1
|
Roncoletta M, de Sales NAA, Rey FSB, Ferraz GC, Morani EDSC. Galectin-1 Used in Assisted Reproduction-Embryo Safety and Toxicology Studies. Molecules 2023; 28:859. [PMID: 36677917 PMCID: PMC9861859 DOI: 10.3390/molecules28020859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Galectin-1 has been cited as a mediator involved in preventing early embryonic death in mammals and is implicated in maternal-fetal tolerance. Galectin-1 is also a reasonable tool to improve fertility in assisted reproduction procedures. As recommended in the ICH guidelines (S5-R2 and S6-R1) and based on bioethical concerns, we chose bovine embryos (BE) to assess in vitro embryo development as part of a larger reproductive safety and toxicology study in progress. The design considered in vitro embryo development using rHGAL-1 supplementations (in three different concentrations) of the in vitro embryo culture (IVP) media. Based on procedures for the commercial in vitro production of BE using oocytes aspirated from slaughterhouse ovaries, rHGAL-1 supplementation was performed in two experiments: In Experiment 1 on oocyte maturation, involving IVM medium supplementation, and in Experiment 2 on culture step IVC, involving supplementation with an SOF medium. IVP commercial procedures were used, with three IVP replicates per experiment, and the oocytes we distributed into four groups of treatment (one control group and three different dosages of rHGAL-1 to supplement both IVM and SOF media using 2, 20, and 40 µg·mL-1, respectively. A total of 967 (Experiment 1) and 1213 (Experiment 2) oocytes were aspirated and submitted to the IVP procedure. There was no damage to the in vitro bovine embryo growth when considering cleavage percentage (%CLE), blastocyst development (Bl, Bx, Bh, and B) at Days 7 and 8, or an amount of rHGAL-1 supplementation ≤20 µg·mL-1. The immunohistochemistry assay with D8 embryos cultivated using rHGAL-1 supplementation on the culture medium (SOF medium) demonstrated the presence of exogenous GAL-1 distributed in mass cell and trophoblastic cells, and the profile observed was dependent on exogenous supplementation, which was most evident in hatched embryos. The findings confirmed the use of a reasonable amount of rHGAL-1 for in vitro embryonic development and would make the use of rHGAL-1 in assisted reproduction in humans more reliable and safer. Even though it was not the objective of the study, we verified that supplementation with 2 µg·mL-1 significantly improved some of the evaluated parameters of embryonic development (%BlD7, %BD7, %BlD8, %BhD8, and %BD8).
Collapse
Affiliation(s)
- Marcelo Roncoletta
- Yoni Group, Inprenha Biotecnologia, Jaboticabal 14897-899, São Paulo, Brazil
| | - Nathali Adrielli Agassi de Sales
- Laboratório de Fisiologia do Exercício e Farmacologia (LAFEQ), Departamento de Morfologia e Fisiologia Animal, Universidade Estadual Paulista (UNESP), Jaboticabal 14884-900, São Paulo, Brazil
| | | | - Guilherme Camargo Ferraz
- Laboratório de Fisiologia do Exercício e Farmacologia (LAFEQ), Departamento de Morfologia e Fisiologia Animal, Universidade Estadual Paulista (UNESP), Jaboticabal 14884-900, São Paulo, Brazil
| | | |
Collapse
|
2
|
Chen JL, Chen Y, Xu DX, Chen DZ. Possible important roles of galectins in the healing of human fetal membranes. Front Endocrinol (Lausanne) 2022; 13:941029. [PMID: 36017312 PMCID: PMC9395672 DOI: 10.3389/fendo.2022.941029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
The fetal membranes healing is a complex and dynamic process of replacing devitalized and missing cellular structures and tissue layers. Multiple cells and extracellular matrices, and cell differentiation, migration and proliferation may participate in restoring the integrity of damaged tissue, however this process still remains unclear. Therefore, there is a need to identify and integrate new ideas and methods to design a more effective dressing to accelerate fetal membrane healing. This review explores the function and role of galectins in the inflammatory, epithelial mesenchymal transition, proliferative migration, and remodeling phases of fetal membrane healing. In conclusion, the preliminary findings are promising. Research on amnion regeneration is expected to provide insight into potential treatment strategies for premature rupture of membranes.
