1
|
You Q, Ke Y, Chen X, Yan W, Li D, Chen L, Wang R, Yu J, Hong H. Loss of Endothelial Annexin A1 Aggravates Inflammation-Induched Vascular Aging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307040. [PMID: 38358087 PMCID: PMC11022713 DOI: 10.1002/advs.202307040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 01/03/2024] [Indexed: 02/16/2024]
Abstract
Chronic inflammation is increasingly considered as the most important component of vascular aging, contributing to the progression of age-related cardiovascular diseases. To delay the process of vascular aging, anti-inflammation may be an effective measure. The anti-inflammatory factor annexin A1 (ANXA1) is shown to participate in several age-related diseases; however, its function during vascular aging remains unclear. Here, an ANXA1 knockout (ANXA1-/-) and an endothelial cell-specific ANXA1 deletion mouse (ANXA1△EC) model are used to investigate the role of ANXA1 in vascular aging. ANXA1 depletion exacerbates vascular remodeling and dysfunction while upregulates age- and inflammation-related protein expression. Conversely, Ac2-26 (a mimetic peptide of ANXA1) supplementation reverses this phenomenon. Furthermore, long-term tumor necrosis factor-alpha (TNF-α) induction of human umbilical vein endothelial cells (HUVECs) increases cell senescence. Finally, the senescence-associated secretory phenotype and senescence-related protein expression, rates of senescence-β-galactosidase positivity, cell cycle arrest, cell migration, and tube formation ability are observed in both ANXA1-knockdown HUVECs and overexpressed ANXA1-TNF-α induced senescent HUVECs. They also explore the impact of formyl peptide receptor 2 (a receptor of ANXA1) in an ANXA1 overexpression inflammatory model. These data provide compelling evidence that age-related inflammation in arteries contributes to senescent endothelial cells that promote vascular aging.
Collapse
Affiliation(s)
- Qinyi You
- Department of Geriatrics, Fujian Medical University Union Hospital, Fujian Key Laboratory of Vascular Aging (Fujian Medical University), Fujian Institute of Geriatrics, Department of Cardiology, Fujian Heart Disease Center, Fujian Clinical Research Center for Vascular and Brain Aging, Fuzhou, Fujian, 350001, China
| | - Yilang Ke
- Department of Geriatrics, Fujian Medical University Union Hospital, Fujian Key Laboratory of Vascular Aging (Fujian Medical University), Fujian Institute of Geriatrics, Department of Cardiology, Fujian Heart Disease Center, Fujian Clinical Research Center for Vascular and Brain Aging, Fuzhou, Fujian, 350001, China
| | - Xiaofeng Chen
- Department of Geriatrics, Fujian Medical University Union Hospital, Fujian Key Laboratory of Vascular Aging (Fujian Medical University), Fujian Institute of Geriatrics, Department of Cardiology, Fujian Heart Disease Center, Fujian Clinical Research Center for Vascular and Brain Aging, Fuzhou, Fujian, 350001, China
| | - Wanhong Yan
- Department of Geriatrics, Fujian Medical University Union Hospital, Fujian Key Laboratory of Vascular Aging (Fujian Medical University), Fujian Institute of Geriatrics, Department of Cardiology, Fujian Heart Disease Center, Fujian Clinical Research Center for Vascular and Brain Aging, Fuzhou, Fujian, 350001, China
| | - Dang Li
- Department of Geriatrics, Fujian Medical University Union Hospital, Fujian Key Laboratory of Vascular Aging (Fujian Medical University), Fujian Institute of Geriatrics, Department of Cardiology, Fujian Heart Disease Center, Fujian Clinical Research Center for Vascular and Brain Aging, Fuzhou, Fujian, 350001, China
| | - Lu Chen
- Department of Geriatrics, Fujian Medical University Union Hospital, Fujian Key Laboratory of Vascular Aging (Fujian Medical University), Fujian Institute of Geriatrics, Department of Cardiology, Fujian Heart Disease Center, Fujian Clinical Research Center for Vascular and Brain Aging, Fuzhou, Fujian, 350001, China
| | - Run Wang
- Department of Geriatrics, Fujian Medical University Union Hospital, Fujian Key Laboratory of Vascular Aging (Fujian Medical University), Fujian Institute of Geriatrics, Department of Cardiology, Fujian Heart Disease Center, Fujian Clinical Research Center for Vascular and Brain Aging, Fuzhou, Fujian, 350001, China
| | - Jie Yu
- Department of Geriatrics, Fujian Medical University Union Hospital, Fujian Key Laboratory of Vascular Aging (Fujian Medical University), Fujian Institute of Geriatrics, Department of Cardiology, Fujian Heart Disease Center, Fujian Clinical Research Center for Vascular and Brain Aging, Fuzhou, Fujian, 350001, China
| | - Huashan Hong
- Department of Geriatrics, Fujian Medical University Union Hospital, Fujian Key Laboratory of Vascular Aging (Fujian Medical University), Fujian Institute of Geriatrics, Department of Cardiology, Fujian Heart Disease Center, Fujian Clinical Research Center for Vascular and Brain Aging, Fuzhou, Fujian, 350001, China
| |
Collapse
|
2
|
Moreli JB, Santos MRD, Calderon IDMP, Hebeda CB, Farsky SHP, Bevilacqua E, Oliani SM. The Role of Annexin A1 in DNA Damage Response in Placental Cells: Impact on Gestational Diabetes Mellitus. Int J Mol Sci 2023; 24:10155. [PMID: 37373303 DOI: 10.3390/ijms241210155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
The functions of annexin A1 (ANXA1), which is expressed on membranes and in cytoplasmic granules, have been fully described. Nonetheless, the role of this protein in protecting against DNA damage in the nucleus is still emerging and requires further investigation. Here, we investigated the involvement of ANXA1 in the DNA damage response in placental cells. Placenta was collected from ANXA1 knockout mice (AnxA1-/-) and pregnant women with gestational diabetes mellitus (GDM). The placental morphology and ANXA1 expression, which are related to the modulation of cellular response markers in the presence of DNA damage, were analyzed. The total area of AnxA1-/- placenta was smaller due to a reduced labyrinth zone, enhanced DNA damage, and impaired base excision repair (BER) enzymes, which resulted in the induction of apoptosis in the labyrinthine and junctional layers. The placentas of pregnant women with GDM showed reduced expression of AnxA1 in the villous compartment, increased DNA damage, apoptosis, and a reduction of enzymes involved in the BER pathway. Our translational data provide valuable insights into the possible involvement of ANXA1 in the response of placental cells to oxidative DNA damage and represent an advancement in investigations into the mechanisms involved in placental biology.
