1
|
Nishigaki A, Ishida M, Tsubokura H, Hisamatsu Y, Hirose Y, Okada H. HIF-1α Promotes Luteinization via NDRG1 Induction in the Human Ovary. Biomedicines 2025; 13:328. [PMID: 40002742 PMCID: PMC11852844 DOI: 10.3390/biomedicines13020328] [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: 12/09/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor that plays a crucial role in various physiological and pathological processes of the ovary. However, the timing of HIF-1α expression and its specific biological function in the follicular development of the human ovary remain unclear. Therefore, in this study, we aimed to examine whether HIF-1α and its downstream gene, N-myc downstream-regulated gene 1 (NDRG1), exhibit stage-specific expression during the follicular development process in the human ovary. Methods: We used ovarian tissues from eight women with regular menstrual cycles who were not undergoing hormonal treatment. We investigated HIF-1α and NDRG1 expression and localization using immunohistochemistry. Further, we transfected human ovarian granulosa (KGN) cells with HIF-1α small interfering RNA (siRNA) to investigate the influence of HIF-1α on NDRG1 expression and progesterone synthesis. Results: The immunohistochemical analysis of human ovarian tissues revealed that HIF-1α was localized in the cytoplasm of granulosa cells (GCs) at both the primary and secondary follicular stages. Conversely, in tertiary and later developmental stages, HIF-1α was observed exclusively in the nucleus of GCs. Furthermore, while NDRG1 was not detected in primary follicles, it was present in all GCs beyond the tertiary stage. Notably, transfection of KGN cells with HIF-1α siRNA significantly decreased NDRG1 expression, at both the mRNA and protein levels, and in progesterone synthesis. Conclusion: Our results indicate that HIF-1α and NDRG1 are integral to follicular development and the early luteinization of pre-ovulatory follicles.
Collapse
Affiliation(s)
- Akemi Nishigaki
- Department of Obstetrics and Gynecology, Kansai Medical University, Hirakata 573-1191, Osaka, Japan; (A.N.); (H.T.); (Y.H.); (H.O.)
| | - Mitsuaki Ishida
- Department of Pathology, Osaka Medical and Pharmaceutical University, Takatsuki 569-8686, Osaka, Japan;
| | - Hiroaki Tsubokura
- Department of Obstetrics and Gynecology, Kansai Medical University, Hirakata 573-1191, Osaka, Japan; (A.N.); (H.T.); (Y.H.); (H.O.)
| | - Yoji Hisamatsu
- Department of Obstetrics and Gynecology, Kansai Medical University, Hirakata 573-1191, Osaka, Japan; (A.N.); (H.T.); (Y.H.); (H.O.)
| | - Yoshinobu Hirose
- Department of Pathology, Osaka Medical and Pharmaceutical University, Takatsuki 569-8686, Osaka, Japan;
| | - Hidetaka Okada
- Department of Obstetrics and Gynecology, Kansai Medical University, Hirakata 573-1191, Osaka, Japan; (A.N.); (H.T.); (Y.H.); (H.O.)
| |
Collapse
|
2
|
Pan Y, Wu G, Chen M, Lu X, Shen M, Li H, Liu H. Lactate Promotes Hypoxic Granulosa Cells' Autophagy by Activating the HIF-1α/BNIP3/Beclin-1 Signaling Axis. Genes (Basel) 2024; 16:14. [PMID: 39858561 PMCID: PMC11765430 DOI: 10.3390/genes16010014] [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: 11/19/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES The avascular nature of the follicle creates a hypoxic microenvironment, establishing a niche where granulosa cells (GCs) rely on glycolysis to produce energy in the form of lactate (L-lactate). Autophagy, an evolutionarily conserved stress-response process, involves the formation of autophagosomes to encapsulate intracellular components, delivering them to lysosomes for degradation. This process plays a critical role in maintaining optimal follicular development. However, whether hypoxia regulates autophagy in GCs via lactate remains unclear. METHODS In this study, we investigated lactate-induced autophagy under hypoxia by utilizing glycolysis inhibitors or silencing related genes. RESULTS We observed a significant increase in autophagy in ovarian GCs under hypoxic conditions, indicated by elevated LC3II levels and reduced P62 levels. Suppressing lactate production through glycolytic inhibitors (2-DG and oxamate) or silencing lactate dehydrogenase (LDHA/LDHB) effectively reduced hypoxia-induced autophagy. Further investigation revealed that the HIF1-α/BNIP3/Beclin-1 axis is essential for lactate-induced autophagy under hypoxic conditions. Inhibiting HIF-1α activity using siRNAs or PX-478 downregulated BNIP3 expression and subsequently suppressed autophagy. Similarly, BNIP3 silencing with siRNAs repressed lactate-induced autophagy in hypoxic conditions. Mechanistically, immunoprecipitation experiments showed that BNIP3 disrupted pre-existing Bcl-2/Beclin-1 complexes by competing with Bcl-2 to form Bcl-2/BNIP3 complexes. This interaction released Beclin-1, which subsequently triggered lactate-induced autophagy under hypoxic conditions. CONCLUSIONS These findings unveil a novel mechanism by which hypoxia regulates GC autophagy through lactate production, highlighting its potential role in sustaining follicular development under hypoxic conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Honglin Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.P.); (G.W.); (M.C.); (X.L.); (M.S.); (H.L.)
| |
Collapse
|
3
|
Szymanska M, Basavaraja R, Meidan R. A tale of two endothelins: the rise and fall of the corpus luteum. Reprod Fertil Dev 2024; 37:RD24158. [PMID: 39680472 DOI: 10.1071/rd24158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Endothelins are small 21 amino acid peptides that interact with G-protein-coupled receptors. They are highly conserved across species and play important roles in vascular biology as well as in disease development and progression. Endothelins, mainly endothelin-1 and endothelin-2, are intricately involved in ovarian function and metabolism. These two peptides differ only in two amino acids but are encoded by different genes, which suggests an independent regulation and a cell-specific mode of expression. This review aims to comprehensively discuss the distinct regulation and roles of endothelin-1 and endothelin-2 regarding corpus luteum function throughout its life span.
Collapse
Affiliation(s)
- Magdalena Szymanska
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; and Present address: Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Olsztyn, Poland
| | - Raghavendra Basavaraja
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; and Present address: Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rina Meidan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| |
Collapse
|
4
|
Berisha B, Thaqi G, Schams D, Rodler D, Sinowatz F, Pfaffl MW. Effect of the gonadotropin surge on steroid receptor regulation in preovulatory follicles and newly formed corpora lutea in the cow. Domest Anim Endocrinol 2024; 89:106876. [PMID: 39047595 DOI: 10.1016/j.domaniend.2024.106876] [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: 05/07/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
The objective of the study was to characterize the mRNA expression patterns of specific steroid hormone receptors namely, estrogen receptors (ESRRA-estrogen related receptor alpha and ESRRB-estrogen related receptor beta) and progesterone receptors (PGR) in superovulation-induced bovine follicles during the periovulation and subsequent corpus luteum (CL) formation. The bovine ovaries (n = 5 cow / group), containing preovulatory follicles or early CL, were collected relative to injection of the gonadotropin-releasing hormone (GnRH) at (I) 0 h, (II) 4 h, (III) 10 h, (IV) 20 h, (V) 25 h (preovulatory follicles) and (VI) 60 h (CL, 2-3 days after induced ovulation). In this experiment, we analyzed the steroid receptor mRNA expression and their localization in the follicle and CL tissue. The high mRNA expression of ESRRA, ESRRB, and PGR analyzed in the follicles before ovulation is significantly reduced in the group of follicles during ovulation (25 h after GnRH), rising again significantly after ovulation in newly formed CL, only for ESRRA and PGR (P < 0.05). Immunohistochemically, the nuclei of antral follicles' granulosa cells showed a positive staining for ESRRA, followed by higher activity in the large luteal cells just after ovulation (early CL). In contrast, the lower PGR immunopresence in preovulatory follicles increased in both small and large luteal cell nuclei after follicle ovulation. Our results of steroid receptor mRNA expression in this experimentally induced gonadotropin surge provide insight into the molecular mechanisms of the effects of steroid hormones on follicular-luteal tissue in the period close to the ovulation and subsequent CL formation in the cow.
Collapse
Affiliation(s)
- Bajram Berisha
- Chair of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Weihenstephan 85354, Germany; Animal Biotechnology, Faculty of Agriculture and Veterinary, University of Prishtina, Prishtinë, Kosovo; Academy of Science of Albania, Tirana, Albania
| | - Granit Thaqi
- Chair of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Weihenstephan 85354, Germany.
| | - Dieter Schams
- Chair of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Weihenstephan 85354, Germany
| | - Daniela Rodler
- Department of Veterinary Sciences, Ludwig Maximilian University of Munich, Munich, Germany
| | - Fred Sinowatz
- Department of Veterinary Sciences, Ludwig Maximilian University of Munich, Munich, Germany
| | - Michael W Pfaffl
- Chair of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Weihenstephan 85354, Germany
| |
Collapse
|
5
|
Tarabeih R, Nemerovsky L, Bar-Joseph H, Eldar-Boock A, Elmechaly CL, Ben-Ami I, Shalgi R. Pigment epithelium-derived factor expression and role in follicular development. Reprod Biomed Online 2024; 49:103981. [PMID: 38870625 DOI: 10.1016/j.rbmo.2024.103981] [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: 12/26/2023] [Revised: 03/01/2024] [Accepted: 03/26/2024] [Indexed: 06/15/2024]
Abstract
RESEARCH QUESTION What is the involvement of pigment epithelium-derived factor (PEDF), expressed in granulosa cells, in folliculogenesis? DESIGN mRNA expression of PEDF and other key factors [Cyp19, anti-Müllerian hormone receptor (AMHR) and vascular endothelial growth factor (VEGF)] in mice follicles was examined in order to typify the expression of PEDF in growing follicles and in human primary granulosa cells (hpGC), and to follow the interplay between PEDF and the other main players in folliculogenesis: FSH and AMH. RESULTS mRNA expression of PEDF increased through folliculogenesis, although the pattern differed from that of the other examined genes, affecting the follicular angiogenic and oxidative balance. In hpGC, prolonged exposure to FSH stimulated the up-regulation of PEDF mRNA. Furthermore, a negative correlation between AMH and PEDF was observed: AMH stimulation reduced the expression of PEDF mRNA and PEDF stimulation reduced the expression of AMHR mRNA. CONCLUSIONS Folliculogenesis, an intricate process that requires close dialogue between the oocyte and its supporting granulosa cells, is mediated by various endocrine and paracrine factors. The current findings suggest that PEDF, expressed in granulosa cells, is a pro-folliculogenesis player that interacts with FSH and AMH in the process of follicular growth. However, the mechanism of this process is yet to be determined.
Collapse
Affiliation(s)
- Rana Tarabeih
- Department of Cell and Developmental Biology, Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Luba Nemerovsky
- Department of Cell and Developmental Biology, Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Hadas Bar-Joseph
- TMCR Unit, Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Anat Eldar-Boock
- TMCR Unit, Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Cindy L Elmechaly
- Department of Cell and Developmental Biology, Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ido Ben-Ami
- IVF and Infertility Unit, Department of Obstetrics and Gynaecology, Shaare Zedek Medical Centre, The Hebrew University Medical School of Jerusalem, Jerusalem, Israel
| | - Ruth Shalgi
- Department of Cell and Developmental Biology, Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
6
|
Bharati J, Kumar S, Mohan NH, Pegu SR, Borah S, Gupta VK, Sarkar M. CRISPR/Cas genome editing revealed non-angiogenic role of VEGFA gene in porcine luteal cells: a preliminary report. Mol Biol Rep 2024; 51:195. [PMID: 38270707 DOI: 10.1007/s11033-023-09115-8] [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: 05/19/2023] [Accepted: 12/06/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND The angiogenic cytokine vascular endothelial growth factor A (VEGFA) also exerts non-angiogenic effects on endocrine functionality of porcine luteal cells critical for progesterone (P4) production. METHOD AND RESULTS The expression dynamics of VEGFA-FLT/KDR system were investigated using RT-qPCR during luteal stages and VEGFA gene knock out (KO) porcine luteal cells were generated using CRISPR/Cas9 technology. The downstream effects of VEGFA ablation were studied using RT-qPCR, Annexin V, MTT, ELISA for P4 estimation and scratch wound assay. Bioinformatics analysis of RNA-Seq data of porcine mid-luteal stage was conducted for exploring protein-protein interaction network, KEGG pathways, transcription factors and kinase mapping for VEGFA-FLT/KDR interactomes. The VEGFA-FLT/KDR system expressed throughout the luteal stages with highest expression during mid- luteal stage. Cellular morphology, structure and oil-red-o staining for lipid droplets did not differ significantly between VEGFA KO and wild type cells, however, VEGFA KO significantly decreased (p < 0.05) viability and proliferation efficiency of edited cells on subsequent passages. Expression of apoptotic gene, CASP3 and hypoxia related gene, HIF1A were significantly (p < 0.05) upregulated in KO cells. The relative mRNA expression of VEGFA and steroidogenic genes STAR, CYP11A1 and HSD3B1 decreased significantly (p < 0.05) upon KO, which was further validated by the significant (p < 0.05) decrease in P4 output from KO cells. Bioinformatics analysis mapped VEGFA-FLT/KDR system to signalling pathways associated with steroidogenic cell functionality and survival, which complemented the findings of the study. CONCLUSION The ablation of VEGFA gene resulted in decreased steroidogenic capability of luteal cells, which suggests that VEGFA exerts additional non-angiogenic regulatory effects in luteal cell functionality.