Collapse
Affiliation(s)
- Jia-Le Chen
- The School of Public Health, Anhui Medical University, Hefei, China
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Yu Chen
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - De-Xiang Xu
- The School of Public Health, Anhui Medical University, Hefei, China
| | - Dao-Zhen Chen
- The School of Public Health, Anhui Medical University, Hefei, China
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
- Department of Laboratory, Haidong No.2 People’s Hospital, Haidong, China
| |
Collapse
|
3
|
Seong J, Frias-Aldeguer J, Holzmann V, Kagawa H, Sestini G, Heidari Khoei H, Scholte Op Reimer Y, Kip M, Pradhan SJ, Verwegen L, Vivié J, Li L, Alemany A, Korving J, Darmis F, van Oudenaarden A, Ten Berge D, Geijsen N, Rivron NC. Epiblast inducers capture mouse trophectoderm stem cells in vitro and pattern blastoids for implantation in utero. Cell Stem Cell 2022; 29:1102-1118.e8. [PMID: 35803228 DOI: 10.1016/j.stem.2022.06.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/21/2022] [Accepted: 06/02/2022] [Indexed: 11/03/2022]
Abstract
The embryo instructs the allocation of cell states to spatially regulate functions. In the blastocyst, patterning of trophoblast (TR) cells ensures successful implantation and placental development. Here, we defined an optimal set of molecules secreted by the epiblast (inducers) that captures in vitro stable, highly self-renewing mouse trophectoderm stem cells (TESCs) resembling the blastocyst stage. When exposed to suboptimal inducers, these stem cells fluctuate to form interconvertible subpopulations with reduced self-renewal and facilitated differentiation, resembling peri-implantation cells, known as TR stem cells (TSCs). TESCs have enhanced capacity to form blastoids that implant more efficiently in utero due to inducers maintaining not only local TR proliferation and self-renewal, but also WNT6/7B secretion that stimulates uterine decidualization. Overall, the epiblast maintains sustained growth and decidualization potential of abutting TR cells, while, as known, distancing imposed by the blastocyst cavity differentiates TR cells for uterus adhesion, thus patterning the essential functions of implantation.
Collapse
Affiliation(s)
- Jinwoo Seong
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| | - Javier Frias-Aldeguer
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Utrecht, the Netherlands; Maastricht University, Maastricht, the Netherlands
| | - Viktoria Holzmann
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| | - Harunobu Kagawa
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| | - Giovanni Sestini
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| | - Heidar Heidari Khoei
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Yvonne Scholte Op Reimer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| | - Maarten Kip
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Utrecht, the Netherlands
| | - Saurabh J Pradhan
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| | - Lucas Verwegen
- Department of Cell Biology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Judith Vivié
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Utrecht, the Netherlands
| | - Linfeng Li
- Maastricht University, Maastricht, the Netherlands
| | - Anna Alemany
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Utrecht, the Netherlands
| | - Jeroen Korving
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Utrecht, the Netherlands
| | - Frank Darmis
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Utrecht, the Netherlands
| | | | - Derk Ten Berge
- Department of Cell Biology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Niels Geijsen
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Utrecht, the Netherlands; Department of Anatomy and Embryology, LUMC, Leiden University, Leiden, the Netherlands
| | - Nicolas C Rivron
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria; Hubrecht Institute for Developmental Biology and Stem Cell Research, Utrecht, the Netherlands; Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
4
|
Menkhorst E, Than NG, Jeschke U, Barrientos G, Szereday L, Dveksler G, Blois SM. Medawar's PostEra: Galectins Emerged as Key Players During Fetal-Maternal Glycoimmune Adaptation. Front Immunol 2022; 12:784473. [PMID: 34975875 PMCID: PMC8715898 DOI: 10.3389/fimmu.2021.784473] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
Lectin-glycan interactions, in particular those mediated by the galectin family, regulate many processes required for a successful pregnancy. Over the past decades, increasing evidence gathered from in vitro and in vivo experiments indicate that members of the galectin family specifically bind to both intracellular and membrane bound carbohydrate ligands regulating angiogenesis, immune-cell adaptations required to tolerate the fetal semi-allograft and mammalian embryogenesis. Therefore, galectins play important roles in fetal development and placentation contributing to maternal and fetal health. This review discusses the expression and role of galectins during the course of pregnancy, with an emphasis on maternal immune adaptions and galectin-glycan interactions uncovered in the recent years. In addition, we summarize the galectin fingerprints associated with pathological gestation with particular focus on preeclampsia.
