Skip to main content
Erschienen in: Archives of Gynecology and Obstetrics 5/2023

Open Access 18.01.2023 | Gynecologic Endocrinology and Reproductive Medicine

Co-cultivation of human granulosa cells with ovarian cancer cells leads to a significant increase in progesterone production

verfasst von: Detlef Pietrowski, Martina Grgic, Isabella Haslinger, Julian Marschalek, Christian Schneeberger

Erschienen in: Archives of Gynecology and Obstetrics | Ausgabe 5/2023

Abstract

Purpose

In humans, granulosa cells (GCs) are part of the follicle and nourish the growing oocyte. GCs produce estrogen and, after ovulation, progesterone. They are embedded in a multicellular tissue structure of the ovary, which consists of a variety of different cell types that are essential for the physiological function of the ovary. However, the extent to which individual ovarian cell types contribute to overall functionality has not yet been fully elucidated. In this study, we aim to investigate the effects of co-culturing human granulosa cells with ovarian cancer cells on their progesterone and estrogen production in an in vitro model.

Methods

After seeding, the cells were stimulated with 200 µM forskolin in DMEM for 72 h and the medium of the different cell culture experiments was collected. Subsequently, progesterone and oestradiol concentrations were determined using an Elisa assay.

Results

Morphologically, it was striking that the cells self-organize and form spatially separated areas. Compared to culturing granulosa cells alone, co-culturing human granulosa cells together with the ovarian cancer cell line OvCar-3 resulted in a significant increase in progesterone production (20.3 ng/ml versus 50.2 ng/ml; p < 0.01).

Conclusions

Using a simple in vitro model, we highlight the importance of cellular crosstalk between different ovarian cells in a complex cellular network and that it strongly influences granulosa cell hormone production. This could have potential implications for the procedure of transplanting endocrine tissues after cryopreservation, as it highlights the importance of survival of all cells for the functionality of the transplanted tissue.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
What does this study add to the clinical work
The proposed in vitro model, using ovarian cells as an example, suggests that when ovarian tissue is re-transplanted, the cell types that are most severely damaged can determine the success or failure of the intervention. Furthermore, it indicates a remarkable influence of ovarian cancer cells on its cellular environment.

Introduction

The ovaries—as part of the female reproductive organs—represent a polymorphic complex structure consisting of a multitude of different cell types, each with different functions. To ensure full functionality of the ovary as an organ, all tissue and cell types must interact with each other in a finely orchestrated manner [1, 2].
An essential feature of a morphologically and physiologically intact ovary is the ability to produce sex steroid hormones under the control of gonadotropins. It is well established that granulosa cells (GC) and theca cells (TC) play a key role in this physiological process, being the sites of estrogen and progesterone production. For in vitro studies on ovarian hormone production, GC-based in vitro cell model systems have been developed and provide a valuable tool for an analysis of molecular pathways in hormone production [3, 4]. The major source of human GCs for in vitro studies is usually derived from patients undergoing artificial reproductive technologies (ART). However, these cells are accessible only in limited amounts, which make it difficult to perform widespread experiments related to detailed molecular analysis. Furthermore, and because of the therapy-specific gonadotropin-stimulation with ovulation induction, clinically obtained GCs are mostly luteinized. These GCs have a restricted life span with a slow proliferation rate, and they do not stay viable in vitro after many passages [3, 5]. In addition, primary GCs derived from specific patients may exhibit very high variability, which further complicates the achievement of reproducible results. Due to these problems, permanent cell models with primary GCs are difficult to establish. Therefore, in in vitro studies, human GC cell lines are often considered an attractive option [3, 6]. For example, the immortalized Granulosa cell lines KGN and HGL- 5 have been shown to be valuable in ovarian functional studies [7]. While the KGN cell line produces significant amounts of progesterone after stimulation with cyclic adenosine monophosphate (cAMP) inducing agents other cell lines do not. The KGN cell line was therefore considered particularly useful for in vitro studies of ovarian steroid hormone production.
Recently, it was shown that cell-to-cell contact is a pre-requisite to improve cellular vitality and that a specific microenvironment is necessary for promotion of endocrine function in various tissues including the ovary [810]. In addition, Fozzatti and co-authors have shown that cancer development and cancer progression depend on the mutual interaction between growing tumor cells and their surrounding cellular microenvironment [11].
In the ovary. the influence of other cells on GC triggered hormonal production has not been extensively studied. We hypothesize that GC surrounding cells may have an impact on hormone production of GCs. To address this question, we developed a simple two-cell model consisting of the human granulosa cell line KGN and the human carcinoma cell line OvCar-3 and analyzed the impact of this ovarian cell lines on hormone production by GCs.

