolness, hypoxia and acidity which are essential for stemness maintenance and regulation. Opportune activation of the MICs via fusion with bone marrow stem cells triggers a frenzy of cellular proliferation and sets them on the move again. This scenario is akin to oocyte fertilization in the Fallopian tube and its subsequent journey towards the decidum. Just as the human reproductive process is plagued with undesirable outcomes so is the cancer metastasis highly inefficient. The climax of the cancer metastatic drama (colonization) is reached when proliferating MIC clusters attempt to settle down on decidum-like premetastatic sites. Successfully colonized clusters blossom into overt macrometastases only after the execution of sophisticated immunomodulation, angiogenesis and vascular remodeling. Similarly, the implanted blastomere needs to orchestrate these feats before flourishing into a new life. What is more, the cancer reproductive drama seems to be directed by a primordial hypothalamus–pituitary–gonad axis. Pursuing this reproductive trail could lead to new frontiers and breakthroughs in cancer research and therapeutics.
Collapse
Affiliation(s)
- Xichun Sun
- Department of Pathology and Laboratory Medicine, McGuire Holmes Veteran Affairs Medical Center, School of Medicine, Virginia Commonwealth University, 1201 Broad Rock Boulevard, Richmond, VA 23249 USA.,Department of Hepatobiliary Surgery, People's Hospital of Hunan Province, Hunan, China
| | - Xiwu Liu
- Department of Pathology and Laboratory Medicine, McGuire Holmes Veteran Affairs Medical Center, School of Medicine, Virginia Commonwealth University, 1201 Broad Rock Boulevard, Richmond, VA 23249 USA.,Department of Hepatobiliary Surgery, People's Hospital of Hunan Province, Hunan, China
| |
Collapse
|
14
|
Navali N, Gassemzadeh A, Farzadi L, Abdollahi S, Nouri M, Hamdi K, Mallah F, Jalilvand F. Intrauterine administration of hCG immediately after oocyte retrieval and the outcome of ICSI: a randomized controlled trial. Hum Reprod 2016; 31:2520-2526. [PMID: 27680029 DOI: 10.1093/humrep/dew236] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 08/16/2016] [Accepted: 08/25/2016] [Indexed: 12/28/2022] Open
Abstract
STUDY QUESTION Does the intrauterine administration of hCG immediately after oocyte retrieval in antagonist cycles with ICSI and fresh embryo transfer (ET) influence the implantation rate or chemical and clinical pregnancy rates? SUMMARY ANSWER The intrauterine administration of hCG after oocyte retrieval increases the implantation rate and chemical and clinical pregnancy rates. WHAT IS KNOWN ALREADY Over half of IVF/ICSI cycles fail due to implantation failure. Intrauterine administration of hCG, a few minutes before ET, increased the implantation and pregnancy rates in most but not in all studies. The effect of intrauterine administration of hCG, after oocyte retrieval, has not yet been studied. STUDY DESIGN, SIZE, DURATION The study was a parallel, triple-blind randomized clinical trial (RCT) performed from September 2015 to February 2016, in a university hospital. We recruited women undergoing antagonist ovarian stimulation, ICSI and ET. For an effect size of 0.2, power of 80% at a significance level of 0.05, we needed 150 participants. Accounting for a 7% dropout rate, a total of 160 women was considered appropriate. A computer-generated randomization list with a block size of 4, with 1:1 allocation was used. The treatment allocation was placed in a sealed, opaque, envelope and picked up consecutively. Immediately after oocyte retrieval, patients in the intervention and control groups were treated with intrauterine injection of hCG and saline, respectively. Participants underwent ET on Day 3. A beta-hCG test was done at 2 weeks. If positive, three transvaginal-ultrasonographies (TVSs) were done at 3, 4 and 10 weeks after ET. The participants were called up thereafter and questioned about the continuity of their pregnancy. PARTICIPANTS/MATERIALS, SETTING, METHOD Of 1990 women attending the infertility clinic of our university hospital, 508 were IVF/ICSI candidates during the study period, and 245 of the patients on an antagonist cycle met the criteria to be invited into our trial. Inclusion criteria were normal ovarian reserve, age ≤41, undergoing ICSI, and fresh ET and normal TSH and prolactin. Uncontrolled chronic disease, severe hydrosalpinx, severe endometriosis, morphologic embryo deficiencies, non-obstructive azospermia and high risk of severe ovarian hyperstimulation syndrome were criteria for exclusion. After taking an informed consent, a total of 158 participants were recruited, of which 80 were randomly allocated to receive intrauterine 