1
|
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
|
2
|
Monaco CF, Plewes MR, Przygrodzka E, George JW, Qiu F, Xiao P, Wood JR, Cupp AS, Davis JS. Basic fibroblast growth factor induces proliferation and collagen production by fibroblasts derived from the bovine corpus luteum†. Biol Reprod 2023; 109:367-380. [PMID: 37283496 PMCID: PMC10502575 DOI: 10.1093/biolre/ioad065] [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: 12/01/2022] [Revised: 05/11/2023] [Indexed: 06/08/2023] Open
Abstract
Cyclic regression of the ovarian corpus luteum, the endocrine gland responsible for progesterone production, involves rapid matrix remodeling. Despite fibroblasts in other systems being known for producing and maintaining extracellular matrix, little is known about fibroblasts in the functional or regressing corpus luteum. Vast transcriptomic changes occur in the regressing corpus luteum, among which are reduced levels of vascular endothelial growth factor A (VEGFA) and increased expression of fibroblast growth factor 2 (FGF2) after 4 and 12 h of induced regression, when progesterone is declining and the microvasculature is destabilizing. We hypothesized that FGF2 activates luteal fibroblasts. Analysis of transcriptomic changes during induced luteal regression revealed elevations in markers of fibroblast activation and fibrosis, including fibroblast activation protein (FAP), serpin family E member 1 (SERPINE1), and secreted phosphoprotein 1 (SPP1). To test our hypothesis, we treated bovine luteal fibroblasts with FGF2 to measure downstream signaling, type 1 collagen production, and proliferation. We observed rapid and robust phosphorylation of various signaling pathways involved in proliferation, such as ERK, AKT, and STAT1. From our longer-term treatments, we determined that FGF2 has a concentration-dependent collagen-inducing effect, and that FGF2 acts as a mitogen for luteal fibroblasts. FGF2-induced proliferation was greatly blunted by inhibition of AKT or STAT1 signaling. Our results suggest that luteal fibroblasts are responsive to factors that are released by the regressing bovine corpus luteum, an insight into the contribution of fibroblasts to the microenvironment in the regressing corpus luteum.
Collapse
Affiliation(s)
- Corrine F Monaco
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michele R Plewes
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
- US Department of Veterans Affairs-Nebraska Western Iowa Healthcare System, Omaha, NE, USA
| | - Emilia Przygrodzka
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jitu W George
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
- US Department of Veterans Affairs-Nebraska Western Iowa Healthcare System, Omaha, NE, USA
| | - Fang Qiu
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Peng Xiao
- Department of Genetics, Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jennifer R Wood
- Department of Animal Science, University of Nebraska—Lincoln, Lincoln, NE, USA
| | - Andrea S Cupp
- Department of Animal Science, University of Nebraska—Lincoln, Lincoln, NE, USA
| | - John S Davis
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
- US Department of Veterans Affairs-Nebraska Western Iowa Healthcare System, Omaha, NE, USA
| |
Collapse
|
3
|
Bhardwaj V, Zhang X, Pandey V, Garg M. Neo-vascularization-based therapeutic perspectives in advanced ovarian cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188888. [PMID: 37001618 DOI: 10.1016/j.bbcan.2023.188888] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/23/2023] [Accepted: 03/02/2023] [Indexed: 03/30/2023]
Abstract
The process of angiogenesis is well described for its potential role in the development of normal ovaries, and physiological functions as well as in the initiation, progression, and metastasis of ovarian cancer (OC). In advanced stages of OC, cancer cells spread outside the ovary to the pelvic, abdomen, lung, or multiple secondary sites. This seriously limits the efficacy of therapeutic options contributing to fatal clinical outcomes. Notably, a variety of angiogenic effectors are produced by the tumor cells to initiate angiogenic processes leading to the development of new blood vessels, which provide essential resources for tumor survival, dissemination, and dormant micro-metastasis of tumor cells. Multiple proangiogenic effectors and their signaling axis have been discovered and functionally characterized for potential clinical utility in OC. In this review, we have provided the current updates on classical and emerging proangiogenic effectors, their signaling axis, and the immune microenvironment contributing to the pathogenesis of OC. Moreover, we have comprehensively reviewed and discussed the significance of the preclinical strategies, drug repurposing, and clinical trials targeting the angiogenic processes that hold promising perspectives for the better management of patients with OC.
Collapse
Affiliation(s)
- Vipul Bhardwaj
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Institute of Biopharmaceutical and Bioengineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China
| | - Xi Zhang
- Shenzhen Bay Laboratory, Shenzhen 518055, PR China
| | - Vijay Pandey
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Institute of Biopharmaceutical and Bioengineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China.
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Sector-125, Noida 201301, India.
| |
Collapse
|
4
|
Estienne A, Relav L, Benkoura M, Monniaux D, Morin F, Fabre S, Price CA. Endothelial cell-derived fibroblast growth factor-18 regulates ovarian function in sheep. J Cell Physiol 2022; 237:2528-2538. [PMID: 35315069 DOI: 10.1002/jcp.30718] [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: 12/02/2021] [Revised: 02/08/2022] [Accepted: 02/28/2022] [Indexed: 11/08/2022]
Abstract
Increasing the efficiency of farm animal reproduction is necessary to reduce the environmental impact of food production systems. One approach is to increase the number of healthy eggs (oocytes) produced per female for fertilization, thus it is important to understand factors that decrease oocyte health. One paracrine factor that decreases ovarian follicle growth is fibroblast growth factor 18 (FGF18) secreted by cells in the theca layer of the ovarian follicle, however the factors that regulate FGF18 secretion are unknown. In this study we hypothesized that FGF18 secretion is controled by intrafollicular factors and is linked to fertility, which we tested by using cell culture and sheep genetic models in vivo. Separation of theca cell populations revealed that FGF18 messenger RNA (mRNA) is located mainly in thecal endothelial rather than endocrine cells, and immunohistochemistry localized FGF18 protein to microvessels in the theca layer in situ. Culture of ovine theca-derived endothelial cells was used to demonstrate stimulation of FGF18 mRNA and protein abundance by bone morphogenetic protein 4 (BMP4), a growth factor derived from theca endocrine cells. Taking advantage of a sheep genetic model, we demonstrate reduced ovarian and peripheral FGF18 concentrations in the hyperprolific Booroola ewe harboring the FecBB mutation in BMPR1B. These data suggest a novel control of fertility by follicular endothelial cells, in which theca endocrine cells secrete BMP4 that stimulates the secretion of FGF18 from thecal endothelial cells, which in turn diffuses into the granulosa cell layer and promotes apoptosis.
Collapse
Affiliation(s)
- Anthony Estienne
- Département de biomédecine vétérinaire, Centre de recherche en reproduction et fertilité (CRRF), Faculté de médecine vétérinaire, Université de Montréal, St-Hyacinthe, Quebec, Canada.,Physiologie Animale et Système d'Elevage, UMR Physiologie de la Reproduction et des Comportements, INRAE Tours, Nouzilly, France
| | - Lauriane Relav
- Département de biomédecine vétérinaire, Centre de recherche en reproduction et fertilité (CRRF), Faculté de médecine vétérinaire, Université de Montréal, St-Hyacinthe, Quebec, Canada
| | - Morad Benkoura
- Département de biomédecine vétérinaire, Centre de recherche en reproduction et fertilité (CRRF), Faculté de médecine vétérinaire, Université de Montréal, St-Hyacinthe, Quebec, Canada
| | - Danielle Monniaux
- Physiologie Animale et Système d'Elevage, UMR Physiologie de la Reproduction et des Comportements, INRAE Tours, Nouzilly, France
| | - Fanny Morin
- Département de biomédecine vétérinaire, Centre de recherche en reproduction et fertilité (CRRF), Faculté de médecine vétérinaire, Université de Montréal, St-Hyacinthe, Quebec, Canada
| | - Stéphane Fabre
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Christopher A Price
- Département de biomédecine vétérinaire, Centre de recherche en reproduction et fertilité (CRRF), Faculté de médecine vétérinaire, Université de Montréal, St-Hyacinthe, Quebec, Canada
| |
Collapse
|
5
|
Rytelewska E, Kiezun M, Zaobidna E, Gudelska M, Kisielewska K, Dobrzyn K, Kaminski T, Smolinska N. CHEMERIN as a modulator of angiogenesis and apoptosis processes in the corpus luteum of pigs: An in vitro study. Biol Reprod 2021; 105:1002-1015. [PMID: 34192738 DOI: 10.1093/biolre/ioab126] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/09/2021] [Accepted: 06/29/2021] [Indexed: 11/12/2022] Open
Abstract
The corpus luteum (CL) undergoes rapid changes, and its functional capabilities are influenced by processes such as angiogenesis and apoptosis. According to the literature, chemerin - a protein which participates in the regulation of energy homeostasis and the immune response, may also affect angiogenesis and apoptosis. Therefore, the aim of this study was to investigate the in vitro effect of chemerin on angiogenesis and apoptosis in porcine luteal cells (Lc) during specific phases related to CL physiology. Luteal cells were harvested from gilts during the early-, mid-, and late-luteal phases of the estrous cycle. The cells were preincubated for 48 h and incubated for 24 h with chemerin or a serum-free medium (controls). The abundance of angiogenesis- and apoptosis-related proteins was determined by ELISA in spent culture media, or by ELISA and Western Blot in protein extracts. The current study demonstrated that chemerin stimulates the production of VEGF-A and bFGF by porcine Lc and increases the protein abundance of angiogenic factors receptors (VEGFR1, VEGFR2, VEGFR3, FGFR1, FGFR2) in these cells. The study also revealed that chemerin exerts a modulatory effect (stimulatory/inhibitory, depending on the phase of the cycle) on the protein abundance of Fas, FasL, Bcl-2 and caspase-3 in porcine Lc. These results imply that chemerin may affect angiogenesis and apoptosis processes in the porcine CL, as evidenced by its modulatory effect of chemerin on the protein abundance of crucial angiogenesis- and apoptosis-related factors, observed in an in vitro study of porcine Lc.