Collapse
Affiliation(s)
- Ellen Menkhorst
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, Australia.,Gynaecological Research Centre, The Women's Hospital, Melbourne, VIC, Australia
| | - Nandor Gabor Than
- Systems Biology of Reproduction Research Group, Institute of Enyzmology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Augsburg, Germany
| | - Gabriela Barrientos
- Laboratorio de Medicina Experimental, Hospital Alemán-Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires, Argentina
| | - Laszlo Szereday
- Medical School, Department of Medical Microbiology and Immunology, University of Pecs, Pecs, Hungary
| | - Gabriela Dveksler
- Department of Pathology, Uniformed Services University, Bethesda, MD, United States
| | - Sandra M Blois
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
5
|
Liu Y, Jones C, Coward K. An investigation of mechanisms underlying mouse blastocyst hatching: a ribonucleic acid sequencing study. F&S SCIENCE 2022; 3:35-48. [PMID: 35559994 DOI: 10.1016/j.xfss.2021.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/17/2021] [Accepted: 12/17/2021] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To investigate the regulatory mechanisms and signaling molecules underlying hatching in mouse embryos. DESIGN Experimental laboratory study using a mouse embryo model. SETTING University-based basic scientific research laboratory. ANIMALS A total of 40 B6C3F1 × B6D2F1 mouse embryos were used in this study. INTERVENTION(S) Frozen/thawed mouse embryos, at the 8-cell stage, were cultured in vitro for 2 days. The resulting hatching and prehatching blastocysts were then used for complementary deoxyribonucleic acid (cDNA) library preparation and ribonucleic acid (RNA) sequencing analysis (n = 8 for each group). Differentially expressed genes were then used for downstream functional analysis. In addition, a list of genes related to developmental progression in humans was used to identify genes that were potentially related to the hatching of human embryos. MAIN OUTCOME MEASURE(S) Differentially expressed genes, enriched Gene Ontology terms and canonical pathways, clustered gene networks, activated upstream regulators, and common genes between a gene list of hatching-related genes in mice and a gene list associated with developmental progression in humans. RESULT(S) A total 275 differentially expressed genes were identified between hatching and prehatching blastocysts: 230 up-regulated and 45 down-regulated genes. Functional enrichment analysis suggested that blastocyst hatching in vitro is an adenosine triphosphate (ATP)-dependent process that involves protein biosynthesis and organization of the cytoskeleton. Furthermore, by regulating cell motility, the RhoA signaling pathway (including Arpc2, Cfl1, Gsn, Pfn1, Tpi1, Grb2, Tmsb10, Enah, and Rnd3 genes) may be a crucial signaling pathway during hatching. We also identified a cluster of genes (Krt8, Krt7, Cldn4, and Aqp3) that exerted functional roles in cell-cell junctions and water homeostasis during hatching. Moreover, some growth factors (angiotensinogen and fibroblast growth factor 2) and endocrine factors (estrogen receptor and prolactin) were predicted to be involved in the regulation of embryo hatching. In addition, we identified 81 potential genes that are potentially involved in the hatching process in human embryos. CONCLUSION(S) Our analysis identified potential genes and molecular regulatory pathways involved in the blastocyst hatching process in mice; we also identified genes that may potentially regulate hatching in human embryos. Our findings enhance our knowledge of embryo development and provide useful information for further exploring the mechanisms underlying embryo hatching.