500 IU hCG in up to 0.5 ml normal saline and 78 to receive intrauterine 0.5 ml normal saline immediately after oocyte retrieval, during general anaesthesia. ICSI was performed conventionally. The 4-8 cell embryos were transferred on the third day after oocyte retrieval. Implantation rate, chemical and clinical pregnancy rates were analysed and compared between the two groups. MAIN RESULTS AND THE ROLE OF CHANCE Patients' demographic and baseline characteristics were comparable. The clinical results showed statistically significant differences between the two groups regarding the biochemical pregnancy rate (59.2 versus 31.3%; P = 0.001; odds ratio (OR) = 1.88; 95% CI, 1.26-2.82; risk difference (RD) = 27.8; 95% CI, 11.2-42.3), implantation rate (37 versus 17%; P = 0.012; OR = 2.29; 95% CI, 1.02-5.14; RD = 20.2; 95% CI, 5.4-33.8), clinical pregnancy rate (50.7 versus 16.4%; P < 0.001; OR = 3.08; 95% CI, 1.71-5.55; RD = 34.3; 95% CI, 18.7-47.6) and ongoing pregnancy rate (40.1 versus 13.4%; P = 0.001; OR = 3.04; 95% CI, 1.55-5.93; RD = 27.4; 95% CI, 12.7-40.6). The abortion and ectopic pregnancy rates were not statistically different between the two groups. LIMITATIONS, REASONS FOR CAUTION The insertion of an intrauterine insemination catheter and the injection of a small amount of saline into the uterine cavity (without hCG) may also have some impact on implantation. This effect could be studied by comparing this intervention with another study group without any intrauterine injection.There are no specific side effects mentioned in the literature for the intrauterine administration of hCG, neither were any observed in our study, but it is best to be cautious about probable side effects, because this type of intervention is relatively new and experimental, and deserves more studies before being entered into routine clinical practice. WIDER IMPLICATIONS OF THE FINDINGS Intrauterine administration of hCG immediately after oocyte pick up increases its effectiveness; however, further investigations are required before this procedure can be recommended for clinical practice. STUDY FUNDING/COMPETING INTERESTS This study was supported by the Women's Health Research Center, Tabriz University of Medical Sciences, Iran. No external funds were used. The authors have no conflicts of interest to declare. TRIAL REGISTRATION NUMBER IRCT201206165485N4. TRIAL REGISTRATION DATE 2 September 2015. DATE OF FIRST PATIENT'S ENROLMENT 2 September 2015.
Collapse
Affiliation(s)
- N Navali
- Women's Reproductive Health Research Center, Alzahra University Hospital, Tabriz University of Medical Sciences, Artesh Road, Tabriz 5138665793, Iran .,Reproductive Medical Center, Alzahra University Hospital, Tabriz University of Medical Sciences, Artesh Road, Tabriz 5138665793, Iran
| | - A Gassemzadeh
- Women's Reproductive Health Research Center, Alzahra University Hospital, Tabriz University of Medical Sciences, Artesh Road, Tabriz 5138665793, Iran.,Reproductive Medical Center, Alzahra University Hospital, Tabriz University of Medical Sciences, Artesh Road, Tabriz 5138665793, Iran
| | - L Farzadi
- Women's Reproductive Health Research Center, Alzahra University Hospital, Tabriz University of Medical Sciences, Artesh Road, Tabriz 5138665793, Iran.,Reproductive Medical Center, Alzahra University Hospital, Tabriz University of Medical Sciences, Artesh Road, Tabriz 5138665793, Iran
| | - S Abdollahi
- Women's Reproductive Health Research Center, Alzahra University Hospital, Tabriz University of Medical Sciences, Artesh Road, Tabriz 5138665793, Iran.,Reproductive Medical Center, Alzahra University Hospital, Tabriz University of Medical Sciences, Artesh Road, Tabriz 5138665793, Iran
| | - M Nouri
- Women's Reproductive Health Research Center, Alzahra University Hospital, Tabriz University of Medical Sciences, Artesh Road, Tabriz 5138665793, Iran.,Reproductive Medical Center, Alzahra University Hospital, Tabriz University of Medical Sciences, Artesh Road, Tabriz 5138665793, Iran
| | - K Hamdi
- Women's Reproductive Health Research Center, Alzahra University Hospital, Tabriz University of Medical Sciences, Artesh Road, Tabriz 5138665793, Iran.,Reproductive Medical Center, Alzahra University Hospital, Tabriz University of Medical Sciences, Artesh Road, Tabriz 5138665793, Iran
| | - F Mallah
- Women's Reproductive Health Research Center, Alzahra University Hospital, Tabriz University of Medical Sciences, Artesh Road, Tabriz 5138665793, Iran.,Reproductive Medical Center, Alzahra University Hospital, Tabriz University of Medical Sciences, Artesh Road, Tabriz 5138665793, Iran
| | - F Jalilvand