Collapse
Affiliation(s)
- Edyta Rytelewska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Marta Kiezun
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Ewa Zaobidna
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Marlena Gudelska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Katarzyna Kisielewska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Kamil Dobrzyn
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Tadeusz Kaminski
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Nina Smolinska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
6
|
Basavaraja R, Drum JN, Sapuleni J, Bibi L, Friedlander G, Kumar S, Sartori R, Meidan R. Downregulated luteolytic pathways in the transcriptome of early pregnancy bovine corpus luteum are mimicked by interferon-tau in vitro. BMC Genomics 2021; 22:452. [PMID: 34134617 PMCID: PMC8207607 DOI: 10.1186/s12864-021-07747-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 05/21/2021] [Indexed: 12/15/2022] Open
Abstract
Background Maintenance of the corpus luteum (CL) beyond the time of luteolysis is essential for establishing pregnancy. Identifying the distinct features of early pregnancy CL remains unresolved, hence we analyzed here the transcriptome of CL on day 18 pregnant (P) and non-pregnant (NP) cows using RNA-Seq. CL of P cows expressed ISGs, verifying exposure to the pregnancy recognition signal, interferon-tau (IFNT), whereas the CL of NP cows had elevated luteal progesterone levels, implying that luteolysis had not yet commenced. Results The DEGs, IPA, and metascape canonical pathways, along with GSEA analysis, differed markedly in the CL of P cows from those of NP cows, at the same day of the cycle. Both metascape and IPA identified similar significantly enriched pathways such as interferon alpha/beta, sonic hedgehog pathway, TNFA, EDN1, TGFB1, and PDGF. However, type-1 interferon and sonic hedgehog pathways were positively enriched whereas most of the enriched pathways were downregulated in the P compared to NP samples. Thirty-four % of these pathways are known to be elevated by PGF2A during luteolysis. Notably, selective DEGs in luteinized granulosa cells were modulated by IFNT in vitro in a similar manner to their regulation in the CL of P cows. Conclusion This study unraveled the unique transcriptomic signature of the IFNT-exposed, early pregnancy CL, highlighting the abundance of downregulated pathways known to be otherwise induced during luteolysis. These and IFNT-regulated in vitro pregnancy-specific DEGs suggest that IFNT contributes to the characteristics and maintenance of early pregnancy CL. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07747-3.
Collapse
Affiliation(s)
- Raghavendra Basavaraja
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 7610001, Rehovot, Israel
| | - Jessica N Drum
- Department of Animal Science, University of São Paulo, Piracicaba, Brazil
| | - Jackson Sapuleni
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 7610001, Rehovot, Israel
| | - Lonice Bibi
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 7610001, Rehovot, Israel
| | - Gilgi Friedlander
- The Mantoux Bioinformatics institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Sai Kumar
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 7610001, Rehovot, Israel
| | - Roberto Sartori
- Department of Animal Science, University of São Paulo, Piracicaba, Brazil
| | - Rina Meidan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 7610001, Rehovot, Israel.
| |
Collapse
|
7
|
Stucker S, De Angelis J, Kusumbe AP. Heterogeneity and Dynamics of Vasculature in the Endocrine System During Aging and Disease. Front Physiol 2021; 12:624928. [PMID: 33767633 PMCID: PMC7987104 DOI: 10.3389/fphys.2021.624928] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
The endocrine system consists of several highly vascularized glands that produce and secrete hormones to maintain body homeostasis and regulate a range of bodily functions and processes, including growth, metabolism and development. The dense and highly vascularized capillary network functions as the main transport system for hormones and regulatory factors to enable efficient endocrine function. The specialized capillary types provide the microenvironments to support stem and progenitor cells, by regulating their survival, maintenance and differentiation. Moreover, the vasculature interacts with endocrine cells supporting their endocrine function. However, the structure and niche function of vasculature in endocrine tissues remain poorly understood. Aging and endocrine disorders are associated with vascular perturbations. Understanding the cellular and molecular cues driving the disease, and age-related vascular perturbations hold potential to manage or even treat endocrine disorders and comorbidities associated with aging. This review aims to describe the structure and niche functions of the vasculature in various endocrine glands and define the vascular changes in aging and endocrine disorders.
Collapse
Affiliation(s)
| | | | - Anjali P. Kusumbe
- Tissue and Tumor Microenvironments Group, Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom
| |
Collapse
|
8
|
Piotrowska-Tomala KK, Jonczyk AW, Kordowitzki P, Jalali BM, Skarzynski DJ. The effect of basic fibroblast growth factor 2 on the bovine corpus luteum depends on the stage of the estrous cycle and modulates prostaglandin F 2α action. Animal 2020; 15:100048. [PMID: 33516003 DOI: 10.1016/j.animal.2020.100048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 08/03/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022] Open
Abstract
The roles of fibroblast growth factor 2 (FGF2) in the corpus luteum (CL) function and its modulatory effect on prostaglandin (PG) F2α during the bovine estrous cycle were studied using the following design of in vivo and in vitro experiments: (1) effects of FGF2 and FGF receptor 1 inhibitor (PD173074) on bovine CL function in the early (PGF2α-resistant) and mid (PGF2α-responsive) luteal stage in vivo, (2) the modulatory effect of FGF2 on PGF2α action during the luteal phase in vivo and (3) effects of FGF2 and PD173074 on bovine CL secretory function in vitro. Cows were treated by injection into the CL with: (1) saline (control), (2) FGF2, (3) PD173074, (4) FGF2 followed by intramuscular (i.m.) PGF2α, (5) PD173074 followed by i.m. PGF2α and (6) i.m. PGF2α as a positive control. For in vitro experiments, CL explants were treated with the aforementioned factors. Progesterone (P4) concentrations of blood samples or culture media were determined by radioimmunoassay. Relative mRNA expressions of the genes involved in angiogenesis and steroidogenesis were determined by quantitative real-time PCR. Although FGF2 treatment on day 4 of the estrous cycle did not change the cycle length, FGF2 with PGF2α decreased the P4 concentrations observed during the estrous cycle compared to the control group (P < 0.001). Moreover, FGF2 treatment on day 10 prolonged CL function as indicated by a significantly greater concentration of P4 on day 21 compared to the control group. In the in vitro study, FGF2 decreased cytochrome P450 family 11 subfamily A member 1 (CYP11A1) and hydroxy-delta-5-steroid dehydrogenase (HSD3B1) mRNA expression (P < 0.01) and decreased P4 production in the early-stage CL (P < 0.001). However, FGF2 + PGF2α or PGF2α alone resulted in an elevation of steroidogenic acute regulatory protein and CYP11A1 mRNA expression and P4 secretion in the early-stage CL (P < 0.01). In the mid-luteal phase, FGF2 upregulated CYP11A1 and HSD3B1 mRNA expression (P < 0.01), while FGF2 + PGF2α increased only HSD3B1 mRNA expression (P < 0.001). In conclusion, FGF2 seems to play a modulatory role in CL development or luteolysis, differentially regulating steroidogenesis and angiogenic factors as well as PGF2α actions.