Collapse
Affiliation(s)
- Jusciele Brogin Moreli
- Post-Graduation in Structural and Functional Biology, Federal University of São Paulo (UNIFESP), São Paulo 04023-062, Brazil
- Faceres School of Medicine (FACERES), São José do Rio Preto 15090-305, Brazil
| | - Mayk Ricardo Dos Santos
- Department of Biology, School of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto 15054-000, Brazil
| | - Iracema de Mattos Paranhos Calderon
- Graduate Program in Gynecology, Obstetrics and Mastology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
| | - Cristina Bichels Hebeda
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo (USP), São Paulo 05508-000, Brazil
| | - Sandra Helena Poliselli Farsky
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo (USP), São Paulo 05508-000, Brazil
| | - Estela Bevilacqua
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-000, Brazil
| | - Sonia Maria Oliani
- Post-Graduation in Structural and Functional Biology, Federal University of São Paulo (UNIFESP), São Paulo 04023-062, Brazil
- Department of Biology, School of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto 15054-000, Brazil
- Advanced Research Center in Medicine (CEPAM), União das Faculdades dos Grandes Lagos (Unilago), São José do Rio Preto 15030-070, Brazil
| |
Collapse
|
3
|
Unravelling the genetics of non-random fertilization associated with gametic incompatibility. Sci Rep 2022; 12:22314. [PMID: 36566278 PMCID: PMC9789956 DOI: 10.1038/s41598-022-26910-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
In the dairy industry, mate allocation is dependent on the producer's breeding goals and the parents' breeding values. The probability of pregnancy differs among sire-dam combinations, and the compatibility of a pair may vary due to the combination of gametic haplotypes. Under the hypothesis that incomplete incompatibility would reduce the odds of fertilization, and complete incompatibility would lead to a non-fertilizing or lethal combination, deviation from Mendelian inheritance expectations would be observed for incompatible pairs. By adding an interaction to a transmission ratio distortion (TRD) model, which detects departure from the Mendelian expectations, genomic regions linked to gametic incompatibility can be identified. This study aimed to determine the genetic background of gametic incompatibility in Holstein cattle. A total of 283,817 genotyped Holstein trios were used in a TRD analysis, resulting in 422 significant regions, which contained 2075 positional genes further investigated for network, overrepresentation, and guilt-by-association analyses. The identified biological pathways were associated with immunology and cellular communication and a total of 16 functional candidate genes were identified. Further investigation of gametic incompatibility will provide opportunities to improve mate allocation for the dairy cattle industry.
Collapse
|
4
|
Hebeda CB, Savioli AC, Scharf P, de Paula-Silva M, Gil CD, Farsky SHP, Sandri S. Neutrophil depletion in the pre-implantation phase impairs pregnancy index, placenta and fetus development. Front Immunol 2022; 13:969336. [PMID: 36248911 PMCID: PMC9558710 DOI: 10.3389/fimmu.2022.969336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Maternal neutrophils cells are players in gestational tolerance and fetus delivery. Nonetheless, their actions in each phase of the pregnancy are unknown. We here investigated the role of maternal neutrophil depletion before the blastocyst implantation phase and outcomes in the pregnancy index, placenta, and fetus development. Neutrophils were pharmacologically depleted by i.p. injection of anti-Gr1 (anti-neutrophils; 200 µg) 24 hours after plug visualization in allogeneic-mated C57BL/6/BALB/c mice. Depletion of peripheral neutrophils lasted until 48 hours after anti-Gr1 injection (gestational day 1.5-3.5). On gestational day 5.5, neutrophil depletion impaired the blastocyst implantation, as 50% of pregnant mice presented reduced implantation sites. On gestational day 18.5, neutrophil depletion reduced the pregnancy rate and index, altered the placenta disposition in the uterine horns, and modified the structure of the placenta, detected by reduced junctional zone, associated with decreased numbers of giant trophoblast cells, spongiotrophoblast. Reduced number of placenta cells labeled for vascular endothelial growth factor (VEGF), platelet-endothelial cell adhesion molecule (PECAM-1), and intercellular cell adhesion molecule (ICAM-1), important markers of angiogenesis and adhesiveness, were detected in neutrophil depleted mice. Furthermore, neutrophil depletion promoted a higher frequency of monocytes, natural killers, and T regulatory cells, and lower frequency of cytotoxic T cells in the blood, and abnormal development of offspring. Associated data obtained herein highlight the pivotal role of neutrophils actions in the early stages of pregnancy, and address further investigations on the imbricating signaling evoked by neutrophils in the trophoblastic interaction with uterine epithelium.