- Women's Reproductive Health Research Center, Alzahra University Hospital, Tabriz University of Medical Sciences, Artesh Road, Tabriz 5138665793, Iran
| |
Collapse
|
15
|
Ulloa-Aguirre A, Lira-Albarrán S. Clinical Applications of Gonadotropins in the Male. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 143:121-174. [PMID: 27697201 DOI: 10.1016/bs.pmbts.2016.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The pituitary gonadotropins, luteinizing hormone (LH), and follicle-stimulating hormone (FSH) play a pivotal role in reproduction. The synthesis and secretion of gonadotropins are regulated by complex interactions among several endocrine, paracrine, and autocrine factors of diverse chemical structure. In men, LH regulates the synthesis of androgens by the Leydig cells, whereas FSH promotes Sertoli cell function and thereby influences spermatogenesis. Gonadotropins are complex molecules composed of two subunits, the α- and β-subunit, that are noncovalently associated. Gonadotropins are decorated with glycans that regulate several functions of the protein including folding, heterodimerization, stability, transport, conformational maturation, efficiency of heterodimer secretion, metabolic fate, interaction with their cognate receptor, and selective activation of signaling pathways. A number of congenital and acquired abnormalities lead to gonadotropin deficiency and hypogonadotropic hypogonadism, a condition amenable to treatment with exogenous gonadotropins. Several natural and recombinant preparations of gonadotropins are currently available for therapeutic purposes. The difference between natural and the currently available recombinant preparations (which are massively produced in Chinese hamster ovary cells for commercial purposes) mainly lies in the abundance of some of the carbohydrates that conform the complex glycans attached to the protein core. Whereas administration of exogenous gonadotropins in patients with isolated congenital hypogonadotropic hypogonadism is a well recognized therapeutic approach, their role in treating men with normogonadotropic idiopathic infertility is still controversial. This chapter concentrates on the main structural and functional features of the gonadotropin hormones and how basic concepts have been translated into the clinical arena to guide therapy for gonadotropin deficit in males.
Collapse
Affiliation(s)
- A Ulloa-Aguirre
- Research Support Network, Universidad Nacional Autónoma de México (UNAM)-National Institutes of Health, Mexico City, Mexico.
| | - S Lira-Albarrán
- Department of Reproductive Biology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
16
|
Su RW, Strug MR, Joshi NR, Jeong JW, Miele L, Lessey BA, Young SL, Fazleabas AT. Decreased Notch pathway signaling in the endometrium of women with endometriosis impairs decidualization. J Clin Endocrinol Metab 2015; 100:E433-42. [PMID: 25546156 PMCID: PMC4333047 DOI: 10.1210/jc.2014-3720] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
CONTEXT Endometriosis is a common gynecological disease affecting one in 10 women of reproductive age and is a major cause of pelvic pain and impaired fertility. Endometrial stromal cells of women with endometriosis exhibit a reduced response to in vitro decidualization. NOTCH1 is critical for decidualization of both mouse and human uterine stromal cells. OBJECTIVE This study aimed to determine whether decidualization failure in women with endometriosis is a consequence of impaired Notch signaling. SETTING AND DESIGN We investigated expression levels of Notch signaling components in the endometrium of women and baboons with or without endometriosis. We identified NOTCH1-regulated genes during decidualization of human uterine fibroblast (HuF) cells by microarray and quantified their expression levels in in vitro-decidualized endometrial stromal cells isolated from women with or without endometriosis. RESULTS Notch signaling receptors NOTCH1 and NOTCH4, ligands JAGGED2 and DLL4, as well as direct target genes HES5 and HEY1 were decreased in the eutopic endometrium of women and baboons with endometriosis. Notch signaling was decreased in stromal cells isolated from women with endometriosis, which was associated with impaired in vitro decidualization. Genes that were down-regulated by NOTCH1 silencing in decidualized HuF cells were also decreased in decidualized endometrial stromal cells of women with endometriosis. FOXO1 acts as a downstream target of Notch signaling and endometriosis is associated with decreased expression of NOTCH1-regulated, FOXO1-responsive genes during decidualization. CONCLUSIONS Decreased Notch signaling is associated with endometriosis and contributes to impaired decidualization through the down-regulation of FOXO1.