Collapse
Affiliation(s)
- K K Piotrowska-Tomala
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-747 Olsztyn, Poland.
| | - A W Jonczyk
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-747 Olsztyn, Poland
| | - P Kordowitzki
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-747 Olsztyn, Poland
| | - B M Jalali
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-747 Olsztyn, Poland
| | - D J Skarzynski
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-747 Olsztyn, Poland
| |
Collapse
|
9
|
Yu B, Yao Y, Zhang X, Xu H, Lu J, Ruan M. Synergic Effect of Ligusticum chuanxiong Hort Extract and Borneol in Protecting Brain Microvascular Endothelial Cells against Oxygen-Glucose Deprivation/Reperfusion Injury. INT J PHARMACOL 2020. [DOI: 10.3923/ijp.2020.447.459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
10
|
Abdulrahman N, Fair T. Contribution of the immune system to follicle differentiation, ovulation and early corpus luteum formation. Anim Reprod 2019; 16:440-448. [PMID: 32435287 PMCID: PMC7234072 DOI: 10.21451/1984-3143-ar2019-0087] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 07/25/2019] [Indexed: 12/30/2022] Open
Abstract
Much of what we know about the involvement of the immune system in periovulatory follicle differentiation, ovulation and subsequent formation of the corpus luteum in cattle is drawn from the findings of studies in several mammalian livestock species. By integrating published histological data from cattle, sheep and pigs and referring back to the more comprehensive knowledge bank that exists for mouse and humans we can sketch out the key cells of the immune system and the cytokines and growth factors that they produce that are involved in follicle differentiation and luteinization, ovulation and early follicle development. These contributions are reviewed and the key findings, discussed.
Collapse
Affiliation(s)
- Noof Abdulrahman
- School of Agriculture & Food Sciences, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Trudee Fair
- School of Agriculture & Food Sciences, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
11
|
Interferon-Tau Exerts Direct Prosurvival and Antiapoptotic Actions in Luteinized Bovine Granulosa Cells. Sci Rep 2019; 9:14682. [PMID: 31605002 PMCID: PMC6789004 DOI: 10.1038/s41598-019-51152-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 09/23/2019] [Indexed: 12/12/2022] Open
Abstract
Interferon-tau (IFNT), serves as a signal to maintain the corpus luteum (CL) during early pregnancy in domestic ruminants. We investigated here whether IFNT directly affects the function of luteinized bovine granulosa cells (LGCs), a model for large-luteal cells. Recombinant ovine IFNT (roIFNT) induced the IFN-stimulated genes (ISGs; MX2, ISG15, and OAS1Y). IFNT induced a rapid and transient (15–45 min) phosphorylation of STAT1, while total STAT1 protein was higher only after 24 h. IFNT treatment elevated viable LGCs numbers and decreased dead/apoptotic cell counts. Consistent with these effects on cell viability, IFNT upregulated cell survival proteins (MCL1, BCL-xL, and XIAP) and reduced the levels of gamma-H2AX, cleaved caspase-3, and thrombospondin-2 (THBS2) implicated in apoptosis. Notably, IFNT reversed the actions of THBS1 on cell viability, XIAP, and cleaved caspase-3. Furthermore, roIFNT stimulated proangiogenic genes, including FGF2, PDGFB, and PDGFAR. Corroborating the in vitro observations, CL collected from day 18 pregnant cows comprised higher ISGs together with elevated FGF2, PDGFB, and XIAP, compared with CL derived from day 18 cyclic cows. This study reveals that IFNT activates diverse pathways in LGCs, promoting survival and blood vessel stabilization while suppressing cell death signals. These mechanisms might contribute to CL maintenance during early pregnancy.
Collapse
|
12
|
Jonczyk AW, Piotrowska-Tomala KK, Kordowitzki P, Skarzynski DJ. Effects of prostaglandin F 2α on angiogenic and steroidogenic pathways in the bovine corpus luteum may depend on its route of administration. J Dairy Sci 2019; 102:10573-10586. [PMID: 31521364 DOI: 10.3168/jds.2019-16644] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/24/2019] [Indexed: 12/13/2022]
Abstract
Prostaglandin (PG) F2α and its analogs (aPGF2α) are used to induce regression of the corpus luteum (CL); their administration during the middle stage of the estrous cycle causes luteolysis in cattle. However, the bovine CL is resistant to the luteolytic actions of aPGF2α in the early stage of the estrous cycle. The mechanisms underlying this differential luteal sensitivity, as well as acquisition of luteolytic sensitivity by the CL, are still not fully understood. Therefore, to characterize possible differences in response to aPGF2α administration, we aimed to determine changes in expression of genes related to (1) angiogenesis-fibroblast growth factor 2 (FGF2), fibroblast growth factor receptor 1 (FGFR1), fibroblast growth factor receptor 2 (FGFR2), vascular endothelial growth factor A (VEGFA), vascular endothelial growth factor receptor 1 (VEGFR1), vascular endothelial growth factor receptor 2 (VEGFR2); and (2) steroidogenesis-steroidogenic acute regulatory protein (STAR), cytochrome P450 family 11 subfamily A member 1 (P450scc), and hydroxy-delta-5-steroid dehydrogenase, 3 β- and steroid delta-isomerase 1 (HSD3B) in early- and middle-stage CL that accompany local (intra-CL) versus systemic (i.m.) aPGF2α injection. Cows at d 4 (early stage) or d 10 (middle stage) of the estrous cycle were treated as follows: (1) systemic saline injection, (2) systemic aPGF2α injection (25 mg), (3) local saline injection, and (4) local aPGF2α injection (2.5 mg). Progesterone (P4) concentration was measured in jugular vein blood samples during the entire set of experiments. After 4 h of treatment, CL were collected by ovariectomy, and mRNA and protein expression levels were determined by reverse transcription quantitative-PCR and Western blotting, respectively. Local and systemic aPGF2α injections upregulated FGF2 expression but decreased expression of VEGFA in both CL stages. Both aPGF2α injections increased the expression of STAR in early-stage CL, but downregulated it in middle-stage CL. In the early-stage CL, local administration of aPGF2α upregulated HSD3B, whereas systemic injection decreased its mRNA expression in early- and middle-stage CL. Moreover, we observed a decrease in the P4 level earlier after local aPGF2α injection than after systemic administration. These results indicate that aPGF2α acting locally may play a luteotrophic role in early-stage CL. The systemic effect of aPGF2α on the mRNA expression of genes participating in steroidogenesis seems to be more substantial than its local effect in middle-stage CL.
Collapse
Affiliation(s)
- A W Jonczyk
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland
| | - K K Piotrowska-Tomala
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland.
| | - P Kordowitzki
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland
| | - D J Skarzynski
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland
| |
Collapse
|
13
|
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: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
14
|
An X, Ma H, Han P, Zhu C, Cao B, Bai Y. Genome-wide differences in DNA methylation changes in caprine ovaries between oestrous and dioestrous phases. J Anim Sci Biotechnol 2018; 9:85. [PMID: 30524725 PMCID: PMC6277999 DOI: 10.1186/s40104-018-0301-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 10/22/2018] [Indexed: 12/22/2022] Open
Abstract
Background DNA methylation plays a vital role in reproduction. Entire genome DNA methylation changes during the oestrous phase (ES) and dioestrous phase (DS) in the ovaries of Guanzhong dairy goats were investigated using bisulphite sequencing to understand the molecular biological mechanisms of these goats’ oestrous cycle. Results We discovered distinct genome-wide DNA methylation patterns in ES and DS ovaries. A total of 26,910 differentially methylated regions were upregulated and 21,453 differentially methylated regions were downregulated in the ES samples compared with the DS samples (P-values ≤0.05 and fold change of methylation ratios ≥2). Differentially methylated region analysis showed hypomethylation in the gene body regions and hypermethylation in the joining region between upstream regions and gene bodies. The methylation ratios of the STAR, FGF2, FGF12, BMP5 and SMAD6 genes in the ES samples were lower than those of the DS samples (P-values ≤0.05 and fold change of methylation ratios ≥2). Conversely, the methylation ratios of the EGFR, TGFBR2, IGF2BP1 and MMD2 genes increased in the ES samples compared with the DS samples. In addition, 223 differentially methylated genes were found in the GnRH signalling pathway (KO04912), ovarian steroidogenesis pathway (KO04913), oestrogen signalling pathway (KO04915), oxytocin signalling pathway (KO04921), insulin secretion pathway (KO04911) and MAPK signalling pathway (KO04010). Conclusions This study is the first large-scale comparison of the high-resolution DNA methylation landscapes of oestrous and dioestrous ovaries from dairy goats. Previous studies and our investigations have shown that the NR5A2, STAR, FGF2 and BMP5 genes might have potential application value in regulating caprine oestrus. Electronic supplementary material The online version of this article (10.1186/s40104-018-0301-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaopeng An
- 1College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 People's Republic of China
| | - Haidong Ma
- 1College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 People's Republic of China
| | - Peng Han
- 1College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 People's Republic of China
| | - Chao Zhu
- 1College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 People's Republic of China
| | - Binyun Cao
- 1College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 People's Republic of China
| | - Yueyu Bai
- Animal Health Supervision Institute of Henan Province, No. 91 Jingsan Road, Zhengzhou, Henan 450008 People's Republic of China
| |
Collapse
|
15
|
Shrestha K, Meidan R. The cAMP-EPAC Pathway Mediates PGE2-Induced FGF2 in Bovine Granulosa Cells. Endocrinology 2018; 159:3482-3491. [PMID: 30085093 DOI: 10.1210/en.2018-00527] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/26/2018] [Indexed: 01/10/2023]
Abstract
During the periovulatory period, the profile of fibroblast growth factor 2 (FGF2) coincides with elevated prostaglandin E2 (PGE2) levels. We investigated whether PGE2 can directly stimulate FGF2 production in bovine granulosa cells and, if so, which prostaglandin E2 receptor (PTGER) type and signaling cascades are involved. PGE2 temporally stimulated FGF2. Accordingly, endoperoxide-synthase2-silenced cells, exhibiting low endogenous PGE2 levels, had reduced FGF2. Furthermore, elevation of viable granulosa cell numbers by PGE2 was abolished with FGF2 receptor 1 inhibitor, suggesting that FGF2 mediates this action of PGE2. Epiregulin (EREG), a known PGE2-inducible gene, was studied alongside FGF2. PTGER2 agonist elevated cAMP as well as FGF2 and EREG levels. However, a marked difference between cAMP-induced downstream signaling was observed for FGF2 and EREG. Whereas FGF2 upregulated by PGE2, PTGER2 agonist, or forskolin was unaffected by the protein kinase A (PKA) inhibitor H89, EREG was significantly inhibited. FGF2 was dose-dependently stimulated by the exchange protein directly activated by cAMP (EPAC) activator; a similar induction was observed for EREG. However, forskolin-stimulated FGF2, but not EREG, was inhibited in EPAC1-silenced cells. These findings ascribe a novel autocrine role for PGE2, namely, elevating FGF2 production in granulosa cells. This study also reveals that cAMP-activated EPAC1, rather than PKA, mediates the effect of PGE2/PTGER2 on the expression of FGF2. Stimulation of EREG by PGE2 is also mediated by PTGER2 but, in contrast to FGF2, EREG was found to be PKA sensitive. PGE2-stimulated FGF2 can act to maintain granulosa cell survival; it can also act on ovarian endothelial cells to promote angiogenesis.