Collapse
Affiliation(s)
- Jaya Bharati
- ICAR-National Research Centre on Pig, Rani, Guwahati, Assam, 781131, India.
- Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, 243122, India.
| | - Satish Kumar
- ICAR-National Research Centre on Pig, Rani, Guwahati, Assam, 781131, India
| | - N H Mohan
- ICAR-National Research Centre on Pig, Rani, Guwahati, Assam, 781131, India
| | - Seema Rani Pegu
- ICAR-National Research Centre on Pig, Rani, Guwahati, Assam, 781131, India
| | - Sanjib Borah
- Lakhimpur College of Veterinary Science, Assam Agricultural University, North Lakhimpur, Assam, India
| | - Vivek Kumar Gupta
- Lakhimpur College of Veterinary Science, Assam Agricultural University, North Lakhimpur, Assam, India
| | - Mihir Sarkar
- ICAR-National Research Centre On Yak, Dirang, Arunachal Pradesh, 790101, India.
| |
Collapse
|
7
|
Höfner M, Eubler K, Herrmann C, Berg U, Berg D, Welter H, Imhof A, Forné I, Mayerhofer A. Reduced oxygen concentrations regulate the phenotype and function of human granulosa cells in vitro and cause a diminished steroidogenic but increased inflammatory cellular reaction. Mol Hum Reprod 2023; 30:gaad049. [PMID: 38128016 DOI: 10.1093/molehr/gaad049] [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: 08/07/2023] [Revised: 12/13/2023] [Indexed: 12/23/2023] Open
Abstract
Oxygen (O2) concentrations have recently been discussed as important regulators of ovarian cells. Human IVF-derived granulosa cells (human GCs) can be maintained in vitro and are a widely used cellular model for the human ovary. Typically, GCs are cultured at atmospheric O2 levels (approximately around 20%), yet the O2 conditions in vivo, especially in the preovulatory follicle, are estimated to be much lower. Therefore, we comprehensively evaluated the consequences of atmospheric versus hypoxic (1% O2) conditions for 4 days on human GCs. We found lower cellular RNA and protein levels but unchanged cell numbers at 1% O2, indicating reduced transcriptional and/or translational activity. A proteomic analysis showed that 391 proteins were indeed decreased, yet 133 proteins were increased under hypoxic conditions. According to gene ontology (GO) enrichment analysis, pathways associated with metabolic processes, for example amino acid-catabolic-processes, mitochondrial protein biosynthesis, and steroid biosynthesis, were downregulated. Pathways associated with glycolysis, chemical homeostasis, cellular response to hypoxia, and actin filament bundle assembly were upregulated. In accordance with lower CYP11A1 (a cholesterol side-chain cleavage enzyme) levels, progesterone release was decreased. A proteome profiler, as well as IL-6 and IL-8 ELISA assays, revealed that hypoxia led to increased secretion of pro-inflammatory and angiogenic factors. Immunofluorescence studies showed nuclear localization of hypoxia-inducible factor 1α (HIF1α) in human GCs upon acute (2 h) exposure to 1% O2 but not in cells exposed to 1% O2 for 4 days. Hence, the role of HIF1α may be restricted to initiation of the hypoxic response in human GCs. The results provide a detailed picture of hypoxia-induced phenotypic changes in human GCs and reveal that chronically low O2 conditions inhibit the steroidogenic but promote the inflammatory phenotype of these cells.
Collapse
Affiliation(s)
- Maria Höfner
- Cell Biology, Anatomy III, Biomedical Center Munich (BMC), Faculty of Medicine, Ludwig-Maximilian-University (LMU), Planegg-Martinsried, Germany
| | - Katja Eubler
- Cell Biology, Anatomy III, Biomedical Center Munich (BMC), Faculty of Medicine, Ludwig-Maximilian-University (LMU), Planegg-Martinsried, Germany
| | - Carola Herrmann
- Cell Biology, Anatomy III, Biomedical Center Munich (BMC), Faculty of Medicine, Ludwig-Maximilian-University (LMU), Planegg-Martinsried, Germany
| | - Ulrike Berg
- Fertility Centre A.R.T., Bogenhausen, Munich, Germany
| | - Dieter Berg
- Fertility Centre A.R.T., Bogenhausen, Munich, Germany
| | - Harald Welter
- Cell Biology, Anatomy III, Biomedical Center Munich (BMC), Faculty of Medicine, Ludwig-Maximilian-University (LMU), Planegg-Martinsried, Germany
| | - Axel Imhof
- Protein Analysis Unit, BMC, Faculty of Medicine, LMU, Planegg-Martinsried, Germany
| | - Ignasi Forné
- Protein Analysis Unit, BMC, Faculty of Medicine, LMU, Planegg-Martinsried, Germany
| | - Artur Mayerhofer
- Cell Biology, Anatomy III, Biomedical Center Munich (BMC), Faculty of Medicine, Ludwig-Maximilian-University (LMU), Planegg-Martinsried, Germany
| |
Collapse
|
8
|
Monaco CF, Davis JS. Mechanisms of angioregression of the corpus luteum. Front Physiol 2023; 14:1254943. [PMID: 37841308 PMCID: PMC10568036 DOI: 10.3389/fphys.2023.1254943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
The corpus luteum is a transient ovarian endocrine gland that produces the progesterone necessary for the establishment and maintenance of pregnancy. The formation and function of this gland involves angiogenesis, establishing the tissue with a robust blood flow and vast microvasculature required to support production of progesterone. Every steroidogenic cell within the corpus luteum is in direct contact with a capillary, and disruption of angiogenesis impairs luteal development and function. At the end of a reproductive cycle, the corpus luteum ceases progesterone production and undergoes rapid structural regression into a nonfunctional corpus albicans in a process initiated and exacerbated by the luteolysin prostaglandin F2α (PGF2α). Structural regression is accompanied by complete regression of the luteal microvasculature in which endothelial cells die and are sloughed off into capillaries and lymphatic vessels. During luteal regression, changes in nitric oxide transiently increase blood flow, followed by a reduction in blood flow and progesterone secretion. Early luteal regression is marked by an increased production of cytokines and chemokines and influx of immune cells. Microvascular endothelial cells are sensitive to released factors during luteolysis, including thrombospondin, endothelin, and cytokines like tumor necrosis factor alpha (TNF) and transforming growth factor β 1 (TGFB1). Although PGF2α is known to be a vasoconstrictor, endothelial cells do not express receptors for PGF2α, therefore it is believed that the angioregression occurring during luteolysis is mediated by factors downstream of PGF2α signaling. Yet, the exact mechanisms responsible for angioregression in the corpus luteum remain unknown. This review describes the current knowledge on angioregression of the corpus luteum and the roles of vasoactive factors released during luteolysis on luteal vasculature and endothelial cells of the microvasculature.
Collapse
Affiliation(s)
- Corrine F. Monaco
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, United States
| | - John S. Davis
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, United States
- US Department of Veterans Affairs Nebraska-Western Iowa Healthcare System, Omaha, NE, United States
| |
Collapse
|
9
|
Jin J, Ren P, Li X, Zhang Y, Yang W, Ma Y, Lai M, Yu C, Zhang S, Zhang YL. Ovulatory signal-triggered chromatin remodeling in ovarian granulosa cells by HDAC2 phosphorylation activation-mediated histone deacetylation. Epigenetics Chromatin 2023; 16:11. [PMID: 37076890 PMCID: PMC10116676 DOI: 10.1186/s13072-023-00485-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 04/07/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND Epigenetic reprogramming is involved in luteinizing hormone (LH)-induced ovulation; however, the underlying mechanisms are largely unknown. RESULTS We here observed a rapid histone deacetylation process between two waves of active transcription mediated by the follicle-stimulating hormone (FSH) and the LH congener human chorionic gonadotropin (hCG), respectively. Analysis of the genome-wide H3K27Ac distribution in hCG-treated granulosa cells revealed that a rapid wave of genome-wide histone deacetylation remodels the chromatin, followed by the establishment of specific histone acetylation for ovulation. HDAC2 phosphorylation activation coincides with histone deacetylation in mouse preovulatory follicles. When HDAC2 was silenced or inhibited, histone acetylation was retained, leading to reduced gene transcription, retarded cumulus expansion, and ovulation defect. HDAC2 phosphorylation was associated with CK2α nuclear translocation, and inhibition of CK2α attenuated HDAC2 phosphorylation, retarded H3K27 deacetylation, and inactivated the ERK1/2 signaling cascade. CONCLUSIONS This study demonstrates that the ovulatory signal erases histone acetylation through activation of CK2α-mediated HDAC2 phosphorylation in granulosa cells, which is an essential prerequisite for subsequent successful ovulation.
Collapse
Affiliation(s)
- Jiamin Jin
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, 310016, China
| | - Peipei Ren
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, 310016, China
| | - Xiang Li
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, 310016, China
| | - Yinyi Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, 310016, China
| | - Weijie Yang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, 310016, China
| | - Yerong Ma
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, 310016, China
| | - Mengru Lai
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, 310016, China
| | - Chao Yu
- College of Life Science, Zhejiang University, Hangzhou, 310058, China
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China.
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, 310016, China.
| | - Yin-Li Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China.
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, 310016, China.
| |
Collapse
|
10
|
He H, Zhang H, Pan Y, Zhang T, Yang S, Liu M, Robert N, Wang J, Zhao T, Zhao L, Fan J, Cui Y, Yu S. Low oxygen concentration improves yak oocyte maturation and inhibits apoptosis through HIF-1 and VEGF. Reprod Domest Anim 2021; 57:381-392. [PMID: 34967955 DOI: 10.1111/rda.14076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 12/28/2021] [Indexed: 11/29/2022]
Abstract
The gas-phase environment of in vitro culture system plays an important role in the development of oocytes, and oxygen concentration is one of the important factors. In the present study, we aimed to explore the effect of different oxygen concentrations (20%, 10%, 5% or 1% O2 ) in yak oocyte maturation and to detect the expression of HIF-1α, VEGF and cell apoptosis in yak COCs. First, the maturation rate of oocytes, cleavage rate and blastocysts rate following parthenogenetic activation in the group with 5% oxygen concentration were significantly higher (p < 0.05) than the other groups. Then, TUNEL analysis showed that the 5% oxygen concentration group significantly inhibited apoptosis of cumulus-oocyte complexes (COCs) compared to the other group, and the transcription and protein levels of pro-apoptotic factor Bax, HIF-1α and VEGF in yak COCs significantly reduced, while anti-apoptotic factor Bcl-2 significantly increased. Furthermore, immunohistochemical staining results indicated that HIF-1α protein was mainly located in theca follicle interna, mural follicular stratum granulosum, corona radiata and ovarian stroma in the follicular ovarian tissue; while VEGF protein was mainly located in the granulosa and theca cell layers. In summary, our findings demonstrate that 5% oxygen concentration may promote maturation and inhibit apoptosis of oocytes through HIF-1α-mediated VEGF expression.
Collapse
Affiliation(s)
- Honghong He
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China.,College of Animal Husbandry and Verterinary Medicine, Southwest Minzu University, Chengdu, 610041, China
| | - Huizhu Zhang
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yangyang Pan
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Tongxiang Zhang
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Shanshan Yang
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Minqing Liu
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Niayale Robert
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Jinglei Wang
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Tian Zhao
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Ling Zhao
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Jiangfeng Fan
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yan Cui
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Sijiu Yu
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
11
|
Atkinson L, Martin F, Sturmey RG. Intraovarian injection of platelet-rich plasma in assisted reproduction: too much too soon? Hum Reprod 2021; 36:1737-1750. [PMID: 33963408 PMCID: PMC8366566 DOI: 10.1093/humrep/deab106] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/09/2021] [Indexed: 12/13/2022] Open
Abstract
The prospect of ovarian rejuvenation offers the tantalising prospect of treating age-related declines in fertility or in pathological conditions such as premature ovarian failure. The concept of ovarian rejuvenation was invigorated by the indication of the existence of oogonial stem cells (OSCs), which have been shown experimentally to have the ability to differentiate into functional follicles and generate oocytes; however, their clinical potential remains unknown. Furthermore, there is now growing interest in performing ovarian rejuvenation in situ. One proposed approach involves injecting the ovary with platelet rich plasma (PRP). PRP is a component of blood that remains after the in vitro removal of red and white blood cells. It contains blood platelets, tiny anucleate cells of the blood, which are responsible for forming athrombus to prevent bleeding. In addition, PRP contains an array of cytokines and growth factors, as well as a number of small molecules.The utility ofPRP has been investigatedin a range of regenerative medicine approaches and has been shown to induce differentiation of a range of cell types, presumably through the action of cytokines. A handful ofcasereports have described the use of PRP injections into the ovaryin the human, and while these clinical data report promising results, knowledge on the mechanisms and safety of PRP injections into the ovary remain limited.In this article, we summarise some of the physiological detail of platelets and PRP, before reviewing the existing emerging literature in this area. We then propose potential mechanisms by which PRP may be eliciting any effects before reflecting on some considerations for future studies in the area. Importantly, on the basis of our existing knowledge, we suggest that immediate use of PRP in clinical applications is perhaps premature and further fundamental and clinical research on the nature of ovarian insufficiency, as well as the mechanism by which PRP may act on the ovary, is needed to fully understand this promising development.