Collapse
Affiliation(s)
- Ren-Wei Su
- Department of Obstetrics, Gynecology and Reproductive Biology (R.-W.S., M.R.S., N.R.J., J.-W.J., A.T.F.), Michigan State University, Grand Rapids, Michigan 49503; Cancer Institute (L.M.), Louisiana State University Health Sciences Center and Stanley S. Scott Cancer Center, New Orleans, Louisiana 70112; Greenville Health Systems (B.A.L.), University of South Carolina, Greenville, South Carolina 29605; and Department of Obstetrics and Gynecology (S.L.Y.), University of North Carolina, Chapel Hill, North Carolina 27599
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Sisinni L, Landriscina M. The Role of Human Chorionic Gonadotropin as Tumor Marker: Biochemical and Clinical Aspects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 867:159-76. [PMID: 26530366 DOI: 10.1007/978-94-017-7215-0_11] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Tumor markers are biological substances that are produced/released mainly by malignant tumor cells, enter the circulation in detectable amounts and are potential indicators of the presence of a tumor. The most useful biochemical markers are the tumor-specific molecules, i.e., receptors, enzymes, hormones, growth factors or biological response modifiers that are specifically produced by tumor cells and not, or minimally, by the normal counterpart (Richard et al. Principles and practice of gynecologic oncology. Wolters Kluwer Health, Philadelphia, 2009). Based on their specificity and sensitivity in each malignancy, biomarkers are used for screening, diagnosis, disease monitoring and therapeutic response assessment in clinical management of cancer patients.This chapter is focused on human chorionic gonadotropin (hCG), a hormone with a variety of functions and widely used as a tumor biomarker in selected tumors. Indeed, hCG is expressed by both trophoblastic and non-trophoblastic human malignancies and plays a role in cell transformation, angiogenesis, metastatization, and immune escape, all process central to cancer progression. Of note, hCG testing is crucial for the clinical management of placental trophoblastic malignancies and germ cell tumors of the testis and the ovary. Furthermore, the production of hCG by tumor cells is accompanied by varying degrees of release of the free subunits into the circulation, and this is relevant for the management of cancer patients (Triozzi PL, Stevens VC, Oncol Rep 6(1):7-17, 1999).The name chorionic gonadotropin was conceived: chorion derives from the latin chordate meaning afterbirth, gonadotropin indicates that the hormone is a gonadotropic molecule, acting on the ovaries and promoting steroid production (Cole LA, Int J Endocrinol Metab 9(2):335-352, 2011). The function, the mechanism of action and the interaction between hCG and its receptor continue to be the subject of intensive investigation, even though many issues about hCG have been well documented (Tegoni M et al., J Mol Biol 289(5):1375-1385, 1999).
Collapse
Affiliation(s)
- Lorenza Sisinni
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Matteo Landriscina
- Clinical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto, 1, 71100, Foggia, Italy.
| |
Collapse
|
18
|
Su RW, Fazleabas AT. Implantation and Establishment of Pregnancy in Human and Nonhuman Primates. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2015; 216:189-213. [PMID: 26450500 PMCID: PMC5098399 DOI: 10.1007/978-3-319-15856-3_10] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Implantation and the establishment of pregnancy are critical for the propagation of the species, but yet remain the limiting steps in human and primate reproduction. Successful implantation requires a competent blastocyst and a receptive endometrium during a specific window of time during the menstrual cycle to initiate the bilateral communication required for the establishment of a successful pregnancy. This chapter provides an overview of these processes and discusses the molecular mechanisms associated with implantation of the blastocyst and decidualization of the uterus in primates.
Collapse
Affiliation(s)
- Ren-Wei Su
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, 49503, USA.