Materials and methods

Except otherwise stated, all chemicals were obtained from Sigma (Sigma Chemical Co., St Louis, USA).

Cell culture and stimulation

The human granulosa cell line KGN was established by Nishi et al., 2001 from a stage III granulosa cell carcinoma [12]. Human KGN cells were cultured as previously described in DMEM (Dulbecco's Modified Eagle Medium), 10% FCS (Fetal calf serum) and a final concentration of 100 units/ml of penicillin and 100 µg/ml of streptomycin. (1% Pen/Strep) at 37 °C and 5% CO2 [13, 14]. Cell passages were 3–5.
The OvCar-3 cell line was cultivated in DMEM, 10% FCS (Fetal calf serum) and 1% Pen/Strep at 37 °C and 5% CO2. Cell passages were 2–5.
In brief, cells were seeded in 6-well plates at a density of 1 × 105 cells per ml. After 24 h, the medium was discarded and the remaining cells were washed twice with PBS and new media were added. Stimulation was carried by adding Forskolin to a final concentration of 1, 10, 50, 100, 200 µM, respectively. After 72 h, the supernatants were collected and stored for hormone quantification. In the case of co-culturing 5 × 104 of each cell line were seeded.
For cell passaging media was discarded and cells were treated with 3 ml Trypsin/EDTA solution containing 0.5 g trypsin and 0.2 g EDTA for 3 min at 37 °C, 5% CO2 after washing with PBS. The reaction was stopped by adding 2 ml of media containing 10% FCS. The detached cells were centrifuged for 5 min at 800 g. The media was discarded and new media was added to the cell pellet. The cell pellet was solved by gentle pipetting and cells were counted after Trypan Blue staining in a counting chamber.

Hormone quantification

Cell culture media was collected after 72 h of treatment and kept frozen at − 80 °C prior to testing.
Enzyme-linked immunosorbent assays (ELISAs) for estrogen (17beta-estradiol) and progesterone were performed using an ELISA kit according to the manufacturer's instructions (IBL international, Hamburg, Germany). Essentially, 25 µl of each sample was added in duplicate to a 96-well ELISA plate and incubated for 1 h at room temperature. Unbound material was washed out three times with washing solution. After addition of substrate solution, plates were incubated for an additional 15 min and stop solution was added. Absorbance (OD) was determined using a microplate reader (LumiStar Optima, BMG, Kumberg, Austria) at 450 nm.
Intra-assay variability and inter-assay variability for the progesterone ELISA ranged from 5.4 to 6.99% CV and from 4.34 to 9.96% CV, respectively. Intra-assay variability and inter-assay variability for the estrogen ELISA ranged from 8.70 to 90.23% CV and from 6.87 to 14.91% CV, respectively.

Statistics

The results of the hormone quantification experiments have been analyzed using ANOVA for independent samples followed by Tukey's HSD test. Differences were considered statistically significant at p < 0.05. Statistical analyses were performed at the statskingdom website at https://​www.​statskingdom.​com.

Results

To investigate the effect on steroid hormone production by co-culturing a granulosa cell line and an epithelial carcinoma cell line, both cell lines were cultured under standard cell culture conditions separately and in a 1:1 ratio for 72 h. Morphologically, Fig. 1 shows that OvCar-3 and KGN together build distinguishable areas and the cell lines do not completely intermix. To investigate the hormone production capacity of the cells after culturing, we stimulated the cells with increasing amounts of forskolin, a cyclic adenosine monophosphate (cAMP)-inducing agent. Although OvCar-3 cells were not known to produce progesterone, we assessed this possibility by measuring progesterone concentration after stimulation with increasing amounts of forskolin. As demonstrated in Fig. 2, the stimulation of the OvCar-3 cells has no detectable effect including a concentration of 200 µM forskolin. Stimulation of the human granulosa cell line KGN demonstrates a concentration-dependent increase in progesterone production from 2.2 ng/ml at a progesterone concentration of 10 µM to an amount of 20.3 ng/ml at a concentration of 200 µM forskolin. When we cultivated both cell lines in a co-culture and a ratio of 1:1, we observed an increase in progesterone production from 2.8 ng/ml at a concentration of 10 µM forskolin up to 50.2 ng/ml at a concentration of 200 µM forskolin. We would like to point out that the amount of progesterone-producing KGN cells in these co-culture experiments is about half the amount of the KGN single-culture experiments. Additionally, we performed the same series of experiments and determined the estrogen production of both cell lines. However, significant amounts of estrogen was not detectable (data not shown).