Collapse
Affiliation(s)
- Lloyd Atkinson
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK
| | - Francesca Martin
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK
| | - Roger G Sturmey
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK.,Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, St Mary's Hospital, Manchester, UK
| |
Collapse
|
12
|
Pereira MM, Mainigi M, Strauss JF. Secretory products of the corpus luteum and preeclampsia. Hum Reprod Update 2021; 27:651-672. [PMID: 33748839 PMCID: PMC8222764 DOI: 10.1093/humupd/dmab003] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 01/18/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Despite significant advances in our understanding of the pathophysiology of preeclampsia (PE), there are still many unknowns and controversies in the field. Women undergoing frozen-thawed embryo transfer (FET) to a hormonally prepared endometrium have been found to have an unexpected increased risk of PE compared to women who receive embryos in a natural FET cycle. The differences in risk have been hypothesized to be related to the absence or presence of a functioning corpus luteum (CL). OBJECTIVE AND RATIONALE To evaluate the literature on secretory products of the CL that could be essential for a healthy pregnancy and could reduce the risk of PE in the setting of FET. SEARCH METHODS For this review, pertinent studies were searched in PubMed/Medline (updated June 2020) using common keywords applied in the field of assisted reproductive technologies, CL physiology and preeclampsia. We also screened the complete list of references in recent publications in English (both animal and human studies) on the topics investigated. Given the design of this work as a narrative review, no formal criteria for study selection or appraisal were utilized. OUTCOMES The CL is a major source of multiple factors regulating reproduction. Progesterone, estradiol, relaxin and vasoactive and angiogenic substances produced by the CL have important roles in regulating its functional lifespan and are also secreted into the circulation to act remotely during early stages of pregnancy. Beyond the known actions of progesterone and estradiol on the uterus in early pregnancy, their metabolites have angiogenic properties that may optimize implantation and placentation. Serum levels of relaxin are almost undetectable in pregnant women without a CL, which precludes some maternal cardiovascular and renal adaptations to early pregnancy. We suggest that an imbalance in steroid hormones and their metabolites and polypeptides influencing early physiologic processes such as decidualization, implantation, angiogenesis and maternal haemodynamics could contribute to the increased PE risk among women undergoing programmed FET cycles. WIDER IMPLICATIONS A better understanding of the critical roles of the secretory products of the CL during early pregnancy holds the promise of improving the efficacy and safety of ART based on programmed FET cycles.
Collapse
Affiliation(s)
- María M Pereira
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Monica Mainigi
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Centre for Research on Reproduction and Women’s Health, University of Pennsylvania, Philadelphia, PA,19104 USA
| | - Jerome F Strauss
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Centre for Research on Reproduction and Women’s Health, University of Pennsylvania, Philadelphia, PA,19104 USA
| |
Collapse
|
13
|
Li C, Liu Z, Zhou J, Meng X, Liu S, Li W, Zhang X, Zhou J, Yao W, Dong C, Cao Y, Li R, Chen B, Jiang A, Jiang Y, Ning C, Zhao F, Wei Y, Sun SC, Tao J, Wu W, Shen M, Liu H. Insulin-like growth factor-I prevents hypoxia-inducible factor-1 alpha-dependent G1/S arrest by activating cyclin E/cyclin-dependent kinase2 via the phoshatidylinositol-3 kinase/AKT/forkhead box O1/Cdkn1b pathway in porcine granulosa cells†. Biol Reprod 2021; 102:116-132. [PMID: 31435642 DOI: 10.1093/biolre/ioz162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/23/2019] [Accepted: 08/12/2019] [Indexed: 11/13/2022] Open
Abstract
As the follicle develops, the thickening of the granulosa compartment leads to progressively deficient supply of oxygen in granulosa cells (GCs) due to the growing distances from the follicular vessels. These conditions are believed to cause hypoxia in GCs during folliculogenesis. Upon hypoxic conditions, several types of mammalian cells have been reported to undergo cell cycle arrest. However, it remains unclear whether hypoxia exerts any impact on cell cycle progression of GCs. On the other hand, although the GCs may live in a hypoxic environment, their mitotic capability appears to be unaffected in growing follicles. It thus raises the question whether there are certain intraovarian factors that might overcome the inhibitory effects of hypoxia. The present study provides the first evidence suggesting that cobalt chloride (CoCl2)-mimicked hypoxia prevented G1-to-S cell cycle progression in porcine GCs. In addition, we demonstrated that the inhibitory effects of CoCl2 on GCs cell cycle are mediated through hypoxia-inducible factor-1 alpha/FOXO1/Cdkn1b pathway. Moreover, we identified insulin-like growth factor-I (IGF-I) as an intrafollicular factor required for cell cycle recovery by binding to IGF-I receptor in GCs suffering CoCl2 stimulation. Further investigations confirmed a role of IGF-I in preserving G1/S progression of CoCl2-treated GCs via activating the cyclin E/cyclin-dependent kinase2 complex through the phoshatidylinositol-3 kinase/protein kinase B (AKT)/FOXO1/Cdkn1b axis. Although the present findings were based on a hypoxia mimicking model by using CoCl2, our study might shed new light on the regulatory mechanism of GCs cell cycle upon hypoxic stimulation.
Collapse
Affiliation(s)
- Chengyu Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhaojun Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jiaqi Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xueqin Meng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Shuo Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Weijian Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xue Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jilong Zhou
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Wang Yao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Chao Dong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yan Cao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Rongyang Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Baobao Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Aiwen Jiang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yi Jiang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Caibo Ning
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Fang Zhao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yinghui Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jingli Tao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wangjun Wu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Ming Shen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Honglin Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
14
|
Lim M, Brown HM, Kind KL, Thompson JG, Dunning KR. Hemoglobin: potential roles in the oocyte and early embryo†. Biol Reprod 2020; 101:262-270. [PMID: 31058953 DOI: 10.1093/biolre/ioz078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/02/2019] [Accepted: 05/04/2019] [Indexed: 12/13/2022] Open
Abstract
Hemoglobin (Hb) is commonly known for its capacity to bind and transport oxygen and carbon dioxide in erythroid cells. However, it plays additional roles in cellular function and health due to its capacity to bind other gases including nitric oxide. Further, Hb acts as a potent antioxidant, quenching reactive oxygen species. Despite its potential roles in cellular function, the preponderance of Hb research remains focused on its role in oxygen regulation. There is increasing evidence that Hb expression is more ubiquitous than previously thought, with Hb and its variants found in a myriad of cell types ranging from macrophages to spermatozoa. The majority of nonerythroid cell types that express Hb are situated within hypoxic environments, suggesting Hb may play a role in hypoxia-inducible factor-regulated gene expression by controlling the level of oxygen available or as an adaptation to low oxygen providing a mechanism to store oxygen. Oocyte maturation and preimplantation embryo development occur within the low oxygen environments of the antral follicle and oviduct/uterus, respectively. Interestingly, Hb was recently found in human cumulus and granulosa cells and murine cumulus-oocyte complexes and preimplantation embryos. Here, we consolidate and analyze the research generated todate on Hb expression in nonerythroid cells with a particular focus on reproductive cell types. We outline future directions of this research to elucidate the role of Hb during oocyte maturation and preimplantation embryo development and finally, we explore the potential clinical applications and benefits of Hb supplementation during the in vitro culture of gametes and embryos.
Collapse
Affiliation(s)
- Megan Lim
- Robinson Research Institute, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia.,Australian Research Council Centre of Excellence for Nanoscale BioPhotonics.,Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, South Australia, Australia
| | - Hannah M Brown
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Karen L Kind
- School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Jeremy G Thompson
- Robinson Research Institute, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia.,Australian Research Council Centre of Excellence for Nanoscale BioPhotonics.,Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, South Australia, Australia
| | - Kylie R Dunning
- Robinson Research Institute, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia.,Australian Research Council Centre of Excellence for Nanoscale BioPhotonics.,Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
15
|
Berisha B, Schams D, Sinowatz F, Rodler D, Pfaffl MW. Hypoxia-inducible factor-1alpha and nitric oxide synthases in bovine follicles close to ovulation and early luteal angiogenesis. Reprod Domest Anim 2020; 55:1573-1584. [PMID: 32869370 DOI: 10.1111/rda.13812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/05/2020] [Accepted: 08/23/2020] [Indexed: 11/30/2022]
Abstract
The objective of the study was to characterize expression patterns of hypoxia-inducible factor-1alpha (HIF1A), inducible nitric oxide synthase (iNOS) and endothelial (eNOS) isoforms in time-defined follicle classes before and after GnRH application in the cow. Ovaries containing pre-ovulatory follicles or corpora lutea were collected by transvaginal ovariectomy (n = 5 cows/group) as follow: (I) before GnRH administration; (II) 4h after GnRH; (III) 10h after GnRH; (IV) 20h after GnRH; (V) 25h after GnRH; and (VI) 60h after GnRH (early corpus luteum). The mRNA abundance of HIF1A in the follicle group before GnRH was high, followed by a significant down regulation afterwards with a minimum level 25h after GnRH (close to ovulation) and significant increase only after ovulation. The mRNA abundance of iNOS before GnRH was high, decreased significantly during LH surge, with minimum levels afterwards. In contrast, the mRNA of eNOS decreased in the follicle group 20h after GnRH, followed by a rapid and significant upregulation just after ovulation. Immunohistochemically, the granulosa cells of antral follicles and the eosinophils of the theca tissue as well of the early corpus luteum showed a strong staining for HIF1A. The location of the eosinophils could be clearly demonstrated by immunostaining with an eosinophil-specific antibody (EMBP) and transmission electron microscopy. In conclusion, the parallel and acute regulated expression patterns of HIF1A and NOS isoforms, specifically during the interval between the LH surge and ovulation, indicate that these paracrine factors are involved in the local mechanisms, regulating final follicle maturation, ovulation and early luteal angiogenesis.
Collapse
Affiliation(s)
- Bajram Berisha
- Animal Biotechnology, Faculty of Agriculture and Veterinary, University of Prishtina, Prishtinë, Kosovo.,Animal Physiology and Immunology Weihenstephan, Technical University of Munich, Freising, Germany
| | - Dieter Schams
- Animal Physiology and Immunology Weihenstephan, Technical University of Munich, Freising, Germany
| | - Fred Sinowatz
- Department of Veterinary Sciences, Ludwig Maximilian University of Munich, Munich, Germany
| | - Daniela Rodler
- Department of Veterinary Sciences, Ludwig Maximilian University of Munich, Munich, Germany
| | - Michael W Pfaffl
- Animal Physiology and Immunology Weihenstephan, Technical University of Munich, Freising, Germany
| |
Collapse
|
16
|
Fan J, Yu Y, Han X, He H, Luo Y, Yu S, Cui Y, Xu G, Wang L, Pan Y. The expression of hypoxia-inducible factor-1 alpha in primary reproductive organs of the female yak (Bos grunniens) at different reproductive stages. Reprod Domest Anim 2020; 55:1371-1382. [PMID: 32706432 DOI: 10.1111/rda.13783] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 07/19/2020] [Indexed: 12/12/2022]
Abstract
The yak (Bos grunniens) is the most important livestock animal in high-altitude regions owing to its prominent adaptability to cold conditions, nutritional deficiencies and hypoxia. The reproductive organs exhibit different histological appearances and physiological processes at different reproductive stages. Hypoxia-inducible factor-1 alpha (HIF-1α) is the regulatory subunit of HIF-1 that crucially regulates the response to hypoxia in mammalian organisms. The goal of our study was to investigate the expression and distribution of HIF-1α in the primary yak reproductive organs at different reproductive stages. Samples of the ovary, oviduct and uterus of 15 adult female yaks were collected and used in the experiment. The expression and localization of HIF-1α proteins and mRNA were investigated using quantitative real-time polymerase chain reaction (qRT-PCR), Western blot (WB) and immunohistochemistry (IHC). The results indicated that the expression of HIF-1α protein in the ovary was higher during the luteal phase than during the follicular phase and gestation period (p < .05). In the oviduct, HIF-1α protein was also more highly expressed during the luteal phase than during the follicular phase and gestation period (p < .01). However, in the uterus, the HIF-1α protein had stronger expression during the gestation period than during the follicular phase (p < .01) and luteal phase (p < .05). The expression of HIF-1α mRNA was similar to that of its protein. Immunohistochemical analysis revealed intense immunostaining of HIF-1α proteins in the follicular granulosa cells, granular luteal cells, villous epithelial cells of the oviduct, endometrial glandular epithelium and luminal epithelium, foetal villous trophoblast, and epithelia of caruncular crypts. This study showed that the expression of HIF-1α in the ovary, oviduct and uterus varies according to the stage of the reproductive cycle. This implies that HIF-1α plays an important role in regulating the stage-specific physiological function of yak reproductive organs under hypoxic environments.