| | - Asgerally T Fazleabas
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, 49503, USA
| |
Collapse
|
19
|
Aaleyasin A, Aghahosseini M, Rashidi M, Safdarian L, Sarvi F, Najmi Z, Mobasseri A, Amoozgar B. In vitro fertilization outcome following embryo transfer with or without preinstillation of human chorionic gonadotropin into the uterine cavity: a randomized controlled trial. Gynecol Obstet Invest 2014; 79:201-5. [PMID: 25531413 DOI: 10.1159/000363235] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 04/24/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND Intrauterine injection of human chorionic gonadotropin (hCG) at embryo transfer (ET) has been shown to improve the outcome of assisted reproductive techniques. The aim of this study was to confirm previous findings. METHODS In this randomized controlled trial, 483 infertile women who were candidates for in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) for the first time were randomly assigned to receive an intrauterine injection of 500 IU hCG or placebo (tissue culture media) before ET. The main outcome measures were implantation and clinical pregnancy rates. RESULTS Both the hCG-treated group (n = 240) and control group (n = 243) were similar at baseline in terms of demographic and obstetrical characteristics. There were significant differences between the two groups regarding the implantation rate (23.6 vs. 12.2%, p < 0.001), pregnancy rate (54.6 vs. 35.8%, p < 0.001), clinical pregnancy rate (50 vs. 32.1%, p < 0.001), ongoing pregnancy rate (15.3 vs. 9.2%, p < 0.001) and live delivery rate (14.3 vs. 8.4%, p < 0.001). The rate of fertilization and abortion rates were not statistically different. CONCLUSION Intrauterine injection of hCG before ET improves implantation and pregnancy rates and may be considered an adjuvant in IVF/ICSI.
Collapse
Affiliation(s)
- Ashraf Aaleyasin
- Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Choi J, Smitz J. Luteinizing hormone and human chorionic gonadotropin: origins of difference. Mol Cell Endocrinol 2014; 383:203-13. [PMID: 24365330 DOI: 10.1016/j.mce.2013.12.009] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/06/2013] [Accepted: 12/12/2013] [Indexed: 01/24/2023]
Abstract
Luteinizing hormone (LH) and human chorionic gonadotropin (hCG) are widely recognized for their roles in ovulation and the support of early pregnancy. Aside from the timing of expression, however, the differences between LH and hCG have largely been overlooked in the clinical realm because of their similar molecular structures and shared receptor. With technologic advancements, including the development of highly purified and recombinant gonadotropins, researchers now appreciate that these hormones are not as interchangeable as once believed. Although they bind to a common receptor, emerging evidence suggests that LH and hCG have disparate effects on downstream signaling cascades. Increased understanding of the inherent differences between LH and hCG will foster more effective diagnostic and prognostic assays for use in a variety of clinical contexts and support the individualization of treatment strategies for conditions such as infertility.
Collapse
Affiliation(s)
- Janet Choi
- The Center for Women's Reproductive Care at Columbia University, New York, NY, United States.
| | - Johan Smitz
- UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
21
|
Feng X, Zhang M, Guan R, Segaloff DL. Heterodimerization between the lutropin and follitropin receptors is associated with an attenuation of hormone-dependent signaling. Endocrinology 2013; 154:3925-30. [PMID: 23825122 PMCID: PMC3776865 DOI: 10.1210/en.2013-1407] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The LH receptor (LHR) and FSH receptor (FSHR) are each G protein-coupled receptors that play critical roles in reproductive endocrinology. Each of these receptors has previously been shown to self-associate into homodimers and oligomers shortly after their biosynthesis. As shown herein using bioluminescence resonance energy transfer to detect protein-protein interactions, our data show that the LHR and FSHR, when coexpressed in the same cells, specifically heterodimerize with each other. Further experiments confirm that at least a portion of the cellular LHR/FSHR heterodimers are present on the cell surface and are functional. We then sought to ascertain what effects, if any, heterodimerization between the LHR and FSHR might have on signaling. It was observed that when the LHR was expressed under conditions promoting the heterodimerization with FSHR, LH or human chorionic gonadotropin (hCG) stimulation of Gs was attenuated. Conversely, when the FSHR was expressed under conditions promoting heterodimerization with the LHR, FSH-stimulated Gs activation was attenuated. These results demonstrate that the coexpression of the LHR and FSHR enables heterodimerizaton between the 2 gonadotropin receptors and results in an attenuation of signaling through each receptor.
Collapse
Affiliation(s)
- Xiuyan Feng
- PhD, Department of Molecular Physiology and Biophysics, 5-470 Bowen Science Building, The University of Iowa, Iowa City, Iowa 52242.