Discussion

In our study, we were able to show that the KGN–OvCar-3 co-culture system is a suitable model for demonstrating a stimulatory paracrine effect of co-culturing these two ovarian cell lines on progesterone production.
The progesterone synthesis of the human granulosa cell line KGN is mediated by a cAMP-triggered mechanism involving protein kinase A (PKA) dependent pathways [15]. Therefore, it is suitable to increase progesterone production either by increasing the amount of cAMP in the cells or by stimulating the enzyme adenylate cyclase with forskolin as we did in our study.
The progesterone amounts measured in our experiments for KGN cell culture after 72 h and stimulation with 200 µM forskolin are in the same physiological range as the values reported by Nishi et al., after (Bu)2cAMP stimulation (20.3 ng/ml vs. 15 ng/ml) (Nishi, Yanase et al. 2001). In the co-culture experiments together with OvCar-3 cells, however, we reached values up to 50.2 ng/ml, which corresponds to an increase by a factor of about five, taking into account that only half of the KGN cells have been seeded. A modulatory effect on cell viability and function by co-culturing different cell types was shown by Lancaster et al., for stem cell based organoid models [16]. In addition, a form of self-organization of granulosa cells and different other cells has already been described by Qiu et al. [17] as well as our group [18, 19]. De Souza and colleagues have shown that mesenchymal stem cells (MSC) increase viability and hormone production capacity of islets cells [20] and Jiang and colleagues reported a regulatory effect by endothelial cells on microglial cells [21]. A stimulatory effect by co-culturing on cell migration of cancer cells and on ovarian differentiation was reported by Dogan et al. [22] and Mackay et al. [23]. Our data are in line with results of other authors that co-cultivation of different cell types from ovarian origin can have a significant effect on hormone production. For example, the data of Qiu et al. have shown in in vitro experiments that co-culturing ovarian cortical stromal cells with primary granulosa cells derived from goat ovaries can stimulate both progesterone synthesis and cell proliferation.
Human granulosa cells are the site of hormone production in the ovary and play a crucial role in oocyte development trough interaction with a variety of other cells and cell types, resulting in a coordinated synergistic physiological process. It is justifiable to state that the failure of a small part of this system can lead to the breakdown of the entire physiological network. Our simple in vitro model of cooperation of granulosa cells and epithelial ovarian cells in hormone production supports the hypothesis that also in ovarian tissue, the cell types adjacent to the granulosa cells make an important contribution to hormone production and, thus, also to functionality. However, a restriction of the validity of our model is that we are dealing with two cell lines and therefore a generalization to the in vivo situation seems to be possible only to a limited extent. Our model uses cancer cells, not primary cells and cannot fully describe the complex interactions, which occur in vivo.
In the special situation of cryopreservation and re-transplantaion of ovarian tissue, fundamentally different cell types must cope with the stressors of freezing and the potentially toxic properties of the cryoprotective solutions. Therefore, special care should be taken in account to ensure that all cell types of the tissue are viable again after warming to return to full functionality after re-transplantation. Cell types that were most severely damaged by the procedure used may be decisive for the success or failure of the therapy in terms of different vitality.

Conclusion

Our model seems to be a simple and “easy to handle” model to study the effects of different cell types on the ability of human granulosa cells to produce progesterone. Furthermore, it highlights the relevance of survival of all cells in endocrine tissues after cryopreservation and subsequent re-transplantation.

Acknowledgements

Human KGN cells were a friendly kind gift by K. Horling Department of Anatomy and Cell Biology, Martin Luther University Faculty of Medicine, Halle (Saale), Germany. The human ovarian cancer cell line OvCar-3 were a friendly gift by Dr. D.C. Castillo-Tong (Medical University Vienna, Vienna, Austria).

Declarations

Conflict of interest

The authors have no relevant financial or non-financial interests to disclose.