Collapse
Affiliation(s)
- Cristina Bichels Hebeda
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, SP, São Paulo, Brazil
- Núcleo de Pesquisa em Ciências Médicas, Fundação Universidade para o Desenvolvimento do Alto Vale do Itajaí – UNIDAVI, Rio do Sul, SC, Brazil
| | - Anna Carolina Savioli
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, SP, São Paulo, Brazil
| | - Pablo Scharf
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, SP, São Paulo, Brazil
| | - Marina de Paula-Silva
- Center for Stem Cells and Regenerative Medicine, King’s College London, London, United Kingdom
| | - Cristiane Damas Gil
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Sandra Helena Poliselli Farsky
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, SP, São Paulo, Brazil
| | - Silvana Sandri
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, SP, São Paulo, Brazil
- *Correspondence: Silvana Sandri,
| |
Collapse
|
5
|
Contribution of plasma, placental, inflammatory and pro-resolving mediators in labor induction. Placenta 2022; 122:9-17. [DOI: 10.1016/j.placenta.2022.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/04/2022] [Accepted: 03/13/2022] [Indexed: 11/23/2022]
|
6
|
Chen SY, Schenkel FS, Melo ALP, Oliveira HR, Pedrosa VB, Araujo AC, Melka MG, Brito LF. Identifying pleiotropic variants and candidate genes for fertility and reproduction traits in Holstein cattle via association studies based on imputed whole-genome sequence genotypes. BMC Genomics 2022; 23:331. [PMID: 35484513 PMCID: PMC9052698 DOI: 10.1186/s12864-022-08555-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 04/12/2022] [Indexed: 02/06/2023] Open
Abstract
Background Genetic progress for fertility and reproduction traits in dairy cattle has been limited due to the low heritability of most indicator traits. Moreover, most of the quantitative trait loci (QTL) and candidate genes associated with these traits remain unknown. In this study, we used 5.6 million imputed DNA sequence variants (single nucleotide polymorphisms, SNPs) for genome-wide association studies (GWAS) of 18 fertility and reproduction traits in Holstein cattle. Aiming to identify pleiotropic variants and increase detection power, multiple-trait analyses were performed using a method to efficiently combine the estimated SNP effects of single-trait GWAS based on a chi-square statistic. Results There were 87, 72, and 84 significant SNPs identified for heifer, cow, and sire traits, respectively, which showed a wide and distinct distribution across the genome, suggesting that they have relatively distinct polygenic nature. The biological functions of immune response and fatty acid metabolism were significantly enriched for the 184 and 124 positional candidate genes identified for heifer and cow traits, respectively. No known biological function was significantly enriched for the 147 positional candidate genes found for sire traits. The most important chromosomes that had three or more significant QTL identified are BTA22 and BTA23 for heifer traits, BTA8 and BTA17 for cow traits, and BTA4, BTA7, BTA17, BTA22, BTA25, and BTA28 for sire traits. Several novel and biologically important positional candidate genes were strongly suggested for heifer (SOD2, WTAP, DLEC1, PFKFB4, TRIM27, HECW1, DNAH17, and ADAM3A), cow (ANXA1, PCSK5, SPESP1, and JMJD1C), and sire (ELMO1, CFAP70, SOX30, DGCR8, SEPTIN14, PAPOLB, JMJD1C, and NELL2) traits. Conclusions These findings contribute to better understand the underlying biological mechanisms of fertility and reproduction traits measured in heifers, cows, and sires, which may contribute to improve genomic evaluation for these traits in dairy cattle. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08555-z.
Collapse
Affiliation(s)
- Shi-Yi Chen
- Department of Animal Sciences, Purdue University, 270 S. Russell Street, West Lafayette, IN, 47907-2041, USA.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Flavio S Schenkel
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Ana L P Melo
- Department of Reproduction and Animal Evaluation, Rural Federal University of Rio de Janeiro, Seropédica, RJ, 23897-000, Brazil
| | - Hinayah R Oliveira
- Department of Animal Sciences, Purdue University, 270 S. Russell Street, West Lafayette, IN, 47907-2041, USA.,Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Victor B Pedrosa
- Department of Animal Sciences, Purdue University, 270 S. Russell Street, West Lafayette, IN, 47907-2041, USA.,Department of Animal Sciences, State University of Ponta Grossa, Ponta Grossa, PR, 84030-900, Brazil
| | - Andre C Araujo
- Department of Animal Sciences, Purdue University, 270 S. Russell Street, West Lafayette, IN, 47907-2041, USA
| | - Melkaye G Melka
- Department of Animal and Food Science, University of Wisconsin River Falls, River Falls, WI, 54022, USA
| | - Luiz F Brito
- Department of Animal Sciences, Purdue University, 270 S. Russell Street, West Lafayette, IN, 47907-2041, USA. .,Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
7
|
Banliat C, Mahé C, Lavigne R, Com E, Pineau C, Labas V, Guyonnet B, Mermillod P, Saint-Dizier M. Dynamic Changes in the Proteome of Early Bovine Embryos Developed In Vivo. Front Cell Dev Biol 2022; 10:863700. [PMID: 35386205 PMCID: PMC8979002 DOI: 10.3389/fcell.2022.863700] [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: 01/27/2022] [Accepted: 03/07/2022] [Indexed: 11/24/2022] Open
Abstract
Early embryo development is a dynamic process involving important molecular and structural changes leading to the embryonic genome activation (EGA) and early cell lineage differentiation. Our aim was to elucidate proteomic changes in bovine embryos developed in vivo. Eleven females were used as embryo donors and pools of embryos at the 4–6 cell, 8–12 cell, morula, compact morula and blastocyst stages were analyzed by nanoliquid chromatography coupled with label free quantitative mass spectrometry. A total of 2,757 proteins were identified, of which 1,950 were quantitatively analyzed. Principal component analysis of data showed a clear separation of embryo pools according to their developmental stage. The hierarchical clustering of differentially abundant proteins evidenced a first cluster of 626 proteins that increased in abundance during development and a second cluster of 400 proteins that decreased in abundance during development, with most significant changes at the time of EGA and blastocyst formation. The main pathways and processes overrepresented among upregulated proteins were RNA metabolism, protein translation and ribosome biogenesis, whereas Golgi vesicle transport and protein processing in endoplasmic reticulum were overrepresented among downregulated proteins. The pairwise comparison between stages allowed us to identify specific protein interaction networks and metabolic pathways at the time of EGA, morula compaction and blastocyst formation. This is the first comprehensive study of proteome dynamics in non-rodent mammalian embryos developed in vivo. These data provide a number of protein candidates that will be useful for further mechanistic and functional studies.