| | | | | | | |
Collapse
|
22
|
Troppmann B, Kleinau G, Krause G, Gromoll J. Structural and functional plasticity of the luteinizing hormone/choriogonadotrophin receptor. Hum Reprod Update 2013; 19:583-602. [DOI: 10.1093/humupd/dmt023] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
23
|
Seow KM, Lee JL, Doong ML, Huang SW, Hwang JL, Huang WJ, Chang FY, Ho LT, Juan CC. Human chorionic gonadotropin regulates gastric emptying in ovariectomized rats. J Endocrinol 2013. [PMID: 23197744 DOI: 10.1530/joe-12-0421] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Prolongation of gastrointestinal transit resulting in nausea and vomiting in pregnancy (NVP) is the most common phenomenon during the first trimester of pregnancy. Increased human chorionic gonadotropin (hCG) concentration during the first trimester is the most likely cause of NVP. The aim of this study was to investigate the effect of hCG on gastrointestinal transit and plasma concentrations of cholecystokinin (CCK) in ovariectomized (Ovx) rats. I.p. injection of hCG was used to evaluate the dose effect of hCG on gastrointestinal transit in Ovx rats. The CCK antagonist lorglumide was used to clarify the role of CCK in regulating gastrointestinal transit. Gastrointestinal transit was assessed 15 min after intragastric gavage of a mixture of 10% charcoal and Na(2)(51)CrO(4) (0.5 μCi/ml). After i.p. administration of hCG, gastric emptying was inhibited in Ovx rats, but intestinal transit was not affected. Plasma CCK concentrations were increased in a dose-dependent manner after hCG treatment, and gastric emptying showed a significant negative correlation with CCK concentrations (P=0.01, r(2)=-0.5104). Peripheral administration (i.p.) of lorglumide, a selective CCK(1) receptor antagonist, attenuated the hCG-induced inhibition of gastric emptying in Ovx rats, whereas central administration via the i.c.v. route did not. hCG treatment of Ovx rats inhibits gastric emptying in a dose-dependent manner via a peripheral mechanism of CCK hypersecretion and activation of CCK(1) receptors.
Collapse
Affiliation(s)
- Kok-Min Seow
- Department of Obstetrics and Gynecology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Tiruthani K, Sarkar P, Rao B. Trophoblast differentiation of human embryonic stem cells. Biotechnol J 2013; 8:421-33. [PMID: 23325630 DOI: 10.1002/biot.201200203] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 11/03/2012] [Accepted: 12/06/2012] [Indexed: 11/08/2022]
Abstract
Molecular mechanisms regulating human trophoblast differentiation remain poorly understood due to difficulties in obtaining primary tissues from very early developmental stages in humans. Therefore, the use of human embryonic stem cells (hESCs) as a source for generating trophoblast tissues is of significant interest. Trophoblast-like cells have been obtained through treatment of hESCs with bone morphogenetic protein (BMP) or inhibitors of activin/nodal/transforming growth factor-β signaling, or through protocols involving formation of embryoid bodies (EBs); however, there is controversy over whether hESC-derived cells are indeed analogous to true trophoblasts found in vivo. In this review, we provide an overview of previously described efforts to obtain trophoblasts from hESCs. We also discuss the merits and limitations of hESCs as a source of trophoblast derivatives.
Collapse
Affiliation(s)
- Karthik Tiruthani
- Department of Chemical and Biomolecular Engineering, North Carolina State University, NC 27695, USA
| | | | | |
Collapse
|
25
|
Bernardini L, Moretti-Rojas I, Brush M, Rojas FJ, Balmaceda JP. Failure of hCG/LH receptors to stimulate the transmembrane effector adenylyl cyclase in human endometrium. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/abb.2013.410126] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
26
|
Shiefa S, Amargandhi M, Bhupendra J, Moulali S, Kristine T. First Trimester Maternal Serum Screening Using Biochemical Markers PAPP-A and Free β-hCG for Down Syndrome, Patau Syndrome and Edward Syndrome. Indian J Clin Biochem 2013; 28:3-12. [PMID: 24381414 PMCID: PMC3547446 DOI: 10.1007/s12291-012-0269-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Accepted: 09/30/2012] [Indexed: 11/24/2022]
Abstract
The first trimester screening programme offers a noninvasive option for the early detection of aneuploidy pregnancies. This screening is done by a combination of two biochemical markers i.e. serum free β-human chorionic gonadotrophin (free β-hCG) and pregnancy associated plasma protein A (PAPP-A), maternal age and fetal nuchal translucency (NT) thickness at 11 + 0-13 + 6 weeks of gestation. A beneficial consequence of screening is the early diagnosis or trisomies 21, 18 and 13. At 11 + 0-13 + 6 weeks, the relative prevalence of trisomies 18 and 13 to trisomy 21 are found to be one to three and one to seven, respectively. All three trisomies are associated with increased maternal age, increased fetal NT and decreased PAPP-A, but in trisomy 21 serum free β-hCG is increased whereas in trisomies 18 and 13 free β-hCG is decreased.