Collapse
Affiliation(s)
- Ketan Shrestha
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Rina Meidan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
16
|
Barreto Amaral Teixeira D, Alves Fernandes Júnior G, Beraldo dos Santos Silva D, Bermal Costa R, Takada L, Gustavo Mansan Gordo D, Bresolin T, Carvalheiro R, Baldi F, Galvão de Albuquerque L. Genomic analysis of stayability in Nellore cattle. PLoS One 2017; 12:e0179076. [PMID: 28591167 PMCID: PMC5462402 DOI: 10.1371/journal.pone.0179076] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/23/2017] [Indexed: 12/15/2022] Open
Abstract
Stayability, which can be defined as the probability of a cow calving at a certain age when given the opportunity, is an important reproductive trait in beef cattle because it is directly related to herd profitability. The objective of this study was to estimate genetic parameters and to identify possible genomic regions associated with the phenotypic expression of stayability in Nellore cows. The variance components were estimated by Bayesian inference using a threshold animal model that included the systematic effects of contemporary group and sexual precocity and the random effects of animal and residual. The SNP effects were estimated by the single-step genomic BLUP method using information of 2,838 animals (2,020 females and 930 sires) genotyped with the Illumina High-Density BeadChip Array (San Diego, CA, USA). The variance explained by windows formed by 200 consecutive SNPs was used to identify genomic regions of largest effect on the expression of stayability. The heritability was 0.11 ± 0.01 when A matrix (pedigree) was used and 0.14 ± 0.01 when H matrix (relationship matrix that combines pedigree information and SNP data) was used. A total of 147 candidate genes for stayability were identified on chromosomes 1, 2, 5, 6, 9 and 20 and on the X chromosome. New candidate regions for stayability were detected, most of them related to reproductive, immunological and central nervous system functions.
Collapse
Affiliation(s)
| | | | | | | | - Luciana Takada
- Faculdade de Ciências Agrárias e Veterinárias, UNESP, Jaboticabal, SP, Brazil
| | | | - Tiago Bresolin
- Faculdade de Ciências Agrárias e Veterinárias, UNESP, Jaboticabal, SP, Brazil
| | - Roberto Carvalheiro
- Faculdade de Ciências Agrárias e Veterinárias, UNESP, Jaboticabal, SP, Brazil
- CNPq Fellowship, Brasília, DF, Brazil
| | - Fernando Baldi
- Faculdade de Ciências Agrárias e Veterinárias, UNESP, Jaboticabal, SP, Brazil
- CNPq Fellowship, Brasília, DF, Brazil
| | - Lucia Galvão de Albuquerque
- Faculdade de Ciências Agrárias e Veterinárias, UNESP, Jaboticabal, SP, Brazil
- CNPq Fellowship, Brasília, DF, Brazil
- * E-mail:
| |
Collapse
|
17
|
Mishra S, Parmar M, Chouhan V, Rajesh G, Yadav V, Bharti M, Bharati J, Mondal T, Reshma R, Paul A, Dangi S, Das B, González L, Sharma G, Singh G, Sarkar M. Expression and localization of fibroblast growth factor (FGF) family in corpus luteum during different stages of estrous cycle and synergistic role of FGF2 and vascular endothelial growth factor (VEGF) on steroidogenesis, angiogenesis and survivability of cultured buffalo luteal cells. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.aggene.2016.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
18
|
Woad KJ, Robinson RS. Luteal angiogenesis and its control. Theriogenology 2016; 86:221-8. [PMID: 27177965 DOI: 10.1016/j.theriogenology.2016.04.035] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/11/2016] [Accepted: 03/14/2016] [Indexed: 12/17/2022]
Abstract
Angiogenesis, the formation of new blood vessels from preexisting ones, is critical to luteal structure and function. In addition, it is a complex and tightly regulated process. Not only does rapid and extensive angiogenesis occur to provide the corpus luteum with an unusually high blood flow and support its high metabolic rate, but in the absence of pregnancy, the luteal vasculature must rapidly regress to enable the next cycle of ovarian activity. This review describes a number of key endogenous stimulatory and inhibitory factors, which act in a delicate balance to regulate luteal angiogenesis and ultimately luteal function. In vitro luteal angiogenesis cultures have demonstrated critical roles for fibroblast growth factor 2 (FGF2) in endothelial cell proliferation and sprouting, although other factors such as vascular endothelial growth factor A (VEGFA) and platelet-derived growth factor were important modulators in the control of luteal angiogenesis. Post-transcriptional regulation by small non-coding microRNAs is also likely to play a central role in the regulation of luteal angiogenesis. Appropriate luteal angiogenesis requires the coordinated activity of numerous factors expressed by several cell types at different times, and this review will also describe the role of perivascular pericytes and the importance of vascular maturation and stability. It is hoped that a better understanding of the critical processes underlying the transition from follicle to corpus luteum and subsequent luteal development will benefit the management of luteal function in the future.
Collapse
Affiliation(s)
- Kathryn J Woad
- School of Veterinary Medicine and Science, Sutton Bonington Campus, University of Nottingham, Leicestershire, UK.
| | - Robert S Robinson
- School of Veterinary Medicine and Science, Sutton Bonington Campus, University of Nottingham, Leicestershire, UK
| |
Collapse
|
19
|
Human vascular tissue models formed from human induced pluripotent stem cell derived endothelial cells. Stem Cell Rev Rep 2016; 11:511-25. [PMID: 25190668 DOI: 10.1007/s12015-014-9549-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Here we describe a strategy to model blood vessel development using a well-defined induced pluripotent stem cell-derived endothelial cell type (iPSC-EC) cultured within engineered platforms that mimic the 3D microenvironment. The iPSC-ECs used here were first characterized by expression of endothelial markers and functional properties that included VEGF responsiveness, TNF-α-induced upregulation of cell adhesion molecules (MCAM/CD146; ICAM1/CD54), thrombin-dependent barrier function, shear stress-induced alignment, and 2D and 3D capillary-like network formation in Matrigel. The iPSC-ECs also formed 3D vascular networks in a variety of engineering contexts, yielded perfusable, interconnected lumen when co-cultured with primary human fibroblasts, and aligned with flow in microfluidics devices. iPSC-EC function during tubule network formation, barrier formation, and sprouting was consistent with that of primary ECs, and the results suggest a VEGF-independent mechanism for sprouting, which is relevant to therapeutic anti-angiogenesis strategies. Our combined results demonstrate the feasibility of using a well-defined, stable source of iPSC-ECs to model blood vessel formation within a variety of contexts using standard in vitro formats.