Collapse
Affiliation(s)
- Charles Banliat
- CNRS, INRAE, Université de Tours, IFCE, UMR PRC, Nouzilly, France.,Union Evolution, Noyal-sur-Vilaine, France
| | - Coline Mahé
- CNRS, INRAE, Université de Tours, IFCE, UMR PRC, Nouzilly, France
| | - Régis Lavigne
- Irset-UMRS 1085, Inserm, University of Rennes, Rennes, France.,Protim, Univ Rennes, Biosit-UMS 3480, US-S 018, Rennes, France
| | - Emmanuelle Com
- Irset-UMRS 1085, Inserm, University of Rennes, Rennes, France.,Protim, Univ Rennes, Biosit-UMS 3480, US-S 018, Rennes, France
| | - Charles Pineau
- Irset-UMRS 1085, Inserm, University of Rennes, Rennes, France.,Protim, Univ Rennes, Biosit-UMS 3480, US-S 018, Rennes, France
| | - Valérie Labas
- CNRS, INRAE, Université de Tours, IFCE, UMR PRC, Nouzilly, France.,Pixanim, INRAE, Université de Tours, CHU de Tours, Nouzilly, France
| | | | - Pascal Mermillod
- CNRS, INRAE, Université de Tours, IFCE, UMR PRC, Nouzilly, France
| | | |
Collapse
|
8
|
Zhu X, Shi G, Lu J, Qian X, Wang D. Potential regulatory mechanism of TNF-α/TNFR1/ANXA1 in glioma cells and its role in glioma cell proliferation. Open Life Sci 2022; 17:208-220. [PMID: 35415239 PMCID: PMC8934857 DOI: 10.1515/biol-2022-0023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/09/2021] [Accepted: 01/03/2022] [Indexed: 11/15/2022] Open
Abstract
The purpose of this study was to explore the regulatory mechanism of Annexin A1 (ANXA1) in glioma cells in the inflammatory microenvironment induced by tumour necrosis factor α (TNF-α) and its effects on glioma cell proliferation. CCK-8 analysis demonstrated that TNF-α stimulation promotes rapid growth in glioma cells. Changes in tumour necrosis factor receptor 1 (TNFR1) and ANXA1 expression in glioma cells stimulated with TNF-α were revealed through western blot analysis and immunofluorescence staining. Coimmunoprecipitation analysis revealed that ANXA1 interacts with TNFR1. Moreover, we found that ANXA1 promotes glioma cell growth by activating the p65 and Akt signalling pathways. Finally, immunohistochemistry analysis showed an obvious correlation between ANXA1 expression and Ki-67 in glioma tissues. In summary, our results indicate that the TNF-α/TNFR1/ANXA1 axis regulates the proliferation of glioma cells and that ANXA1 plays a regulatory role in the inflammatory microenvironment.