Ethical approval

Not applicable as this is a basic research study using established cell lines.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Gynäkologie

Kombi-Abonnement

Mit e.Med Gynäkologie erhalten Sie Zugang zu CME-Fortbildungen der beiden Fachgebiete, den Premium-Inhalten der Fachzeitschriften, inklusive einer gedruckten gynäkologischen oder urologischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Fan X, Bialecka M, Moustakas I, Lam E, Torrens-Juaneda V, Borggreven NV et al (2019) Single-cell reconstruction of follicular remodeling in the human adult ovary. Nat Commun 10:3164CrossRefPubMedPubMedCentral Fan X, Bialecka M, Moustakas I, Lam E, Torrens-Juaneda V, Borggreven NV et al (2019) Single-cell reconstruction of follicular remodeling in the human adult ovary. Nat Commun 10:3164CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Nelson SM, Telfer EE, Anderson RA (2013) The ageing ovary and uterus: new biological insights. Hum Reprod Update 19:67–83CrossRefPubMed Nelson SM, Telfer EE, Anderson RA (2013) The ageing ovary and uterus: new biological insights. Hum Reprod Update 19:67–83CrossRefPubMed
3.
Zurück zum Zitat Havelock JC, Rainey WE, Carr BR (2004) Ovarian granulosa cell lines. Mol Cell Endocrinol 228:67–78CrossRefPubMed Havelock JC, Rainey WE, Carr BR (2004) Ovarian granulosa cell lines. Mol Cell Endocrinol 228:67–78CrossRefPubMed
4.
Zurück zum Zitat Almeida CP, Ferreira MCF, Silveira CO, Campos JR, Borges IT, Baeta PG et al (2018) Clinical correlation of apoptosis in human granulosa cells-a review. Cell Biol Int 42:1276–1281CrossRefPubMed Almeida CP, Ferreira MCF, Silveira CO, Campos JR, Borges IT, Baeta PG et al (2018) Clinical correlation of apoptosis in human granulosa cells-a review. Cell Biol Int 42:1276–1281CrossRefPubMed
5.
Zurück zum Zitat Lie BL, Leung E, Leung PC, Auersperg N (1996) Long-term growth and steroidogenic potential of human granulosa-lutein cells immortalized with SV40 large T antigen. Mol Cell Endocrinol 120:169–176CrossRefPubMed Lie BL, Leung E, Leung PC, Auersperg N (1996) Long-term growth and steroidogenic potential of human granulosa-lutein cells immortalized with SV40 large T antigen. Mol Cell Endocrinol 120:169–176CrossRefPubMed
6.
Zurück zum Zitat Rainey WH, Sawetawan C, Shay JW, Michael MD, Mathis JM, Kutteh W et al (1994) Transformation of human granulosa cells with the E6 and E7 regions of human papillomavirus. J Clin Endocrinol Metab 78:705–710PubMed Rainey WH, Sawetawan C, Shay JW, Michael MD, Mathis JM, Kutteh W et al (1994) Transformation of human granulosa cells with the E6 and E7 regions of human papillomavirus. J Clin Endocrinol Metab 78:705–710PubMed
7.
Zurück zum Zitat Ernst J, Grabiec U, Greither T, Fischer B, Dehghani F (2016) The endocannabinoid system in the human granulosa cell line KGN. Mol Cell Endocrinol 423:67–76CrossRefPubMed Ernst J, Grabiec U, Greither T, Fischer B, Dehghani F (2016) The endocannabinoid system in the human granulosa cell line KGN. Mol Cell Endocrinol 423:67–76CrossRefPubMed
9.
Zurück zum Zitat Li Y, Zhang D, Xu L, Dong L, Zheng J, Lin Y et al (2019) Cell-cell contact with proinflammatory macrophages enhances the immunotherapeutic effect of mesenchymal stem cells in two abortion models. Cell Mol Immunol 16:908–920CrossRefPubMedPubMedCentral Li Y, Zhang D, Xu L, Dong L, Zheng J, Lin Y et al (2019) Cell-cell contact with proinflammatory macrophages enhances the immunotherapeutic effect of mesenchymal stem cells in two abortion models. Cell Mol Immunol 16:908–920CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Sugimura S, Kobayashi N, Okae H, Yamanouchi T, Matsuda H, Kojima T et al (2017) Transcriptomic signature of the follicular somatic compartment surrounding an oocyte with high developmental competence. Sci Rep 7:6815CrossRefPubMedPubMedCentral Sugimura S, Kobayashi N, Okae H, Yamanouchi T, Matsuda H, Kojima T et al (2017) Transcriptomic signature of the follicular somatic compartment surrounding an oocyte with high developmental competence. Sci Rep 7:6815CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Fozzatti L, Cheng SY (2020) Tumor cells and cancer-associated fibroblasts: a synergistic crosstalk to promote thyroid cancer. Endocrinol Metab (Seoul) 35:673–680CrossRefPubMed Fozzatti L, Cheng SY (2020) Tumor cells and cancer-associated fibroblasts: a synergistic crosstalk to promote thyroid cancer. Endocrinol Metab (Seoul) 35:673–680CrossRefPubMed
12.
Zurück zum Zitat Nishi Y, Yanase T, Mu Y, Oba K, Ichino I, Saito M et al (2001) Establishment and characterization of a steroidogenic human granulosa-like tumor cell line, KGN, that expresses functional follicle-stimulating hormone receptor. Endocrinology 142:437–445CrossRefPubMed Nishi Y, Yanase T, Mu Y, Oba K, Ichino I, Saito M et al (2001) Establishment and characterization of a steroidogenic human granulosa-like tumor cell line, KGN, that expresses functional follicle-stimulating hormone receptor. Endocrinology 142:437–445CrossRefPubMed
13.