Collapse
Affiliation(s)
- Yaqiong Liu
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Celine Jones
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Kevin Coward
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Headington, Oxford, United Kingdom.
| |
Collapse
|
6
|
Borowski S, Freitag N, Urban I, Michel G, Barrientos G, Blois SM. Examination of the Contributions of Maternal/Placental-Derived Galectin-1 to Pregnancy Outcome. Methods Mol Biol 2022; 2442:603-619. [PMID: 35320548 DOI: 10.1007/978-1-0716-2055-7_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Galectin-1 (gal-1), a member of a family of evolutionarily conserved glycan-binding proteins, is differentially expressed at the feto-maternal interface and appears to be functionally polyvalent, with a wide range of biological activities. However, the contributions of maternal and/or feto-placental gal-1 to the signaling networks promoting a healthy pregnancy are still being elucidated. This chapter discusses the methods commonly employed to study the maternal or feto-placental contribution of gal-1 during pregnancy in mice. The methods described here can be used to decipher the specific role of each source, e.g., maternal and/or feto-placental derived gal-1 in the orchestration of pregnancy-associated processes.
Collapse
Affiliation(s)
- Sophia Borowski
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, and the Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Nancy Freitag
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, and the Charité-Universitätsmedizin Berlin, Berlin, Germany
- Division of General Internal and Psychosomatic Medicine, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Iris Urban
- Core Facility Transgene Technology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Geert Michel
- Core Facility Transgene Technology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gabriela Barrientos
- Laboratorio de Medicina Experimental, Hospital Alemán-Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Sandra M Blois
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
7
|
Chaney HL, Grose LF, LaBarbara JM, Sirk AW, Blancke AM, Sánchez JM, Passaro C, Lonergan P, Mathew DJ. Galectin-1 Confers Endometrial Gene Expression and Protein Related to Maternal-Conceptus Immune Tolerance in Cattle. Biol Reprod 2021; 106:487-502. [PMID: 34792096 DOI: 10.1093/biolre/ioab215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/25/2021] [Accepted: 11/12/2021] [Indexed: 11/13/2022] Open
Abstract
Conceptus secretory factors include galectins, a family of carbohydrate binding proteins that elicit cell adhesion and immune suppression by interacting with intracellular and extracellular glycans. In rodents, galectin-1 (LGALS1) promotes maternal-fetal immune tolerance in the decidua through expansion of tolerogenic CD11c+ dendritic cells, increased anti-inflammatory IL-10, and activation of FOXP3+ regulatory T cells (Treg). This study characterized galectin expression in early ruminant conceptuses and endometrium. We also tested the effect of recombinant bovine LGALS1 (rbLGALS1) and progesterone (P4) on endometrial expression of genes and protein related to maternal-fetal immune tolerance in cattle. Elongating bovine and ovine conceptuses expressed several galectins, particularly, LGALS1, LGALS3 and LGALS8. Within bovine endometrium, expression of LGALS3, LGALS7 and LGALS9 was greater on Day 16 of pregnancy compared to the estrous cycle. Within ovine endometrium, LGALS7 was greater during pregnancy compared to the estrous cycle and endometrium of pregnant sheep tended to have greater LGALS9 and LGALS15. Expression of endometrial LGALS4 was less during pregnancy in sheep. Treating bovine endometrium with rbLGALS1 increased endometrial expression of CD11c, IL-10 and FOXP3, within 24 h. Specifically, within caruncular endometrium, both rbLGALS1 and P4 increased FOXP3, suggesting that both ligands may promote Treg expansion. Using IHC, FOXP3+ cells with a leukocyte phenotype were localized to the bovine uterine stratum compactum near the uterine surface and increased in response to rbLGALS1. We hypothesize that galectins have important functions during establishment of pregnancy in ruminants and bovine conceptus LGALS1 and luteal P4 confer mechanisms of maternal-conceptus immune tolerance in cattle.