Collapse
Affiliation(s)
- Jiangfeng Fan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yiteng Yu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Xiaohong Han
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Honghong He
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yuzhu Luo
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Sijiu Yu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yan Cui
- Technology and Research Center of Gansu Province for Embryonic Engineering of Bovine and Sheep & Goat, Lanzhou, China
| | - Gengquan Xu
- Technology and Research Center of Gansu Province for Embryonic Engineering of Bovine and Sheep & Goat, Lanzhou, China
| | - Libin Wang
- Technology and Research Center of Gansu Province for Embryonic Engineering of Bovine and Sheep & Goat, Lanzhou, China
| | - Yangyang Pan
- Technology and Research Center of Gansu Province for Embryonic Engineering of Bovine and Sheep & Goat, Lanzhou, China
| |
Collapse
|
17
|
Madu CO, Wang S, Madu CO, Lu Y. Angiogenesis in Breast Cancer Progression, Diagnosis, and Treatment. J Cancer 2020; 11:4474-4494. [PMID: 32489466 PMCID: PMC7255381 DOI: 10.7150/jca.44313] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/04/2020] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is a significant event in a wide range of healthy and diseased conditions. This process frequently involves vasodilation and an increase in vascular permeability. Numerous players referred to as angiogenic factors, work in tandem to facilitate the outgrowth of endothelial cells (EC) and the consequent vascularity. Conversely, angiogenic factors could also feature in pathological conditions. Angiogenesis is a critical factor in the development of tumors and metastases in numerous cancers. An increased level of angiogenesis is associated with decreased survival in breast cancer patients. Therefore, a good understanding of the angiogenic mechanism holds a promise of providing effective treatments for breast cancer progression, thereby enhancing patients' survival. Disrupting the initiation and progression of this process by targeting angiogenic factors such as vascular endothelial growth factor (Vegf)-one of the most potent member of the VEGF family- or by targeting transcription factors, such as Hypoxia-Inducible Factors (HIFs) that act as angiogenic regulators, have been considered potential treatment options for several types of cancers. The objective of this review is to highlight the mechanism of angiogenesis in diseases, specifically its role in the progression of malignancy in breast cancer, as well as to highlight the undergoing research in the development of angiogenesis-targeting therapies.
Collapse
Affiliation(s)
- Chikezie O. Madu
- Departments of Biological Sciences, University of Memphis, Memphis, TN 38152. USA
| | - Stephanie Wang
- Departments of Biology and Advanced Placement Biology, White Station High School, Memphis, TN 38117. USA
| | - Chinua O. Madu
- Departments of Biology and Advanced Placement Biology, White Station High School, Memphis, TN 38117. USA
| | - Yi Lu
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN 38163. USA
| |
Collapse
|
18
|
Abstract
There has been increasing interest in the role of hypoxia in the microenvironment of organs, because of the discovery of hypoxia-inducible factor-1 (HIF1), which acts as a transcription factor for many genes activated specifically under hypoxic conditions. The ovary changes day by day during the estrous cycle as it goes through phases of follicular growth, ovulation, and formation and regression of the corpus luteum (CL). These phenomena are regulated by hypothalamic and pituitary hormones, sex steroids, peptides and cytokines, as well as oxygen conditions. Hypoxia strongly induces angiogenesis via transcription of a potent angiogenic factor, vascular endothelial growth factor (VEGF), that is regulated by HIF1. A CL forms with a rapid increase of angiogenesis that is mainly induced by HIF1-VEGF signaling. Hypoxia also contributes to luteolysis by down-regulating progesterone synthesis and by up-regulating apoptosis of luteal cells. This review focuses on recent studies on the roles of hypoxia- and HIF1-regulated genes in the regulation of bovine CL function.
Collapse
Affiliation(s)
- Ryo Nishimura
- Laboratory of Theriogenology, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori 680-8553, Japan
| | - Kiyoshi Okuda
- Obihiro University of Agriculture & Veterinary Medicine, Hokkaido 080-8555, Japan
| |
Collapse
|
19
|
Lu E, Li C, Wang J, Zhang C. Inflammation and angiogenesis in the corpus luteum. J Obstet Gynaecol Res 2019; 45:1967-1974. [PMID: 31373134 DOI: 10.1111/jog.14076] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/15/2019] [Indexed: 12/15/2022]
Abstract
Angiogenesis is a very important process that helps establish and maintain the normal structure and function of the corpus luteum (CL). Early luteal development can be considered a kind of physiological injury with an inflammatory response; therefore, the inflammatory response may play an important role in the luteal angiogenesis. The inflammatory response is companied by activated leukocytes and their mediators. For luteal tissue, numerous activated leukocytes such as macrophages, neutrophils and eosinophils are present in the early luteal phase and are widely involved in neovascularization. The objective of this review is to describe the role of the inflammatory factors in the angiogenesis and to discuss their mechanism. Knowledge of action and mechanism of these inflammatory factors on angiogenic activity will be beneficial for the understanding of luteal function.
Collapse
Affiliation(s)
- Enhang Lu
- Joint Programme of Nanchang University and Queen Mary University of London, School of Medicine, Nanchang University, Nanchang, China
| | - Chunjie Li
- Forth Clinical College, School of Medicine, Nanchang University, Nanchang, China
| | - Jing Wang
- Department of Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Chunping Zhang
- Department of Cell Biology, School of Medicine, Nanchang University, Nanchang, China
| |
Collapse
|
20
|
C/EBPβ regulates Vegf gene expression in granulosa cells undergoing luteinization during ovulation in female rats. Sci Rep 2019; 9:714. [PMID: 30679486 PMCID: PMC6345775 DOI: 10.1038/s41598-018-36566-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 11/23/2018] [Indexed: 11/08/2022] Open
Abstract
The ovulatory LH-surge increases Vegf gene expression in granulosa cells (GCs) undergoing luteinization during ovulation. To understand the factors involved in this increase, we examined the roles of two transcription factors and epigenetic mechanisms in rat GCs. GCs were obtained from rats treated with eCG before, 4 h, 8 h, 12 h and 24 h after hCG injection. Vegf mRNA levels gradually increased after hCG injection and reached a peak at 12 h. To investigate the mechanism by which Vegf is up-regulated after hCG injection, we focused on C/EBPβ and HIF1α. Their protein expression levels were increased at 12 h. The binding activity of C/EBPβ to the Vegf promoter region increased after hCG injection whereas that of HIF1α did not at this time point. The C/EBPβ binding site had transcriptional activities whereas the HIF1α binding sites did not have transcriptional activities under cAMP stimulation. The levels of H3K9me3 and H3K27me3, which are transcriptional repression markers, decreased in the C/EBPβ binding region after hCG injection. The chromatin structure of this region becomes looser after hCG injection. These results show that C/EBPβ regulates Vegf gene expression with changes in histone modifications and chromatin structure of the promoter region in GCs undergoing luteinization during ovulation.
Collapse
|
21
|
Zhang Z, Huang Y, Zhang J, Liu Z, Lin Q, Wang Z. Activation of NF-κB signaling pathway during HCG-induced VEGF expression in luteal cells. Cell Biol Int 2019; 43:344-349. [PMID: 30597662 DOI: 10.1002/cbin.11090] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 12/21/2018] [Indexed: 12/25/2022]
Abstract
Vascular endothelial growth factor (VEGF) plays an essential role in luteal angiogenesis, the present study therefore utilized luteal cells cultured in vitro to further investigate the activation and contribution of nuclear factor (NF)-κB to VEGF expression induced by human chorionic gonadotrophin (HCG). The present results showed HCG induced VEGF expression as well as hypoxia-inducible factor (HIF)-1α mRNA and protein expressions, which was blocked by NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC). Further analysis found that these increases of VEGF and HIF-1α mRNA induced by HCG were also blocked by NF-κB siRNA transfection, which was consistent with PDTC treatment. However, HIF-1α siRNA treatment significantly decreased HCG induced-VEGF expression with no effect on NF-κB mRNA expression. Furthermore, combination of HIF-1α siRNA and PDTC treatment did not further decrease VEGF mRNA expression, and the result of chromatin immunoprecipitation indicated NF-κB may regulate HIF-1α transcription through binding with its promoter. Taken together, the present results clearly demonstrated that NF-κB was activated to regulate VEGF expression by increasing HIF-1α transcription in luteal cells treated with HCG. Therefore, the present study provided a new and important mechanism of luteal angiogenesis during the formation of corpus luteum in mammals.
Collapse
Affiliation(s)
- Zhenghong Zhang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, No. 8, Shangsan Road, Fuzhou 350007, P. R. China
| | - Yuxiu Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, P. R. China
| | - Jingwei Zhang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, No. 8, Shangsan Road, Fuzhou 350007, P. R. China
| | - Zhaoyuan Liu
- Provincial Key Laboratory for Developmental Biology and Neurosciences, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, No. 8, Shangsan Road, Fuzhou 350007, P. R. China
| | - Qingqiang Lin
- Provincial Key Laboratory for Developmental Biology and Neurosciences, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, No. 8, Shangsan Road, Fuzhou 350007, P. R. China
| | - Zhengchao Wang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, No. 8, Shangsan Road, Fuzhou 350007, P. R. China
| |
Collapse
|
22
|
Dao DT, Anez-Bustillos L, Jabbouri SS, Pan A, Kishikawa H, Mitchell PD, Fell GL, Baker MA, Watnick RS, Chen H, Rogers MS, Bielenberg DR, Puder M. A paradoxical method to enhance compensatory lung growth: Utilizing a VEGF inhibitor. PLoS One 2018; 13:e0208579. [PMID: 30566445 PMCID: PMC6300284 DOI: 10.1371/journal.pone.0208579] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 11/20/2018] [Indexed: 12/11/2022] Open
Abstract
Exogenous vascular endothelial growth factor (VEGF) accelerates compensatory lung growth (CLG) in mice after unilateral pneumonectomy. In this study, we unexpectedly discovered a method to enhance CLG with a VEGF inhibitor, soluble VEGFR1. Eight-week-old C57BL/6 male mice underwent left pneumonectomy, followed by daily intraperitoneal (ip) injection of either saline (control) or 20 μg/kg of VEGFR1-Fc. On post-operative day (POD) 4, mice underwent pulmonary function tests (PFT) and lungs were harvested for volume measurement and analyses of the VEGF signaling pathway. To investigate the role of hypoxia in mediating the effects of VEGFR1, experiments were repeated with concurrent administration of PT-2385, an inhibitor of hypoxia-induced factor (HIF)2α, via orogastric gavage at 10 mg/kg every 12 hours for 4 days. We found that VEGFR1-treated mice had increased total lung capacity (P = 0.006), pulmonary compliance (P = 0.03), and post-euthanasia lung volume (P = 0.049) compared to control mice. VEGFR1 treatment increased pulmonary levels of VEGF (P = 0.008) and VEGFR2 (P = 0.01). It also stimulated endothelial proliferation (P < 0.0001) and enhanced pulmonary surfactant production (P = 0.03). The addition of PT-2385 abolished the increase in lung volume and endothelial proliferation in response to VEGFR1. By paradoxically stimulating angiogenesis and enhancing lung growth, VEGFR1 could represent a new treatment strategy for neonatal lung diseases characterized by dysfunction of the HIF-VEGF pathway.
Collapse
Affiliation(s)
- Duy T. Dao
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States of America
- Department of Surgery, Boston Children’s Hospital, Boston, MA, United States of America
| | - Lorenzo Anez-Bustillos
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States of America
- Department of Surgery, Boston Children’s Hospital, Boston, MA, United States of America
| | - Sahir S. Jabbouri
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States of America
- Department of Surgery, Boston Children’s Hospital, Boston, MA, United States of America
| | - Amy Pan
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States of America
- Department of Surgery, Boston Children’s Hospital, Boston, MA, United States of America
| | - Hiroko Kishikawa
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States of America
- Department of Surgery, Boston Children’s Hospital, Boston, MA, United States of America
| | - Paul D. Mitchell
- Institutional Centers for Clinical and Translational Research, Boston Children’s Hospital, Boston, MA, United States of America
| | - Gillian L. Fell
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States of America
- Department of Surgery, Boston Children’s Hospital, Boston, MA, United States of America
| | - Meredith A. Baker
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States of America
- Department of Surgery, Boston Children’s Hospital, Boston, MA, United States of America
| | - Randolph S. Watnick
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States of America
- Department of Surgery, Boston Children’s Hospital, Boston, MA, United States of America
| | - Hong Chen
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States of America
- Department of Surgery, Boston Children’s Hospital, Boston, MA, United States of America
| | - Michael S. Rogers
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States of America
- Department of Surgery, Boston Children’s Hospital, Boston, MA, United States of America
| | - Diane R. Bielenberg
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States of America
- Department of Surgery, Boston Children’s Hospital, Boston, MA, United States of America
| | - Mark Puder
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States of America
- Department of Surgery, Boston Children’s Hospital, Boston, MA, United States of America
| |
Collapse
|
23
|
Baddela VS, Sharma A, Viergutz T, Koczan D, Vanselow J. Low Oxygen Levels Induce Early Luteinization Associated Changes in Bovine Granulosa Cells. Front Physiol 2018; 9:1066. [PMID: 30131718 PMCID: PMC6090175 DOI: 10.3389/fphys.2018.01066] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/17/2018] [Indexed: 11/29/2022] Open
Abstract
During follicle maturation, oxygen levels continuously decrease in the follicular fluid and reach lowest levels in the preovulatory follicle. The current study was designed to comprehensively understand effects of low oxygen levels on bovine granulosa cells (GC) using our established estrogen active GC culture model. As evident from flow cytometry analysis the viability of GC was not found to be affected at severely low oxygen condition (1% O2) compared to normal (atmospheric) oxygen condition (21% O2). Estimations of hormone concentrations using competitive radioimmunoassay revealed that the production of estradiol and progesterone was significantly reduced at low oxygen condition. To understand the genome-wide changes of gene expression, mRNA microarray analysis was performed using Affymetrix's Bovine Gene 1.0 ST Arrays. This resulted in the identification of 1104 differentially regulated genes of which 505 were up- and 599 down-regulated under low oxygen conditions. Pathway analysis using Ingenuity pathway analyzer (IPA) identified 36 significantly affected (p < 0.05) canonical pathways. Importantly, pathways like "Estrogen-mediated S-phase Entry" and "Cyclins and Cell Cycle Regulation" were found to be greatly down-regulated at low oxygen levels. This was experimentally validated using flow cytometry based cell cycle analysis. Up-regulation of critical genes associated with angiogenesis, inflammation, and glucose metabolism, and down-regulation of FSH signaling, steroidogenesis and cell proliferation indicated that low oxygen levels induced early luteinization associated changes in granulosa cells. Identification of unmethylated CpG sites in the CYP19A1 promoter region suggests that granulosa cells were not completely transformed into luteal cells under the present low oxygen in vitro condition. In addition, the comparison with earlier published in vivo microarray data indicated that 1107 genes showed a similar expression pattern in granulosa cells at low oxygen levels (in vitro) as found in preovulatory follicles after the LH surge (in vivo). Overall, our findings demonstrate for the first time that low oxygen levels in preovulatory follicles may play an important role in supporting early events of luteinization in granulosa cells.