Collapse
Affiliation(s)
- Xiaotian Zhu
- Department of Pathology, Medical College, Nantong University, No. 19 Qixiu Road , Nantong 226001 , Jiangsu Province , P.R. China
| | - Guanhui Shi
- Department of Pathology, Jiangyin People’s Hospital, No. 163, Shoshan Road , Jiangyin 214400, Jiangsu Province , P.R. China
| | - Jinbiao Lu
- Department of Pathology, Medical College, Nantong University, No. 19 Qixiu Road , Nantong 226001 , Jiangsu Province , P.R. China
| | - Xin Qian
- Department of Pathology, Medical College, Nantong University, No. 19 Qixiu Road , Nantong 226001 , Jiangsu Province , P.R. China
| | - Donglin Wang
- Department of Pathology, Medical College, Nantong University, No. 19 Qixiu Road , Nantong 226001 , Jiangsu Province , P.R. China
| |
Collapse
|
9
|
González-Alvarez ME, McGuire BC, Keating AF. Obesity alters the ovarian proteomic response to zearalenone exposure†. Biol Reprod 2021; 105:278-289. [PMID: 33855340 PMCID: PMC8256104 DOI: 10.1093/biolre/ioab069] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/10/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022] Open
Abstract
Zearalenone (ZEN), a nonsteroidal estrogenic mycotoxin, is detrimental to female reproduction. Altered chemical biotransformation, depleted primordial follicles and a blunted genotoxicant response have been discovered in obese female ovaries, thus, this study investigated the hypothesis that obesity would enhance ovarian sensitivity to ZEN exposure. Seven-week-old female wild-type nonagouti KK.Cg-a/a mice (lean) and agouti lethal yellow KK.Cg-Ay/J mice (obese) received food and water ad libitum, and either saline or ZEN (40 μg/kg) per os for 15 days. Body and organ weights, and estrous cyclicity were recorded, and ovaries collected posteuthanasia for protein analysis. Body and liver weights were increased (P < 0.05) in the obese mice, but obesity did not affect (P > 0.05) heart, kidney, spleen, uterus, or ovary weight and there was no impact (P > 0.05) of ZEN exposure on body or organ weight in lean or obese mice. Obese mice had shorter proestrus (P < 0.05) and a tendency (P = 0.055) for longer metestrus/diestrus. ZEN exposure in obese mice increased estrus but shortened metestrus/diestrus length. Neither obesity nor ZEN exposure impacted (P > 0.05) circulating progesterone, or ovarian abundance of EPHX1, GSTP1, CYP2E1, ATM, BRCA1, DNMT1, HDAC1, H4K16ac, or H3K9me3. Lean mice exposed to ZEN had a minor increase in γH2AX abundance (P < 0.05). In lean and obese mice, LC-MS/MS identified alterations to proteins involved in chemical metabolism, DNA repair and reproduction. These data identify ZEN-induced adverse ovarian modes of action and suggest that obesity is additive to ZEN-induced ovotoxicity.
Collapse
Affiliation(s)
- M Estefanía González-Alvarez
- Department of Animal Science and Interdepartmental Toxicology Graduate Program, Iowa State University, Ames IA, USA
| | - Bailey C McGuire
- Department of Animal Science and Interdepartmental Toxicology Graduate Program, Iowa State University, Ames IA, USA
| | - Aileen F Keating
- Department of Animal Science and Interdepartmental Toxicology Graduate Program, Iowa State University, Ames IA, USA
| |
Collapse
|
10
|
Zhou S, Guo Y, Sun H, Liu L, Yao L, Liu C, He Y, Cao S, Zhou C, Li M, Cao Y, Wang C, Lu Q, Li W, Guo X, Huo R. Maternal RNF114-mediated target substrate degradation regulates zygotic genome activation in mouse embryos. Development 2021; 148:269079. [PMID: 34104941 DOI: 10.1242/dev.199426] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 05/27/2021] [Indexed: 12/16/2022]
Abstract
Zygotic genomic activation (ZGA) is a landmark event in the maternal-to-zygotic transition (MZT), and the regulation of ZGA by maternal factors remains to be elucidated. In this study, the depletion of maternal ring finger protein 114 (RNF114), a ubiquitin E3 ligase, led to developmental arrest of two-cell mouse embryos. Using immunofluorescence and transcriptome analysis, RNF114 was proven to play a crucial role in major ZGA. To study the underlying mechanism, we performed protein profiling in mature oocytes and found a potential substrate for RNF114, chromobox 5 (CBX5), ubiquitylation and degradation of which was regulated by RNF114. The overexpression of CBX5 prevented embryonic development and impeded major ZGA. Furthermore, TAB1 was abnormally accumulated in mutant two-cell embryos, which was consistent with the result of in vitro knockdown of Rnf114. Knockdown of Cbx5 or Tab1 in maternal RNF114-depleted embryos partially rescued developmental arrest and the defect of major ZGA. In summary, our study reveals that maternal RNF114 plays a precise role in degrading some important substrates during the MZT, the misregulation of which may impede the appropriate activation of major ZGA in mouse embryos.
Collapse
Affiliation(s)
- Shuai Zhou
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing 211166, China.,Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Yueshuai Guo
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing 211166, China
| | - Haifeng Sun
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing 211166, China
| | - Lu Liu
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing 211166, China
| | - Liping Yao
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing 211166, China
| | - Chao Liu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuanlin He
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing 211166, China
| | - Shanren Cao
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Cheng Zhou
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing 211166, China
| | - Mingrui Li
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing 211166, China
| | - Yumeng Cao
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing 211166, China
| | - Congjing Wang
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing 211166, China
| | - Qianneng Lu
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing 211166, China
| | - Wei Li
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing 211166, China
| | - Ran Huo
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
11
|
Poh QH, Rai A, Carmichael II, Salamonsen LA, Greening DW. Proteome reprogramming of endometrial epithelial cells by human trophectodermal small extracellular vesicles reveals key insights into embryo implantation. Proteomics 2021; 21:e2000210. [PMID: 33860638 DOI: 10.1002/pmic.202000210] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/29/2021] [Accepted: 04/12/2021] [Indexed: 01/02/2023]
Abstract
Embryo implantation into the receptive endometrium is critical in pregnancy establishment, initially requiring reciprocal signalling between outer layer of the blastocyst (trophectoderm cells) and endometrial epithelium; however, factors regulating this crosstalk remain poorly understood. Although endometrial extracellular vesicles (EVs) are known to signal to the embryo during implantation, the role of embryo-derived EVs remains largely unknown. Here, we provide a comprehensive proteomic characterisation of a major class of EVs, termed small EVs (sEVs), released by human trophectoderm cells (Tsc-sEVs) and their capacity to reprogram protein landscape of endometrial epithelium in vitro. Highly purified Tsc-sEVs (30-200 nm, ALIX+ , TSG101+ , CD9/63/81+ ) were enriched in known players of implantation (LIFR, ICAM1, TAGLN2, WNT5A, FZD7, ROR2, PRICKLE2), antioxidant activity (SOD1, PRDX1/4/6), tissue integrity (EZR, RAC1, RHOA, TNC), and focal adhesions (FAK, ITGA2/V, ITGB1/3). Functionally, Tsc-sEVs were taken up by endometrial cells, altered transepithelial electrical resistance, and upregulated proteins implicated in embryo attachment (ITGA2/V, ITGB1/3), immune regulation (CD59, CD276, LGALS3), and antioxidant activity (GPX1/3/4, PRDX1/2/4/5/6): processes that are critical for successful implantation. Collectively, we provide critical insights into Tsc-sEV-mediated regulation of endometrial function that contributes to our understanding of the molecular basis of implantation.