Collapse
Affiliation(s)
- Heather L Chaney
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, USA
| | - Lindsay F Grose
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, USA
| | - Jeanna M LaBarbara
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, USA
| | - Adam W Sirk
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, USA
| | - Alyssa M Blancke
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, USA
| | - Jose M Sánchez
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin, Ireland
| | - Claudia Passaro
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin, Ireland
| | - Patrick Lonergan
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin, Ireland
| | - Daniel J Mathew
- Department of Animal Science, University of Tennessee, Knoxville, TN, USA
| |
Collapse
|
8
|
Sun J, Song B, Ban Y, Ma C, Sun J, Ai D, Nan Z, Wang L, Qu X. Whole transcriptome analysis of trophoblasts under hypoxia. Placenta 2021; 117:13-20. [PMID: 34768163 DOI: 10.1016/j.placenta.2021.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/22/2021] [Accepted: 10/04/2021] [Indexed: 10/20/2022]
Abstract
INTRODUCTION A physiological hypoxia environment exists at maternal-fetal interface during early pregnancy. In addition, there is a pathological hypoxic microenvironment in patients with preeclampsia. Therefore, investigating the hypoxic adaptation and the effects of hypoxia on trophoblasts transcriptome is helpful to better understand the function and regulatory mechanism of trophoblasts at the maternal-fetal interface. METHODS Trophoblast cell line HTR-8/SVneo was cultured under normoxia and hypoxia for 24 h, the full transcriptome was analyzed via RNA-Seq. GO and KEGG enrichment were performed on differentially expressed mRNA, adjacent genes of differentially expressed lncRNA, host genes of differentially expressed circRNA and target genes of differential expressed miRNA. RESULTS The results showed that hypoxia differentially regulated 373 mRNAs, 334 lncRNAs, 71 circRNAs and 33 miRNAs. GO and KEGG enrichment showed that hypoxia negatively regulated TLR3 and PI3K-Akt signaling pathways. Consistently, we found hypoxia significantly inhibited TLR3 agonist-induced cytokines expression and the phosphorylation of Akt and mTOR. DISCUSSION Our study obtained the full transcriptome data and potential regulatory network of trophoblasts under hypoxia, providing supportive data for revealing the function of trophoblasts.
Collapse
Affiliation(s)
- Jintang Sun
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China.
| | - Bingfeng Song
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Yanli Ban
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Chao Ma
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Jia Sun
- Department of Neurology, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Dan Ai
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Zhaodi Nan
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Lijie Wang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Xun Qu
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
9
|
Galectins in Cancer and the Microenvironment: Functional Roles, Therapeutic Developments, and Perspectives. Biomedicines 2021; 9:biomedicines9091159. [PMID: 34572346 PMCID: PMC8465754 DOI: 10.3390/biomedicines9091159] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 12/15/2022] Open
Abstract
Changes in cell growth and metabolism are affected by the surrounding environmental factors to adapt to the cell’s most appropriate growth model. However, abnormal cell metabolism is correlated with the occurrence of many diseases and is accompanied by changes in galectin (Gal) performance. Gals were found to be some of the master regulators of cell–cell interactions that reconstruct the microenvironment, and disordered expression of Gals is associated with multiple human metabolic-related diseases including cancer development. Cancer cells can interact with surrounding cells through Gals to create more suitable conditions that promote cancer cell aggressiveness. In this review, we organize the current understanding of Gals in a systematic way to dissect Gals’ effect on human disease, including how Gals’ dysregulated expression affects the tumor microenvironment’s metabolism and elucidating the mechanisms involved in Gal-mediated diseases. This information may shed light on a more precise understanding of how Gals regulate cell biology and facilitate the development of more effective therapeutic strategies for cancer treatment by targeting the Gal family.