Collapse
Affiliation(s)
- Vijay S. Baddela
- Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Arpna Sharma
- Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Torsten Viergutz
- Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Dirk Koczan
- Institute of Immunology, University of Rostock, Rostock, Germany
| | - Jens Vanselow
- Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| |
Collapse
|
24
|
Shrestha K, Onasanya AE, Eisenberg I, Wigoda N, Yagel S, Yalu R, Meidan R, Imbar T. miR-210 and GPD1L regulate EDN2 in primary and immortalized human granulosa-lutein cells. Reproduction 2018; 155:197-205. [PMID: 29301980 DOI: 10.1530/rep-17-0574] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/22/2017] [Accepted: 12/04/2017] [Indexed: 11/08/2022]
Abstract
Endothelin-2 (EDN2), expressed at a narrow window during the periovulatory period, critically affects ovulation and corpus luteum (CL) formation. LH (acting mainly via cAMP) and hypoxia are implicated in CL formation; therefore, we aimed to elucidate how these signals regulate EDN2 using human primary (hGLCs) and immortalized (SVOG) granulosa-lutein cells. The hypoxiamiR, microRNA-210 (miR-210) was identified as a new essential player in EDN2 expression. Hypoxia (either mimetic compound-CoCl2, or low O2) elevated hypoxia-inducible factor 1A (HIF1A), miR-210 and EDN2 Hypoxia-induced miR-210 was suppressed in HIF1A-silenced SVOG cells, suggesting that miR-210 is HIF1A dependent. Elevated miR-210 levels in hypoxia or by miR-210 overexpression, increased EDN2 Conversely, miR-210 inhibition reduced EDN2 levels, even in the presence of CoCl2, indicating the importance of miR-210 in the hypoxic induction of EDN2 A molecule that destabilizes HIF1A protein, glycerol-3-phosphate dehydrogenase 1-like gene-GPD1L, was established as a miR-210 target in both cell types. It was decreased by miR-210-mimic and was increased by miR-inhibitor. Furthermore, reducing GPD1L by endogenously elevated miR-210 (in hypoxia), miR-210-mimic or by GPD1L siRNA resulted in elevated HIF1A protein and EDN2 levels, implying a vital role for GPD1L in the hypoxic induction of EDN2 Under normoxic conditions, forskolin (adenylyl cyclase activator) triggered changes typical of hypoxia. It elevated HIF1A, EDN2 and miR-210 while inhibiting GPD1L Furthermore, HIF1A silencing greatly reduced forskolin's ability to elevate EDN2 and miR-210. This study highlights the novel regulatory roles of miR-210 and its gene target, GPD1L, in hypoxia and cAMP-induced EDN2 by human granulosa-lutein cells.
Collapse
Affiliation(s)
- Ketan Shrestha
- Department of Animal SciencesThe Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Adepeju Esther Onasanya
- Department of Animal SciencesThe Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Iris Eisenberg
- The Magda and Richard Hoffman Center for Human Placenta ResearchHadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Noa Wigoda
- Department of Animal SciencesThe Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Simcha Yagel
- The Magda and Richard Hoffman Center for Human Placenta ResearchHadassah Hebrew University Medical Center, Jerusalem, Israel.,Department of Obstetrics and GynecologyHadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Ronit Yalu
- Department of Animal SciencesThe Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Rina Meidan
- Department of Animal SciencesThe Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Tal Imbar
- The Magda and Richard Hoffman Center for Human Placenta ResearchHadassah Hebrew University Medical Center, Jerusalem, Israel.,Department of Obstetrics and GynecologyHadassah Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
25
|
Chen X, Liu Y, Shan Y, Jin X, Shi Q, Jia C. Oxidized low-density lipoprotein suppresses mouse granulosa cell differentiation through disruption of the hypoxia-inducible factor 1 pathway. Mol Reprod Dev 2017; 84:1306-1313. [PMID: 29155477 DOI: 10.1002/mrd.22933] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 11/06/2017] [Indexed: 12/22/2022]
Abstract
Obesity predisposes women to reproductive disorders. One symptom of obesity in women is higher levels of oxidized Low-density lipoprotein (oxLDL) in serum and preovulatory follicles. The present study was designed to test the hypothesis that oxLDL might impair follicle differentiation and luteinization. Given that Hypoxia-inducible factor 1 (HIF1) plays crucial roles in supporting follicle differentiation and luteinization in mammals, we focused on oxLDL-mediated events that may affect the HIF1 pathway. We report that exposure to oxLDL diminished the expression of HIF1α and its target genes and suppressed the differentiation of mouse luteinized granulosa cells following induction by human Chorionic gonadotophin (hCG) under hypoxic conditions (1% oxygen). Significantly, the proteasome inhibitor MG-132 prevented this oxLDL-attenuation differentiation phenotype by blocking HIF1α degradation. Together, these findings suggest that suppression of granulosa cell differentiation by oxLDL, via HIF1α down-regulation, may contribute the negative effects of obesity on female fertility.
Collapse
Affiliation(s)
- Xiaoliang Chen
- Departmentof Urology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yanhong Liu
- Centre for Reproductive Medicine, Centre for Prenatal Diagnosis, First Hospital, Jilin University, Changchun, China
| | - Yanhong Shan
- Department of Obstetrics, First Hospital, Jilin University, Changchun, China
| | - Xingyi Jin
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Qingyang Shi
- Centre for Reproductive Medicine, Centre for Prenatal Diagnosis, First Hospital, Jilin University, Changchun, China
| | - Chunshu Jia
- Centre for Reproductive Medicine, Centre for Prenatal Diagnosis, First Hospital, Jilin University, Changchun, China
| |
Collapse
|
26
|
Abstract
Endocrine is an important and tightly regulated system for maintaining body homeostasis. Endocrine glands produce hormones, which are released into blood stream to guide the target cells responding to all sorts of stimulations. For maintaining body homeostasis, the secretion and activity of a particular hormone needs to be adjusted in responding to environmental challenges such as changes in nutritional status or chronic stress. Hypoxia, a status caused by reduced oxygen availability or imbalance of oxygen consumption/supply in an organ or within a cell, is a stress that affects many physiological and pathological processes. Hypoxic stress in endocrine organs is especially critical because endocrine glands control body homeostasis. Local hypoxia affects not only the particular gland but also the downstream cells/organs regulated by hormones secreted from this gland. Hypoxia-inducible factors (HIFs) are transcription factors that function as master regulators of oxygen homeostasis. Recent studies report that aberrant expression of HIFs in endocrine organs may result in the development and/or progression of diseases including diabetes, endometriosis, infertility and cancers. In this article, we will review recent findings in HIF-mediated endocrine organ dysfunction and the systemic syndromes caused by these disorders.
Collapse
Affiliation(s)
- Hsiu-Chi Lee
- Institute of Basic Medical SciencesCollege of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shaw-Jenq Tsai
- Institute of Basic Medical SciencesCollege of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of PhysiologyCollege of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
27
|
Fadhillah, Yoshioka S, Nishimura R, Yamamoto Y, Kimura K, Okuda K. Hypoxia-inducible factor 1 mediates hypoxia-enhanced synthesis of progesterone during luteinization of granulosa cells. J Reprod Dev 2016; 63:75-85. [PMID: 27840375 PMCID: PMC5320433 DOI: 10.1262/jrd.2016-068] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Hypoxia has been suggested to enhance progesterone (P4) synthesis in luteinizing granulosa cells (GCs), but the mechanism is unclear. The present study was designed to test the hypothesis that the hypoxia-induced increase in P4 synthesis during luteinization in bovine GCs is mediated by hypoxia-inducible factor 1 (HIF-1). GCs obtained from small antral follicles were cultured with 2 µg/ml insulin in combination with 10 µM forskolin for 24 h as a model of luteinizing GCs. To examine the influence of HIF-1 on P4 synthesis, we determined the effect of changes in protein expression of the α-subunit of HIF-1 (HIF1A) on P4 production and on the expression levels of StAR, P450scc, and 3β-HSD. CoCl2 (100 µM), a hypoxia-mimicking chemical, increased HIF-1α protein expression in luteinizing GCs. After the upregulation of HIF-1α, we observed an increase in P4 production and in the gene and protein expression levels of StAR in CoCl2-treated luteinizing GCs. In contrast, CoCl2 did not affect the expression of either P450scc or 3β-HSD. Echinomycin, a small-molecule inhibitor of HIF-1's DNA-binding activity, attenuated the effects of CoCl2 and of low oxygen tension (10% O2) on P4 production and StAR expression in luteinizing GCs. Overall, these findings suggest that HIF-1 is one of the factors that upregulate P4 in GCs during luteinization.
Collapse
Affiliation(s)
- Fadhillah
- Laboratory of Reproductive Physiology, Graduate School of Environmental and Life Sciences, Okayama University, Okayama 700-8530, Japan
| | | | | | | | | | | |
Collapse
|
28
|
Berisha B, Schams D, Rodler D, Sinowatz F, Pfaffl MW. Expression pattern of HIF1alpha and vasohibins during follicle maturation and corpus luteum function in the bovine ovary. Reprod Domest Anim 2016; 52:130-139. [PMID: 27862406 DOI: 10.1111/rda.12867] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 09/10/2016] [Indexed: 10/20/2022]
Abstract
The aim of this study was to characterize expression patterns of hypoxia-inducible factor-1alpha (HIF1A) and vasohibin family members (VASH1 and VASH2) during different stages of ovarian function in cow. Experiment 1: Antral follicle classification occurred by follicle size and estradiol-17beta (E2) concentration in the follicular fluid into 5 groups (<0.5, 0.5-5, 5-40, 40-180 and >180 E2 ng/ml). Experiment 2: Corpora lutea (CL) were assigned to the following stages: days 1-2, 3-4, 5-7, 8-12, 13-16 and >18 (after regression) of oestrous cycle and of pregnancy (months 1-2, 3-4, 6-7, >8). Experiment 3: Cows on days 8-12 were injected with a prostaglandin F2alpha (PGF) analogue and CL were collected before and 0.5, 2, 4, 12, 24, 48 and 64 hr after PGF injection. Expression of mRNA was measured by qPCR, steroid hormone concentration by EIA and localization by immunohistochemistry. HIF1A mRNA expression in our study increases significantly in follicles during final maturation. The highest HIF1A mRNA expression was detected during the early luteal phase, followed by a significant decrease afterwards. In contrast, the mRNA of vasohibins in small follicle was high, followed by a continuous and significant downregulation in preovulatory follicles. The obtained results show a remarkable inverse expression and localization pattern of HIF1A and vasohibins during different stages of ovarian function in cow. These results lead to the assumption that the examined factors are involved in the local mechanisms regulating angiogenesis and that the interactions between proangiogenic (HIF1A) and antiangiogenic (vasohibins) factors impact all stages of bovine ovary function.
Collapse
Affiliation(s)
- B Berisha
- Faculty of Agriculture and Veterinary, University of Prishtina, Prishtinë, Kosovo.,Institute of Animal Physiology and Immunology Weihenstephan, Technical University of Munich, Freising, Germany
| | - D Schams
- Institute of Animal Physiology and Immunology Weihenstephan, Technical University of Munich, Freising, Germany
| | - D Rodler
- Department of Veterinary Sciences, Institute of Anatomy, Histology and Embryology, Ludwig Maximilian University of Munich, Munich, Germany
| | - F Sinowatz
- Department of Veterinary Sciences, Institute of Anatomy, Histology and Embryology, Ludwig Maximilian University of Munich, Munich, Germany
| | - M W Pfaffl
- Institute of Animal Physiology and Immunology Weihenstephan, Technical University of Munich, Freising, Germany
| |
Collapse
|
29
|
Yang YL, Ren LR, Sun LF, Huang C, Xiao TX, Wang BB, Chen J, Zabel BA, Ren P, Zhang JV. The role of GPR1 signaling in mice corpus luteum. J Endocrinol 2016; 230:55-65. [PMID: 27149986 PMCID: PMC5064765 DOI: 10.1530/joe-15-0521] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/04/2016] [Indexed: 01/17/2023]
Abstract
Chemerin, a chemokine, plays important roles in immune responses, inflammation, adipogenesis, and carbohydrate metabolism. Our recent research has shown that chemerin has an inhibitory effect on hormone secretion from the testis and ovary. However, whether G protein-coupled receptor 1 (GPR1), the active receptor for chemerin, regulates steroidogenesis and luteolysis in the corpus luteum is still unknown. In this study, we established a pregnant mare serum gonadotropin-human chorionic gonadotropin (PMSG-hCG) superovulation model, a prostaglandin F2α (PGF2α) luteolysis model, and follicle and corpus luteum culture models to analyze the role of chemerin signaling through GPR1 in the synthesis and secretion of gonadal hormones during follicular/luteal development and luteolysis. Our results, for the first time, show that chemerin and GPR1 are both differentially expressed in the ovary over the course of the estrous cycle, with highest levels in estrus and metestrus. GPR1 has been localized to granulosa cells, cumulus cells, and the corpus luteum by immunohistochemistry (IHC). In vitro, we found that chemerin suppresses hCG-induced progesterone production in cultured follicle and corpus luteum and that this effect is attenuated significantly by anti-GPR1 MAB treatment. Furthermore, when the phosphoinositide 3-kinase (PI3K) pathway was blocked, the attenuating effect of GPR1 MAB was abrogated. Interestingly, PGF2α induces luteolysis through activation of caspase-3, leading to a reduction in progesterone secretion. Treatment with GPR1 MAB blocked the PGF2α effect on caspase-3 expression and progesterone secretion. This study indicates that chemerin/GPR1 signaling directly or indirectly regulates progesterone synthesis and secretion during the processes of follicular development, corpus luteum formation, and PGF2α-induced luteolysis.