Collapse
Affiliation(s)
- Qi Hui Poh
- Baker Heart and Diabetes Institute, Molecular Proteomics, Melbourne, Victoria, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Alin Rai
- Baker Heart and Diabetes Institute, Molecular Proteomics, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Irena Iśka Carmichael
- Monash Micro Imaging, Monash, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Lois A Salamonsen
- Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - David W Greening
- Baker Heart and Diabetes Institute, Molecular Proteomics, Melbourne, Victoria, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
12
|
Grewal T, Rentero C, Enrich C, Wahba M, Raabe CA, Rescher U. Annexin Animal Models-From Fundamental Principles to Translational Research. Int J Mol Sci 2021; 22:ijms22073439. [PMID: 33810523 PMCID: PMC8037771 DOI: 10.3390/ijms22073439] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Routine manipulation of the mouse genome has become a landmark in biomedical research. Traits that are only associated with advanced developmental stages can now be investigated within a living organism, and the in vivo analysis of corresponding phenotypes and functions advances the translation into the clinical setting. The annexins, a family of closely related calcium (Ca2+)- and lipid-binding proteins, are found at various intra- and extracellular locations, and interact with a broad range of membrane lipids and proteins. Their impacts on cellular functions has been extensively assessed in vitro, yet annexin-deficient mouse models generally develop normally and do not display obvious phenotypes. Only in recent years, studies examining genetically modified annexin mouse models which were exposed to stress conditions mimicking human disease often revealed striking phenotypes. This review is the first comprehensive overview of annexin-related research using animal models and their exciting future use for relevant issues in biology and experimental medicine.
Collapse
Affiliation(s)
- Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
- Correspondence: (T.G.); (U.R.); Tel.: +61-(0)2-9351-8496 (T.G.); +49-(0)251-83-52121 (U.R.)
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (C.R.); (C.E.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (C.R.); (C.E.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Mohamed Wahba
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
| | - Carsten A. Raabe
- Research Group Regulatory Mechanisms of Inflammation, Center for Molecular Biology of Inflammation (ZMBE) and Cells in Motion Interfaculty Center (CiM), Institute of Medical Biochemistry, University of Muenster, 48149 Muenster, Germany;
| | - Ursula Rescher
- Research Group Regulatory Mechanisms of Inflammation, Center for Molecular Biology of Inflammation (ZMBE) and Cells in Motion Interfaculty Center (CiM), Institute of Medical Biochemistry, University of Muenster, 48149 Muenster, Germany;
- Correspondence: (T.G.); (U.R.); Tel.: +61-(0)2-9351-8496 (T.G.); +49-(0)251-83-52121 (U.R.)
| |
Collapse
|
13
|
Hebeda CB, Sandri S, Benis CM, de Paula-Silva M, Loiola RA, Reutelingsperger C, Perretti M, Farsky SHP. Annexin A1/Formyl Peptide Receptor Pathway Controls Uterine Receptivity to the Blastocyst. Cells 2020; 9:cells9051188. [PMID: 32403233 PMCID: PMC7291299 DOI: 10.3390/cells9051188] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/22/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022] Open
Abstract
Embryo implantation into the uterine wall is a highly modulated, complex process. We previously demonstrated that Annexin A1 (AnxA1), which is a protein secreted by epithelial and inflammatory cells in the uterine microenvironment, controls embryo implantation in vivo. Here, we decipher the effects of recombinant AnxA1 in this phenomenon by using human trophoblast cell (BeWo) spheroids and uterine epithelial cells (Ishikawa; IK). AnxA1-treated IK cells demonstrated greater levels of spheroid adherence and upregulation of the tight junction molecules claudin-1 and zona occludens-1, as well as the glycoprotein mucin-1 (Muc-1). The latter effect of AnxA1 was not mediated through IL-6 secreted from IK cells, a known inducer of Muc-1 expression. Rather, these effects of AnxA1 involved activation of the formyl peptide receptors FPR1 and FPR2, as pharmacological blockade of FPR1 or FPR1/FPR2 abrogated such responses. The downstream actions of AnxA1 were mediated through the ERK1/2 phosphorylation pathway and F-actin polymerization in IK cells, as blockade of ERK1/2 phosphorylation reversed AnxA1-induced Muc-1 and claudin-1 expression. Moreover, FPR2 activation by AnxA1 induced vascular endothelial growth factor (VEGF) secretion by IK cells, and the supernatant of AnxA1-treated IK cells evoked angiogenesis in vitro. In conclusion, these data highlight the role of the AnxA1/FPR1/FPR2 pathway in uterine epithelial control of blastocyst implantation.