Collapse
|
10
|
Screening Candidate Genes Regulating Placental Development from Trophoblast Transcriptome at Early Pregnancy in Dazu Black Goats ( Capra hircus). Animals (Basel) 2021; 11:ani11072132. [PMID: 34359260 PMCID: PMC8300351 DOI: 10.3390/ani11072132] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary The trophoblast is an original placental tissue whose normal proliferation, differentiation, migration, adhesion, and angiopoiesis are essential for placenta formation and fetal survival during early pregnancy. However, the key genes and molecular mechanisms involved in placenta development in goats are unknown. Herein, the morphology and histological structures of trophoblast tissues from day 20 to 30 of pregnancy were determined. RNA-sequencing was used to screen potential functional genes in common highly expressed and differentially expressed genes. RAP1 signaling pathway was used as the contact center and coordinated with other pathways to regulate placenta development. This study could provide insights into the molecular mechanisms underlying ruminant placentation. Abstract This study explored the trophoblast transcriptome to understand potential functional genes involved in early placental development in goats and their enriched signaling pathways. Trophoblast samples were collected from nine Dazu Black goats on days 20, 25, and 30 of pregnancy (D20, D25, and D30). As the pregnancy progressed, the morphology and histological structures showed significant growth, adhesion, and angiogenesis. A total of 23,253 commonly expressed genes (CEGs) and 4439 differently expressed genes (DEGs) were detected by RNA sequencing. The common highly expressed genes (ChEGs) (the top 100 CEGs) with the highest FPKM percentage (29.9%) of all CEGs were annotated to the ribosome pathway and maintain pregnancy. DEGs were abundant in D30 vs. D20 (3715 DEGs). Besides, the DEGs were associated with the inhibition of oxidative phosphorylation and activation of PI3K-Akt, focal adhesion, ECM–receptor interaction, Rap1, and CAM signaling pathways. The RAP1 may be a central pathway since it coordinates with others to regulate the cell proliferation, invasion, migration, and fusion of trophoblasts. qRT-PCR and Western blot analysis confirmed the transcriptional expression in IGF1, VEGFC, RAPGEF3, PIK3CA, AKT3, ITGB3, ITGA11, SPP1, NOS1, and ATP6V0B genes and protein levels in VEGF, RAPGEF3, and Akt. This is the first study of transcriptome profiling in goat placenta and provides diverse genetic resources for further research on placenta development.
Collapse
|
11
|
Su MT, Tsai PY, Wang CY, Tsai HL, Kuo PL. Aspirin facilitates trophoblast invasion and epithelial-mesenchymal transition by regulating the miR-200-ZEB1 axis in preeclampsia. Biomed Pharmacother 2021; 139:111591. [PMID: 33865015 DOI: 10.1016/j.biopha.2021.111591] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/19/2022] Open
Abstract
Preeclampsia is a severe gestational hypertensive disorder that occurs after 20 weeks' of gestation. It involves several maternal systems, such as cardiovascular, renal, coagulatory systems, and poses a major threat to the maternal and fetal health. Recent clinical evidence showed that aspirin is an effective preventative treatment for reducing the incidence of premature preeclampsia among high-risk pregnant women, however, the mechanism of drug action is not clear. miR-200 family has been shown to be associated with preeclampsia and upregulated in the plasma and placenta of preeclamptic patients. Here we revealed that miR-200 family inhibited trophoblast invasion and epithelial-mesenchymal transition (EMT) process by stimulating epithelial marker expression (E-cadherin and ZO-1) and repressing mesenchymal marker expression (ZEB1 and TGFβ1). Similarly, EMT markers in the placenta of preeclamptic patients showed higher E-cadherin and lower ZEB1 and TGF-β1 protein expression. Moreover, aspirin was shown to suppress miR-200 family and these miR-200 family-mediated cell functions, including cell invasion and EMT changes, were completely reversed. In conclusion, this study demonstrates the effect of miR-200 family on trophoblast invasion and EMT. For the first time, aspirin was shown to fully reverse miR-200-mediated trophoblast biology and act through the network signaling of TGF-β1/ZEB1/miR-200. These results provide a plausible mechanism explaining aspirin's effect on preeclampsia prevention and a therapeutic target for disease intervention.