Collapse
Affiliation(s)
- Ya-Li Yang
- Research Laboratory for Reproductive HealthShenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Li-Rong Ren
- Shenzhen Key Laboratory of Birth DefectsShenzhen Baoan Maternal and Child Health Hospital, Shenzhen, Guangdong, China
| | - Li-Feng Sun
- Research Laboratory for Reproductive HealthShenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chen Huang
- Research Laboratory for Reproductive HealthShenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China University of Chinese Academy of SciencesShenzhen, China
| | - Tian-Xia Xiao
- Research Laboratory for Reproductive HealthShenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bao-Bei Wang
- Research Laboratory for Reproductive HealthShenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jie Chen
- Research Laboratory for Reproductive HealthShenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Brian A Zabel
- Laboratory of Immunology and Vascular BiologyDepartment of Pathology, Stanford University School of Medicine, Stanford, California, USA, and Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Peigen Ren
- Research Laboratory for Reproductive HealthShenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jian V Zhang
- Research Laboratory for Reproductive HealthShenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
30
|
Zhang Y, Wang SF, Zheng JD, Zhao CB, Zhang YN, Liu LL, Huang JH. Effects of testosterone on the expression levels of AMH, VEGF and HIF-1α in mouse granulosa cells. Exp Ther Med 2016; 12:883-888. [PMID: 27446291 PMCID: PMC4950823 DOI: 10.3892/etm.2016.3436] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 10/22/2015] [Indexed: 01/01/2023] Open
Abstract
The present study aimed to investigate the effects of testosterone on mouse granulosa cell morphology, and the expression levels of anti-Müllerian hormone (AMH), vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1α (HIF-1α). Mouse granulosa cells were isolated and identified, and their morphology was examined using hematoxylin and eosin, F-actin, and follicle-stimulating hormone receptor staining. The mRNA expression levels of AMH, VEGF and HIF-1α were examined using reverse transcription-quantitative polymerase chain reaction, and their protein secretion levels were investigated using enzyme-linked immunosorbent assays. Testosterone treatment did not affect granulosa cell morphology; however, it significantly increased the mRNA expression levels of AMH and VEGF, and the protein secretion levels of AMH, VEGF and HIF-1α. These results suggested that testosterone was able to regulate the functions of granulosa cells by upregulating the expression levels of AMH, VEGF and HIF-1α.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China; International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200030, P.R. China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Jiao Tong University, School of Medicine, Shanghai 200030, P.R. China
| | - Shan-Feng Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Jin-Dan Zheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Chun-Bo Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China; Department of Obstetrics and Gynecology, The Second Affiliated Hospital (Jiande Branch), Medical School of Zhejiang University, The First People's Hospital of Jiande, Hangzhou, Zhejiang 311600, P.R. China
| | - Yi-Nan Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Li-Li Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Jian-Hua Huang
- Department of Key Laboratory of Tissue Engineering, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| |
Collapse
|
31
|
Kind KL, Tam KKY, Banwell KM, Gauld AD, Russell DL, Macpherson AM, Brown HM, Frank LA, Peet DJ, Thompson JG. Oxygen-regulated gene expression in murine cumulus cells. Reprod Fertil Dev 2015; 27:407-18. [PMID: 24388334 DOI: 10.1071/rd13249] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 11/15/2013] [Indexed: 12/19/2022] Open
Abstract
Oxygen is an important component of the environment of the cumulus-oocyte complex (COC), both in vivo within the ovarian follicle and during in vitro oocyte maturation (IVM). Cumulus cells have a key role in supporting oocyte development, and cumulus cell function and gene expression are known to be altered when the environment of the COC is perturbed. Oxygen-regulated gene expression is mediated through the actions of the transcription factors, the hypoxia-inducible factors (HIFs). In the present study, the effect of oxygen on cumulus cell gene expression was examined following in vitro maturation of the murine COC at 2%, 5% or 20% oxygen. Increased expression of HIF-responsive genes, including glucose transporter-1, lactate dehydrogenase A and BCL2/adenovirus E1B interacting protein 3, was observed in cumulus cells matured at 2% or 5%, compared with 20% oxygen. Stabilisation of HIF1α protein in cumulus cells exposed to low oxygen was confirmed by western blot and HIF-mediated transcriptional activity was demonstrated using a transgenic mouse expressing green fluorescent protein under the control of a promoter containing hypoxia response elements. These results indicate that oxygen concentration influences cumulus cell gene expression and support a role for HIF1α in mediating the cumulus cell response to varying oxygen.
Collapse
Affiliation(s)
- Karen L Kind
- The Robinson Institute, Research Centre for Reproductive Health, University of Adelaide, Adelaide, SA 5005, Australia
| | - Kimberley K Y Tam
- The Robinson Institute, Research Centre for Reproductive Health, University of Adelaide, Adelaide, SA 5005, Australia
| | - Kelly M Banwell
- The Robinson Institute, Research Centre for Reproductive Health, University of Adelaide, Adelaide, SA 5005, Australia
| | - Ashley D Gauld
- The Robinson Institute, Research Centre for Reproductive Health, University of Adelaide, Adelaide, SA 5005, Australia
| | - Darryl L Russell
- The Robinson Institute, Research Centre for Reproductive Health, University of Adelaide, Adelaide, SA 5005, Australia
| | - Anne M Macpherson
- The Robinson Institute, Research Centre for Reproductive Health, University of Adelaide, Adelaide, SA 5005, Australia
| | - Hannah M Brown
- The Robinson Institute, Research Centre for Reproductive Health, University of Adelaide, Adelaide, SA 5005, Australia
| | - Laura A Frank
- The Robinson Institute, Research Centre for Reproductive Health, University of Adelaide, Adelaide, SA 5005, Australia
| | - Daniel J Peet
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA 5005, Australia
| | - Jeremy G Thompson
- The Robinson Institute, Research Centre for Reproductive Health, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
32
|
Induction of chemokines and prostaglandin synthesis pathways in luteinized human granulosa cells: potential role of luteotropin withdrawal and prostaglandin F2α in regression of the human corpus luteum. Reprod Biol 2015; 15:247-56. [PMID: 26679166 DOI: 10.1016/j.repbio.2015.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/29/2015] [Accepted: 10/12/2015] [Indexed: 01/08/2023]
Abstract
Our objective was to determine the effects of prostaglandin F2α (PGF2α) and withdrawal of luteotropic stimulants (forskolin or hCG) on expression of chemokines and prostaglandin-endoperoxide synthase 2 (PTGS2) in luteinized human granulosa cells. Human granulosa cells were collected from 12 women undergoing oocyte retrieval and were luteinized in vitro with forskolin or hCG. In first experiment, granulosa-lutein cells were treated with PGF2α, the primary luteolytic hormone in most species. In second experiment, granulosa cells that had been luteinized for 8 d had luteotropins withdrawn for 1, 2, or 3 d. Treatment with PGF2α induced mRNA for chemokine (c-x-c motif) ligand 2 (CXCL2) and CXC ligand 8 (CXCL8; also known as interleukin-8) in granulosa cells luteinized for 8 d but not in cells that were only luteinized for 2 d. Similarly, luteinization of human granulosa cells for 8 d with forskolin or hCG followed by withdrawal of luteotropic stimulants, not only decreased P4 production, but also increased mRNA concentrations for CXCL8, CXCL-2 (after forskolin withdrawal), and PTGS2. These results provide evidence for two key steps in differentiation of luteolytic capability in human granulosa cells. During 8 d of luteinization, granulosa cells acquire the ability to respond to luteolytic factors, such as PGF2α, with induction of genes involved in immune function and PG synthesis. Finally, a decline in luteotropic stimuli triggers similar pathways leading to induction of PTGS2 and possibly intraluteal PGF2α production, chemokine expression, leukocyte infiltration and activation, and ultimately luteal regression.
Collapse
|
33
|
Wu L, Zhang Z, Pan X, Wang Z. Expression and contribution of the HIF-1α/VEGF signaling pathway to luteal development and function in pregnant rats. Mol Med Rep 2015; 12:7153-9. [PMID: 26323652 DOI: 10.3892/mmr.2015.4268] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 08/04/2015] [Indexed: 11/06/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is vital in normal and abnormal angiogenesis in the ovary, particularly during the early development of the corpus luteum in the ovary. However, the molecular regulation of the expression VEGF during luteal development in vivo remains to be fully elucidated. As the expression of VEGF is mediated by hypoxia‑inducible factor (HIF)‑1α in luteal cells cultured in vitro, determined in our previous study, the present study was performed to confirm the hypothesis that HIF‑1α is induced and then regulates the expression of VEGF and VEGF‑dependent luteal development/function in vivo. This was investigated using a pregnant rat model treated with a small‑molecule inhibitor of HIF‑1α, echinomycin (Ech). The development of the corpus luteum in the pregnant rat ovary was identified via performing assays of the serum progesterone, testosterone and estradiol concentrations by radioimmunoassay, accompanied with determination of the changes in the expression levels of HIF‑1α and VEGF by reverse transcription‑quantitative polymerase chain reaction at different days of the developmental process. On day 5, serum progesterone levels were markedly increased, whereas serum levels of testosterone and estradiol did not change significantly. On day 17, the highest level of serum progesterone was observed, however, this was not the case for testosterone and estradiol. Further analysis of the expression levels of HIF‑1α and VEGF revealed that their changes were consistent with the changes in serum levels of progesterone, which occurred in the development of the corpus luteum in the ovaries of pregnant rats. Further investigation demonstrated that Ech inhibited luteal development through inhibiting the expression of VEGF, mediated by HIF‑1α, and subsequent luteal function, which was determined by detecting changes in serum progesterone on days 8 and 14. Taken together, these results demonstrated that HIF‑1α‑mediated expression of VEGF may be one of the important mechanisms regulating ovarian luteal development in mammals in vivo, which may provide novel strategies in treatment for fertility control and for certain types of ovarian dysfunction, including polycystic ovarian syndrome, ovarian hyperstimulation syndrome and ovarian neoplasia.
Collapse
Affiliation(s)
- Lixiang Wu
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian 350108, P.R. China
| | - Zhenghong Zhang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian 350108, P.R. China
| | - Xiaoyan Pan
- Department of Histology and Embryology, Jilin Medical College, Jilin, Jilin 132013, P.R. China
| | - Zhengchao Wang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian 350108, P.R. China
| |
Collapse
|
34
|
Yalu R, Oyesiji AE, Eisenberg I, Imbar T, Meidan R. HIF1A-dependent increase in endothelin 2 levels in granulosa cells: role of hypoxia, LH/cAMP, and reactive oxygen species. Reproduction 2015; 149:11-20. [PMID: 25433027 DOI: 10.1530/rep-14-0409] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hypoxia-inducible factor 1 alpha (HIF1A) and endothelin 2 (EDN2) are transiently expressed during the same time window in the developing corpus luteum (CL). In this study, we sought to investigate the involvement of LH/cAMP, reactive oxygen species (ROS), and a hypoxia-mimetic compound (CoCl2) on HIF1A expression and how it affected EDN2 levels, using transformed human granulosa cells (thGCs) and primary bovine granulosa cells (GCs). CoCl2 elevated HIF1A protein levels in thGCs in a dose-dependent manner. Forskolin alone had no significant effect; however, forskolin and CoCl2 together further induced HIF1A protein and EDN2 mRNA expression in thGCs. Similarly, in primary GCs, LH with CoCl2 synergistically augmented HIF1A protein levels, which resulted in higher expression of EDN2 and another well-known hypoxia-inducible gene, VEGF (VEGFA). Importantly, LH alone elevated HIF1A mRNA but not its protein. The successful knockdown of HIF1A in thGCs using siRNA abolished hypoxia-induced EDN2 and also the additive effect of forskolin and CoCl2. We then examined the roles of ROS in thGCs: hydrogen peroxide (20 and 50 μM) elevated HIF1A protein as well as the expression of EDN2, implying that induction of HIF1A protein levels is sufficient to stimulate the expression of EDN2 (and VEGF) in normoxia. A broad-range ROS scavenger, butylated hydroxyanisole, inhibited CoCl2-induced HIF1A protein with a concomitant reduction in the mRNA expression of EDN2 and VEGF in thGCs. The results obtained in this study suggest that HIF1A, induced by various stimuli, is an essential mediator of EDN2 mRNA expression. The results may also explain the rise in the levels of HIF1A-dependent genes (EDN2 and VEGF) in the developing CL.