Collapse
|
20
|
Kang BJ, Wang Y, Zhang L, Xiao Z, Li SW. bFGF and VEGF improve the quality of vitrified-thawed human ovarian tissues after xenotransplantation to SCID mice. J Assist Reprod Genet 2015; 33:281-9. [PMID: 26712576 DOI: 10.1007/s10815-015-0628-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 11/29/2015] [Indexed: 01/25/2023] Open
Abstract
PURPOSE The aim of this research is to study whether basic fibroblast growth factor (bFGF) alone or in combination with vascular endothelial growth factor (VEGF) could improve the quality of vitrified-thawed human ovarian tissue xenotransplanted to severe combined immune deficiency (SCID) mice. METHODS After collection and cryopreservation, thawed human ovarian tissue were cultured in vitro for 2 days and then xenografted to severe combined immune deficiency (SCID) mice for 7 days. The in vitro culture medium was separated into six groups, including (A) the blank control group, (B) the human recombinant bFGF (150 ng/ml) group, (C) the bFGF (150 ng/ml)+human recombinant VEGF (25 ng/ml) group, (D) bFGF (150 ng/ml)+VEGF (50 ng/ml) group, (E) bFGF (150 ng/ml)+ VEGF (75 ng/ml) group and (F) bFGF (150 ng/ml) + VEGF (100 ng/ml) group. In addition, eight pieces of thawed ovarian tissue were transplanted without in vitro culture, which serve as the fresh control group. The effect of transplantation was assessed by histological analysis, immunohistochemical staining for CD34, Ki-67, and AC-3 expression, and microvessel density (MVD). RESULTS There was no significant difference between the fresh and blank control group. Compared to the blank control group, the number of follicles, MVD, and rate of Ki-67-positive cells increased significantly in groups B, C, D, E, and F, while apoptosis decreased significantly. Compared to the bFGF treatment group, no significant difference appeared in group C, D, E, and F. CONCLUSIONS The administration of bFGF alone or in combination with VEGF improved the quality of postgraft human ovarian tissue, though VEGF, regardless of different concentrations, did not influence effect of bFGF.
Collapse
Affiliation(s)
- Bei-Jia Kang
- Reproductive Medical Center of West China 2nd University Hospital, Sichuan University, Ren Min Nan Lu, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yan Wang
- Reproductive Medical Center of West China 2nd University Hospital, Sichuan University, Ren Min Nan Lu, Chengdu, Sichuan, 610041, People's Republic of China
| | - Long Zhang
- Reproductive Medical Center of West China 2nd University Hospital, Sichuan University, Ren Min Nan Lu, Chengdu, Sichuan, 610041, People's Republic of China
| | - Zhun Xiao
- Reproductive Medical Center of West China 2nd University Hospital, Sichuan University, Ren Min Nan Lu, Chengdu, Sichuan, 610041, People's Republic of China
| | - Shang-Wei Li
- Reproductive Medical Center of West China 2nd University Hospital, Sichuan University, Ren Min Nan Lu, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
21
|
Lutwak E, Price CA, Abramovich SS, Rabinovitz S, Granot I, Dekel N, Ron D. Expression and regulation of the tumor suppressor, SEF, during folliculogenesis in humans and mice. Reproduction 2014; 148:507-17. [PMID: 25118304 DOI: 10.1530/rep-14-0070] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Similar expression to FGF (Sef or IL17-RD), is a tumor suppressor and an inhibitor of growth factors as well as of pro-inflammatory cytokine signaling. In this study, we examined the regulation of Sef expression by gonadotropins during ovarian folliculogenesis. In sexually immature mice, in situ hybridization (ISH) localized Sef gene expression to early developing oocytes and granulosa cells (GC) but not to theca cells. Sef was also expressed in mouse ovarian endothelial cells, in the fallopian tube epithelium as well as in adipose tissue venules. SEF protein expression, determined by immunohistochemistry (IHC), correlated well with Sef mRNA expression in GC, while differential expression was noticed in oocytes. High Sef mRNA but undetectable SEF protein levels were observed in the oocytes of primary/secondary follicles, while an inverse correlation was found in the oocytes of preantral and small antral follicles. Sef mRNA expression dropped after pregnant mare's serum gonadotropin (PMSG) administration, peaked at 6-8 h after human chorionic gonadotropin (hCG) treatment, and declined by 12 h after this treatment. ISH and IHC localized the changes to oocytes and mural GC following PMSG treatment, whereas Sef expression increased in mural GC and declined in granulosa-lutein cells upon hCG treatment. The ovarian expression of SEF was confirmed using human samples. ISH localized SEF transcripts to human GC of antral follicles but not to corpora lutea. Furthermore, SEF mRNA was detected in human GC recovered from preovulatory follicles. These results are the first to demonstrate SEF expression in a healthy ovary during folliculogenesis. Hormonal regulation of its expression suggests that SEF may be an important factor involved in intra-ovarian control mechanisms.
Collapse
Affiliation(s)
- Ela Lutwak
- Department of BiologyTechnion, Israel Institute of Technology, 32000 Haifa, IsraelFaculté de Médecine VétérinaireCentre de Recherche en Reproduction Animale, Université de Montréal, St-Hyacinthe, Quebec, CanadaDepartment of Biological RegulationThe Weizmann Institute of Science, Rehovot, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Kaplan Medical Center (Affiliated to the Medical School of the Hebrew University and Hadassah, Jerusalem), Rehovot, Israel
| | - Christopher A Price
- Department of BiologyTechnion, Israel Institute of Technology, 32000 Haifa, IsraelFaculté de Médecine VétérinaireCentre de Recherche en Reproduction Animale, Université de Montréal, St-Hyacinthe, Quebec, CanadaDepartment of Biological RegulationThe Weizmann Institute of Science, Rehovot, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Kaplan Medical Center (Affiliated to the Medical School of the Hebrew University and Hadassah, Jerusalem), Rehovot, Israel
| | - Sagit-Sela Abramovich
- Department of BiologyTechnion, Israel Institute of Technology, 32000 Haifa, IsraelFaculté de Médecine VétérinaireCentre de Recherche en Reproduction Animale, Université de Montréal, St-Hyacinthe, Quebec, CanadaDepartment of Biological RegulationThe Weizmann Institute of Science, Rehovot, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Kaplan Medical Center (Affiliated to the Medical School of the Hebrew University and Hadassah, Jerusalem), Rehovot, Israel
| | - Shiri Rabinovitz
- Department of BiologyTechnion, Israel Institute of Technology, 32000 Haifa, IsraelFaculté de Médecine VétérinaireCentre de Recherche en Reproduction Animale, Université de Montréal, St-Hyacinthe, Quebec, CanadaDepartment of Biological RegulationThe Weizmann Institute of Science, Rehovot, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Kaplan Medical Center (Affiliated to the Medical School of the Hebrew University and Hadassah, Jerusalem), Rehovot, Israel
| | - Irit Granot
- Department of BiologyTechnion, Israel Institute of Technology, 32000 Haifa, IsraelFaculté de Médecine VétérinaireCentre de Recherche en Reproduction Animale, Université de Montréal, St-Hyacinthe, Quebec, CanadaDepartment of Biological RegulationThe Weizmann Institute of Science, Rehovot, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Kaplan Medical Center (Affiliated to the Medical School of the Hebrew University and Hadassah, Jerusalem), Rehovot, Israel
| | - Nava Dekel
- Department of BiologyTechnion, Israel Institute of Technology, 32000 Haifa, IsraelFaculté de Médecine VétérinaireCentre de Recherche en Reproduction Animale, Université de Montréal, St-Hyacinthe, Quebec, CanadaDepartment of Biological RegulationThe Weizmann Institute of Science, Rehovot, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Kaplan Medical Center (Affiliated to the Medical School of the Hebrew University and Hadassah, Jerusalem), Rehovot, Israel
| | - Dina Ron
- Department of BiologyTechnion, Israel Institute of Technology, 32000 Haifa, IsraelFaculté de Médecine VétérinaireCentre de Recherche en Reproduction Animale, Université de Montréal, St-Hyacinthe, Quebec, CanadaDepartment of Biological RegulationThe Weizmann Institute of Science, Rehovot, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Kaplan Medical Center (Affiliated to the Medical School of the Hebrew University and Hadassah, Jerusalem), Rehovot, Israel
| |
Collapse
|
22
|
Miyamoto A, Shirasuna K, Haneda S, Shimizu T, Matsui M. CELL BIOLOGY SYMPOSIUM: perspectives: possible roles of polymorphonuclear neutrophils in angiogenesis and lymphangiogenesis in the corpus luteum during development and early pregnancy in ruminants. J Anim Sci 2014; 92:1834-9. [PMID: 24663155 DOI: 10.2527/jas.2013-7332] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The establishment of pregnancy requires well-balanced regulation of the endocrine and immune systems and involves interactions among the conceptus, oviduct-uterus, and corpus luteum (CL). In particular, a rapid increase in plasma progesterone during the first week after ovulation is critical for the growth of the conceptus and successful pregnancy in cattle. Events involved in maternal recognition of pregnancy (MRP) may commence within 1 wk from AI, when interferon-stimulated gene expression in circulating polymorphonuclear neutrophils (PMN) increases in pregnant cows. To regulate optimal endocrine conditions within this time, the CL must develop rapidly, with active angiogenesis and lymphangiogenesis. The major angiogenic factors, vascular endothelial growth factor and fibroblast growth factor 2, contribute to the development of the CL but may also act as chemoattractants for PMN. Indeed, the number of PMN is greatest in the new CL, where PMN together with IL-8 induce active angiogenesis and lymphangiogenesis. During MRP, the conceptus secretes interferon tau (IFNT), which prevents CL regression by inhibiting luteolytic release of PGF2α from uterine endometrium. In addition, IFNT and PGE2 reach the CL and may contribute to desensitizing the CL to the luteolytic effects of PGF2α. In the bovine CL, lymphangiogenesis, stimulated by IFNT, may occur during MRP, and thus a shift of local immunity might occur at this timing. The aforementioned evidence supports the possible involvement of PMN in the establishment of pregnancy via CL regulation. Further investigation could expand our understanding of the communication between zygotes, PMN, and reproductive organs during early pregnancy. This should provide new insight into the contribution of neutrophils to CL function and immune tolerance during early pregnancy in ruminants.