Collapse
Affiliation(s)
- S. Shiefa
- SRL Diagnostics, Fortis Healthcare Enterprise, Clinical Biochemist, 64, Al Razi Unit 1007, Block A, P.O. Box 505143, Dubai Health Care City, United Arab Emirates
| | - M. Amargandhi
- SRL Diagnostics, Fortis Healthcare Enterprise, Clinical Biochemist, 64, Al Razi Unit 1007, Block A, P.O. Box 505143, Dubai Health Care City, United Arab Emirates
| | - J. Bhupendra
- SRL Diagnostics, Fortis Healthcare Enterprise, Clinical Biochemist, 64, Al Razi Unit 1007, Block A, P.O. Box 505143, Dubai Health Care City, United Arab Emirates
| | - S. Moulali
- SRL Diagnostics, Fortis Healthcare Enterprise, Clinical Biochemist, 64, Al Razi Unit 1007, Block A, P.O. Box 505143, Dubai Health Care City, United Arab Emirates
| | - T. Kristine
- SRL Diagnostics, Fortis Healthcare Enterprise, Clinical Biochemist, 64, Al Razi Unit 1007, Block A, P.O. Box 505143, Dubai Health Care City, United Arab Emirates
| |
Collapse
|
27
|
Dukic-Stefanovic S, Walther J, Wosch S, Zimmermann G, Wiedemann P, Alexander H, Claudepierre T. Chorionic gonadotropin and its receptor are both expressed in human retina, possible implications in normal and pathological conditions. PLoS One 2012; 7:e52567. [PMID: 23285091 PMCID: PMC3526580 DOI: 10.1371/journal.pone.0052567] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 11/20/2012] [Indexed: 11/18/2022] Open
Abstract
Extra-gonadal role of gonadotropins has been re-evaluated over the last 20 years. In addition to pituitary secretion of luteinizing hormone (LH) and follicle stimulating hormone (FSH), the CNS has been clearly identified as a source of hCG acting locally to influence behaviour. Here we demonstrated that human retina is producing this gonadotropin that acts as a neuroactive molecule. Müller glial and retinal pigmented epithelial (RPE) cells are producing hCG that may affects neighbour cells expressing its receptor, namely cone photoreceptors. It was previously described that amacrine and retinal ganglion (RGC) cells are targets of the gonadotropin releasing hormone that control the secretion of all gonadotropins. Therefore our findings suggest that a complex neuroendocrine circuit exists in the retina, involving hCG secreting cells (glial and RPE), hCG targets (photoreceptors) and hCG-release controlling cells (amacrine and RGC). The exact physiological functions of this circuit have still to be identified, but the proliferation of photoreceptor-derived tumor induced by hCG demonstrated the need to control this neuroendocrine loop.
Collapse
MESH Headings
- Blotting, Western
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Chorionic Gonadotropin, beta Subunit, Human/genetics
- Chorionic Gonadotropin, beta Subunit, Human/metabolism
- Chorionic Gonadotropin, beta Subunit, Human/pharmacology
- Gene Expression Regulation
- Humans
- Protein Transport
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, LH/metabolism
- Retina/metabolism
- Retina/pathology
- Retinoblastoma/metabolism
- Retinoblastoma/pathology
Collapse
Affiliation(s)
| | - Jan Walther
- Department of Ophthalmology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Sebastian Wosch
- Department of Ophthalmology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Gerolf Zimmermann
- Department of Obstetrics and Gynecology, Division of Human Reproduction and Endocrinology, University of Leipzig, Leipzig, Germany
| | - Peter Wiedemann
- Department of Ophthalmology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Henry Alexander
- Department of Obstetrics and Gynecology, Division of Human Reproduction and Endocrinology, University of Leipzig, Leipzig, Germany
| | - Thomas Claudepierre
- Department of Ophthalmology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| |
Collapse
|
28
|
Chambers AE, Griffin C, Naif SA, Mills I, Mills WE, Syngelaki A, Nicolaides KH, Banerjee S. Quantitative ELISAs for serum soluble LHCGR and hCG-LHCGR complex: potential diagnostics in first trimester pregnancy screening for stillbirth, Down's syndrome, preterm delivery and preeclampsia. Reprod Biol Endocrinol 2012; 10:113. [PMID: 23245345 PMCID: PMC3570453 DOI: 10.1186/1477-7827-10-113] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 12/14/2012] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Soluble LH/hCG receptor (sLHCGR) released from placental explants and transfected cells can be detected in sera from pregnant women. To determine whether sLHCGR has diagnostic potential, quantitative ELISAs were developed and tested to examine the correlation between pregnancy outcome and levels of serum sLHCGR and hCG-sLHCGR