1
|
Cadenas J, Adrados CS, Kumar A, Kalra B, Mamsen LS, Andersen CY. Regulating human oocyte maturation in vitro: a hypothesis based on oocytes retrieved from small antral follicles during ovarian tissue cryopreservation. J Assist Reprod Genet 2025:10.1007/s10815-025-03483-9. [PMID: 40261459 DOI: 10.1007/s10815-025-03483-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/10/2025] [Indexed: 04/24/2025] Open
Abstract
PURPOSE To characterize the hormonal environment in spent medium and cumulus cell gene expression during human IVM using oocytes from small antral follicles (SAFs) retrieved from surplus medulla tissue after ovarian tissue cryopreservation. METHODS Immature oocytes from surplus medulla tissue underwent 42-h IVM in media with varying FSH and LH concentrations (0, 10, 100 IU/L FSH, and 100 IU/L FSH + 100 IU/L LH). Oocyte maturation was assessed by germinal vesicle (GV), metaphase I (MI), or metaphase II (MII) stages. Gene expression of FSHR, LHCGR, AMH, CYP19 A1, and INHA in cumulus cells was analyzed by RT-qPCR, and GDF9, AMH, inhibin-B, inhibin-A, and total inhibin were measured in the spent media by ELISA. RESULTS Increased FSH concentrations downregulated FSHR expression and upregulated LHCGR, which correlated with MII transition. GDF9 concentrations in the spent medium significantly decreased with higher FSH, as did GDF9, AMH, and inhibin-B in MII oocytes. Inhibin-A levels tended to be higher in the media of MII oocytes. FSHR expression was positively associated with inhibin-B and negatively with inhibin-A, while LHCGR showed the opposite pattern and was also negatively linked to GDF9 concentration. CONCLUSION FSH-induced LHCGR expression, along with FSHR downregulation, is closely linked to oocyte maturation. Reduced GDF9 secretion from oocytes facilitates LHCGR expression on cumulus cells, while FSH and LH collectively induced hormones like inhibin-A, which likely support oocyte maturation.
Collapse
Affiliation(s)
- Jesús Cadenas
- Laboratory of Reproductive Biology, section 5701, University Hospital of Copenhagen, Rigshospitalet, Henrik Harpestrengsvej 6A, 2100, Copenhagen, Denmark.
| | - Cristina Subiran Adrados
- Laboratory of Reproductive Biology, section 5701, University Hospital of Copenhagen, Rigshospitalet, Henrik Harpestrengsvej 6A, 2100, Copenhagen, Denmark
| | - Ajay Kumar
- Ansh Labs LLC, 445 W. Medical Center Blvd, Webster, TX, 77598, USA
| | - Bhanu Kalra
- Ansh Labs LLC, 445 W. Medical Center Blvd, Webster, TX, 77598, USA
| | - Linn Salto Mamsen
- Laboratory of Reproductive Biology, section 5701, University Hospital of Copenhagen, Rigshospitalet, Henrik Harpestrengsvej 6A, 2100, Copenhagen, Denmark
| | - Claus Yding Andersen
- The Fertility Clinic, Copenhagen University Hospital Herlev, Herlev, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Albeitawi S, Bani-Mousa SU, Jarrar B, Aloqaily I, Al-Shlool N, Alsheyab G, Kassab A, Qawasmi B, Awaisheh A. Associations Between Follicular Fluid Biomarkers and IVF/ICSI Outcomes in Normo-Ovulatory Women-A Systematic Review. Biomolecules 2025; 15:443. [PMID: 40149979 PMCID: PMC11940193 DOI: 10.3390/biom15030443] [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] [Received: 01/12/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
(1) Background: The follicular fluid (FF) comprises a large portion of ovarian follicles, and serves as both a communication and growth medium for oocytes, and thus should be representative of the metabolomic status of the follicle. This review aims to explore FF biomarkers as well as their effects on fertilization, oocyte, and embryo development, and later on implantation and maintenance of pregnancy. (2) Methods: This review was registered in the PROSPERO database with the ID: CRD42025633101. We parsed PubMed, Scopus, and Google Scholar for research on the effects of different FF biomarkers on IVF/ICSI outcomes in normo-ovulatory women. Included studies were assessed for risk of bias using the NOS scale. Data were extracted and tabulated by two independent researchers. (3) Results: 22 included articles, with a sample size range of 31 to 414 and a median of 60 participants, contained 61 biomarkers, including proteins, growth factors, steroid and polypeptide hormones, inflammation and oxidative stress markers, amino acids, vitamins, lipids of different types, and miRNAs. Most of the biomarkers studied had significant effects on IVF/ICSI outcomes, and seem to have roles in various cellular pathways responsible for oocyte and embryo growth, implantation, placental formation, and maintenance of pregnancy. The FF metabolome also seems to be interconnected, with its various components influencing the levels and activities of each other through feedback loops. (4) Conclusions: FF biomarkers can be utilized for diagnostic and therapeutic purposes in IVF; however, further studies are required for choosing the most promising ones due to heterogeneity of results. Widespread adoption of LC-MS and miRNA microarrays can help quantify a representative FF metabolome, and we see great potential for in vitro supplementation (IVS) of some FF biomarkers in improving IVF/ICSI outcomes.
Collapse
Affiliation(s)
- Soha Albeitawi
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | | | - Baraa Jarrar
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Ibrahim Aloqaily
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Nour Al-Shlool
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Ghaida Alsheyab
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Ahmad Kassab
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Baha’a Qawasmi
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Abdalrahman Awaisheh
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| |
Collapse
|
3
|
Jo M, Brännström M, Akins JW, Curry TE. New insights into the ovulatory process in the human ovary. Hum Reprod Update 2025; 31:21-47. [PMID: 39331957 PMCID: PMC11696709 DOI: 10.1093/humupd/dmae027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/02/2024] [Indexed: 09/29/2024] Open
Abstract
BACKGROUND Successful ovulation is essential for natural conception and fertility. Defects in the ovulatory process are associated with various conditions of infertility or subfertility in women. However, our understanding of the intra-ovarian biochemical mechanisms underlying this process in women has lagged compared to our understanding of animal models. This has been largely due to the limited availability of human ovarian samples that can be used to examine changes across the ovulatory period and delineate the underlying cellular/molecular mechanisms in women. Despite this challenge, steady progress has been made to improve our knowledge of the ovulatory process in women by: (i) collecting granulosa cells across the IVF interval, (ii) creating a novel approach to collecting follicular cells and tissues across the periovulatory period from normally cycling women, and (iii) developing unique in vitro models to examine the LH surge or hCG administration-induced ovulatory changes in gene expression, the regulatory mechanisms underlying the ovulatory changes, and the specific functions of the ovulatory factors. OBJECTIVE AND RATIONALE The objective of this review is to summarize findings generated using in vivo and in vitro models of human ovulation, with the goal of providing new insights into the mechanisms underlying the ovulatory process in women. SEARCH METHODS This review is based on the authors' own studies and a search of the relevant literature on human ovulation to date using PubMed search terms such as 'human ovulation EGF-signaling', 'human ovulation steroidogenesis', 'human ovulation transcription factor', 'human ovulation prostaglandin', 'human ovulation proteinase', 'human ovulation angiogenesis' 'human ovulation chemokine', 'human ovulatory disorder', 'human granulosa cell culture'. Our approach includes comparing the data from the authors' studies with the existing microarray or RNA-seq datasets generated using ovarian cells obtained throughout the ovulatory period from humans, monkeys, and mice. OUTCOMES Current findings from studies using in vivo and in vitro models demonstrate that the LH surge or hCG administration increases the expression of ovulatory mediators, including EGF-like factors, steroids, transcription factors, prostaglandins, proteolytic systems, and other autocrine and paracrine factors, similar to those observed in other animal models such as rodents, ruminants, and monkeys. However, the specific ovulatory factors induced, their expression pattern, and their regulatory mechanisms vary among different species. These species-specific differences stress the necessity of utilizing human samples to delineate the mechanisms underlying the ovulatory process in women. WIDER IMPLICATIONS The data from human ovulation in vivo and in vitro models have begun to fill the gaps in our understanding of the ovulatory process in women. Further efforts are needed to discover novel ovulatory factors. One approach to address these gaps is to improve existing in vitro models to more closely mimic in vivo ovulatory conditions in humans. This is critically important as the knowledge obtained from these human studies can be translated directly to aid in the diagnosis of ovulation-associated pathological conditions, for the development of more effective treatment to help women with anovulatory infertility or, conversely, to better manage ovulation for contraceptive purposes. REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Misung Jo
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Mats Brännström
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Stockholm IVF-EUGIN, Stockholm, Sweden
| | | | - Thomas E Curry
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
4
|
Shi L, Ying H, Dai Y, Rong Y, Chen J, Zhou F, Wang S, Xu S, Tong X, Zhang S. Upregulated let-7 expression in the follicular fluid of patients with endometriomas leads to dysfunction of granulosa cells through targeting of IGF1R. Hum Reprod 2025; 40:119-137. [PMID: 39521729 DOI: 10.1093/humrep/deae247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/07/2024] [Indexed: 11/16/2024] Open
Abstract
STUDY QUESTION What molecular mechanisms underlie the decline in ovarian reserve as the number and quality of oocytes decrease in patients with ovarian endometriomas (OEM)? SUMMARY ANSWER Elevated expression of the let-7 micro(mi)RNAs in the follicular microenvironment of OEM-affected ovaries targets the expression of type 1 insulin-like growth factor receptor (IGF1R) in granulosa cell (GC) and disrupts their proliferation, steroid hormone secretion levels, adenosine triphosphate (ATP) energy metabolism, and reactive oxygen species (ROS) oxidative stress levels. WHAT IS KNOWN ALREADY Patients with OEM exhibit diminished ovarian reserve, characterized by reduced oocyte quantity and quality. Fibrotic changes in the ovarian tissue surrounding the OEM create a disruptive microenvironment for follicular growth and development. STUDY DESIGN, SIZE, DURATION This is a cross-sectional study aimed to elucidate the molecular mechanisms underlying the impact of OEM on follicular development. Initially, miRNA expression profiles in follicular fluid (FF) samples were sequenced from patients with infertility related to OEM (N = 3) and male factor (MF) infertility (N = 3), with the latter serving as the control group. Differentially expressed miRNAs were validated in additional samples from each group (N = 55 in OEM group and N = 45 in MF group) to confirm candidate miRNAs. The study also investigated indicators associated with GCs dysfunction in vitro on rat GCs. Subsequently, rat models of OEM were established through endometrial allogeneic transplantation, and fertility experiments were conducted to assess the let-7/IGF1R axis response to OEM in vivo. Patient samples were collected between May 2018 and April 2019, and the mechanistic study was conducted over the subsequent three years. PARTICIPANTS/MATERIALS, SETTING, METHODS FF and GC samples were obtained from infertile patients undergoing IVF treatment for OEM and MF related infertility. miRNA expression profiles in FF samples were analyzed using second-generation high-throughput sequencing technology, and candidate miRNAs were validated through quantitative PCR (qPCR). In the in vitro experiments conducted with rat GCs, cell proliferation was assessed using the CCK-8 assay, while steroid hormone concentrations were measured using chemiluminescence. ATP content was determined with an ATP assay kit, and levels of ROS were quantified using flow cytometry. A dual luciferase reporter gene assay was employed to identify the target gene of let-7 based on the construction of a IGF1R reporter gene plasmid using 293T cells. Western blotting was utilized to evaluate the expression of IGF1R in GCs, as well as its downstream proteins, and changes in signaling pathways following let-7 agomir/antagomir transfection and/or Igf1r silencing. In the in vivo OEM rat models, alterations in ovarian structure and cyst morphology were observed using hematoxylin and eosin staining. The expressions of let-7 and Igf1r in GCs were evaluated through qPCR, while variations in IGF1R expression were investigated with immunohistochemistry. MAIN RESULTS AND THE ROLE OF CHANCE The cohort of patients with ovarian OEM in this study exhibited significantly decreased antral follicle counts, oocyte retrieval numbers, and normal fertilization rates compared to the control group with MF. The expression of the let-7 miRNA family was markedly upregulated in the FF and GCs of OEM patients. Transfection of rat GCs with let-7 agonists diminished the functions of GCs, including disrupted cell proliferation, mitochondrial oxidative phosphorylation, and steroid hormone secretion, while transfection of rat GCs with let-7 antagonists caused the opposite effects. Luciferase reporter gene experiments confirmed that let-7 complementarily bound to the 3'-untranslated regions of IGF1R. Stimulation of let-7 expression in rat GCs led to a significant decrease in IGF1R expression, while inhibition of let-7 increased IGF1R expression. The expression of IGF1R in the GCs of OEM patients was also significantly reduced compared to MF patients. Silencing of Igf1r led to the dysfunction of GCs, similar to the effects of let-7 agonization, as demonstrated by the downregulation of key proteins involved in cell proliferation (CCND2 and CCND3) and oestradiol synthesis, as well as an increase in progesterone synthesis (StAR), while implicating the PI3K-Akt and MAPK signaling pathways. The antagonistic effect of let-7 on GCs was ineffective when Igf1r was silenced. Conversely, the agonistic effect of let-7 on GCs could be reversed by stimulation with the IGF1R ligand IGF-1. These findings suggested that let-7 regulated the proliferation, differentiation, and ATP synthesis of GCs through targeting IGF1R. The OEM rat model demonstrated alterations in ovarian morphology and structure, along with reduced fertility. Let-7 expression was significantly upregulated in GCs of OEM rats compared to normal rats, while Igf1r and IGF1R expression in pre-ovulatory follicular GCs were notably downregulated, supporting the notion that elevated let-7 expression in the follicular microenvironment of OEM inhibited IGF1R, leading to abnormal GC function and impacting fertility at the molecular level. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION The synthesis and secretion mechanisms of steroid hormones are intricate and complex. Some enzymes that regulate oestrogen synthesis also play a role in progesterone synthesis. Moreover, certain receptors can respond to multiple hormone signals. Therefore, in this study, the expression patterns of key enzymes such as CYP17A, CYP11A1, HSD3B2, StAR, and receptors including AR, LHCGR, FSHR, ESR2, might be influenced by various factors and might not demonstrate complete consistency. WIDER IMPLICATIONS OF THE FINDINGS Future research will concentrate on investigating the potential impact of ovarian stromal cells on the external microenvironment of follicle growth. Additionally, screening for small molecule drugs that target let-7 and IGF1R actions can be conducted to intervene and modify the ovarian microenvironment, ultimately enhancing ovarian function. STUDY FUNDING/COMPETING INTEREST(S) This study received funding from the National Natural Science Foundation of China (grant number 82301851 to L.B.S., grant numbers U23A20403 and U20A20349 to S.Y.Z., and grant number 82371637 to Y.D.D.) and the Natural Science Foundation of Zhejiang Province (grant LTGY23H040010 to F.Z.). The authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
- Libing Shi
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Hanqi Ying
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Yongdong Dai
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Yan Rong
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Jianmin Chen
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Feng Zhou
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Shasha Wang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Shiqian Xu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Xiaomei Tong
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| |
Collapse
|
5
|
Chaves MA, Ferst JG, Fiorenza MF, Vit FF, da Silveira JC. The Influence of Ovarian-Derived Extracellular Vesicles in Reproduction. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2025. [PMID: 39741214 DOI: 10.1007/102_2024_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
In this chapter, we explore the multifaceted roles of extracellular vesicles (EVs) in ovarian biology, focusing on their contributions to folliculogenesis, oocyte competence, corpus luteum function, and immune response regulation. EVs, particularly those derived from follicular fluid (ffEVs), are crucial mediators of cell-to-cell communication within the ovarian follicle, influencing processes such as meiotic progression, stress response, and hormonal regulation. We review preexisting literature, highlighting key findings on the molecular cargo of EVs, such as miRNAs and proteins, and their involvement in regulating the function of the follicle cells. Additionally, the influence of EVs on the immune responses within the ovary was also addressed. Some attention is given to the potential of EVs as non-invasive biomarkers and therapeutic tools, particularly in addressing conditions like premature ovarian insufficiency and polycystic ovary syndrome. By discussing the existing challenges and emerging research, we hope that this chapter will provide a deeper understanding of EVs' therapeutic potential and offer insights or suggestions for advancing assisted reproductive technologies.
Collapse
Affiliation(s)
- Matheus A Chaves
- Laboratory of Molecular Morphophysiology and Development, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | - Juliana G Ferst
- Laboratory of Molecular Morphophysiology and Development, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | - Mariani F Fiorenza
- Laboratory of Molecular Morphophysiology and Development, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | - Franciele F Vit
- Laboratory of Molecular Morphophysiology and Development, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | - Juliano C da Silveira
- Laboratory of Molecular Morphophysiology and Development, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
6
|
Shaji A, Kumaresan A, Sinha MK, Nag P, Patil S, Jeyakumar S, Gowdar Veerappa V, Manimaran A, Ramesha K. Identification of potential differences in salivary proteomic profiles between estrus and diestrus stage of estrous cycle in dairy cows. Syst Biol Reprod Med 2024; 70:204-217. [PMID: 39008339 DOI: 10.1080/19396368.2024.2370328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 06/15/2024] [Indexed: 07/16/2024]
Abstract
In the present study, a comparative global high-throughput proteomic analysis strategy was used to identify proteomic differences between estrus and diestrus stage of estrous cycle in dairy cows. Saliva was collected from cows during estrus and diestrus, and subjected to LC-MS/MS-based proteomic analysis. A total of 2842 proteins were detected in the saliva of cows, out of which, 2437 and 1428 non-redundant proteins were identified in estrous and diestrous saliva, respectively. Further, it was found that 1414 and 405 salivary proteins were specific to estrus and diestrus, respectively while 1023 proteins were common to both groups. Among the significantly dysregulated proteins, the expression of 56 proteins was down-regulated (abundance ratio <0.5) while 40 proteins were up-regulated (abundance ratio > 2) in estrous compared to diestrous saliva. The proteins, such as HSD17B12, INHBA, HSP70, ENO1, SRD5A1, MOS, AMH, ECE2, PDGFA, OPRK1, SYN1, CCNC, PLIN5, CETN1, AKR1C4, NMNAT1, CYP2E1, and CYP19A1 were detected only in the saliva samples derived from estrous cows. Considerable number of proteins detected in the saliva of estrous cows were found to be involved in metabolic pathway, PI3K-Akt signaling pathway, toll-like receptor signaling pathway, steroid biosynthesis pathway, insulin signaling pathway, calcium signaling pathway, estrogen signaling pathway, oxytocin signaling pathway, TGF-β signaling pathway and oocyte meiosis. On the other hand, proteins detected in saliva of diestrous cows were involved mainly in metabolic pathway. Collectively, these data provide preliminary evidence of a potential difference in salivary proteins at different stages of estrous cycle in dairy cows.
Collapse
Affiliation(s)
- Arsha Shaji
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Arumugam Kumaresan
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Manish Kumar Sinha
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Pradeep Nag
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Shivanagouda Patil
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Sakthivel Jeyakumar
- Dairy Production Section, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Vedamurthy Gowdar Veerappa
- Dairy Production Section, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Ayyasamy Manimaran
- Dairy Production Section, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Kerekoppa Ramesha
- Dairy Production Section, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| |
Collapse
|
7
|
Couty N, Estienne A, Le Lay S, Rame C, Chevaleyre C, Allard-Vannier E, Péchoux C, Guerif F, Vasseur C, Aboulouard S, Salzet M, Dupont J, Froment P. Human ovarian extracellular vesicles proteome from polycystic ovary syndrome patients associate with follicular development alterations. FASEB J 2024; 38:e70113. [PMID: 39436214 DOI: 10.1096/fj.202400521rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 09/18/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024]
Abstract
The development of the ovarian follicle requires the presence of several factors that come from the blood and follicular cells. Among these factors, extracellular vesicles (EVs) represent an original communication pathway inside the ovarian follicle. Recently, EVs have been shown to play potential roles in follicular development and reproduction-related disorders, including the polycystic ovary syndrome (PCOS). The proteomic analysis of sEVs isolated from FF in comparison to sEVs purified from plasma has shown a specific pattern of proteins secreted by ovarian steroidogenic cells such as granulosa cells. Thus, a human granulosa cell line exposed to sEVs from FF of normal patients increased their progesterone, estradiol, and testosterone secretion. However, if the sEVs were derived from FF of PCOS patients, the activity of stimulating progesterone production was lost. Stimulation of steroidogenesis by sEVs was associated with an increase in the expression of the StAR gene. In addition, sEVs from FF increased cell proliferation and migration of granulosa cells, and this phenomenon was amplified if sEVs were derived from PCOS patients. Interestingly, STAT3 is a protein overexpressed in sEVs from PCOS patients interacting with most of the cluster of proteins involved in the phenotype observed (cell proliferation, migration, and steroid production) in granulosa cells. In conclusion, this study has demonstrated that sEVs derived from FF could regulate directly the granulosa cell activity. The protein content in sEVs from FF is different in the case of PCOS syndrome and could perturb the granulosa cell functions, including inflammation, steroidogenesis, and cytoskeleton architecture.
Collapse
Affiliation(s)
- Noemie Couty
- INRAE, CNRS, Université de Tours, PRC, Nouzilly, France
| | | | - Soazig Le Lay
- L'Institut du Thorax, CNRS, INSERM, Nantes Université, Nantes, France
- Université Angers, SFR ICAT, Angers, France
| | | | | | | | - Christine Péchoux
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | | | - Claudine Vasseur
- Centre de fertilité, Pôle Santé Léonard de Vinci, Chambray-lès-Tours, France
| | - Soulaimane Aboulouard
- INSERM U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse (PRISM), Université Lille 1, Villeneuve d'Ascq, France
| | - Michel Salzet
- INSERM U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse (PRISM), Université Lille 1, Villeneuve d'Ascq, France
| | - Joelle Dupont
- INRAE, CNRS, Université de Tours, PRC, Nouzilly, France
| | | |
Collapse
|
8
|
Zheng M, Poulsen LC, Wang NF, Mamsen LS, Johannsen ML, Styrishave B, Grøndahl ML, Løssl K, Englund ALM, Skouby SO, Andersen CY. Progesterone and 17-hydroxy-progesterone concentrations in follicular fluid and serum reflect their production in granulosa and theca cells. Reprod Biomed Online 2024; 49:103853. [PMID: 38865783 DOI: 10.1016/j.rbmo.2024.103853] [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] [Received: 11/02/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 06/14/2024]
Abstract
RESEARCH QUESTION How is the production of progesterone (P4) and 17-hydroxy-P4 (17-OH-P4) regulated between theca cells and granulosa cells during the follicular phase, during ovulation and after transformation into a corpus luteum? DESIGN Three cohorts were examined: (i) 31 women undergoing natural and stimulated cycles, with serum hormone measurements taken every 3 days; (ii) 50 women undergoing ovarian stimulation, with hormone concentrations in serum and follicular fluid assessed at five time points during final follicle maturation; and (iii) 12 women undergoing fertility preservation, with hormone concentrations evaluated via the follicular fluid of small antral follicles. RESULTS In the early follicular phase, theca cells primarily synthesized 17-OH-P4 while granulosa cells produced limited P4, maintaining the P4:17-OH-P4 ratio <1. As follicles reached follicle selection at a diameter of approximately 10 mm, P4 synthesis in granulosa cells was up-regulated, but P4 was mainly accumulated in follicular fluid. During final maturation, enhanced activity of the enzyme HSD3B2 in granulosa cells enhanced P4 production, with the P4:17-OH-P4 ratio increasing to >1. The concentration of 17-OH-P4 in the luteal phase was similar to that in the follicular phase, but P4 production increased in the luteal phase, yielding a P4:17-OH-P4 ratio significantly >1. CONCLUSIONS The P4:17-OH-P4 ratio reflects the activity of granulosa cells and theca cells during the follicular phase and following luteinization in the corpus luteum. Managing the function of granulosa cells is key for reducing the concentration of P4 during ovarian stimulation, but the concerted action of FSH and LH on granulosa cells during the second half of the follicular phase makes this complex.