Collapse
Affiliation(s)
- Ronit Yalu
- Department of Animal SciencesThe Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Herzl Street, Rehovot 76100, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Adepeju Esther Oyesiji
- Department of Animal SciencesThe Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Herzl Street, Rehovot 76100, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Iris Eisenberg
- Department of Animal SciencesThe Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Herzl Street, Rehovot 76100, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Tal Imbar
- Department of Animal SciencesThe Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Herzl Street, Rehovot 76100, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Rina Meidan
- Department of Animal SciencesThe Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Herzl Street, Rehovot 76100, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| |
Collapse
|
35
|
Bunel A, Nivet AL, Blondin P, Vigneault C, Richard FJ, Sirard MA. Cumulus cell gene expression associated with pre-ovulatory acquisition of developmental competence in bovine oocytes. Reprod Fertil Dev 2015; 26:855-65. [PMID: 23827322 DOI: 10.1071/rd13061] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Accepted: 06/04/2013] [Indexed: 12/24/2022] Open
Abstract
The final days before ovulation impact significantly on follicular function and oocyte quality. This study investigated the cumulus cell (CC) transcriptomic changes during the oocyte developmental competence acquisition period. Six dairy cows were used for 24 oocyte collections and received FSH twice daily over 3 days, followed by FSH withdrawal for 20, 44, 68 and 92 h in four different oestrous cycles for each of the six cows. Half of the cumulus-oocyte complexes were subjected to in vitro maturation, fertilisation and culture to assess blastocyst rate. The other half of the CC underwent microarray analysis (n=3 cows, 12 oocyte collections) and qRT-PCR (n=3 other cows, 12 oocyte collections). According to blastocyst rates, 20 h of FSH withdrawal led to under-differentiated follicles (49%), 44 and 68 h to the most competent follicles (71% and 61%) and 92 h to over-differentiated ones (51%). Ten genes, from the gene lists corresponding to the three different follicular states, were subjected to qRT-PCR. Interestingly, CYP11A1 and NSDHL gene expression profiles reflected the blastocyst rate. However most genes were associated with the over-differentiated status: GATM, MAN1A1, VNN1 and NRP1. The early period of FSH withdrawal has a minimal effect on cumulus gene expression, whereas the longest period has a very significant one and indicates the beginning of the atresia process.
Collapse
Affiliation(s)
- A Bunel
- Centre de Recherche en Biologie de la Reproduction, Faculté des Sciences de l'Agriculture et de l'Alimentation, Département des Sciences Animales, Université Laval, Québec, QC G1V 0A6, Canada
| | - A L Nivet
- Centre de Recherche en Biologie de la Reproduction, Faculté des Sciences de l'Agriculture et de l'Alimentation, Département des Sciences Animales, Université Laval, Québec, QC G1V 0A6, Canada
| | - P Blondin
- L'Alliance Boviteq, 19320 Grand rang St-François, Saint-Hyacinthe, Québec, QC J2T 5H1, Canada
| | - C Vigneault
- L'Alliance Boviteq, 19320 Grand rang St-François, Saint-Hyacinthe, Québec, QC J2T 5H1, Canada
| | - F J Richard
- Centre de Recherche en Biologie de la Reproduction, Faculté des Sciences de l'Agriculture et de l'Alimentation, Département des Sciences Animales, Université Laval, Québec, QC G1V 0A6, Canada
| | - M A Sirard
- Centre de Recherche en Biologie de la Reproduction, Faculté des Sciences de l'Agriculture et de l'Alimentation, Département des Sciences Animales, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
36
|
Thompson JG, Brown HM, Kind KL, Russell DL. The Ovarian Antral Follicle: Living on the Edge of Hypoxia or Not?1. Biol Reprod 2015; 92:153. [DOI: 10.1095/biolreprod.115.128660] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/22/2015] [Indexed: 11/01/2022] Open
|
37
|
Zhang Z, Pang X, Tang Z, Yin D, Wang Z. Overexpression of hypoxia-inducible factor prolyl hydoxylase-2 attenuates hypoxia-induced vascular endothelial growth factor expression in luteal cells. Mol Med Rep 2015; 12:3809-3814. [PMID: 25975603 DOI: 10.3892/mmr.2015.3788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 04/15/2015] [Indexed: 11/06/2022] Open
Abstract
Vascular endothelial growth factor (VEGF)-dependent angiogenesis has a crucial role in the corpus luteum formation and their functional maintenances in mammalian ovaries. A previous study by our group reported that activation of hypoxia‑inducible factor (HIF)‑1α signaling contributes to the regulation of VEGF expression in the luteal cells (LCs) in response to hypoxia and human chorionic gonadotropin. The present study was designed to test the hypothesis that HIF prolyl‑hydroxylases (PHDs) are expressed in LCs and overexpression of PHD2 attenuates the expression of VEGF induced by hypoxia in LCs. PHD2-overexpressing plasmid was transfected into LC2 cells, and successful plasmid transfection and expression was confirmed by reverse transcription quantitative polymerase chain reaction and western blot analysis. In addition, the present study investigated changes of HIF‑1α and VEGF expression after incubation under hypoxic conditions and PHD2 transfection. PHD2 expression was significantly higher expressed than the other two PHD isoforms, indicating its major role in LCs. Moreover, a significant increase of VEGF mRNA expression was identified after incubation under hypoxic conditions, which was, however, attenuated by PHD2 overexpression in LCs. Further analysis also indicated that this hypoxia‑induced increase in the mRNA expression of VEGF was consistent with increases in the protein levels of HIF‑1α, which is regulated by PHD-mediated degradation. In conclusion, the results of the present study indicated that PHD2 is the main PHD expressed in LCs and hypoxia‑induced VEGF expression can be attenuated by PHD2 overexpression through HIF‑1α‑mediated mechanisms in LCs. This PHD2-mediated transcriptional activation may be one of the mechanisms regulating VEGF expression in LCs during mammalian corpus luteum development.
Collapse
Affiliation(s)
- Zhenghong Zhang
- Provincial Key Laboratory for Developmental Biology and Neurobiology, College of Life Science, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| | - Xunsheng Pang
- Provincial Key Laboratory for Developmental Biology and Neurobiology, College of Life Science, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| | - Zonghao Tang
- Provincial Key Laboratory for Developmental Biology and Neurobiology, College of Life Science, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| | - Dingzhong Yin
- Provincial Key Laboratory for Developmental Biology and Neurobiology, College of Life Science, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| | - Zhengchao Wang
- Provincial Key Laboratory for Developmental Biology and Neurobiology, College of Life Science, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| |
Collapse
|
38
|
Nishimura R, Okuda K. Multiple roles of hypoxia in ovarian function: roles of hypoxia-inducible factor-related and -unrelated signals during the luteal phase. Reprod Fertil Dev 2015; 28:RD15010. [PMID: 25940685 DOI: 10.1071/rd15010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 03/13/2015] [Indexed: 02/28/2024] Open
Abstract
There is increasing interest in the role of oxygen conditions in the microenvironment of organs because of the discovery of a hypoxia-specific transcription factor, namely hypoxia-inducible factor (HIF) 1. Ovarian function has several phases that change day by day, including ovulation, follicular growth and corpus luteum formation and regression. These phases are regulated by many factors, including pituitary hormones and local hormones, such as steroids, peptides and cytokines, as well as oxygen conditions. Hypoxia strongly induces angiogenesis because transcription of the potent angiogenic factor vascular endothelial growth factor (VEGF) is regulated by HIF1. Follicular development and luteal formation are accompanied by a marked increase in angiogenesis assisted by HIF1-VEGF signalling. Hypoxia is also one of the factors that induces luteolysis by suppressing progesterone synthesis and by promoting apoptosis of luteal cells. The present review focuses on recent studies of hypoxic conditions, as well as HIF1-regulated genes and proteins, in the regulation of ovarian function.
Collapse
|
39
|
Brown HM, Anastasi MR, Frank LA, Kind KL, Richani D, Robker RL, Russell DL, Gilchrist RB, Thompson JG. Hemoglobin: a gas transport molecule that is hormonally regulated in the ovarian follicle in mice and humans. Biol Reprod 2014; 92:26. [PMID: 25395682 DOI: 10.1095/biolreprod.114.124594] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
An increasing number of nonerythroid tissues are found to express hemoglobin mRNA and protein. Hemoglobin is a well-described gas transport molecule, especially for O2, but also for NO, CO2, and CO, and also acts as a reactive oxygen species scavenger. We previously found Hba-a1 and Hbb mRNA and protein at high levels within mouse periovulatory cumulus cells, but not in cumulus following in vitro maturation. This led us to investigate the temporal and spatial regulation in follicular cells during the periovulatory period. Cumulus-oocyte complexes were collected from equine chorionic gonadotropin/human chorionic gonadotropin-treated peripubertal SV129 female mice and collected and analyzed for gene expression and protein localization at a variety of time points over the periovulatory period. A further cohort matured in vitro with different forms of hemoglobin (ferro- and ferrihemoglobin) under different O2 atmospheric conditions (2%, 5%, and 20% O2) were subsequently fertilized in vitro and cultured to the blastocyst stage. Murine mRNA transcripts for hemoglobin were regulated by stimulation of the ovulatory cascade, in both granulosa and cumulus cells, and expression of HBA1 and HBB was highly significant in human granulosa and cumulus, but erythrocyte cell marker genes were not. Several other genes involved in hemoglobin function were similarly luteinizing hormone-regulated, including genes for heme biosynthesis. Immunohistochemistry revealed a changing localization pattern of HBA-A1 protein in murine cumulus cells and oocytes following the ovulatory signal. Significantly, no positive staining for HBA-A1 protein was observed within in vitro-matured oocytes, but, if coincubated with ferro- or ferrihemoglobin, cytoplasmic HBA-A1 was observed, similar to in vivo-derived oocytes. Addition of ferro-, but not ferrihemoglobin, had a small, positive effect on blastocyst yield, but only under either 2% or 20% O2 gas atmosphere. The identification of hemoglobin within granulosa and cumulus cells poses many questions as to its function in these cells. There are several possible roles, the most likely of which is either an O2 or NO sequestering molecule; perhaps both roles are engaged. The strong endocrine regulation during the periovulatory period suggests to us that one potential function of hemoglobin is to provide a short-lived hypoxic environment by binding very tightly any available O2. This, in turn, facilitates the differentiation of the follicle towards corpus luteum formation by enabling the stabilization of a key transcription factor known to initiate such differentiation: hypoxia inducible factor.
Collapse
Affiliation(s)
- Hannah M Brown
- School of Pediatrics and Reproductive Health, School of Animal and Veterinary Sciences, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Marie R Anastasi
- School of Pediatrics and Reproductive Health, School of Animal and Veterinary Sciences, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Laura A Frank
- School of Pediatrics and Reproductive Health, School of Animal and Veterinary Sciences, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Karen L Kind
- School of Pediatrics and Reproductive Health, School of Animal and Veterinary Sciences, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Dulama Richani
- School of Pediatrics and Reproductive Health, School of Animal and Veterinary Sciences, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Rebecca L Robker
- School of Pediatrics and Reproductive Health, School of Animal and Veterinary Sciences, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Darryl L Russell
- School of Pediatrics and Reproductive Health, School of Animal and Veterinary Sciences, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Robert B Gilchrist
- School of Pediatrics and Reproductive Health, School of Animal and Veterinary Sciences, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Jeremy G Thompson
- School of Pediatrics and Reproductive Health, School of Animal and Veterinary Sciences, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
40
|
Tamura K, Yoshie M, Hashimoto K, Tachikawa E. Inhibitory effect of insulin-like growth factor-binding protein-7 (IGFBP7) on in vitro angiogenesis of vascular endothelial cells in the rat corpus luteum. J Reprod Dev 2014; 60:447-53. [PMID: 25212428 PMCID: PMC4284319 DOI: 10.1262/jrd.2014-069] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Angiogenesis in the developing corpus luteum (CL) is a prerequisite for establishment and maintenance of an early pregnancy. To explore the physiological significance of insulin-like growth factor-binding protein-7 (IGFBP7) in the developing CL, the effects of IGFBP7 on vascular endothelial growth factor (VEGFA)- and luteinizing hormone (LH)-induced in vitro tube formation were tested using isolated luteal microvascular endothelial cells (LECs). Capillary-like tube formation of LECs and their proliferation were stimulated by both VEGFA and LH. IGFBP7 treatment suppressed VEGFA- or LH-induced tube formation. The proliferation and migration of LECs, and phosphorylation of mitogen-activated protein kinase kinase and extracellular signal-regulated kinase 1/2 were inhibited by IGFBP7. Furthermore, IGFBP7 attenuated VEGFA-enhanced cyclooxygenase (COX)-2 mRNA expression and prostaglandin E2 secretion. These findings suggest the possibility that
luteal IGFBP7 secretion may suppress the stimulatory effect of VEGFA on angiogenesis in the early CL.