Zurück zum Zitat Xu Y, Zagoura D, Keck C, Pietrowski D (2006) Expression of Eph receptor tyrosine kinases and their ligands in human granulosa lutein cells and human umbilical vein endothelial cells. Exp Clin Endocrinol Diabetes 114:590–595CrossRefPubMed Xu Y, Zagoura D, Keck C, Pietrowski D (2006) Expression of Eph receptor tyrosine kinases and their ligands in human granulosa lutein cells and human umbilical vein endothelial cells. Exp Clin Endocrinol Diabetes 114:590–595CrossRefPubMed
14.
Zurück zum Zitat Hovemann I, Keck C, Fiebich BL, Pietrowski D (2008) Effects of selenium and mistletoe extracts on the expression of angiogenic growth factors and sprout formation in a spheroid cell culture system of human granulosa lutein cells. Geburtshilfe Frauenheilkd 68:620–624CrossRef Hovemann I, Keck C, Fiebich BL, Pietrowski D (2008) Effects of selenium and mistletoe extracts on the expression of angiogenic growth factors and sprout formation in a spheroid cell culture system of human granulosa lutein cells. Geburtshilfe Frauenheilkd 68:620–624CrossRef
15.
Zurück zum Zitat Fang L, Guo Y, Li Y, Jia Q, Han X, Liu B et al (2022) Epigallocatechin-3-gallate stimulates StAR expression and progesterone production in human granulosa cells through the 67-kDa laminin receptor-mediated CREB signaling pathway. J Cell Physiol 237:687–695CrossRefPubMed Fang L, Guo Y, Li Y, Jia Q, Han X, Liu B et al (2022) Epigallocatechin-3-gallate stimulates StAR expression and progesterone production in human granulosa cells through the 67-kDa laminin receptor-mediated CREB signaling pathway. J Cell Physiol 237:687–695CrossRefPubMed
16.
Zurück zum Zitat Lancaster MA, Knoblich JA (2014) Organogenesis in a dish: modeling development and disease using organoid technologies. Science 345:1247125CrossRefPubMed Lancaster MA, Knoblich JA (2014) Organogenesis in a dish: modeling development and disease using organoid technologies. Science 345:1247125CrossRefPubMed
17.
Zurück zum Zitat Qiu M, Liu J, Han C, Wu B, Yang Z, Su F et al (2014) The influence of ovarian stromal/theca cells during in vitro culture on steroidogenesis, proliferation and apoptosis of granulosa cells derived from the goat ovary. Reprod Domest Anim 49:170–6CrossRefPubMed Qiu M, Liu J, Han C, Wu B, Yang Z, Su F et al (2014) The influence of ovarian stromal/theca cells during in vitro culture on steroidogenesis, proliferation and apoptosis of granulosa cells derived from the goat ovary. Reprod Domest Anim 49:170–6CrossRefPubMed
18.
Zurück zum Zitat Becker J, Walz A, Daube S, Keck C, Pietrowski D (2007) Distinct responses of human granulosa lutein cells after hCG or LH stimulation in a spheroidal cell culture system. Mol Reprod Dev 74:1312–1316CrossRefPubMed Becker J, Walz A, Daube S, Keck C, Pietrowski D (2007) Distinct responses of human granulosa lutein cells after hCG or LH stimulation in a spheroidal cell culture system. Mol Reprod Dev 74:1312–1316CrossRefPubMed
19.
Zurück zum Zitat Walz A, Keck C, Weber H, Kissel C, Pietrowski D (2005) Effects of luteinizing hormone and human chorionic gonadotropin on corpus luteum cells in a spheroid cell culture system. Mol Reprod Dev 72:98–104CrossRefPubMed Walz A, Keck C, Weber H, Kissel C, Pietrowski D (2005) Effects of luteinizing hormone and human chorionic gonadotropin on corpus luteum cells in a spheroid cell culture system. Mol Reprod Dev 72:98–104CrossRefPubMed
20.
Zurück zum Zitat de Souza BM, Bouças AP, Oliveira FD, Reis KP, Ziegelmann P, Bauer AC et al (2017) Effect of co-culture of mesenchymal stem/stromal cells with pancreatic islets on viability and function outcomes: a systematic review and meta-analysis. Islets 9:30–42CrossRefPubMedPubMedCentral de Souza BM, Bouças AP, Oliveira FD, Reis KP, Ziegelmann P, Bauer AC et al (2017) Effect of co-culture of mesenchymal stem/stromal cells with pancreatic islets on viability and function outcomes: a systematic review and meta-analysis. Islets 9:30–42CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Jiang L, Womble T, Saporta S, Chen N, Sanberg CD, Sanberg PR et al (2010) Human umbilical cord blood cells decrease microglial survival in vitro. Stem Cells Dev 19:221–228CrossRefPubMedPubMedCentral Jiang L, Womble T, Saporta S, Chen N, Sanberg CD, Sanberg PR et al (2010) Human umbilical cord blood cells decrease microglial survival in vitro. Stem Cells Dev 19:221–228CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Mackay S, Willerton L, Ballingall CL, Henderson NJ, Smith RA (2004) Developing mouse Sertoli cells in vitro: effects on developing ovaries in co-culture and production of anti-Müllerian hormone. Cells Tissues Organs 177:79–86CrossRefPubMed Mackay S, Willerton L, Ballingall CL, Henderson NJ, Smith RA (2004) Developing mouse Sertoli cells in vitro: effects on developing ovaries in co-culture and production of anti-Müllerian hormone. Cells Tissues Organs 177:79–86CrossRefPubMed
Metadaten
Titel
Co-cultivation of human granulosa cells with ovarian cancer cells leads to a significant increase in progesterone production
verfasst von
Detlef Pietrowski
Martina Grgic
Isabella Haslinger
Julian Marschalek
Christian Schneeberger
Publikationsdatum
18.01.2023
Verlag
Springer Berlin Heidelberg
Erschienen in
Archives of Gynecology and Obstetrics / Ausgabe 5/2023
Print ISSN: 0932-0067
Elektronische ISSN: 1432-0711
DOI
https://doi.org/10.1007/s00404-023-06914-z