Collapse
Affiliation(s)
- M Zheng
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark.
| | - L C Poulsen
- The Fertility Clinic, Herlev Hospital, Herlev, Denmark
| | - N F Wang
- The Fertility Clinic, Copenhagen University Hospital, Copenhagen, Denmark
| | - L S Mamsen
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Copenhagen, Denmark
| | - M L Johannsen
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Copenhagen, Denmark; Toxicology and Drug Metabolism Group, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - B Styrishave
- Toxicology and Drug Metabolism Group, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - M L Grøndahl
- The Fertility Clinic, Herlev Hospital, Herlev, Denmark
| | - K Løssl
- The Fertility Clinic, Copenhagen University Hospital, Copenhagen, Denmark
| | - A L M Englund
- The Fertility Clinic, Zealand University Hospital, Køge, Denmark
| | - S O Skouby
- The Fertility Clinic, Herlev Hospital, Herlev, Denmark
| | - C Y Andersen
- Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark; The Fertility Clinic, Herlev Hospital, Herlev, Denmark.
| |
Collapse
|
9
|
Mousaei Ghasroldasht M, Liakath Ali F, Park HS, Hadizadeh M, Weng SHS, Huff A, Vafaei S, Al-Hendy A. A Comparative Analysis of Naïve Exosomes and Enhanced Exosomes with a Focus on the Treatment Potential in Ovarian Disorders. J Pers Med 2024; 14:482. [PMID: 38793064 PMCID: PMC11122298 DOI: 10.3390/jpm14050482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/17/2024] [Accepted: 04/27/2024] [Indexed: 05/26/2024] Open
Abstract
Exosome-based therapy has emerged as a promising strategy for addressing diverse disorders, indicating the need for further exploration of the potential therapeutic effects of the exosome cargos. This study introduces "enhanced exosomes", a novel type of exosomes developed through a novel cell culture system. These specific exosomes may become potent therapeutic agents for treating ovarian disorders. In this study, we conducted a comparative analysis of the protein and miRNA cargo compositions of enhanced exosomes and naïve exosomes. Our findings revealed distinct cargo compositions in enhanced exosomes, featuring upregulated proteins such as EFEMP1, HtrA1, PAM, and SDF4, suggesting their potential for treating ovarian disorders. MicroRNA profiling revealed that miR-1-3p, miR-103a-3p, miR-122-5p, miR-1271-5p, miR-133a-3p, miR-184, miR-203a-3p, and miR-206 are key players in regulating ovarian cancer and chemosensitivity by affecting cell cycle progression, cell proliferation, and cell development. We examined polycystic ovary syndrome and premature ovarian insufficiency and identified the altered expression of various miRNAs, such as miR-125b-5p and miR-130b-3p, for diagnostic insights. This study highlights the potential of enhanced exosomes as new therapeutic agents for women's reproductive health, offering a detailed understanding of the impact of their cargo on ovarian disorders.
Collapse
Affiliation(s)
- Mohammad Mousaei Ghasroldasht
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (M.M.G.); (F.L.A.); (H.-S.P.); (S.V.)
| | - Farzana Liakath Ali
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (M.M.G.); (F.L.A.); (H.-S.P.); (S.V.)
| | - Hang-Soo Park
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (M.M.G.); (F.L.A.); (H.-S.P.); (S.V.)
- Department of Biomedical Science, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Morteza Hadizadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 76198-13159, Iran
| | - Shao Huan Samuel Weng
- Proteomics Platform, Office of Shared Research Facilities, Biological Sciences Division, University of Chicago, Chicago, IL 60637, USA; (S.H.S.W.); (A.H.)
| | - Allen Huff
- Proteomics Platform, Office of Shared Research Facilities, Biological Sciences Division, University of Chicago, Chicago, IL 60637, USA; (S.H.S.W.); (A.H.)
| | - Somayeh Vafaei
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (M.M.G.); (F.L.A.); (H.-S.P.); (S.V.)
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (M.M.G.); (F.L.A.); (H.-S.P.); (S.V.)
| |
Collapse
|
10
|
Johannsen ML, Poulsen LC, Mamsen LS, Grøndahl ML, Englund ALM, Lauritsen NL, Carstensen EC, Styrishave B, Yding Andersen C. The intrafollicular concentrations of biologically active cortisol in women rise abruptly shortly before ovulation and follicular rupture. Hum Reprod 2024; 39:578-585. [PMID: 38268234 DOI: 10.1093/humrep/deae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/19/2023] [Indexed: 01/26/2024] Open
Abstract
STUDY QUESTION What is the temporal activity and the concentration in follicular fluid (FF) of the anti-inflammatory steroid cortisol during the ovulatory process in humans? SUMMARY ANSWER Intrafollicular concentrations of cortisol become massively upregulated close to ovulation concomitant with an exceptionally high biological activity securing a timely and efficient termination of inflammatory processes. WHAT IS KNOWN ALREADY Ovulation has been described as a local, controlled inflammatory process resulting in the degeneration of the follicle wall which facilitate oocyte extrusion. Ovulation also affects the glucocorticoid metabolism of granulosa cells (GCs) and although de novo synthesis of cortisol only occurs in the adrenal cortex, the mid-cycle surge has been shown to induce a change from high expression of HSD11B2, inactivating cortisol to cortisone, to high expression of HSD11B1 which reversibly catalyses cortisol production from cortisone. Furthermore, high concentrations of progesterone and 17OH-progesterone within follicles may cause dislodging of cortisol from cortisol binding protein (CBP) thereby activating the biological activity of cortisol. STUDY DESIGN, SIZE, DURATION This prospective cohort study included 50 women undergoing fertility treatment according to a standard antagonist protocol at a university hospital-affiliated fertility clinic in Denmark. PARTICIPANTS/MATERIALS, SETTING, METHODS Women donated FF and GCs from one follicle for research purpose aspirated at one of four time points during the process of final maturation of follicles: T = 0 h, T = 12 h, T = 17 h, T = 32 h. A second sample was collected at oocyte pick up at T = 36 h. The concentration of cortisol and cortisone together with a range of sex steroids was measured by LC-MS/MS in FF collected at the five time points mentioned above. Whole genome microarray data, validated by q-PCR analysis, was used to evaluate gene expression of CYP11B1, CYP21A2, HSD11B1, HSD11B2, and NR3C1 in GCs at the same time points. MAIN RESULTS AND THE ROLE OF CHANCE The concentration of cortisol was significantly increased from a few nM at 0 h to around 100-140 nM (P ≤ 0.0001) at 32-36 h, whilst cortisone was almost constant from 0 to 17 h at a concentration of between 90 and 100 nM being significantly reduced to 25-40 nM (P ≤ 0.0001) at 32-36 h. This was paralleled by a 690-fold upregulation of HSD11B1 from 0 to 12 h increasing to a more than 20.000-fold change at 36 h. HSD11B2 was quickly downregulated 15- to 20-fold after ovulation induction. Concentrations of progesterone and 17OH-progesterone increased during the ovulatory process to high levels which in essence displaces cortisol from its binding protein CBP due to similar binding affinities. Furthermore, a significant decrease in 11-deoxycortisol expression was seen, but CYP11B1 expression was below detection limit in GCs. LIMITATIONS, REASONS FOR CAUTION The study included women undergoing ovarian stimulation and results may differ from the natural cycle. More observations at each specific time point may have strengthened the conclusions. Furthermore, we have not been able to measure the actual active biological concentration of cortisol. WIDER IMPLICATIONS OF THE FINDINGS For the first time, this study collectively evaluated the temporal pattern of cortisol and cortisone concentrations during human ovulation, rendering a physiological framework for understanding potential dysregulations in the inflammatory reaction of ovulation. STUDY FUNDING/COMPETING INTEREST(S) This research was supported by the University Hospital of Copenhagen, Rigshospitalet, and Novo Nordisk Foundation grant number NNF21OC00700556. Interreg V ÔKS through ReproUnion (www.reprounion.eu); Region Zealand Research Foundation. The funders had no role in study design, collection of data, analyses, writing of the article, or the decision to submit it for publication. The authors have no conflicts of interest to declare. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- M L Johannsen
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Copenhagen Ø, Denmark
- Toxicolgy and Drug Metabolism group, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Ø, Denmark
| | - L C Poulsen
- Fertility Clinic, University Hospital of Copenhagen, Herlev and Gentofte Hospital, Herlev, Denmark
| | - L S Mamsen
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Copenhagen Ø, Denmark
| | - M L Grøndahl
- Fertility Clinic, University Hospital of Copenhagen, Herlev and Gentofte Hospital, Herlev, Denmark
| | - A L M Englund
- Fertility Clinic, Zealand University Hospital, Køge, Denmark
| | - N L Lauritsen
- Toxicolgy and Drug Metabolism group, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Ø, Denmark
| | - E C Carstensen
- Toxicolgy and Drug Metabolism group, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Ø, Denmark
| | - B Styrishave
- Toxicolgy and Drug Metabolism group, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Ø, Denmark
| | - C Yding Andersen
- Faculty of Health and Medical Science, Institute for Clinical Medicine, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
11
|
Cadenas J, Pors SE, Hansen CP, Olufsen SM, Subiran C, Bøtkjær JA, La Cour Poulsen L, Fedder J, Dueholm M, Colmorn LB, Kristensen SG, Mamsen LS, Andersen CY. Midkine characterization in human ovaries: potential new variants in follicles. F&S SCIENCE 2023; 4:294-301. [PMID: 37739342 DOI: 10.1016/j.xfss.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/15/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
OBJECTIVE To characterize the growth factor midkine (MDK) in the human ovary to determine whether MDK is produced locally within the ovary, examine whether different ovarian cell types are more likely to produce MDK, and determine whether there are any stage-specific variations during follicle growth. Previous studies have revealed that MDK potentially affects human follicle growth and oocyte maturation. Proteomic analyses in follicular fluid (FF) have identified MDK to functionally cluster together and follow a similar expression profile to that of well-known proteins involved in ovarian follicle development. Midkine has not yet been characterized in the human ovary. DESIGN Descriptive study. SETTING University Hospital. PATIENTS The study included samples from 121 patients: 71 patients (aged 17-37 years) who underwent ovarian tissue cryopreservation provided granulosa cells (GC), cumulus cells, ovarian cortex, medulla tissue, and FF from small antral follicles (SAF); and 50 patients (aged 20-35 years) receiving in vitro fertilization treatment provided FF from preovulatory follicles before and after induction of final follicle maturation. INTERVENTIONS None. MAIN OUTCOME MEASURES MDK relative gene expression was quantified using a real-time quantitative polymerase chain reaction in cumulus cells, GC, and medulla tissue. Additionally, immunostaining and western blotting assays were used to detect MDK protein in the ovarian cortex, which contains preantral follicles, SAF, and medulla tissue. Furthermore, enzyme-linked immunosorbent assay analyses were performed to measure the concentration of MDK in FF aspirated from SAF and preovulatory follicles both before and 36 hours after inducing the final maturation of follicles. RESULTS Immunostaining and reverse transcription-quantitative polymerase chain reaction revealed a more prominent expression of MDK in GC compared with other ovarian cell types. Intrafollicular MDK concentration was significantly higher in SAF compared with preovulatory follicles. In addition, different molecular weight species of MDK were detected using western blotting in various ovarian sample types: GC and FF samples presented primarily one band of approximately 15 kDa and an additional band of approximately 13 kDa, although other bands with higher molecular weight (between 30 and 38 kDa) were detected in medulla tissue. CONCLUSIONS This is the first time that MDK has been immunolocalized in human ovarian cells at the protein level and that potentially different MDK variants have been detected in human FF, GC, and ovarian medulla tissue. Future studies are needed to sequence and identify the different potential MDK variants found to determine their functional importance for ovary and oocyte competence.