Collapse
Affiliation(s)
- Kazuhiro Tamura
- Department of Endocrine and Neural Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | | | | | | |
Collapse
|
41
|
Stimulatory effect of vascular endothelial growth factor on progesterone production and survivability of cultured bubaline luteal cells. Anim Reprod Sci 2014; 148:251-9. [DOI: 10.1016/j.anireprosci.2014.06.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 05/14/2014] [Accepted: 06/19/2014] [Indexed: 11/17/2022]
|
42
|
Developmental study of the distribution of hypoxia-induced factor-1 alpha and microtubule-associated protein 2 in children’s brainstem: Comparison between controls and cases with signs of perinatal hypoxia. Neuroscience 2014; 271:77-98. [DOI: 10.1016/j.neuroscience.2014.04.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 03/21/2014] [Accepted: 04/08/2014] [Indexed: 11/20/2022]
|
43
|
Fadhillah, Yoshioka S, Nishimura R, Okuda K. Hypoxia promotes progesterone synthesis during luteinization in bovine granulosa cells. J Reprod Dev 2014; 60:194-201. [PMID: 24583842 PMCID: PMC4085383 DOI: 10.1262/jrd.2014-014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To determine whether hypoxia has an effect on luteinization, we examined the influence of hypoxia on a model of bovine luteinizing and non-luteinizing granulosa cell culture. The granulosa cells were obtained from small antral follicles (≤ 6 mm in diameter). To induce luteinization, the cells were treated for 24 h with insulin (2 µg/ml), forskolin (10 µM) or insulin in combination with forskolin at 20% O2. After 24 h, progesterone (P4) production was higher in the treated cells, which we defined as luteinizing granulosa cells, than in non-treated cells, which we defined as non-luteinizing granulosa cells. P4 production by non-luteinizing granulosa cells was not affected by hypoxia (24 h at 10% and 5% O2), while P4 production by granulosa cells treated with insulin in combination with forskolin was significantly increased under hypoxia (24 h at 10% and 5% O2). Because hypoxia affected P4 production by the luteinizing granulosa cells but not by the non-luteinizing granulosa cells, hypoxia seems to promote P4 production during, rather than before, luteinization. In the cells treated with insulin in combination with forskolin, mRNA and protein expression of steroidogenic acute regulatory protein (StAR) and protein expression of 3β-hydroxysteroid dehydrogenase (3β-HSD) increased under 10% O2, while mRNA and protein expressions of key protein and enzymes in P4 biosynthesis did not increase under 5% O2. The overall results suggest that hypoxia plays a role in progressing and completing the luteinization by enhancing P4 production through StAR as well as 3β-HSD expressions in the early time of establishing the corpus luteum.
Collapse
Affiliation(s)
- Fadhillah
- Laboratory of Reproductive Physiology, Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan
| | | | | | | |
Collapse
|
44
|
Mohamed A, El-Rayes B, Khuri FR, Saba NF. Targeted therapies in metastatic esophageal cancer: advances over the past decade. Crit Rev Oncol Hematol 2014; 91:186-96. [PMID: 24582516 DOI: 10.1016/j.critrevonc.2014.01.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 01/14/2014] [Accepted: 01/17/2014] [Indexed: 12/29/2022] Open
Abstract
Esophageal cancer is one of the most aggressive malignancies of the upper aerodigestive tract. Despite advances in surgical techniques and multi-modality therapies, the 5-year survival rate remains poor (14%). Over the past decade, efforts have been focused on the field of drug development with the advancement of novel molecularly targeted therapeutic agents. These agents target a variety of cancer relevant pathways such as vascular endothelial growth factor (VEGF) or its receptor, the cyclooxygenase-2 (COX-2), epidermal growth factor receptor (EGFR), and mammalian target of rapamycin (mTOR) pathways. The number of approved targeted agents remains few, with HER-2 inhibitors leading the list for treatment of HER-2 expressing metastatic adenocarcinomas. Novel agents have not yet been widely explored in esophageal cancer. In this review, we will provide a concise and systematic overview of the development of novel targeted therapies currently under investigation for the treatment of metastatic esophageal disease.
Collapse
Affiliation(s)
- Amr Mohamed
- Department of Medicine, Morehouse School of Medicine, Atlanta, GA, USA
| | - Bassel El-Rayes
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA, USA
| | - Fadlo R Khuri
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA, USA
| | - Nabil F Saba
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA, USA.
| |
Collapse
|
45
|
Papa PDC, Sousa LMMDC, Silva RDS, de Fátima LA, da Fonseca VU, do Amaral VC, Hoffmann B, Alves-Wagner AB, Machado UF, Kowalewski MP. Glucose transporter 1 expression accompanies hypoxia sensing in the cyclic canine corpus luteum. Reproduction 2014; 147:81-9. [DOI: 10.1530/rep-13-0398] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The canine corpus luteum (CL) functions as a source of progesterone (P4) and 17β-oestradiol (E2); however, the transport of energy substrates to maintain its high hormonal output has not yet been characterised. This study involved the localisation and temporal distribution of the facilitative glucose transporter 1 and the quantification of the corresponding protein (GLUT1) and gene (SLC2A1) expression. Some GLUT1/SLC2A1 regulatory proteins, such as hypoxia-inducible factor 1α (HIF1A) and fibroblast growth factor 2 (FGF2); mRNAs, such as HIF1A, FGF2 and vascular endothelial growth factor A (VEGFA); and VEGFA receptors 1 and 2 (FLT1 and KDR) were also analysed from days 10 to 70 after ovulation. Additionally, plasma P4 and E2 levels were assessed via chemiluminescence. Moreover, the canine KDR sequence has been cloned, thereby enabling subsequent semi-quantitative PCR analysis. Our results demonstrate time-dependent variations in the expression profile of SLC2A1 during dioestrus, which were accompanied by highly correlated changes (0.84<r<0.98; P<0.03) in the gene expression of HIF1A, VEGF and FLT1 as well as in P4 plasma concentrations. FGF2 mRNA correlated with E2 plasma concentrations (r=0.61; P=0.01). Our data reveal that the glucose transporter is regulated throughout the CL lifespan and suggest that CL depends on the sensing of hypoxia and the status of luteal vascularisation. Moreover, time-dependent expression of GLUT1/SLC2A1 may lie underneath increased metabolic and energetic requirements for sustaining P4 production.
Collapse
|
46
|
Collet G, Lamerant-Fayel N, Tertil M, El Hafny-Rahbi B, Stepniewski J, Guichard A, Foucault-Collet A, Klimkiewicz K, Petoud S, Matejuk A, Grillon C, Jozkowicz A, Dulak J, Kieda C. Hypoxia-regulated overexpression of soluble VEGFR2 controls angiogenesis and inhibits tumor growth. Mol Cancer Ther 2013; 13:165-78. [PMID: 24170768 DOI: 10.1158/1535-7163.mct-13-0637] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
VEGFs are found at high levels in hypoxic tumors. As major components directing pathologic neovascularization, they regulate stromal reactions. Consequently, novel strategies targeting and inhibiting VEGF overproduction upon hypoxia offer considerable potential for modern anticancer therapies controlling rather than destroying tumor angiogenesis. Here, we report the design of a vector expressing the soluble form of VEGF receptor-2 (sVEGFR2) driven by a hypoxia-responsive element (HRE)-regulated promoter. To enable in vivo imaging by infrared visualization, mCherry and IFP1.4 coding sequences were built into the vector. Plasmid construction was validated through transfection into embryonic human kidney HEK293 and murine B16F10 melanoma cells. sVEGFR2 was expressed in hypoxic conditions only, confirming that the gene was regulated by the HRE promoter. sVEGFR2 was found to bind efficiently and specifically to murine and human VEGF-A, reducing the growth of tumor and endothelial cells as well as impacting angiogenesis in vitro. The hypoxia-conditioned sVEGFR2 expression was shown to be functional in vivo: Tumor angiogenesis was inhibited and, on stable transfection of B16F10 melanoma cells, tumor growth was reduced. Enhanced expression of sVEGFR2 was accompanied by a modulation in levels of VEGF-A. The resulting balance reflected the effect on tumor growth and on control of angiogenesis. A concomitant increase of intratumor oxygen tension also suggested an influence on vessel normalization. The possibility to express an angiogenesis regulator as sVEGFR2, in a hypoxia-conditioned manner, significantly opens new strategies for tumor vessel-controlled normalization and the design of adjuvants for combined cancer therapies.
Collapse
Affiliation(s)
- Guillaume Collet
- Corresponding Authors: Claudine Kieda, CNRS, rue Charles Sadron, Orleans 45071, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Akiyama I, Yoshino O, Osuga Y, Shi J, Harada M, Koga K, Hirota Y, Hirata T, Fujii T, Saito S, Kozuma S. Bone morphogenetic protein 7 increased vascular endothelial growth factor (VEGF)-a expression in human granulosa cells and VEGF receptor expression in endothelial cells. Reprod Sci 2013; 21:477-82. [PMID: 24023033 DOI: 10.1177/1933719113503411] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The formation of an individual capillary network in the theca cell layer is required for ovarian folliculogenesis. Although vascular endothelial growth factor (VEGF) is critical for this process, the regulation of VEGF has been unclear. In the present study, the relationship between VEGF and intraovarian cytokine, bone morphogenetic protein 7 (BMP-7) was investigated. Granulosa cells (GC), obtained from in vitro fertilization patients, were cultured with BMP-7 followed by RNA extraction. Human umbilical vein endothelial cells (HUVECs) were also cultured with BMP-7 followed by RNA extraction, tube formation assay, or cell count analysis. The BMP-7 stimulated VEGF messenger RNA (mRNA) and protein expression in GC significantly. In HUVEC, BMP-7 increased an approximately 1.8-fold in the cell number and induced the tube formation significantly compared to control. The BMP-7 also induced a 2-fold increase in VEGF receptor mRNA transcript relative abundance in HUVEC. The BMP-7, a theca cell-derived factor, may stimulate endothelial cell to form vasculature in the follicle via 2 distinct mechanisms, induction of VEGF expression in GC and increased sensitivity of endothelial cells to VEGF.
Collapse
Affiliation(s)
- Ikumi Akiyama
- 1Department of Obstetrics and Gynecology, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Meidan R, Klipper E, Zalman Y, Yalu R. The role of hypoxia-induced genes in ovarian angiogenesis. Reprod Fertil Dev 2013; 25:343-50. [PMID: 22950963 DOI: 10.1071/rd12139] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 07/26/2012] [Indexed: 01/23/2023] Open
Abstract
The hypoxic microenvironment that occurs in fast-growing tissue such as the corpus luteum (CL) is a major contributor to its ability to survive via the induction of an intricate vascular network. Cellular responses to hypoxia are mediated by hypoxia-inducible factor-1 (HIF-1), an oxygen-regulated transcriptional activator. HIF-1, a heterodimer consisting of a constitutively-expressed β subunit and an oxygen-regulated α subunit, binds to the hypoxia responsive element (HRE) present in the promoter regions of responsive genes. This review summarises evidence for the involvement of hypoxia and HIF-1α in CL development and function. Special emphasis is given to hypoxia-induced, luteal cell-specific expression of multiple genes (vascular endothelial growth factor A (VEGFA), fibroblast growth factor 2 (FGF-2), prokineticin receptor 2 (PK-R2), stanniocalcin 1 (STC-1) and endothelin 2 (EDN-2) that participate in the angiogenic process during CL formation.
Collapse
Affiliation(s)
- Rina Meidan
- Department of Animal Sciences, The Hebrew University of Jerusalem, The Robert H. Smith Faculty of Agriculture, Food and Environment, Rehovot 76100, Israel.
| | | | | | | |
Collapse
|
49
|
Duncan WC, Nio-Kobayashi J. Targeting angiogenesis in the pathological ovary. Reprod Fertil Dev 2013; 25:362-71. [PMID: 22951108 DOI: 10.1071/rd12112] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 07/13/2012] [Indexed: 12/17/2022] Open
Abstract
The ovary is a key tissue in the study of physiological neo-vascularisation in the adult and its study has highlighted important molecules involved in the regulation of angiogenesis in vivo. These include vascular endothelial growth factor, delta-like ligand 4, thrombospondin-1, prokineticin-1 and prostaglandin E2. Targeting these molecular pathways has therapeutic potential and their manipulation has an increasing preclinical and clinical role in the management of the pathological ovary. Targeting angiogenic pathways has utility in the promotion of ovarian angiogenesis to improve tissue and follicle survival and function as well as the prevention and management of ovarian hyperstimulation syndrome. There is a theoretical possibility that targeting angiogenesis may improve the function of the polycystic ovary and a real role for targeting angiogenesis in ovarian cancer.
Collapse
Affiliation(s)
- W Colin Duncan
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | | |
Collapse
|
50
|
Cytokines and angiogenesis in the corpus luteum. Mediators Inflamm 2013; 2013:420186. [PMID: 23840095 PMCID: PMC3693155 DOI: 10.1155/2013/420186] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 05/22/2013] [Accepted: 05/22/2013] [Indexed: 12/24/2022] Open
Abstract
In adults, physiological angiogenesis is a rare event, with few exceptions as the vasculogenesis needed for tissue growth and function in female reproductive organs. Particularly in the corpus luteum (CL), regulation of angiogenic process seems to be tightly controlled by opposite actions resultant from the balance between pro- and antiangiogenic factors. It is the extremely rapid sequence of events that determines the dramatic changes on vascular and nonvascular structures, qualifying the CL as a great model for angiogenesis studies. Using the mare CL as a model, reports on locally produced cytokines, such as tumor necrosis factor α (TNF), interferon gamma (IFNG), or Fas ligand (FASL), pointed out their role on angiogenic activity modulation throughout the luteal phase. Thus, the main purpose of this review is to highlight the interaction between immune, endothelial, and luteal steroidogenic cells, regarding vascular dynamics/changes during establishment and regression of the equine CL.
Collapse
|