Collapse
Affiliation(s)
- A Miyamoto
- Graduate School of Animal and Food Hygiene, Obihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, Japan
| | | | | | | | | |
Collapse
|
23
|
Ever-changing cell interactions during the life span of the corpus luteum: Relevance to luteal regression. Reprod Biol 2014; 14:75-82. [DOI: 10.1016/j.repbio.2013.12.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Accepted: 12/17/2013] [Indexed: 11/22/2022]
|
24
|
Fátima LA, Evangelista MC, Silva RS, Cardoso APM, Baruselli PS, Papa PC. FSH up-regulates angiogenic factors in luteal cells of buffaloes. Domest Anim Endocrinol 2013; 45:224-37. [PMID: 24209507 DOI: 10.1016/j.domaniend.2013.09.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 09/11/2013] [Accepted: 09/12/2013] [Indexed: 02/05/2023]
Abstract
Follicle-stimulating hormone has been widely used to induce superovulation in buffaloes and cows and usually triggers functional and morphologic alterations in the corpus luteum (CL). Several studies have shown that FSH is involved in regulating vascular development and that adequate angiogenesis is essential for normal luteal development. Angiogenesis is regulated by many growth factors, of which vascular endothelial growth factor (VEGF) and fibroblast growth factor 2 (FGF2) have an established central role. Therefore, we have used a combination of in vitro and in vivo studies to assess the effects of FSH on the expression of VEGF and FGF2 and their receptors in buffalo luteal cells. The in vivo model consisted of 12 buffalo cows, divided into control (n = 6) and superovulated (n = 6) groups, and CL samples were collected on day 6 after ovulation. In this model, we analyzed the gene and protein expression of FGF2 and its receptors and the protein expression of VEGFA systems with the use of real-time PCR, Western blot analysis, and immunohistochemistry. In the in vitro model, granulosa cells were collected from small follicles (diameter, 4-6 mm) of buffaloes and cultured for 4 d in serum-free medium with or without FSH (10 ng/mL). To induce in vitro luteinization, LH (250 ng/mL) and fetal bovine serum (10%) were added to the medium, and granulosa cells were maintained in culture for 4 d more. The progesterone concentration in the medium was measured at days 4, 5, and 8 after the beginning of cell culture. Cells were collected at day 8 and subjected to real-time PCR, Western blot analysis, and immunofluorescence for assessment of the expression of FGF2, VEGF, and their receptors. To address the percentage of steroidogenic and growth factor-expressing cells in the culture, flow cytometry was performed. We observed that in superovulated buffalo CL, the FGF2 system mRNA expression was decreased even as protein expression was increased and that the VEGF protein was increased (P < 0.05). In vitro experiments with granulosa cells showed an increase in the mRNA expression of VEGF and FGF2 and its receptors 1 and 2 and protein expression of VEGF, kinase insert domain receptor, FGF receptor 2, and FGF receptor 3 in cells treated with FSH (P < 0.05), in contrast to the in vivo experiments. Moreover, the progesterone production by FSH-treated cells was elevated compared with untreated cells (P < 0.05). Our findings indicate that VEGF, FGF2, and their receptors were differentially regulated by FSH in vitro and in vivo in buffalo luteal cells, which points toward a role of CL environment in modulating cellular answers to gonadotropins.
Collapse
MESH Headings
- Angiogenic Proteins/analysis
- Angiogenic Proteins/genetics
- Animals
- Buffaloes/metabolism
- Cells, Cultured
- Female
- Fibroblast Growth Factor 2/analysis
- Fibroblast Growth Factor 2/genetics
- Fluorescent Antibody Technique
- Follicle Stimulating Hormone/pharmacology
- Granulosa Cells/chemistry
- Granulosa Cells/drug effects
- Granulosa Cells/metabolism
- Luteal Cells/chemistry
- Luteal Cells/metabolism
- Luteinizing Hormone/pharmacology
- Male
- Progesterone/biosynthesis
- RNA, Messenger/analysis
- Real-Time Polymerase Chain Reaction/veterinary
- Receptor, Fibroblast Growth Factor, Type 2/analysis
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 3/analysis
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Superovulation/physiology
- Up-Regulation
- Vascular Endothelial Growth Factor A/analysis
- Vascular Endothelial Growth Factor A/genetics
Collapse
Affiliation(s)
- L A Fátima
- Department of Surgery, Sector of Anatomy, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Prof. Dr Orlando Marques Paiva, 87, São Paulo, SP, 05508-270, Brazil.
| | | | | | | | | | | |
Collapse
|
25
|
Miyamoto A, Shirasuna K, Shimizu T, Matsui M. Impact of angiogenic and innate immune systems on the corpus luteum function during its formation and maintenance in ruminants. Reprod Biol 2013; 13:272-8. [PMID: 24287035 DOI: 10.1016/j.repbio.2013.09.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 09/27/2013] [Accepted: 09/28/2013] [Indexed: 12/25/2022]
Abstract
The corpus luteum (CL) is formed from an ovulated follicle, and grows rapidly to secrete progesterone (P4) thereby supporting implantation and maintenance of pregnancy. It is now evident that angiogenesis is necessary to form the structure of the developing CL as well as to acquire the steroidogenic capacity to secrete large amounts of P4. It is of interest that the increases in CL size, plasma P4 concentration and luteal blood flow are occurring in parallel during the first seven days after ovulation. Angiogenic factors, such as vascular endothelial growth factor-A (VEGFA) and basic fibroblast growth factor (FGF2), play a central role in promoting cell proliferation and angiogenesis in the developing CL. Angiopoietins regulate the stability of blood vessels, which directly affects angiogenesis or angiolysis via angiogenic factors. Vasohibin-1 is a novel negative feedback regulator, which inhibits VEGF-based vasculogenesis. It became evident that the immune cells, i.e., macrophages, eosinophils and neutrophils are recruited into the CL - using the innate immune system - just after ovulation which is accompanied by bleeding. The immune cells support active angiogenesis and thus the growth of the CL. In cows, the lymphatic system, but not blood vascular system, is reconstituted during early pregnancy, and embryonic trophoblast-derived interferon tau could play a crucial role in inducing lymphangiogenesis. This novel phenomenon may support a maternal recognition of pregnancy in shifting the local systems in such a way that they ensure a long-term supply of P4 over the period of pregnancy. Overall, the current findings support the concept that several major components involved in the regulation of the CL development and maintenance overlap in stimulating steroidogenesis, angiogenesis, vascular function and the innate immune system.
Collapse
Affiliation(s)
- Akio Miyamoto
- Graduate School of Animal and Food Hygiene, Obihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, Japan.