Collapse
Affiliation(s)
- Jesús Cadenas
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark.
| | - Susanne Elisabeth Pors
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | | | - Sarah Maria Olufsen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Cristina Subiran
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Jane Alrø Bøtkjær
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Liv La Cour Poulsen
- The Fertility Clinic, Copenhagen University Hospital, Herlev Hospital, Borgmester Ib Juuls Vej 9, Herlev, Denmark
| | - Jens Fedder
- Centre of Andrology and Fertility Clinic, Odense University Hospital, Odense, Denmark; Research Unit of Human Reproduction, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Margit Dueholm
- Department of Gynecology and Obstetrics, Aarhus University Hospital, Aarhus, Denmark
| | | | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Linn Salto Mamsen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Claus Yding Andersen
- The Department of Clinical Medicine, Faculty of Health and Medical Science, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
12
|
Manríquez-Treviño Y, Sánchez-Ramírez B, Grado-Ahuir JA, Castro-Valenzuela B, González-Horta C, Burrola-Barraza M. Human TIMP1 Is a Growth Factor That Improves Oocyte Developmental Competence. BIOTECH 2023; 12:60. [PMID: 37873882 PMCID: PMC10594479 DOI: 10.3390/biotech12040060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/24/2023] [Accepted: 10/05/2023] [Indexed: 10/25/2023] Open
Abstract
Oocyte developmental competence is the ability of a mature oocyte to be fertilized and subsequently support embryonic development. Such competence is gained during folliculogenesis and is facilitated by the bidirectional communication into a compacted cumulus-oocyte complex (COC). Human tissue inhibitor of metalloproteinases-1 (TIMP1) participates in biological processes, including cell growth, differentiation, and apoptosis. This study aimed to evaluate the influence of TIMP1 as a growth factor on the in vitro maturation (IVM) culture of bovine COCs to improve oocyte developmental competence. All TIMP1 treatments (50, 100, and 150 ng/mL) favored the COCs' compaction structure (p < 0.05). TIMP1 at 150 ng/mL produced more oocytes in metaphase II compared to the other treatments (p < 0.05). The 150 ng/mL TIMP1 generated oocytes with the most (p < 0.05) cortical granules below the plasma membrane (pattern I). In a parthenogenesis assay, oocyte IVM in 50 ng/mL of TIMP1 produced the most blastocyst compared to the other treatments (p < 0.05). The Principal Component Analysis (PCA) showed that 50 ng/mL of TIMP1 was the best condition to develop oocyte competence because it was associated with the COC compact and cortical granule pattern I. TIMP1 influences the development of oocyte competence when added to the IVM culture medium of COCs.
Collapse
Affiliation(s)
- Yolanda Manríquez-Treviño
- Facultad de Zootecnia y Ecología, Universidad Autónoma de Chihuahua (UACH), Perif. Fco. R. Almada Km. 1, Chihuahua 31453, Chihuahua, Mexico; (Y.M.-T.); (J.A.G.-A.); (B.C.-V.)
| | - Blanca Sánchez-Ramírez
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua (UACH), Campus Universitario #2, Chihuahua 31125, Chihuahua, Mexico; (B.S.-R.); (C.G.-H.)
| | - Juan Alberto Grado-Ahuir
- Facultad de Zootecnia y Ecología, Universidad Autónoma de Chihuahua (UACH), Perif. Fco. R. Almada Km. 1, Chihuahua 31453, Chihuahua, Mexico; (Y.M.-T.); (J.A.G.-A.); (B.C.-V.)
| | - Beatriz Castro-Valenzuela
- Facultad de Zootecnia y Ecología, Universidad Autónoma de Chihuahua (UACH), Perif. Fco. R. Almada Km. 1, Chihuahua 31453, Chihuahua, Mexico; (Y.M.-T.); (J.A.G.-A.); (B.C.-V.)
| | - Carmen González-Horta
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua (UACH), Campus Universitario #2, Chihuahua 31125, Chihuahua, Mexico; (B.S.-R.); (C.G.-H.)
| | - M.Eduviges Burrola-Barraza
- Facultad de Zootecnia y Ecología, Universidad Autónoma de Chihuahua (UACH), Perif. Fco. R. Almada Km. 1, Chihuahua 31453, Chihuahua, Mexico; (Y.M.-T.); (J.A.G.-A.); (B.C.-V.)
| |
Collapse
|
13
|
Kanaka V, Drakakis P, Loutradis D, Tsangaris GT. Proteomics in the study of female fertility: an update. Expert Rev Proteomics 2023; 20:319-330. [PMID: 37874610 DOI: 10.1080/14789450.2023.2275683] [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] [Received: 03/09/2023] [Accepted: 10/20/2023] [Indexed: 10/25/2023]
Abstract
INTRODUCTION Female fertility has been a field of interest for the scientific community throughout the years. The contribution of proteomics in the study of female fertility as well as female infertility and in vitro fertilization (IVF) has been significant. Proteomics is a recently developed field, extensively applied to the identification and quantification of proteins, which could be used as potential biomarkers in a diagnostic, prognostic, or predictive manner in a variety of medical conditions. AREAS COVERED The present review focuses on proteomic studies of the oocyte and endometrial environment as well as on conditions related to infertility, such as polycystic ovarian syndrome, endometriosis, obesity, and unexplained infertility. Moreover, this review presents studies that have been done in an effort to search for fertility biomarkers in individuals following the IVF procedure. EXPERT OPINION The comprehension of the molecular pathways behind female fertility and infertility could contribute to the diagnosis, prognosis, and prediction of infertility. Moreover, the identification of proteomic biomarkers for IVF cycles could predict the possible outcome of an IVF cycle, prevent an unsuccessful IVF, and monitor the IVF cycle in a personalized manner, leading to increased success rates. [Figure: see text].
Collapse
Affiliation(s)
- Vasiliki Kanaka
- First Department of Obstetrics and Gynecology, School of Medicine, National and Kapodistrian University of Athens, Alexandra Hospital, Athens, Greece
- Proteomics Research Unit, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Petros Drakakis
- Third Department of Obstetrics and Gynecology, School of Medicine, National and Kapodistrian University of Athens, Attikon Hospital, Athens, Greece
| | - Dimitrios Loutradis
- First Department of Obstetrics and Gynecology, School of Medicine, National and Kapodistrian University of Athens, Alexandra Hospital, Athens, Greece
| | - George Th Tsangaris
- Proteomics Research Unit, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
14
|
Babu A, Ramanathan G. Multi-omics insights and therapeutic implications in polycystic ovary syndrome: a review. Funct Integr Genomics 2023; 23:130. [PMID: 37079114 DOI: 10.1007/s10142-023-01053-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/04/2023] [Accepted: 04/08/2023] [Indexed: 04/21/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a common gynecological disease that causes adverse effects in women in their reproductive phase. Nonetheless, the molecular mechanisms remain unclear. Over the last decade, sequencing and omics approaches have advanced at an increased pace. Omics initiatives have come to the forefront of biomedical research by presenting the significance of biological functions and processes. Thus, multi-omics profiling has yielded important insights into understanding the biology of PCOS by identifying potential biomarkers and therapeutic targets. Multi-omics platforms provide high-throughput data to leverage the molecular mechanisms and pathways involving genetic alteration, epigenetic regulation, transcriptional regulation, protein interaction, and metabolic alterations in PCOS. The purpose of this review is to outline the prospects of multi-omics technologies in PCOS research by revealing novel biomarkers and therapeutic targets. Finally, we address the knowledge gaps and emerging treatment strategies for the management of PCOS. Future PCOS research in multi-omics at the single-cell level may enhance diagnostic and treatment options.
Collapse
Affiliation(s)
- Achsha Babu
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Gnanasambandan Ramanathan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
15
|
Cadenas J, Poulsen LC, Nikiforov D, Grøndahl ML, Kumar A, Bahnu K, Englund ALM, Malm J, Marko-Varga G, Pla I, Sanchez A, Pors SE, Andersen CY. Regulation of human oocyte maturation in vivo during the final maturation of follicles. Hum Reprod 2023; 38:686-700. [PMID: 36762771 DOI: 10.1093/humrep/dead024] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/03/2023] [Indexed: 02/11/2023] Open
Abstract
STUDY QUESTION Which substances and signal transduction pathways are potentially active downstream to the effect of FSH and LH in the regulation of human oocyte maturation in vivo? SUMMARY ANSWER The regulation of human oocyte maturation appears to be a multifactorial process in which several different signal transduction pathways are active. WHAT IS KNOWN ALREADY Many studies in animal species have provided insight into the mechanisms that govern the final maturation of oocytes. Currently, these studies have identified several different mechanisms downstream to the effects of FSH and LH. Some of the identified mechanisms include the regulation of cAMP/cGMP levels in oocytes involving C-type natriuretic peptide (CNP), effects of epidermal growth factor (EGF)-related peptides such as amphiregulin (AREG) and/or epiregulin (EREG), effect of TGF-β family members including growth differentiation factor 9 (GDF9) and morphogenetic protein 15 (BMP15), activins/inhibins, follicular fluid meiosis activating sterol (FF-MAS), the growth factor midkine (MDK), and several others. However, to what extent these pathways and mechanisms are active in humans in vivo is unknown. STUDY DESIGN, SIZE, DURATION This prospective cohort study included 50 women undergoing fertility treatment in a standard antagonist protocol at a university hospital affiliated fertility clinic in 2016-2018. PARTICIPANTS/MATERIALS, SETTING, METHODS We evaluated the substances and signalling pathways potentially affecting human oocyte maturation in follicular fluid (FF) and granulosa cells (GCs) collected at five time points during the final maturation of follicles. Using ELISA measurement and proteomic profiling of FF and whole genome gene expression in GC, the following substances and their signal transduction pathways were collectively evaluated: CNP, the EGF family, inhibin-A, inhibin-B, activins, FF-MAS, MDK, GDF9, and BMP15. MAIN RESULTS AND THE ROLE OF CHANCE All the evaluated substances and signal transduction pathways are potentially active in the regulation of human oocyte maturation in vivo except for GDF9/BMP15 signalling. In particular, AREG, inhibins, and MDK were significantly upregulated during the first 12-17 h after initiating the final maturation of follicles and were measured at significantly higher concentrations than previously reported. Additionally, the genes regulating FF-MAS synthesis and metabolism were significantly controlled in favour of accumulation during the first 12-17 h. In contrast, concentrations of CNP were low and did not change during the process of final maturation of follicles, and concentrations of GDF9 and BMP15 were much lower than reported in small antral follicles, suggesting a less pronounced influence from these substances. LARGE SCALE DATA None. LIMITATIONS, REASONS FOR CAUTION Although GC and cumulus cells have many similar features, it is a limitation of the current study that information for the corresponding cumulus cells is not available. However, we seldom recovered a cumulus-oocyte complex during the follicle aspiration from 0 to 32 h. WIDER IMPLICATIONS OF THE FINDINGS Delineating the mechanisms governing the regulation of human oocyte maturation in vivo advances the possibility of developing a platform for IVM that, as for most other mammalian species, results in healthy offspring with good efficacy. Mimicking the intrafollicular conditions during oocyte maturation in vivo in small culture droplets during IVM may enhance oocyte nuclear and cytoplasmic maturation. The primary outlook for such a method is, in the context of fertility preservation, to augment the chances of achieving biological children after a cancer treatment by subjecting oocytes from small antral follicles to IVM. Provided that aspiration of oocytes from small antral follicles in vivo can be developed with good efficacy, IVM may be applied to infertile patients on a larger scale and can provide a cheap alternative to conventional IVF treatment with ovarian stimulation. Successful IVM has the potential to change current established techniques for infertility treatment. STUDY FUNDING/COMPETING INTEREST(S) This research was supported by the University Hospital of Copenhagen, Rigshospitalet, the Independent Research Fund Denmark (grant number 0134-00448), and the Interregional EU-sponsored ReproUnion network. There are no conflicts of interest to be declared.