Weitere Artikel der Ausgabe 5/2023

Archives of Gynecology and Obstetrics 5/2023 Zur Ausgabe

Alter der Mutter beeinflusst Risiko für kongenitale Anomalie

28.05.2024 Kinder- und Jugendgynäkologie Nachrichten

Welchen Einfluss das Alter ihrer Mutter auf das Risiko hat, dass Kinder mit nicht chromosomal bedingter Malformation zur Welt kommen, hat eine ungarische Studie untersucht. Sie zeigt: Nicht nur fortgeschrittenes Alter ist riskant.

Fehlerkultur in der Medizin – Offenheit zählt!

28.05.2024 Fehlerkultur Podcast

Darüber reden und aus Fehlern lernen, sollte das Motto in der Medizin lauten. Und zwar nicht nur im Sinne der Patientensicherheit. Eine negative Fehlerkultur kann auch die Behandelnden ernsthaft krank machen, warnt Prof. Dr. Reinhard Strametz. Ein Plädoyer und ein Leitfaden für den offenen Umgang mit kritischen Ereignissen in Medizin und Pflege.

Mammakarzinom: Brustdichte beeinflusst rezidivfreies Überleben

26.05.2024 Mammakarzinom Nachrichten

Frauen, die zum Zeitpunkt der Brustkrebsdiagnose eine hohe mammografische Brustdichte aufweisen, haben ein erhöhtes Risiko für ein baldiges Rezidiv, legen neue Daten nahe.

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.