| | | | | | | |
Collapse
|
26
|
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: 2.9] [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
|
27
|
Skarzynski DJ, Piotrowska-Tomala KK, Lukasik K, Galvão A, Farberov S, Zalman Y, Meidan R. Growth and Regression in Bovine Corpora Lutea: Regulation by Local Survival and Death Pathways. Reprod Domest Anim 2013; 48 Suppl 1:25-37. [DOI: 10.1111/rda.12203] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Accepted: 05/20/2013] [Indexed: 11/30/2022]
Affiliation(s)
- DJ Skarzynski
- Department of Reproductive Immunology and Pathology; Institute of Animal Reproduction and Food Research; Polish Academy of Sciences; Olsztyn; Poland
| | - KK Piotrowska-Tomala
- Department of Reproductive Immunology and Pathology; Institute of Animal Reproduction and Food Research; Polish Academy of Sciences; Olsztyn; Poland
| | - K Lukasik
- Department of Reproductive Immunology and Pathology; Institute of Animal Reproduction and Food Research; Polish Academy of Sciences; Olsztyn; Poland
| | - A Galvão
- Department of Reproductive Immunology and Pathology; Institute of Animal Reproduction and Food Research; Polish Academy of Sciences; Olsztyn; Poland
| | - S Farberov
- Department of Animal Sciences; The Robert H. Smith Faculty of Agriculture, Food and Environment; The Hebrew University of Jerusalem; Rehovot; Israel
| | - Y Zalman
- Department of Animal Sciences; The Robert H. Smith Faculty of Agriculture, Food and Environment; The Hebrew University of Jerusalem; Rehovot; Israel
| | - R Meidan
- Department of Animal Sciences; The Robert H. Smith Faculty of Agriculture, Food and Environment; The Hebrew University of Jerusalem; Rehovot; Israel
| |
Collapse
|
28
|
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: 3.2] [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
|
29
|
Laird M, Woad KJ, Hunter MG, Mann GE, Robinson RS. Fibroblast growth factor 2 induces the precocious development of endothelial cell networks in bovine luteinising follicular cells. Reprod Fertil Dev 2013; 25:372-86. [DOI: 10.1071/rd12182] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2012] [Accepted: 10/11/2012] [Indexed: 11/23/2022] Open
Abstract
The transition from follicle to corpus luteum represents a period of intense angiogenesis; however, the exact roles of angiogenic factors during this time remain to be elucidated. Thus, the roles of vascular endothelial growth factor (VEGF) A, fibroblast growth factor (FGF) 2 and LH in controlling angiogenesis were examined in the present study. A novel serum-free luteinising follicular angiogenesis culture system was developed in which progesterone production increased during the first 5 days and was increased by LH (P < 0.01). Blockade of signalling from FGF receptors (SU5402; P < 0.001) and, to a lesser extent, VEGF receptors (SU1498; P < 0.001) decreased the development of endothelial cell (EC) networks. Conversely, FGF2 dose-dependently (P < 0.001) induced the precocious transition of undeveloped EC islands into branched networks associated with a twofold increase in the number of branch points (P < 0.001). In contrast, VEGFA had no effect on the area of EC networks or the number of branch points. LH had no effect on the area of EC networks, but it marginally increased the number of branch points (P < 0.05) and FGF2 production (P < 0.001). Surprisingly, progesterone production was decreased by FGF2 (P < 0.01) but only on Day 5 of culture. Progesterone production was increased by SU5402 (P < 0.001) and decreased by SU1498 (P < 0.001). These results demonstrate that FGF and VEGF receptors play a fundamental role in the formation of luteal EC networks in vitro, which includes a novel role for FGF2 in induction of EC sprouting.
Collapse
|
30
|
Macchiarelli G, Palmerini MG, Nottola SA, Cecconi S, Tanemura K, Sato E. Restoration of corpus luteum angiogenesis in immature hypothyroid rdw rats after thyroxine treatment: morphologic and molecular evidence. Theriogenology 2012; 79:116-26. [PMID: 23122683 DOI: 10.1016/j.theriogenology.2012.09.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 09/16/2012] [Accepted: 09/17/2012] [Indexed: 01/23/2023]
Abstract
Thyroxine (T4) plus gonadotropins might stimulate ovarian follicular angiogenesis in immature infertile hypothyroid rdw rats by upregulating mRNA expression of major angiogenic factors. Development of growing corpus luteum (CL) is strongly related to angiogenesis and to morphofunctional development of microcirculation. Our aim was to investigate if T4 is involved in CL angiogenesis and in the activation of capillary cells and angiogenic factors after ovulation in a spontaneous model of hypothyroidism, the rdw rat. Rdw rats were treated with T4 plus gonadotropins (equine chorionic gonadotropin plus human chorionic gonadotropin; eCG+hCG) or gonadotropins alone in order to evaluate the effects of T4 on early luteal angiogenesis, on microvascular cells and on expression of major growth factors which are involved in the regulation of angiogenesis. Wistar-Imamichi rats treated with gonadotropins were used as controls. The ovaries were collected 4 days after hCG administration and analyzed using morphologic and molecular approaches. Thyroxine plus gonadotropins stimulated the growth of CLs and follicles as in controls, differently from rdw rats treated only with gonadotropins, in which CLs were not found and only small follicles, often atretic, could be recognized. In T4 plus gonadotropin-treated rdw rats CLs showed increased microvasculature, numerous activated capillaries characterized by sprouting and other angiogenic figures, and associated pericytes. Quantitative analysis revealed that the number of pericytes in T4 plus gonadotropin-treated rdw rats was comparable with that found in control rats and was significantly higher than that found in gonadotropin-treated rdw rats. The mRNA expression of vascular endothelial growth factor and basic fibroblast growth factor was significantly higher in control rats and in T4 plus gonadotropin-treated rdw rats than in gonadotropin-treated rdw rats. mRNA expression of tumor necrosis factor α, transforming growth factor β, and epidermal growth factor did not show significant changes. Our data originally demonstrated that T4 promoted the growth of an active microcirculation in developing CLs of gonadotropin-primed hypothyroid rdw rats, mainly by inducing sprouting angiogenesis, pericyte recruitment, and upregulation of mRNA expression of vascular endothelial growth factor and basic fibroblast growth factor. In conclusion, we suggest that T4 plays a key role in restoring luteal angiogenesis in ovaries of immature hypothyroid rdw rats.
Collapse
Affiliation(s)
- Guido Macchiarelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| | | | | | | | | | | |
Collapse
|
31
|
Joseph C, Hunter MG, Sinclair KD, Robinson RS. The expression, regulation and function of secreted protein, acidic, cysteine-rich in the follicle–luteal transition. Reproduction 2012; 144:361-72. [DOI: 10.1530/rep-12-0099] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The role of the tissue remodelling protein, secreted protein, acidic, cysteine-rich (SPARC), in key processes (e.g. cell reorganisation and angiogenesis) that occur during the follicle–luteal transition is unknown. Hence, we investigated the regulation of SPARC in luteinsing follicular cells and potential roles of SPARC peptide 2.3 in a physiologically relevant luteal angiogenesis culture system. SPARC protein was detected mainly in the theca layer of bovine pre-ovulatory follicles, but its expression was considerably greater in the corpus haemorrhagicum. Similarly, SPARC protein (western blotting) was up-regulated in luteinising granulosa but not in theca cells during a 6-day culture period. Potential regulatory candidates were investigated in luteinising granulosa cells: LH did not affect SPARC (P>0.05); transforming growth factor (TGF) B1 (P<0.001) dose dependently induced the precocious expression of SPARC and increased final levels: this effect was blocked (P<0.001) by SB505124 (TGFB receptor 1 inhibitor). Additionally, fibronectin, which is deposited during luteal development, increased SPARC (P<0.01). In luteal cells, fibroblast growth factor 2 decreased SPARC (P<0.001) during the first 5 days of culture, while vascular endothelial growth factor A increased its expression (P<0.001). Functionally, KGHK peptide, a SPARC proteolytic fragment, stimulated the formation of endothelial cell networks in a luteal cell culture system (P<0.05) and increased progesterone production (P<0.05). Collectively, these findings indicate that SPARC is intricately regulated by pro-angiogenic and other growth factors together with components of the extracellular matrix during the follicle–luteal transition. Thus, it is possible that SPARC plays an important modulatory role in regulating angiogenesis and progesterone production during luteal development.
Collapse
|
32
|
Shirasuna K, Nitta A, Sineenard J, Shimizu T, Bollwein H, Miyamoto A. Vascular and immune regulation of corpus luteum development, maintenance, and regression in the cow. Domest Anim Endocrinol 2012; 43:198-211. [PMID: 22560178 DOI: 10.1016/j.domaniend.2012.03.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 03/18/2012] [Accepted: 03/28/2012] [Indexed: 01/05/2023]
Abstract
The bovine corpus luteum (CL) is a unique, transient organ with well-coordinated mechanisms by which its development, maintenance, and regression are effectively controlled. Angiogenic factors, such as vascular endothelial growth factor A and basic fibroblast growth factor, play an essential role in promoting progesterone secretion, cell proliferation, and angiogenesis. These processes are critically regulated, through both angiogenic and immune systems, by the specific immune cells, including macrophages, eosinophils, and neutrophils, that are recruited into the developing CL. The bovine luteolytic cascade appears to be similar to that of general acute inflammation in terms of time-dependent infiltration by immune cells (neutrophils, macrophages, and T lymphocytes) and drastic changes in vascular tonus and blood flow, which are regulated by luteal nitric oxide and the vasoconstrictive factors endothelin-1 and angiotensin II. Over the period of maternal recognition of pregnancy, the maternal immune system should be well controlled to accept the semiallograft fetus. The information on the presence of the developing embryo in the genital tract is suggested to be transmitted to the ovary by both the endocrine system and the circulating immune cells. In the bovine CL, the lymphatic system, but not the blood vascular system, is reconstituted during early pregnancy, and interferon tau from the embryo could trigger this novel phenomenon. Collectively, the angiogenic and vasoactive factors produced by luteal cells and the time-dependently recruited immune cells within the CL and their interactions appear to play critical roles in regulating luteal functions throughout the life span of the CL.