Collapse
Affiliation(s)
- Mei-Tsz Su
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 704, Taiwan; Department of Obstetrics and Gynecology, Tainan Hospital, Ministry of Health and Welfare, Tainan, Taiwan.
| | - Pei-Yin Tsai
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 704, Taiwan
| | - Chia-Yih Wang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hui-Ling Tsai
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 704, Taiwan
| | - Pao-Lin Kuo
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 704, Taiwan
| |
Collapse
|
12
|
Dinh HQ, Lin X, Abbasi F, Nameki R, Haro M, Olingy CE, Chang H, Hernandez L, Gayther SA, Wright KN, Aspuria PJ, Karlan BY, Corona RI, Li A, Rimel BJ, Siedhoff MT, Medeiros F, Lawrenson K. Single-cell transcriptomics identifies gene expression networks driving differentiation and tumorigenesis in the human fallopian tube. Cell Rep 2021; 35:108978. [PMID: 33852846 PMCID: PMC10108902 DOI: 10.1016/j.celrep.2021.108978] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/30/2020] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
The human fallopian tube harbors the cell of origin for the majority of high-grade serous "ovarian" cancers (HGSCs), but its cellular composition, particularly the epithelial component, is poorly characterized. We perform single-cell transcriptomic profiling of around 53,000 individual cells from 12 primary fallopian specimens to map their major cell types. We identify 10 epithelial subpopulations with diverse transcriptional programs. Based on transcriptional signatures, we reconstruct a trajectory whereby secretory cells differentiate into ciliated cells via a RUNX3high intermediate. Computational deconvolution of advanced HGSCs identifies the "early secretory" population as a likely precursor state for the majority of HGSCs. Its signature comprises both epithelial and mesenchymal features and is enriched in mesenchymal-type HGSCs (p = 6.7 × 10-27), a group known to have particularly poor prognoses. This cellular and molecular compendium of the human fallopian tube in cancer-free women is expected to advance our understanding of the earliest stages of fallopian epithelial neoplasia.
Collapse
Affiliation(s)
- Huy Q Dinh
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Xianzhi Lin
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Forough Abbasi
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Robbin Nameki
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Marcela Haro
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Claire E Olingy
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Heidi Chang
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lourdes Hernandez
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Simon A Gayther
- Center for Bioinformatics and Functional Genomics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kelly N Wright
- Division of Minimally Invasive Gynecologic Surgery, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Paul-Joseph Aspuria
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Beth Y Karlan
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Rosario I Corona
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Andrew Li
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - B J Rimel
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Matthew T Siedhoff
- Division of Minimally Invasive Gynecologic Surgery, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Fabiola Medeiros
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kate Lawrenson
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Center for Bioinformatics and Functional Genomics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
13
|
Wang M, Xu Y, Wang P, Xu Y, Jin P, Wu Z, Qian Y, Bai L, Dong M. Galectin-14 Promotes Trophoblast Migration and Invasion by Upregulating the Expression of MMP-9 and N-Cadherin. Front Cell Dev Biol 2021; 9:645658. [PMID: 33796532 PMCID: PMC8007908 DOI: 10.3389/fcell.2021.645658] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/16/2021] [Indexed: 01/05/2023] Open
Abstract
Galectin-14 is specifically expressed in placental trophoblasts, and its expression is reduced in trophoblasts retrieved from the cervix of women destined to develop early pregnancy loss. However, the roles of galectin-14 in regulating trophoblasts and in the pathogenesis of pregnancy complication have never been investigated. In the current research, we aimed to investigate the roles of galectin-14 in the regulation of trophoblasts. Tissues of the placenta and villi were collected. Primary trophoblasts and human trophoblast cell line HTR-8/SVneo were used. Western blotting and RT-PCR were used to quantify gene expression. The siRNA-mediated galectin-14 knockdown and lentivirus-mediated overexpression were performed to manipulate the gene expression in trophoblasts. Transwell migration and invasion assays were used to evaluate cell migration and invasion capacity. Gelatin zymography was used to determine the gelatinase activity. Galectin-14 was significantly decreased in the villi of early pregnancy loss and the placenta of preeclampsia. Knockdown of galectin-14 in primary trophoblasts inhibited cell migration and invasion, downregulated the expression of matrix metalloproteinase (MMP)-9 and N-cadherin, the activity of MMP-9, and decreased the phosphorylation of Akt. Meanwhile, the overexpression of galectin-14 in HTR-8/SVneo promoted cell migration and invasion, upregulated the expression of MMP-9 and N-cadherin, the activity of MMP-9, and increased the phosphorylation of Akt. Increased Akt phosphorylation promoted cell migration and invasion and upregulated the expression and activity of MMP-9, while decreased Akt phosphorylation inhibited cell migration and invasion and downregulated the expression and activity of MMP-9. Thus, galectin-14 promotes trophoblast migration and invasion by enhancing the expression of MMP-9 and N-cadherin through Akt phosphorylation. The dysregulation of galectin-14 is involved in the pathogenesis of early pregnancy loss and preeclampsia.