Collapse
Affiliation(s)
- J Cadenas
- Laboratory of Reproductive Biology, Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - L C Poulsen
- Zealand Fertility Clinic, Zealand University Hospital, Køge, Denmark
| | - D Nikiforov
- Laboratory of Reproductive Biology, Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - M L Grøndahl
- The Fertility Clinic, Copenhagen University Hospital, Herlev Hospital, Herlev, Denmark
| | - A Kumar
- Ansh Labs LLC, Webster, TX, USA
| | - K Bahnu
- Ansh Labs LLC, Webster, TX, USA
| | - A L M Englund
- Zealand Fertility Clinic, Zealand University Hospital, Køge, Denmark
| | - J Malm
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, Malmö, Sweden.,Department of Biomedical Engineering, Clinical Protein Science & Imaging, Biomedical Centre, Lund University, Lund, Sweden
| | - G Marko-Varga
- Department of Biomedical Engineering, Clinical Protein Science & Imaging, Biomedical Centre, Lund University, Lund, Sweden
| | - I Pla
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, Malmö, Sweden.,Department of Biomedical Engineering, Clinical Protein Science & Imaging, Biomedical Centre, Lund University, Lund, Sweden
| | - A Sanchez
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, Malmö, Sweden.,Department of Biomedical Engineering, Clinical Protein Science & Imaging, Biomedical Centre, Lund University, Lund, Sweden
| | - S E Pors
- Laboratory of Reproductive Biology, Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - C Yding Andersen
- Laboratory of Reproductive Biology, Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, Rigshospitalet, Denmark.,Faculty of Health and Medical Science, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
16
|
Future potential of in vitro maturation including fertility preservation. Fertil Steril 2023; 119:550-559. [PMID: 36702341 DOI: 10.1016/j.fertnstert.2023.01.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023]
Abstract
In several mammalian species, oocytes from small antral follicles after in vitro maturation (IVM) are successfully used for procreation. Humans are the exception, mainly because of limited access to immature oocytes and because oocyte maturation is uniquely regulated in women. With the introduction of cryopreservation of the ovarian cortex for fertility preservation, immature oocytes from small antral follicles in the medulla are now available for developing IVM on the basis of actual human studies. This review presents recent findings in favor of developing human IVM, including the oocyte diameter, follicle size from which the immature oocytes are collected, necessary level of follicle-stimulating hormone and luteinizing hormone to accelerate IVM, and secretion of factors from the cumulus-oocyte complex that affect the way oocyte maturation takes place. Furthermore, on the basis of studies in human granulosa cells and follicle fluid collected during the final maturation of follicles in vivo, a number of signal transduction pathways and hormone levels active during physiological conditions have been identified, providing new candidates and ways to improve the current IVM platform. Furthermore, it is suggested that the small droplet of culture medium in which IVM is performed mimics the hormonal milieu within a follicle created by the somatic cells and oocyte in vivo and may be used to advance oocyte nuclear and cytoplasmic maturation. Collectively, we envision that a continued research effort will develop a human IVM platform equally effective as for other mammalian species.
Collapse
|
17
|
Grzeczka A, Graczyk S, Skowronska A, Skowronski MT, Kordowitzki P. Relevance of Vitamin D and Its Deficiency for the Ovarian Follicle and the Oocyte: An Update. Nutrients 2022; 14:nu14183712. [PMID: 36145088 PMCID: PMC9502977 DOI: 10.3390/nu14183712] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/30/2022] [Accepted: 09/07/2022] [Indexed: 11/25/2022] Open
Abstract
For many years, vitamin D (VD) has been known to be an essential micronutrient with important relevance not only for the skeletal system, but also for numerous other mammalian organ systems. Low levels of VD result in a VD deficiency, which is a global health problem. Moreover, VD deficiencies are linked to several pathologies, for instance, diseases of the cardiovascular system, diabetes mellitus, or sub- and infertility. In the past two decades, an increasing body of evidence has shown that adequate physiological levels of VD are crucial for the female gamete and its microenvironment, and VD deficiency has been associated with decreased live birth rates among women undergoing in vitro fertilization (IVF). With regard to the female reproductive tract, VD receptors (VDRs) have been detected in the ovary, endometrium, and the placenta. Although it has been reported that VD seems to be relevant for both calcium-dependent and independent pathways, its relevance for the oocyte’s developmental competence and life span remains elusive. Therefore, herein, we aim to provide an update on the importance of VD and VD deficiency for the oocyte and the follicular microenvironment.
Collapse
Affiliation(s)
- Arkadiusz Grzeczka
- Department of Preclinical and Basic Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Gagarina Street 1, 87-100 Torun, Poland
| | - Szymon Graczyk
- Department of Preclinical and Basic Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Gagarina Street 1, 87-100 Torun, Poland
| | - Agnieszka Skowronska
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum of the University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Mariusz T. Skowronski
- Department of Preclinical and Basic Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Gagarina Street 1, 87-100 Torun, Poland
| | - Paweł Kordowitzki
- Department of Preclinical and Basic Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Gagarina Street 1, 87-100 Torun, Poland
- Correspondence:
| |
Collapse
|
18
|
Kanaka V, Proikakis S, Drakakis P, Loutradis D, Tsangaris GT. Implementing a preimplantation proteomic approach to advance assisted reproduction technologies in the framework of predictive, preventive, and personalized medicine. EPMA J 2022; 13:237-260. [PMID: 35719135 PMCID: PMC9203609 DOI: 10.1007/s13167-022-00282-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/05/2022] [Indexed: 10/28/2022]
Abstract
The evolution of the field of assisted reproduction technology (ART) in the last 40 years has significantly contributed to the management of global infertility. Despite the great numbers of live births that have been achieved through ART, there is still potential for increasing the success rates. As a result, there is a need to create optimum conditions in order to increase ART efficacy. The selection of the best sperm, oocyte, and embryo, as well as the achievement of optimal endometrial receptivity, through the contribution of new diagnostic and treatment methods, based on a personalized proteomic approach, may assist in the attainment of this goal. Proteomics represent a powerful new technological development, which seeks for protein biomarkers in human tissues. These biomarkers may aid to predict the outcome, prevent failure, and monitor in a personalized manner in vitro fertilization (IVF) cycles. In this review, we will present data from studies that have been conducted in the search for such biomarkers in order to identify proteins related to good sperm, oocyte, and embryo quality, as well as optimal endometrial receptivity, which may later lead to greater results and the desirable ART outcome.
Collapse
Affiliation(s)
- Vasiliki Kanaka
- First Department of Obstetrics and Gynecology, School of Medicine, National and Kapodistrian University of Athens, Alexandra Hospital, Athens, Greece
- Proteomics Research Unit, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Stavros Proikakis
- Proteomics Research Unit, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Petros Drakakis
- Third Department of Obstetrics and Gynecology, School of Medicine, National and Kapodistrian University of Athens, Attikon Hospital, Athens, Greece
| | - Dimitrios Loutradis
- First Department of Obstetrics and Gynecology, School of Medicine, National and Kapodistrian University of Athens, Alexandra Hospital, Athens, Greece
| | - George Th. Tsangaris
- Proteomics Research Unit, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| |
Collapse
|
19
|
Proteomic Alterations in Follicular Fluid of Human Small Antral Follicles Collected from Polycystic Ovaries—A Pilot Study. Life (Basel) 2022; 12:life12030391. [PMID: 35330141 PMCID: PMC8954146 DOI: 10.3390/life12030391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/03/2022] [Accepted: 03/05/2022] [Indexed: 11/25/2022] Open
Abstract
Polycystic ovaries (PCO) contain antral follicles that arrest growing around 3–11 mm in diameter, perturbing the dominant follicle’s selection and the subsequent ovulatory process. Proteomic alterations of PCO follicular fluid (FF) (i.e., microenvironment in which the oocyte develops until ovulation) have been studied from large follicles in connection with oocyte pickup during ovarian stimulation. The present study aimed to detect proteomic alterations in FF from unstimulated human small antral follicles (hSAF) obtained from PCO. After performing deep-sequencing label-free proteomics on 10 PCO and 10 non-PCO FF samples from unstimulated hSAF (4.6–9.8 mm), 1436 proteins were identified, of which 115 were dysregulated in PCO FF samples. Pathways and processes related to the immune system, inflammation, and oxidative stress appeared to be upregulated in PCO, while extracellular matrix receptors interactions, the collagens-containing extracellular matrix, and the regulation of signaling were downregulated. The secreted proteins SFRP1, THBS4, and C1QC significantly decreased their expression in PCO FF, and this downregulation was suggested to affect future oocyte competence. In conclusion, our study revealed, for the first time, evidence of proteomic alterations occurring in the FF of PCO hSAF that may be related to the dysfunction of follicular growth and subsequent oocyte competence.
Collapse
|
20
|
Almeida N, Rodriguez J, Pla Parada I, Perez-Riverol Y, Woldmar N, Kim Y, Oskolas H, Betancourt L, Valdés JG, Sahlin KB, Pizzatti L, Szasz AM, Kárpáti S, Appelqvist R, Malm J, B. Domont G, C. S. Nogueira F, Marko-Varga G, Sanchez A. Mapping the Melanoma Plasma Proteome (MPP) Using Single-Shot Proteomics Interfaced with the WiMT Database. Cancers (Basel) 2021; 13:6224. [PMID: 34944842 PMCID: PMC8699267 DOI: 10.3390/cancers13246224] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/30/2021] [Accepted: 12/08/2021] [Indexed: 12/26/2022] Open
Abstract
Plasma analysis by mass spectrometry-based proteomics remains a challenge due to its large dynamic range of 10 orders in magnitude. We created a methodology for protein identification known as Wise MS Transfer (WiMT). Melanoma plasma samples from biobank archives were directly analyzed using simple sample preparation. WiMT is based on MS1 features between several MS runs together with custom protein databases for ID generation. This entails a multi-level dynamic protein database with different immunodepletion strategies by applying single-shot proteomics. The highest number of melanoma plasma proteins from undepleted and unfractionated plasma was reported, mapping >1200 proteins from >10,000 protein sequences with confirmed significance scoring. Of these, more than 660 proteins were annotated by WiMT from the resulting ~5800 protein sequences. We could verify 4000 proteins by MS1t analysis from HeLA extracts. The WiMT platform provided an output in which 12 previously well-known candidate markers were identified. We also identified low-abundant proteins with functions related to (i) cell signaling, (ii) immune system regulators, and (iii) proteins regulating folding, sorting, and degradation, as well as (iv) vesicular transport proteins. WiMT holds the potential for use in large-scale screening studies with simple sample preparation, and can lead to the discovery of novel proteins with key melanoma disease functions.
Collapse
Affiliation(s)
- Natália Almeida
- Laboratory of Proteomics/LADETEC, Universidade Federal Do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil;
- Proteomics Unit, Institute of Chemistry, Universidade Federal Do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil;
- Clinical Protein Science & Imaging, Biomedical Center, Department of Biomedical Engineering, Lund University, BMC D13, 22184 Lund, Sweden; (N.W.); (K.B.S.); (G.M.-V.)
| | - Jimmy Rodriguez
- Division of Chemistry I, Department of Biochemistry and Biophysics, Karolinska Institute, 17165 Stockholm, Sweden;
| | - Indira Pla Parada
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 20502 Malmö, Sweden; (I.P.P.); (J.M.)
| | - Yasset Perez-Riverol
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK;
| | - Nicole Woldmar
- Clinical Protein Science & Imaging, Biomedical Center, Department of Biomedical Engineering, Lund University, BMC D13, 22184 Lund, Sweden; (N.W.); (K.B.S.); (G.M.-V.)
- Laboratory of Molecular Biology and Blood Proteomics—LADETEC, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro 21941-598, Brazil;
| | - Yonghyo Kim
- Data Convergence Drug Research Center, Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea;
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, 22185 Lund, Sweden; (H.O.); (L.B.); (J.G.V.); (R.A.)
| | - Henriett Oskolas
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, 22185 Lund, Sweden; (H.O.); (L.B.); (J.G.V.); (R.A.)
| | - Lazaro Betancourt
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, 22185 Lund, Sweden; (H.O.); (L.B.); (J.G.V.); (R.A.)
| | - Jeovanis Gil Valdés
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, 22185 Lund, Sweden; (H.O.); (L.B.); (J.G.V.); (R.A.)
| | - K. Barbara Sahlin
- Clinical Protein Science & Imaging, Biomedical Center, Department of Biomedical Engineering, Lund University, BMC D13, 22184 Lund, Sweden; (N.W.); (K.B.S.); (G.M.-V.)
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 20502 Malmö, Sweden; (I.P.P.); (J.M.)
| | - Luciana Pizzatti
- Laboratory of Molecular Biology and Blood Proteomics—LADETEC, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro 21941-598, Brazil;
| | | | - Sarolta Kárpáti
- Department of Dermatology, Venereology and Dermatooncology, Semmelweis University, 1085 Budapest, Hungary;
| | - Roger Appelqvist
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, 22185 Lund, Sweden; (H.O.); (L.B.); (J.G.V.); (R.A.)
| | - Johan Malm
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 20502 Malmö, Sweden; (I.P.P.); (J.M.)
| | - Gilberto B. Domont
- Proteomics Unit, Institute of Chemistry, Universidade Federal Do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil;
| | - Fábio C. S. Nogueira
- Laboratory of Proteomics/LADETEC, Universidade Federal Do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil;
- Proteomics Unit, Institute of Chemistry, Universidade Federal Do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil;
| | - György Marko-Varga
- Clinical Protein Science & Imaging, Biomedical Center, Department of Biomedical Engineering, Lund University, BMC D13, 22184 Lund, Sweden; (N.W.); (K.B.S.); (G.M.-V.)