Collapse
Affiliation(s)
- Cristina B. Hebeda
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
| | - Silvana Sandri
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
| | - Cláudia M. Benis
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
| | - Marina de Paula-Silva
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
| | - Rodrigo A. Loiola
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
| | - Chris Reutelingsperger
- Faculty of Health, Medicine and Life Sciences, Part of Maastricht University Medical Center, Part of Maastricht University, 6211 LK Maastricht, The Netherlands;
| | - Mauro Perretti
- The William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK;
| | - Sandra H. P. Farsky
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
- Correspondence: ; Tel.: +55-(11)-3091-2197
| |
Collapse
|
14
|
Banliat C, Tsikis G, Labas V, Teixeira-Gomes AP, Com E, Lavigne R, Pineau C, Guyonnet B, Mermillod P, Saint-Dizier M. Identification of 56 Proteins Involved in Embryo-Maternal Interactions in the Bovine Oviduct. Int J Mol Sci 2020; 21:ijms21020466. [PMID: 31940782 PMCID: PMC7013689 DOI: 10.3390/ijms21020466] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/08/2020] [Accepted: 01/10/2020] [Indexed: 01/18/2023] Open
Abstract
The bovine embryo develops in contact with the oviductal fluid (OF) during the first 4–5 days of pregnancy. The aim of this study was to decipher the protein interactions occurring between the developing embryo and surrounding OF. In-vitro produced 4–6 cell and morula embryos were incubated or not (controls) in post-ovulatory OF (OF-treated embryos) and proteins were then analyzed and quantified by high resolution mass spectrometry (MS) in both embryo groups and in OF. A comparative analysis of MS data allowed the identification and quantification of 56 embryo-interacting proteins originated from the OF, including oviductin (OVGP1) and several annexins (ANXA1, ANXA2, ANXA4) as the most abundant ones. Some embryo-interacting proteins were developmental stage-specific, showing a modulating role of the embryo in protein interactions. Three interacting proteins (OVGP1, ANXA1 and PYGL) were immunolocalized in the perivitelline space and in blastomeres, showing that OF proteins were able to cross the zona pellucida and be taken up by the embryo. Interacting proteins were involved in a wide range of functions, among which metabolism and cellular processes were predominant. This study identified for the first time a high number of oviductal embryo-interacting proteins, paving the way for further targeted studies of proteins potentially involved in the establishment of pregnancy in cattle.
Collapse
Affiliation(s)
- Charles Banliat
- INRAE, CNRS, Université de Tours, IFCE, UMR PRC, 37380 Nouzilly, France; (C.B.); (G.T.); (V.L.); (P.M.)
- Union Evolution, 35530 Noyal-sur-Vilaine, France;
| | - Guillaume Tsikis
- INRAE, CNRS, Université de Tours, IFCE, UMR PRC, 37380 Nouzilly, France; (C.B.); (G.T.); (V.L.); (P.M.)
| | - Valérie Labas
- INRAE, CNRS, Université de Tours, IFCE, UMR PRC, 37380 Nouzilly, France; (C.B.); (G.T.); (V.L.); (P.M.)
- INRAE, Université de Tours, CHU de Tours, Plate-forme CIRE, PAIB, 37380 Nouzilly, France;
| | - Ana-Paula Teixeira-Gomes
- INRAE, Université de Tours, CHU de Tours, Plate-forme CIRE, PAIB, 37380 Nouzilly, France;
- INRAE, UMR 1282 ISP, 37380 Nouzilly, France
| | - Emmanuelle Com
- Inserm, University of Rennes, EHESP, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, 35000 Rennes, France; (E.C.); (R.L.); (C.P.)
- Protim, Inserm U1085, Irset, Campus de Beaulieu, University of Rennes 1, Proteomics Core Facility, 35000 Rennes, France
| | - Régis Lavigne
- Inserm, University of Rennes, EHESP, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, 35000 Rennes, France; (E.C.); (R.L.); (C.P.)
- Protim, Inserm U1085, Irset, Campus de Beaulieu, University of Rennes 1, Proteomics Core Facility, 35000 Rennes, France
| | - Charles Pineau
- Inserm, University of Rennes, EHESP, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, 35000 Rennes, France; (E.C.); (R.L.); (C.P.)
- Protim, Inserm U1085, Irset, Campus de Beaulieu, University of Rennes 1, Proteomics Core Facility, 35000 Rennes, France
| | | | - Pascal Mermillod
- INRAE, CNRS, Université de Tours, IFCE, UMR PRC, 37380 Nouzilly, France; (C.B.); (G.T.); (V.L.); (P.M.)
| | - Marie Saint-Dizier
- INRAE, CNRS, Université de Tours, IFCE, UMR PRC, 37380 Nouzilly, France; (C.B.); (G.T.); (V.L.); (P.M.)