Collapse
Affiliation(s)
- Miaomiao Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuqing Xu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Peng Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanfei Xu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Pengzhen Jin
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zaigui Wu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yeqing Qian
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Women's Reproductive Health of Zhejiang Province, Hangzhou, China.,Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, Hangzhou, China
| | - Long Bai
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Women's Reproductive Health of Zhejiang Province, Hangzhou, China
| | - Minyue Dong
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Women's Reproductive Health of Zhejiang Province, Hangzhou, China.,Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, Hangzhou, China
| |
Collapse
|
14
|
Tazhitdinova R, Timoshenko AV. The Emerging Role of Galectins and O-GlcNAc Homeostasis in Processes of Cellular Differentiation. Cells 2020; 9:cells9081792. [PMID: 32731422 PMCID: PMC7465113 DOI: 10.3390/cells9081792] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 02/07/2023] Open
Abstract
Galectins are a family of soluble β-galactoside-binding proteins with diverse glycan-dependent and glycan-independent functions outside and inside the cell. Human cells express twelve out of sixteen recognized mammalian galectin genes and their expression profiles are very different between cell types and tissues. In this review, we summarize the current knowledge on the changes in the expression of individual galectins at mRNA and protein levels in different types of differentiating cells and the effects of recombinant galectins on cellular differentiation. A new model of galectin regulation is proposed considering the change in O-GlcNAc homeostasis between progenitor/stem cells and mature differentiated cells. The recognition of galectins as regulatory factors controlling cell differentiation and self-renewal is essential for developmental and cancer biology to develop innovative strategies for prevention and targeted treatment of proliferative diseases, tissue regeneration, and stem-cell therapy.
Collapse
|
15
|
Blois SM, Dveksler G, Vasta GR, Freitag N, Blanchard V, Barrientos G. Pregnancy Galectinology: Insights Into a Complex Network of Glycan Binding Proteins. Front Immunol 2019; 10:1166. [PMID: 31231368 PMCID: PMC6558399 DOI: 10.3389/fimmu.2019.01166] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 05/08/2019] [Indexed: 12/15/2022] Open
Abstract
Galectins are a phylogenetically conserved family of soluble β-galactoside binding proteins, consisting of 15 different types, each with a specific function. Galectins contribute to placentation by regulating trophoblast development, migration, and invasion during early pregnancy. In addition, galectins are critical players regulating maternal immune tolerance to the embedded embryo. Recently, the role of galectins in angiogenesis during decidualization and in placenta formation has gained attention. Altered expression of galectins is associated with abnormal pregnancies and infertility. This review focuses on the role of galectins in pregnancy-associated processes and discusses the relevance of galectin-glycan interactions as potential therapeutic targets in pregnancy disorders.
Collapse
Affiliation(s)
- Sandra M Blois
- Reproductive Medicine Research Group, Division of General Internal and Psychosomatic Medicine, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Gabriela Dveksler
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Gerardo R Vasta
- Department of Microbiology and Immunology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, UMB, Baltimore, MD, United States
| | - Nancy Freitag
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Véronique Blanchard
- Berlin Institute of Health, Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gabriela Barrientos
- Laboratory of Experimental Medicine, Hospital Alemán, School of Medicine, University of Buenos Aires, CONICET, Buenos Aires, Argentina
| |
Collapse
|