- Chemical Genomics Global Research Lab, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
- Department of Surgery, Tokyo Medical University, 6-7-1 Nishishinjiku Shinjiku-ku, Tokyo 160-0023, Japan
| | - Aniel Sanchez
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 20502 Malmö, Sweden; (I.P.P.); (J.M.)
| |
Collapse
|
21
|
Lewandowska AE, Fel A, Thiel M, Czaplewska P, Łukaszuk K, Wiśniewski JR, Ołdziej S. Compatibility of Distinct Label-Free Proteomic Workflows in Absolute Quantification of Proteins Linked to the Oocyte Quality in Human Follicular Fluid. Int J Mol Sci 2021; 22:7415. [PMID: 34299044 PMCID: PMC8304916 DOI: 10.3390/ijms22147415] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 01/02/2023] Open
Abstract
We present two separate label-free quantitative workflows based on different high-resolution mass spectrometers and LC setups, which are termed after the utilized instrument: Quad-Orbitrap (nano-LC) and Triple Quad-TOF (micro-LC) and their directed adaptation toward the analysis of human follicular fluid proteome. We identified about 1000 proteins in each distinct workflow using various sample preparation methods. With assistance of the Total Protein Approach, we were able to obtain absolute protein concentrations for each workflow. In a pilot study of twenty samples linked to diverse oocyte quality status from four donors, 455 and 215 proteins were quantified by the Quad-Orbitrap and Triple Quad-TOF workflows, respectively. The concentration values obtained from both workflows correlated to a significant degree. We found reasonable agreement of both workflows in protein fold changes between tested groups, resulting in unified lists of 20 and 22 proteins linked to oocyte maturity and blastocyst development, respectively. The Quad-Orbitrap workflow was best suited for an in-depth analysis without the need of extensive fractionation, especially of low abundant proteome, whereas the Triple Quad-TOF workflow allowed a more robust approach with a greater potential to increase in effectiveness with the growing number of analyzed samples after the initial effort of building a comprehensive spectral library.
Collapse
Affiliation(s)
- Aleksandra E. Lewandowska
- Intercollegiate Faculty of Biotechnology UG&MUG, University of Gdańsk, Abrahama 58, 80-307 Gdańsk, Poland; (A.F.); (M.T.); (P.C.)
| | - Anna Fel
- Intercollegiate Faculty of Biotechnology UG&MUG, University of Gdańsk, Abrahama 58, 80-307 Gdańsk, Poland; (A.F.); (M.T.); (P.C.)
| | - Marcel Thiel
- Intercollegiate Faculty of Biotechnology UG&MUG, University of Gdańsk, Abrahama 58, 80-307 Gdańsk, Poland; (A.F.); (M.T.); (P.C.)
| | - Paulina Czaplewska
- Intercollegiate Faculty of Biotechnology UG&MUG, University of Gdańsk, Abrahama 58, 80-307 Gdańsk, Poland; (A.F.); (M.T.); (P.C.)
| | - Krzysztof Łukaszuk
- INVICTA Fertility and Reproductive Center, Polna 64, 81-740 Sopot, Poland;
- Department of Obstetrics and Gynecological Nursing, Faculty of Health Sciences, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Jacek R. Wiśniewski
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany;
| | - Stanisław Ołdziej
- Intercollegiate Faculty of Biotechnology UG&MUG, University of Gdańsk, Abrahama 58, 80-307 Gdańsk, Poland; (A.F.); (M.T.); (P.C.)
| |
Collapse
|
22
|
Dell'Aversana C, Cuomo F, Longobardi S, D'Hooghe T, Caprio F, Franci G, Santonastaso M, Colacurci N, Barone S, Pisaturo V, Valerio D, Altucci L. Age-related miRNome landscape of cumulus oophorus cells during controlled ovarian stimulation protocols in IVF cycles. Hum Reprod 2021; 36:1310-1325. [PMID: 33454781 PMCID: PMC8058597 DOI: 10.1093/humrep/deaa364] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 12/06/2020] [Indexed: 12/26/2022] Open
Abstract
STUDY QUESTION Is the microRNA (miRNA) expression pattern of cumulus oophorus cells (COCs) in women undergoing medically assisted reproduction (MAR) procedures differentially modulated according to patient age and gonadotropin treatment strategy? SUMMARY ANSWER Maternal age is an independent factor impacting miRNA expression in COCs while gonadotropin treatment may affect follicular miRNA expression and IVF efficacy. WHAT IS KNOWN ALREADY Epigenetic mechanisms in female infertility are complex and poorly studied. DNA methylation, histone modifications, miRNAs and nucleosome positioning influence cellular machinery through positive and negative feedback mechanisms either alone or interactively. miRNAs are important regulators during oogenesis, spermatogenesis and early embryogenesis, and are reported to play a role in regulating crosstalk between the oocyte and COCs. Although miRNome analysis has been performed in female human reproductive tissues (endometrium, myometrium, cervix and ovaries), epigenetic modifications in women with infertility have not been explored in detail. In addition, the impact of gonadotropin treatments during MAR on miRNA expression in COCs has not been fully investigated. STUDY DESIGN, SIZE, DURATION This study was carried out in 53 COC samples obtained from mature metaphase II (MII) oocytes in 53 women undergoing MAR treatment. A total of 38 samples for assay development were pooled by maternal age and gonadotropin treatment into four predetermined subgroups: ≥36 years and recombinant human FSH (r-hFSH), n = 10; ≥36 years and r-hFSH+ recombinant human-luteinizing hormone (r-hLH), n = 10; ≤35 years and r-hFSH, n = 9; ≤35 years and r-hFSH+r-hLH, n = 9. miRNome profiles were determined and compared between subgroups. Expression of defined miRNAs was validated in the remaining fifteen samples, representative of each subgroup, by quantitative polymerase chain reaction (PCR). PARTICIPANTS/MATERIALS, SETTING, METHODS COCs were processed for miRNA-enriched total RNA extraction and pooled in homogeneous subgroups to obtain a sufficient amount and quality of starting material to perform the analysis. Each pooled sample underwent miRNA profiling using PCR assay system to examine expression of 752 human miRNAs without pre-amplification. Data were analyzed using the delta-delta Ct method for relative quantitation and prediction of target genes (with at least four algorithms predicting the same miRNA-gene interaction pair (HIT)>4). The miRSystem database provided functional annotation enrichment (raw P-value <0.05) of co-expressed miRNAs. MAIN RESULTS AND THE ROLE OF CHANCE We found distinctive miRNA expression profiles in each subgroup correlating with age and MAR stimulation. In addition, a number of selective and co-expressed miRNAs were revealed by comparative analysis. A cluster of 37 miRNAs were commonly but differentially expressed in all four pools. Significant differences were observed in expression regulation of 37 miRNAs between age groups (≤35 or ≥36) in women receiving r-hFSH+r-hLH compared to those receiving r-hFSH alone. Higher concentrations and increased numbers of miRNAs were recorded in younger than in older patients, regardless of treatment. Functional and expression studies performed to retrieve common miRNome profiles revealed an enrichment of biological functions in oocyte growth and maturation, embryo development, steroidogenesis, ovarian hyperstimulation, apoptosis and cell survival, glucagon and lipid metabolism, and cell trafficking. The highest scored pathways of target genes of the 37 common miRNAs were associated with mitogen-activated protein kinase (MAPK) signaling pathways, G alpha signaling, transcription regulation, tight junctions, RNA polymerase I and III, and mitochondrial transcription. We identified a potential age- and MAR stimulation-dependent signature in the miRNA landscape of COCs. LIMITATIONS, REASONS FOR CAUTION We cannot rule out the possibility that other unknown individual genetic or clinical factors may have interfered with the reported results. Since miRNA profiling was conducted with a predefined array of target probes, other miRNA molecules, potentially modulated by age and hormonal stimulation, may have been missed in this study. WIDER IMPLICATIONS OF THE FINDINGS miRNA expression in COCs is modulated by gonadotropin treatment and correlates strongly with age. A better understanding of the expression patterns and functions of miRNAs may lead to the development of novel therapeutics to treat ovarian dysfunction and improve fertility in older women. STUDY FUNDING/COMPETING INTEREST This study was funded by Merck KGaA, Darmstadt, Germany. All authors declared no competing interest, except SL and TD who are fully employed by Merck KGaA. TRIAL REGISTRATION NUMBER N/A
Collapse
Affiliation(s)
- C Dell'Aversana
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples 80138, Italy.,Institute of Experimental Endocrinology and Oncology 'Gaetano Salvatore' (IEOS)-National Research Council (CNR), Naples 80131, Italy
| | - F Cuomo
- EPI-C S.r.l., Naples 80138, Italy
| | | | | | - F Caprio
- Outpatient Fertility Unit, University of Campania 'Luigi Vanvitelli', Naples 80138, Italy
| | - G Franci
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples 80138, Italy.,Department of Medicine, Surgery and Dentistry "ScuolaMedicaSalernitana", University of Salerno, Baronissi, SA 84081, Italy
| | - M Santonastaso
- Department of Woman, Child and General and Special Surgery, University of Campania 'Luigi Vanvitelli', Naples 80138, Italy
| | - N Colacurci
- Outpatient Fertility Unit, University of Campania 'Luigi Vanvitelli', Naples 80138, Italy.,Department of Woman, Child and General and Special Surgery, University of Campania 'Luigi Vanvitelli', Naples 80138, Italy
| | - S Barone
- Department of Assisted Reproduction, Versilia Hospital, Lido di Camaiore, Lucca 55049, Italy
| | - V Pisaturo
- Department of Reproductive Medicine, International Evangelical Hospital, Genoa 16122, Italy
| | - D Valerio
- Merck Serono S.p.A, Rome 00176, Italy.,Institute of Genetic Research (IRG), Naples 80143, Italy
| | - L Altucci
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples 80138, Italy
| |
Collapse
|
23
|
Pla I, Sanchez A, Pors SE, Pawlowski K, Appelqvist R, Sahlin KB, Poulsen LLC, Marko-Varga G, Andersen CY, Malm J. Proteome of fluid from human ovarian small antral follicles reveals insights in folliculogenesis and oocyte maturation. Hum Reprod 2021; 36:756-770. [PMID: 33313811 PMCID: PMC7891813 DOI: 10.1093/humrep/deaa335] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
STUDY QUESTION Is it possible to identify by mass spectrometry a wider range of proteins and key proteins involved in folliculogenesis and oocyte growth and development by studying follicular fluid (FF) from human small antral follicles (hSAF)? SUMMARY ANSWER The largest number of proteins currently reported in human FF was identified in this study analysing hSAF where several proteins showed a strong relationship with follicular developmental processes. WHAT IS KNOWN ALREADY Protein composition of human ovarian FF constitutes the microenvironment for oocyte development. Previous proteomics studies have analysed fluids from pre-ovulatory follicles, where large numbers of plasma constituents are transferred through the follicular basal membrane. This attenuates the detection of low abundant proteins, however, the basal membrane of small antral follicles is less permeable, making it possible to detect a large number of proteins, and thereby offering further insights in folliculogenesis. STUDY DESIGN, SIZE, DURATION Proteins in FF from unstimulated hSAF (size 6.1 ± 0.4 mm) were characterised by mass spectrometry, supported by high-throughput and targeted proteomics and bioinformatics. The FF protein profiles from hSAF containing oocytes, capable or not of maturing to metaphase II of the second meiotic division during an IVM (n = 13, from 6 women), were also analysed. PARTICIPANTS/MATERIALS, SETTING, METHODS We collected FF from hSAF of ovaries that had been surgically removed from 31 women (∼28.5 years old) undergoing unilateral ovariectomy for fertility preservation. MAIN RESULTS AND THE ROLE OF CHANCE In total, 2461 proteins were identified, of which 1108 identified for the first time in FF. Of the identified proteins, 24 were related to follicular regulatory processes. A total of 35 and 65 proteins were down- and up-regulated, respectively, in fluid from hSAF surrounding oocytes capable of maturing (to MII). We found that changes at the protein level occur already in FF from small antral follicles related to subsequent oocyte maturation. LIMITATIONS, REASONS FOR CAUTION A possible limitation of our study is the uncertainty of the proportion of the sampled follicles that are undergoing atresia. Although the FF samples were carefully aspirated and processed to remove possible contaminants, we cannot ensure the absence of some proteins derived from cellular lysis provoked by technical reasons. WIDER IMPLICATIONS OF THE FINDINGS This study is, to our knowledge, the first proteomics characterisation of FF from hSAF obtained from women in their natural menstrual cycle. We demonstrated that the analysis by mass spectrometry of FF from hSAF allows the identification of a greater number of proteins compared to the results obtained from previous analyses of larger follicles. Significant differences found at the protein level in hSAF fluid could predict the ability of the enclosed oocyte to sustain meiotic resumption. If this can be confirmed in further studies, it demonstrates that the viability of the oocyte is determined early on in follicular development and this may open up new pathways for augmenting or attenuating subsequent oocyte viability in the pre-ovulatory follicle ready to undergo ovulation. STUDY FUNDING/COMPETING INTEREST(S) The authors thank the financial support from ReproUnion, which is funded by the Interreg V EU programme. No conflict of interest was reported by the authors. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Indira Pla