complex. METHODS Anti-LHCGR poly- and monoclonal antibodies recognizing defined LHCGR epitopes, commerical anti-hCGbeta antibody, together with recombinant LHCGR and yoked hCGbeta-LHCGR standard calibrators were used to develop two ELISAs. These assays were employed to quantify serum sLHCGR and hCG-sLHCGR at first trimester human pregnancy. RESULTS Two ELISAs were developed and validated. Unlike any known biomarker, sLHCGR and hCG-sLHCGR are unique because Down's syndrome (DS), preeclampsia and preterm delivery are linked to both low (less than or equal to 5 pmol/mL), and high (equal to or greater than 170 pmol/mL) concentrations. At these cut-off values, serum hCG-sLHCGR together with PAPP-A detected additional DS pregnancies (21%) which were negative by free hCGbeta plus PAPP-A screening procedure. Therefore, sLHCGR/hCG-sLHCGR has an additive effect on the current primary biochemical screening of aneuploid pregnancies. More than 88% of pregnancies destined to end in fetal demise (stillbirth) exhibited very low serum hCG-sLHCGR(less than or equal to 5 pmol/mL) compared to controls (median 16.15 pmol/mL, n = 390). The frequency of high hCG-sLHCGR concentrations (equal to or greater than 170 pmol/mL) in pathological pregnancies was at least 3-6-fold higher than that of the control, suggesting possible modulation of the thyrotropic effect of hCG by sLHCGR. CONCLUSIONS Serum sLHCGR/hCG-sLHCGR together with PAPP-A, have significant potential as first trimester screening markers for predicting pathological outcomes in pregnancy.
Collapse
Affiliation(s)
- Anne E Chambers
- Department of Clinical Biochemistry, Heartlands Hospital, Birmingham, B9 5SS, UK
- Present address: Origin Biomarkers, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire, AL7 3AX, UK
| | | | | | - Ian Mills
- Birmingham Women’s Hospital, Edgbaston, Birmingham, UK
| | - Walter E Mills
- Department of Clinical Biochemistry, Heartlands Hospital, Birmingham, B9 5SS, UK
- Present address: Origin Biomarkers, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire, AL7 3AX, UK
| | - Argyro Syngelaki
- Harris Birthright Research Centre for Fetal Medicine, King’s College Hospital, London, UK
| | - Kypros H Nicolaides
- Harris Birthright Research Centre for Fetal Medicine, King’s College Hospital, London, UK
| | - Subhasis Banerjee
- Department of Clinical Biochemistry, Heartlands Hospital, Birmingham, B9 5SS, UK
- Present address: Origin Biomarkers, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire, AL7 3AX, UK
| |
Collapse
|
29
|
Comparison of splenic transcriptome activity of two rainbow trout strains differing in robustness under regional aquaculture conditions. Mol Biol Rep 2012; 40:1955-66. [DOI: 10.1007/s11033-012-2252-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 10/10/2012] [Indexed: 10/27/2022]
|
30
|
Abstract
The LH receptor (LHR) and FSH receptor (FSHR), collectively termed the gonadotropin receptors, are members of the Family A of GPCRs. The gonadotropin receptors each contain N-linked carbohydrates that are not directly involved in hormone binding, but contribute to the proper folding, and therefore, cell surface expression of the receptor. Loss-of-function mutations of an LHR or FSHR results in decreased target cell responsiveness. Most inactivating mutations cause receptor misfolding, resulting in the retention of the mutant in its immature form in the endoplasmic reticulum. A membrane-permeable allosteric agonist of the LHR has been shown to serve as a pharmacological chaperone for misfolded and intracellularly retained LHRs by promoting their cell surface expression. Wild-type LHR and FSHR each form homodimers and heterodimers while in the ER. Therefore, when wild-type receptor is co-expressed with a misfolded mutant, the misfolded receptor dimerizes with immature wild-type receptor in the ER, causing a dominant-negative effect on cell surface expression of the mature wild-type receptor. Notably, the propensity for homodimerization is not affected by the activation status of the receptor. However, within a receptor dimer, the activity of one protomer may allosterically regulate the other protomer. Therefore, the dimerization of the gonadotropin receptors appears to be an obligate process that is part of the normal itinerary for trafficking to the cell surface and, once there, the dimerized receptors allow for additional modulations of cell signaling.
Collapse
Affiliation(s)
- Deborah L Segaloff
- Department of Molecular Physiology and Biophysics, The University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA,
| |
Collapse
|