Collapse
Affiliation(s)
- K Shirasuna
- Graduate School of Animal and Food Hygiene, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | | | | | | | | | | |
Collapse
|
33
|
Kliem H, Rodler D, Ulbrich SE, Sinowatz F, Berisha B, Meyer HHD, Schams D. Dexamethasone-induced eosinopenia is associated with lower progesterone production in cattle. Reprod Domest Anim 2012; 48:137-48. [PMID: 22621206 DOI: 10.1111/j.1439-0531.2012.02116.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Eosinophilic cells accumulate in the capillaries of the bovine Graafian follicle shortly before ovulation and in the early developing corpus luteum (CL). Suppressing the migration of these eosinophilic cells by dexamethasone allowed us to evaluate their possible function in the CL development. Brown Swiss cows (n = 10) were randomly subdivided into two groups (n = 5). Every group was used once as control group and once as experimental group with two oestrous cycles between each treatment. Eighteen hours (h) after oestrus synchronization, dexamethasone or saline was given. Ovulation was induced 24 h later with gonadotropin-releasing hormone. Another injection of dexamethasone or saline was given 12 h later. Eosinophilic cells in the blood were counted daily until day 7 after the first dexamethasone injection. The collection of ovaries took place at days 1, 2 and 5. Gene expression, protein concentration and location of angiogenic factors, chemokines, insulin-like growth factor 1 (IGF1) and eosinophilic cells were studied. No eosinophilic cells were found in the CL of the treatment group. Blood progesterone decreased significantly in the dexamethasone group from day 8 to 17. The protein concentration of FGF2 increased significantly in CL tissue at day 2 and VEGFA decreased. Local IGF1 gene expression in the CL was not regulated. We assume from our data that the migration of eosinophilic cells into the early CL is not an essential, but an important stimulus for angiogenesis during early CL development in cattle.
Collapse
Affiliation(s)
- H Kliem
- Physiology Weihenstephan, Technische Universität München, Freising, Germany
| | | | | | | | | | | | | |
Collapse
|
34
|
Zalman Y, Klipper E, Farberov S, Mondal M, Wee G, Folger JK, Smith GW, Meidan R. Regulation of angiogenesis-related prostaglandin f2alpha-induced genes in the bovine corpus luteum. Biol Reprod 2012; 86:92. [PMID: 22174022 DOI: 10.1095/biolreprod.111.095067] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
We recently compared prostaglandin F2alpha (PG)-induced global gene expression profiles in PG-refractory, bovine corpus luteum (CL) collected on Day 4 of the estrous cycle, versus PG-responsive, Day 11 CL. Transcriptome analyses led us to study the regulation of angiogenesis-related genes by PG and their functions in luteal endothelial cells (ECs). We found that PG regulated angiogenesis-modulating factors in a luteal stage-dependent way. A robust increase in FGF2 expression (mRNA and protein) occurred in the PG-refractory Day 4 CL promoting CL survival and function. Inhibitors of FGF2 action, thrombospondin 1 and 2, their receptor (CD36), and PTX3 were upregulated by PG specifically in Day 11 CL undergoing luteolysis. VEGF mRNA decreased 4 h post-PG in both Day 4 and Day 11 CL. The resulting destabilization of blood vessels in Day 11 CL is expected to weaken the gland and reduce its hormonal output. These genes were expressed in dispersed luteal ECs and steroidogenic cells; however, thrombospondin 1 and FGF2 were more abundant in luteal ECs. Expression of such genes and their ability to modulate FGF2 actions were investigated. Similar to its in vivo effect, PG, in vitro, stimulated the expression of thrombospondins and PTX3 genes in several luteal cell models. Importantly, these factors influenced the angiogenic properties of luteal ECs. FGF2 dose-dependently enhanced cell migration and proliferation, whereas thrombospondin 1 and PTX3 inhibited FGF2 actions in luteal ECs. Collectively, the data presented here suggest that, by tilting the balance between pro- and antiangiogenic factors, PG can potentially control the ability of the CL to resist or advance toward luteolysis.
Collapse
Affiliation(s)
- Yulia Zalman
- Department of Animal Sciences, the Robert H. Smith Faculty of Agriculture, Food and Environment, the Hebrew University of Jerusalem, Rehovot, Israel
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Woad KJ, Hunter MG, Mann GE, Laird M, Hammond AJ, Robinson RS. Fibroblast growth factor 2 is a key determinant of vascular sprouting during bovine luteal angiogenesis. Reproduction 2012; 143:35-43. [DOI: 10.1530/rep-11-0277] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Fibroblast growth factor (FGF) 2 and vascular endothelial growth factor (VEGF) A are thought to be key controllers of luteal angiogenesis; however, their precise roles in the regulation and coordination of this complex process remain unknown. Thus, the temporal and spatial patterns of endothelial network formation were determined by culturing mixed cell types from early bovine corpora lutea on fibronectin in the presence of FGF2 and VEGFA (6 h to 9 days). Endothelial cells, as determined by von Willebrand factor immunohistochemistry, initially grew in cell islands (days 0–3), before undergoing a period of vascular sprouting to display a more tubule-like appearance (days 3–6), and after 9 days in culture had formed extensive intricate networks. Mixed populations of luteal cells were treated with SU1498 (VEGF receptor 2 inhibitor) or SU5402 (FGF receptor 1 inhibitor) or control on days 0–3, 3–6 or 6–9 to determine the role of FGF2 and VEGFA during these specific windows. The total area of endothelial cells was unaffected by SU1498 treatment during any window. In contrast, SU5402 treatment caused maximal reduction in the total area of endothelial cell networks on days 3–6 vs controls (mean reduction 81%;P<0.001) during the period of tubule initiation. Moreover, SU5402 treatment on days 3–6 dramatically reduced the total number of branch points (P<0.001) and degree of branching per endothelial cell island (P<0.05) in the absence of changes in mean island area. This suggests that FGF2 is a key determinant of vascular sprouting and hence critical to luteal development.
Collapse
|
36
|
Nitta A, Shirasuna K, Haneda S, Matsui M, Shimizu T, Matsuyama S, Kimura K, Bollwein H, Miyamoto A. Possible involvement of IFNT in lymphangiogenesis in the corpus luteum during the maternal recognition period in the cow. Reproduction 2011; 142:879-92. [PMID: 21933879 DOI: 10.1530/rep-11-0157] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The corpus luteum (CL), which secretes large amounts of progesterone and is thus essential for establishing pregnancy, contains various types of immune cells that may play essential roles in CL function by generating immune responses. The lymphatic system is the second circulation system and is necessary for immune function, but the lymphatic system of the bovine CL has not been characterized in detail. We collected bovine CLs on days 12 and 16 of the estrous cycle (C12 and C16) and days 16 and 40 of early pregnancy (P16 and P40). Lymphatic endothelial hyaluronan receptor 1 (LYVE1) protein was detected in the CL by immunohistochemistry and western blotting and increased at P40 compared with C16. The mRNA expression levels of lymphangiogenic factors, such as vascular endothelial growth factor-C (VEGFC), VEGFD, and their common receptor VEGFR3, as well as the lymphatic endothelial cell (LyEC) marker podoplanin, increased in P16 and P40 CLs. Thus, it is suggested that the lymphatic system of the bovine CL reconstitutes during early pregnancy. Interferon tau (IFNT) from the conceptus in the uterus is a candidate for activating luteal lymphangiogenesis during the maternal recognition period (MRP). We found that treatment of LyECs isolated from internal iliac lymphatic vessels with IFNT stimulated LyEC proliferation and significantly increased mRNA expression of VEGFC and IFN-stimulated gene 15. Moreover, both IFNT and VEGFC induced LyECs to form capillary-like tubes in vitro. In conclusion, it is suggested that new lymphangiogenesis in the bovine CL begins during the MRP and that IFNT may mediate this novel phenomenon.
Collapse
Affiliation(s)
- Akane Nitta
- Graduate School of Animal and Food Hygiene, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Robinson RS, Woad KJ, Hammond AJ, Laird M, Hunter MG, Mann GE. Angiogenesis and vascular function in the ovary. Reproduction 2009; 138:869-81. [DOI: 10.1530/rep-09-0283] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Ovarian function is dependent on the establishment and continual remodelling of a complex vascular system. This enables the follicle and/or corpus luteum (CL) to receive the required supply of nutrients, oxygen and hormonal support as well as facilitating the release of steroids. Moreover, the inhibition of angiogenesis results in the attenuation of follicular growth, disruption of ovulation and drastic effects on the development and function of the CL. It appears that the production and action of vascular endothelial growth factor A (VEGFA) is necessary at all these stages of development. However, the expression of fibroblast growth factor 2 (FGF2) in the cow is more dynamic than that of VEGFA with a dramatic upregulation during the follicular–luteal transition. This upregulation is then likely to initiate intense angiogenesis in the presence of high VEGFA levels. Recently, we have developed a novel ovarian physiological angiogenesis culture system in which highly organised and intricate endothelial cell networks are formed. This system will enable us to elucidate the complex inter-play between FGF2 and VEGFA as well as other angiogenic factors in the regulation of luteal angiogenesis. Furthermore, recent evidence indicates that pericytes might play an active role in driving angiogenesis and highlights the importance of pericyte–endothelial interactions in this process. Finally, the targeted promotion of angiogenesis may lead to the development of novel strategies to alleviate luteal inadequacy and infertility.
Collapse
|