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 205 02 Malmö, Sweden.,Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84 Lund, Sweden
| | - Aniel Sanchez
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 205 02 Malmö, Sweden.,Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84 Lund, Sweden
| | - Susanne Elisabeth Pors
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, 2100 Copenhagen, Denmark
| | - Krzysztof Pawlowski
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84 Lund, Sweden.,Department of Experimental Design and Bioinformatics, Faculty of Agriculture and Biology, Warsaw University of Life Sciences SGGW, Warszawa 02-787, Poland
| | - Roger Appelqvist
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84 Lund, Sweden
| | - K Barbara Sahlin
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 205 02 Malmö, Sweden.,Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84 Lund, Sweden
| | - Liv La Cour Poulsen
- Fertility Clinic, Department of Gynaecology and Obstetrics, Zealand University Hospital, Lykkebækvej 14, 4600 Køge, Denmark
| | - György Marko-Varga
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84 Lund, Sweden.,First Department of Surgery, Tokyo Medical University, Shinjiku-ku, Japan
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, 2100 Copenhagen, Denmark
| | - Johan Malm
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 205 02 Malmö, Sweden.,Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84 Lund, Sweden
| |
Collapse
|
24
|
Poulsen LC, Englund ALM, Andersen AS, Bøtkjær JA, Mamsen LS, Damdimopoulou P, Østrup O, Grøndahl ML, Yding Andersen C. Follicular hormone dynamics during the midcycle surge of gonadotropins in women undergoing fertility treatment. Mol Hum Reprod 2021; 26:256-268. [PMID: 32023345 DOI: 10.1093/molehr/gaaa013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/21/2020] [Indexed: 12/16/2022] Open
Abstract
Changes in concentrations of intra-follicular hormones during ovulation are important for final oocyte maturation and endometrial priming to ensure reproductive success. As no human studies have investigated these changes in detail, our objective was to describe the dynamics of major follicular fluid (FF) hormones and transcription of steroidogenic enzymes and steroid receptors in human granulosa cells (GCs) during ovulation. We conducted a prospective cohort study at a public fertility clinic in 2016-2018. Fifty women undergoing ovarian stimulation for fertility treatment were included. From each woman, FF and GCs were collected by transvaginal ultrasound-guided follicle puncture of one follicle at two specific time points during ovulation, and the study covered a total of five time points: before ovulation induction (OI), 12, 17, 32 and 36 h after OI. Follicular fluid concentrations of oestradiol, progesterone, androstenedione, testosterone, 17-hydroxyprogesterone, anti-Mullerian hormone, inhibin A and inhibin B were measured using ELISA assays, and a statistical mixed model was used to analyse differences in hormone levels between time points. Gene expression of 33 steroidogenic enzymes and six hormone receptors in GCs across ovulation were assessed by microarray analysis, and selected genes were validated by quantitative reverse transcription PCR. We found that concentrations of oestradiol, testosterone, progesterone, AMH, inhibin A and inhibin B (P < 0.001) and gene expression of 12 steroidogenic enzymes and five receptors (false discovery rate < 0.0001) changed significantly during ovulation. Furthermore, we found parallel changes in plasma hormones. The substantial changes in follicular hormone production during ovulation highlight their importance for reproductive success.
Collapse
Affiliation(s)
- L C Poulsen
- Fertility Clinic, Zealand University Hospital, Lykkebækvej 14, 4600 Køge, Denmark
| | - A L M Englund
- Fertility Clinic, Zealand University Hospital, Lykkebækvej 14, 4600 Køge, Denmark
| | - A S Andersen
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| | - J A Bøtkjær
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| | - L S Mamsen
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| | - P Damdimopoulou
- Swedish Toxicology Sciences Research Centre (Swetox), Karolinska Institute, Unit of Toxicology Sciences, 15136 Södertälje, Sweden.,Department of Clinical Science, Intervention and Technology, Karolinska Institute, SE-141 83 Stockholm, Sweden
| | - O Østrup
- Center for Genomic Medicine, Microarray Core Facility, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| | - M L Grøndahl
- Fertility Clinic, University Hospital of Copenhagen, Herlev and Gentofte Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - C Yding Andersen
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| |
Collapse
|
25
|
Poulsen LC, Bøtkjær JA, Østrup O, Petersen KB, Andersen CY, Grøndahl ML, Englund ALM. Two waves of transcriptomic changes in periovulatory human granulosa cells. Hum Reprod 2021; 35:1230-1245. [PMID: 32378719 DOI: 10.1093/humrep/deaa043] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 02/05/2020] [Indexed: 11/14/2022] Open
Abstract
STUDY QUESTION How does the human granulosa cell (GC) transcriptome change during ovulation? SUMMARY ANSWER Two transcriptional peaks were observed at 12 h and at 36 h after induction of ovulation, both dominated by genes and pathways known from the inflammatory system. WHAT IS KNOWN ALREADY The crosstalk between GCs and the oocyte, which is essential for ovulation and oocyte maturation, can be assessed through transcriptomic profiling of GCs. Detailed transcriptional changes during ovulation have not previously been assessed in humans. STUDY DESIGN, SIZE, DURATION This prospective cohort study comprised 50 women undergoing fertility treatment in a standard antagonist protocol at a university hospital-affiliated fertility clinic in 2016-2018. PARTICIPANTS/MATERIALS, SETTING, METHODS From each woman, one sample of GCs was collected by transvaginal ultrasound-guided follicle aspiration either before or 12 h, 17 h or 32 h after ovulation induction (OI). A second sample was collected at oocyte retrieval, 36 h after OI. Total RNA was isolated from GCs and analyzed by microarray. Gene expression differences between the five time points were assessed by ANOVA with a random factor accounting for the pairing of samples, and seven clusters of protein-coding genes representing distinct expression profiles were identified. These were used as input for subsequent bioinformatic analyses to identify enriched pathways and suggest upstream regulators. Subsets of genes were assessed to explore specific ovulatory functions. MAIN RESULTS AND THE ROLE OF CHANCE We identified 13 345 differentially expressed transcripts across the five time points (false discovery rate, <0.01) of which 58% were protein-coding genes. Two clusters of mainly downregulated genes represented cell cycle pathways and DNA repair. Upregulated genes showed one peak at 12 h that resembled the initiation of an inflammatory response, and one peak at 36 h that resembled the effector functions of inflammation such as vasodilation, angiogenesis, coagulation, chemotaxis and tissue remodelling. Genes involved in cell-matrix interactions as a part of cytoskeletal rearrangement and cell motility were also upregulated at 36 h. Predicted activated upstream regulators of ovulation included FSH, LH, transforming growth factor B1, tumour necrosis factor, nuclear factor kappa-light-chain-enhancer of activated B cells, coagulation factor 2, fibroblast growth factor 2, interleukin 1 and cortisol, among others. The results confirmed early regulation of several previously described factors in a cascade inducing meiotic resumption and suggested new factors involved in cumulus expansion and follicle rupture through co-regulation with previously described factors. LARGE SCALE DATA The microarray data were deposited to the Gene Expression Omnibus (www.ncbi.nlm.nih.gov/gds/, accession number: GSE133868). LIMITATIONS, REASONS FOR CAUTION The study included women undergoing ovarian stimulation and the findings may therefore differ from a natural cycle. However, the results confirm significant regulation of many well-established ovulatory genes from a series of previous studies such as amphiregulin, epiregulin, tumour necrosis factor alfa induced protein 6, tissue inhibitor of metallopeptidases 1 and plasminogen activator inhibitor 1, which support the relevance of the results. WIDER IMPLICATIONS OF THE FINDINGS The study increases our understanding of human ovarian function during ovulation, and the publicly available dataset is a valuable resource for future investigations. Suggested upstream regulators and highly differentially expressed genes may be potential pharmaceutical targets in fertility treatment and gynaecology. STUDY FUNDING/COMPETING INTEREST(S) The study was funded by EU Interreg ÔKS V through ReproUnion (www.reprounion.eu) and by a grant from the Region Zealand Research Foundation. None of the authors have any conflicts of interest to declare.
Collapse
Affiliation(s)
- L C Poulsen
- Fertility Clinic, Zealand University Hospital, Lykkebækvej 14, 4600 Køge, Denmark
| | - J A Bøtkjær
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| | - O Østrup
- Center for Genomic Medicine, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| | - K B Petersen
- Fertility Clinic, Zealand University Hospital, Lykkebækvej 14, 4600 Køge, Denmark
| | - C Yding Andersen
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| | - M L Grøndahl
- Fertility Clinic, University Hospital of Copenhagen, Herlev and Gentofte Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - A L M Englund
- Fertility Clinic, Zealand University Hospital, Lykkebækvej 14, 4600 Køge, Denmark
| |
Collapse
|
26
|
Wang C, Fei X, Zhang H, Zhou W, Cheng Z, Feng Y. Proteomic Analysis of the Alterations in Follicular Fluid Proteins During Oocyte Maturation in Humans. Front Endocrinol (Lausanne) 2021; 12:830691. [PMID: 35185790 PMCID: PMC8850365 DOI: 10.3389/fendo.2021.830691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/31/2021] [Indexed: 11/13/2022] Open
Abstract
Many components in ovarian follicles (follicular fluid, cumulus cells, granular cells, etc.) dynamically change during folliculogenesis and play a positive or negative role in oocyte maturation. Infertile women who underwent intracytoplasmic sperm injection (ICSI) treatment in the reproductive medicine centre of Hangzhou Women's Hospital between October 2018 and October 2021 were included. The ovarian follicular fluid and cumulus cells of diminished ovarian response (DOR) patients and control subjects with medical records of clinical data were collected. In total, 31 differentially expressed proteins, including 10 upregulated proteins (>1.50-fold, P<0.05) and 21 downregulated proteins (<0.67-fold, P<0.05), were identified in mature vs. immature oocytes by iTRAQ labelling coupled with 2D LC-MS/MS. GO analysis revealed that 'cell population proliferation' was the most diverse enrichment trend between up/downregulated proteins, while phagosome process and the PI3K-Akt signaling pathway were the two most significant pathways revealed by KEGG enrichment classification. Human prostatic acid phosphatase (PAP, ACPP) and CD5 antigen-like (CD5L) were two proteins verified by ELISA to be differentially expressed between MII and Gv oocytes (P<0.0001 and P<0.0001, respectively). Further measurement found significantly lower level of ACPP in follicular fluids and cumulus cells of DOR patients (P=0.028 and P=0.004, respectively), as an indicator of oocyte quality. Otherwise, CD5L level is upregulated in follicular fluid of DOR patients (P<0.0001). Our study provided experimental data to establish the objective indicator of oocyte maturation in the microenvironment of ovarian follicles, and also provided new insight into the measurement of oocyte quality.