- Faculty of Sciences and Techniques, Department Agrosciences, University of Tours, 37000 Tours, France
- Correspondence: ; Tel.: +33-2-47-42-75-08
| |
Collapse
|
15
|
Munuce MJ, Marini PE, Teijeiro JM. Expression profile and distribution of Annexin A1, A2 and A5 in human semen. Andrologia 2019; 51:e13224. [DOI: 10.1111/and.13224] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/17/2018] [Accepted: 11/29/2018] [Indexed: 12/25/2022] Open
Affiliation(s)
- María José Munuce
- Laboratorio de Medicina Reproductiva, Facultad de Ciencias Bioquímicas y Farmacéuticas; CONICET, Universidad Nacional de Rosario; Rosario Argentina
| | - Patricia Estela Marini
- Laboratorio de Medicina Reproductiva, Facultad de Ciencias Bioquímicas y Farmacéuticas; CONICET, Universidad Nacional de Rosario; Rosario Argentina
- Consejo de Investigaciones de la Universidad Nacional de Rosario and Instituto de Biología Molecular y Celular de Rosario, IBR-CONICET; Rosario Argentina
| | - Juan Manuel Teijeiro
- Laboratorio de Medicina Reproductiva, Facultad de Ciencias Bioquímicas y Farmacéuticas; CONICET, Universidad Nacional de Rosario; Rosario Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET; Rosario Argentina
| |
Collapse
|
16
|
Jiang X, Xue S, Kang T, Liu H, Ren H, Hua R, Ni D, Lei M. Annexin A8 (ANXA8) regulates proliferation of porcine endometrial cells via Akt signalling pathway. Reprod Domest Anim 2019; 54:3-10. [PMID: 30040162 DOI: 10.1111/rda.13280] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 06/29/2018] [Indexed: 12/22/2022]
Abstract
Annexin A8 (ANXA8) gene, a member of the annexin family, encodes an anticoagulant protein involved in blood coagulation cascade and acts as an indirect inhibitor of the thromboplastin-specific complex. However, little is known about the function of ANXA8 in porcine endometrial cells so far. Here, ANXA8 mRNA was found to be abundant in porcine endometrium on days 11-13 of pregnancy. Real-time RT-PCR analysis indicated that the mRNA expression of the leukaemia inhibitory factor (LIF) and the epidermal growth factor (EGF) was upregulated by ANXA8 in porcine endometrial cells. Immunofluorescence technology and cell cycle analysis revealed that ANXA8 promoted the proliferation of endometrial cells, as evidenced by the abundant proliferating cell nuclear antigen (PCNA) expression and an increase in the S phase. Western blot analysis results indicated that ANXA8 activated the phosphorylation of the target protein kinase B (Akt) protein. Immunofluorescence technology results showed that the PCNA protein had no significant change in porcine endometrial cells with both ANXA8 overexpression and the addition of Akt inhibitor. Furthermore, the number of implantation sites was significantly reduced by injection of mus-siRNA-ANXA8 into the uterine horn of mice. Collectively, these results suggest that ANXA8 promotes the proliferation of endometrial cells through the Akt signalling pathway.
Collapse
Affiliation(s)
- Xiaona Jiang
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Songyi Xue
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Tingting Kang
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Huijing Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Huihui Ren
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Renwu Hua
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Debin Ni
- National Engineering Research Center for Livestock, Huazhong Agricultural University, Wuhan, China
| | - Minggang Lei
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- National Engineering Research Center for Livestock, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| |
Collapse
|
17
|
Marei WFA, Khalil WA, Pushpakumara APG, El-Harairy MA, Abo El-Atta AMA, Wathes DC, Fouladi-Nashta A. Polyunsaturated fatty acids influence offspring sex ratio in cows. Int J Vet Sci Med 2018; 6:S36-S40. [PMID: 30761319 PMCID: PMC6161865 DOI: 10.1016/j.ijvsm.2018.01.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/14/2018] [Accepted: 01/30/2018] [Indexed: 01/08/2023] Open
Abstract
Dietary polyunsaturated fatty acids (PUFAs) can influence fertility in farm animals. Some evidence in mice and sheep have suggested that PUFAs may influence offspring sex ratio, which may have significant value for cattle production. To test this hypothesis, three groups of Holstein cows were supplemented with either 0%, 3% or 5% protected fat (PF) in the form of calcium salt of fatty acids (rich in omega-6) from 14–21 days pre-partum until conception. Proven-fertile frozen semen from the same ejaculate was used for insemination. Calf sex recorded at birth was 8/19 (42.1%) male offspring in the control group, increasing to 14/20 (70%, P > 0.05) and 17/20 (85%, P < 0.05) in 3% and 5% PF, respectively. To test if this effect was caused by a direct influence on the oocyte, we supplemented bovine cumulus oocyte complexes during in vitro maturation with either omega-3 alpha-linolenic acid (ALA), omega-6 linoleic acid (LA) or trans-10, cis-12 conjugated linoleic acid (CLA). Sex ratio of the produced transferable embryos was determined using PCR of SRY gene. Similar to the in vivo results, sex ratio was skewed to the male side in the embryos derived from LA- and CLA-treated oocytes (79% and 71%) compared to control and ALA-treated oocytes (44% and 54%, respectively). These results indicate that both dietary and in vitro supplementation of omega-6 PUFAs can skew the sex ratio towards the male side in cattle. Further experiments are required to confirm this effect on a larger scale and to study the mechanisms of action that might be involved.
Collapse
Affiliation(s)
- Waleed F A Marei
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Wael A Khalil
- Department of Animal Production, Faculty of Agriculture, Mansoura University, Mansoura 35516, Egypt
| | - Anil P G Pushpakumara
- Department of Farm Animal Production and Health, Faculty of Veterinary Medicine and Animal Science, University of Peradeniya, Sri Lanka
| | - Mostafa A El-Harairy
- Department of Animal Production, Faculty of Agriculture, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed M A Abo El-Atta
- Department of Animal Production, Faculty of Agriculture, Mansoura University, Mansoura 35516, Egypt
| | - D Claire Wathes
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hatfield AL9 7TA, UK
| | - Ali Fouladi-Nashta
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Hatfield AL9 7TA, UK
| |
Collapse
|