Collapse
Affiliation(s)
- Chong Wang
- Reproductive Medicine Center, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, China
- Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyang Fei
- Reproductive Medicine Center, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, China
| | - Hongyan Zhang
- Reproductive Medicine Center, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, China
| | - Wenjing Zhou
- Reproductive Medicine Center, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, China
| | - Zhaojun Cheng
- Reproductive Medicine Center, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, China
| | - Ying Feng
- Reproductive Medicine Center, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, China
- *Correspondence: Ying Feng,
| |
Collapse
|
27
|
Andersen CY, Kelsey T, Mamsen LS, Vuong LN. Shortcomings of an unphysiological triggering of oocyte maturation using human chorionic gonadotropin. Fertil Steril 2020; 114:200-208. [PMID: 32654823 DOI: 10.1016/j.fertnstert.2020.05.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 12/20/2022]
Abstract
Final maturation of follicles has, in connection with ovarian stimulation and infertility treatment, traditionally been achieved by the administration of a human chorionic gonadotropin (hCG) bolus trigger of 5,000 to 10,000 IU. This trigger serves two purposes: induce oocyte maturation; and serve as luteal phase support owing to its long half-life. It now appears that the hCG bolus trigger is unable to support both these two purposes optimally. In particular, after an hCG trigger, the early luteal phase is hormonally abnormal and different from conditions observed in the natural menstrual cycle: the timing of the initiation of hCG and progesterone rise is much faster after an hCG trigger than in a natural menstrual cycle; the maximal concentrations of hCG and progesterone considerably exceed those naturally observed; and the timing of the peak progesterone concentration after an hCG trigger is advanced several days compared with the natural cycle. Furthermore, the hCG trigger without any follicle-stimulating hormone activity may induce oocyte maturation less efficiently than the combined luteinizing hormone and follicle-stimulating hormone surge normally seen. Collectively, the endometrium is likely to be advanced after an hCG trigger, and the implantation potential is probably not optimal. The precise effect on pregnancy rates after the different progressions of hCG and progesterone concentrations during the early luteal phase has not yet been determined, but more individualized methods using more physiological approaches are likely to improve reproductive outcomes.
Collapse
Affiliation(s)
- Claus Yding Andersen
- Laboratory of Reproductive Biology, The Copenhagen University Hospital, Copenhagen, Denmark; Faculty of Health and Medical Science, Copenhagen University, Copenhagen, Denmark.
| | - Thomas Kelsey
- School of Computer Science, University of St Andrews, St. Andrews, Scotland
| | - Linn Salto Mamsen
- Laboratory of Reproductive Biology, The Copenhagen University Hospital, Copenhagen, Denmark
| | - Lan Ngoc Vuong
- Department of Obstetrics and Gynecology, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam; IVFMD, My Duc Hospital, Ho Chi Minh City, Vietnam; HOPE Research Center, My Duc Hospital, Ho Chi Minh City, Vietnam
| |
Collapse
|
28
|
Moura MT, Paula-Lopes FF. Thermoprotective molecules to improve oocyte competence under elevated temperature. Theriogenology 2020; 156:262-271. [PMID: 32784066 DOI: 10.1016/j.theriogenology.2020.06.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/12/2020] [Accepted: 06/13/2020] [Indexed: 02/06/2023]
Abstract
Heat stress is an environmental factor that challenges livestock by disturbing animal homeostasis. Despite the broad detrimental effects of heat stress on reproductive function, the germline and the early preimplantation embryo are particularly prone. There is extensive evidence that elevated temperature reduces oocyte developmental competence through a series of cellular and molecular damages. Further research revealed that the oocyte respond to stress by activating cellular mechanisms such as heat shock response, unfolded protein response and autophagy to improve survival under heat shock. Such knowledge paved the way for the identification of thermoprotective molecules that alleviate heat-induced oocyte oxidative stress, organelle damage, and apoptosis. Therefore, this review depicts the deleterious effects of heat shock on oocyte developmental competence, heat-induced cellular and molecular changes, outlines pro-survival cellular mechanisms and explores thermoprotective molecules to improve oocyte competence.
Collapse
Affiliation(s)
- Marcelo T Moura
- Department of Biological Sciences, Federal University of São Paulo - UNIFESP, Diadema, SP, Brazil
| | - Fabíola F Paula-Lopes
- Department of Biological Sciences, Federal University of São Paulo - UNIFESP, Diadema, SP, Brazil.
| |
Collapse
|
29
|
Zhang Y, Zhu Z, Li H, Zhu M, Peng X, Xin A, Qu R, He W, Fu J, Sun X. Resolvin E1 in Follicular Fluid Acts as a Potential Biomarker and Improves Oocyte Developmental Competence by Optimizing Cumulus Cells. Front Endocrinol (Lausanne) 2020; 11:210. [PMID: 32373069 PMCID: PMC7176900 DOI: 10.3389/fendo.2020.00210] [Citation(s) in RCA: 9] [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: 01/05/2020] [Accepted: 03/25/2020] [Indexed: 12/15/2022] Open
Abstract
Metabolic profile of follicular fluid (FF) has been investigated to look for biomarkers for oocyte quality. Resolvin E1 (RvE1), a potent pro-resolving mediator, was reported to have protective action in cell function. The study aimed to examine the predictive value of RvE1 for oocyte quality and to explore the cellular mechanism of RvE1 in improving oocyte competence. Metabolic profiles of 80 FF samples showed a higher level of RvE1 in group A (blastocysts scored ≥ B3BC and B3CB according to Gardner's blastocyst scoring system, N = 36) than that of group B (blastocysts scored < B3BC and B3CB, N = 44, P = 0.0018). The receiver operating characteristic (ROC) curve analysis showed that RvE1 level in FF below 8.96 pg/ml (AUC:0.75; 95%CI: 0.64-0.86; P = 0.00012) could predict poor oocyte quality with specificity of 97.22%, suggesting RvE1 as a potential biomarker to exclude inferior oocytes. Besides, the level of RvE1 was found to be significantly lower in FF than in serum (57.49 to 17.62 pg/ml; P=.0037) and was gradually accumulated in the culture medium of cumulus cells (CCs) during cell culture, which indicated that RvE1 came from both blood exudates and local secretion. The in vitro experiment revealed thecellular mechanism of RvE1 in improvingoocyte qualityby decreasing the cumulus cellapoptotic rate and increasing cell viability and proliferation. It is the first time thatthe role of RvE1 in reproduction is explored. In conclusion, RvE1 is valuable as a potential exclusive biomarker for oocyte selection andplays a role in improving oocyte quality.
Collapse
Affiliation(s)
- Yijing Zhang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai JIAI Genetics & IVF Institute, Shanghai, China
- Key Laboratory of Female Reproductive Endocrine Related Diseases of Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Zhongyi Zhu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Key Laboratory of Female Reproductive Endocrine Related Diseases of Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - He Li
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai JIAI Genetics & IVF Institute, Shanghai, China
| | - Mingjiang Zhu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, China
| | - Xiandong Peng
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai JIAI Genetics & IVF Institute, Shanghai, China
| | - Aijie Xin
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai JIAI Genetics & IVF Institute, Shanghai, China
| | - Ronggui Qu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai JIAI Genetics & IVF Institute, Shanghai, China
| | - Wen He
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai JIAI Genetics & IVF Institute, Shanghai, China
- Key Laboratory of Female Reproductive Endocrine Related Diseases of Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jing Fu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai JIAI Genetics & IVF Institute, Shanghai, China
| | - Xiaoxi Sun
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai JIAI Genetics & IVF Institute, Shanghai, China
- Key Laboratory of Female Reproductive Endocrine Related Diseases of Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
30
|
Ma Y, Jin J, Tong X, Yang W, Ren P, Dai Y, Pan Y, Zhang Y, Zhang S. ADAMTS1 and HSPG2 mRNA levels in cumulus cells are related to human oocyte quality and controlled ovarian hyperstimulation outcomes. J Assist Reprod Genet 2020; 37:657-667. [PMID: 31974739 PMCID: PMC7125252 DOI: 10.1007/s10815-019-01659-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 12/12/2019] [Indexed: 12/17/2022] Open
Abstract
PURPOSE The study investigated potential correlations between the expression levels of ADAMTS1 and HSPG2 in cumulus cells (CCs) and controlled ovarian hyperstimulation (COH) outcomes. METHODS RT-PCR was used to determine ADAMTS1 and HSPG2 mRNA levels in mice CCs at different timepoints (0, 4, 8, 12, and 16 h) after human chorionic gonadotropin (hCG) injection, and in CCs after RNAi treatment. Women with polycystic ovary syndrome (PCOS) (n = 45) and normal ovulatory controls (n = 103) undergoing IVF/ICSI were recruited. Relative ADAMTS1 and HSPG2 mRNA levels were measured by RT-PCR. Moreover, correlations of ADAMTS1 and HSPG2 levels with COH outcomes were analyzed. RESULTS At different timepoints after hCG treatment, ADAMTS1 mRNA had the highest level at 12 h, whereas HSPG2 showed opposite profiles to ADAMTS1 with the lowest level at 12 h. HSPG2 expression was upregulated after ADAMTS1 RNAi treatment The PCOS group had higher HSPG2 and lower ADAMTS1 expression levels than controls. In normal ovulatory women (control group), a higher expression of ADAMTS1 and lower expression of HSPG2 were associated with more mature oocytes, transplantable embryos, and good quality embryos, whereas higher transplantable embryo rates and good quality embryo rates were obtained only with lower HSPG2 expression. ROC curves showed the co-measurement of ADAMTS1 and HSPG2 had a better predictive power than separate analyses. CONCLUSION The dynamic profiles of ADAMTS1 and HSPG2 were inversely correlated in CCs. In PCOS and normal ovulatory patients, higher ADAMTS1 and lower HSPG2 expression levels in CCs were related to better COH outcomes.
Collapse
Affiliation(s)
- Yerong Ma
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Jiamin Jin
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Xiaomei Tong
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Weijie Yang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Peipei Ren
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Yongdong Dai
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Yibin Pan
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - YinLi Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China.
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China.
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China.
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China.
| |
Collapse
|
31
|
Rispoli LA, Edwards JL, Pohler KG, Russell S, Somiari RI, Payton RR, Schrick FN. Heat-induced hyperthermia impacts the follicular fluid proteome of the periovulatory follicle in lactating dairy cows. PLoS One 2019; 14:e0227095. [PMID: 31887207 PMCID: PMC6936800 DOI: 10.1371/journal.pone.0227095] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 12/12/2019] [Indexed: 12/27/2022] Open
Abstract
We hypothesized that heat-induced perturbations in cumulus cells surrounding the maturing oocyte may extend to the mural granulosa of the periovulatory follicle in the heat-stressed cow to subsequently the follicular fluid proteome. Lactating Holsteins were pharmacologically stimulated to have a dominant follicle that was capable of responding to a gonadotropin releasing hormone-induced luteinizing hormone surge. Following gonadotropin releasing hormone administration, cows were maintained at ~67 temperature humidity index (THI; thermoneutral conditions) or exposed to conditions simulating an acute heat stress event (71 to 86 THI; heat stress for ~12 h). Dominant follicle collection was conducted in the periovulatory period ~16 h after gonadotropin releasing hormone. Follicular fluid proteome from thermoneutral (n = 5) and hyperthermic (n = 5) cows was evaluated by quantitative tandem mass spectrometry (nano LC-MS/MS). We identified 35 differentially-abundant proteins. Functional annotation revealed numerous immune-related proteins. Subsequent efforts revealed an increase in levels of the proinflammatory mediator bradykinin in follicular fluid (P = 0.0456) but not in serum (P = 0.9319) of hyperthermic cows. Intrafollicular increases in transferrin (negative acute phase protein) in hyperthermic cows (P = 0.0181) coincided with a tendency for levels to be increased in the circulation (P = 0.0683). Nine out of 15 cytokines evaluated were detected in follicular fluid. Heat stress increased intrafollicular interleukin 6 levels (P = 0.0160). Whether hyperthermia-induced changes in the heat-stressed cow's follicular fluid milieu reflect changes in mural granulosa, cumulus, other cell types secretions, and/or transudative changes from circulation remains unclear. Regardless of origin, heat stress/hyperthermia related changes in the follicular fluid milieu may have an impact on components important for ovulation and competence of the cumulus-oocyte complex contained within the periovulatory follicle.
Collapse
Affiliation(s)
- Louisa A. Rispoli
- Department of Animal Science, The University of Tennessee, Institute of Agriculture, AgResearch, Knoxville, TN, United States of America
| | - J. Lannett Edwards
- Department of Animal Science, The University of Tennessee, Institute of Agriculture, AgResearch, Knoxville, TN, United States of America
| | - Ky G. Pohler
- Department of Animal Science, The University of Tennessee, Institute of Agriculture, AgResearch, Knoxville, TN, United States of America
| | - Stephen Russell
- ITSI–Biosciences, LLC, Johnstown, PA, United States of America
| | | | - Rebecca R. Payton
- Department of Animal Science, The University of Tennessee, Institute of Agriculture, AgResearch, Knoxville, TN, United States of America
| | - F. Neal Schrick
- Department of Animal Science, The University of Tennessee, Institute of Agriculture, AgResearch, Knoxville, TN, United States of America
| |
Collapse
|