1
|
Yusuf ANM, Amri MF, Ugusman A, Hamid AA, Abd Rahman IZ, Mokhtar MH. Dysregulation of Leukaemia Inhibitory Factor (LIF) Signalling Pathway by Supraphysiological Dose of Testosterone in Female Sprague Dawley Rats During Development of Endometrial Receptivity. Biomedicines 2025; 13:289. [PMID: 40002703 PMCID: PMC11853574 DOI: 10.3390/biomedicines13020289] [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: 11/05/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
OBJECTIVE This study investigated the effects of a supraphysiological dose of testosterone on uterine morphology and the regulation of the leukaemia inhibitory factor (LIF) signalling pathway during endometrial receptivity. METHODS In this study, 30 adult female Sprague-Dawley rats were divided into treatment and control groups. The treatment groups received subcutaneous injections of 1 mg/kg/day of testosterone from gestational day 1 to day 3, either testosterone alone or in combination with inhibitors (anastrozole, finasteride, or both). A control group of six untreated rats was maintained for comparison. Rats were euthanised on the evening of gestational day 4 to examine uterine morphological changes, gene expression and the distribution of proteins associated with the LIF signalling pathway (LIF, LIFR, JAK1 and STAT3) and MUC1 by quantitative polymerase chain reaction (qPCR) and immunohistochemistry (IHC), respectively. RESULTS The results of this study showed that the thickness of the endometrium and myometrium, as well as the number of glands, markedly decreased in all testosterone-treated rats. In addition, the mRNA levels of LIF, LIFR, JAK1 and STAT3 were significantly downregulated in response to supraphysiological testosterone treatment, while the mRNA of MUC1 was significantly upregulated. The IHC results were consistent with the mRNA data and confirmed the changes in protein distribution in all treatment groups. CONCLUSIONS A supraphysiological dose of testosterone may impair endometrial receptivity through dysregulation of the LIF signalling pathway, potentially affecting fertility.
Collapse
Affiliation(s)
- Allia Najmie Muhammad Yusuf
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (A.N.M.Y.); (A.U.); (A.A.H.); (I.Z.A.R.)
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Mohd Fariz Amri
- Department of Pathology and Microbiology, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia;
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (A.N.M.Y.); (A.U.); (A.A.H.); (I.Z.A.R.)
| | - Adila A Hamid
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (A.N.M.Y.); (A.U.); (A.A.H.); (I.Z.A.R.)
| | - Izzat Zulhilmi Abd Rahman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (A.N.M.Y.); (A.U.); (A.A.H.); (I.Z.A.R.)
| | - Mohd Helmy Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (A.N.M.Y.); (A.U.); (A.A.H.); (I.Z.A.R.)
| |
Collapse
|
2
|
Manrique-Caballero CL, Barasch J, Zaidi SK, Bates CM, Ray EC, Kleyman TR, Al-Bataineh MM. Expression and distribution of MUC1 in the developing and adult kidney. Am J Physiol Renal Physiol 2025; 328:F107-F120. [PMID: 39588770 PMCID: PMC11918333 DOI: 10.1152/ajprenal.00206.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 12/28/2024] Open
Abstract
Mucin 1 (or MUC1) is a heterodimeric transmembrane glycoprotein expressed on the apical surface of polarized epithelial cells in several tissues including the kidney. Recent studies have revealed several novel roles of MUC1 in the kidney, potentially including bacterial infection, mineral balance, and genetic interstitial kidney disease, even though MUC1 levels are reduced not only in the kidney but also in all tissues due to MUC1 mutations. A careful localization of MUC1 in discrete segments of the nephron is the first step in understanding the multiple functional roles of MUC1 in the kidney. Most published reports of MUC1 expression to date have been largely confined to cultured cells, tumor tissues, and selective nephron segments of experimental rodents, and very few studies have been performed using human kidney tissues. Given the rising attention to the role of MUC1 in differing components of renal physiology, we carefully examined the kidney distribution of MUC1 in both human and mouse kidney sections using well-defined markers for different nephron segments or cell types. We further extended our investigation to include sections of early stages of mouse kidney development and upon injury in humans. We included staining for MUC1 in urothelial cells, the highly specialized epithelial cells lining the renal pelvis and bladder. These data implicate a role for MUC1 in antimicrobial defense. Our study provides the groundwork to test the physiological relevance of MUC1 in the urinary tract.NEW & NOTEWORTHY MUC1 is a transmembrane glycoprotein expressed on the apical surface of polarized epithelial tissues and most carcinomas. MUC1 may play novel roles in the kidney including defense against infections. Here, we examine the expression of MUC1 in mouse and human kidneys. We show that the distal nephron and the urinary system are the predominant sites of expression of both message and protein, implicating segment-specific roles including distal nephron defense against ascending bacteria.
Collapse
Affiliation(s)
- Carlos L Manrique-Caballero
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Jonathan Barasch
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, United States
| | - Syed K Zaidi
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Carlton M Bates
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Evan C Ray
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Mohammad M Al-Bataineh
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
3
|
Zahir M, Tavakoli B, Zaki-Dizaji M, Hantoushzadeh S, Majidi Zolbin M. Non-coding RNAs in Recurrent implantation failure. Clin Chim Acta 2024; 553:117731. [PMID: 38128815 DOI: 10.1016/j.cca.2023.117731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
Recurrent implantation failure (RIF), defined as the inability to achieve conception following multiple consecutive in-vitro fertilization (IVF) attempts, represents a complex and multifaceted challenge in reproductive medicine. The emerging role of non-coding RNAs in RIF etiopathogenesis has only gained prominence over the last decade, illustrating a new dimension to our understanding of the intricate network underlying RIF. Successful embryo implantation demands a harmonious synchronization between an adequately decidualized endometrium, a competent blastocyst, and effective maternal-embryonic interactions. Emerging evidence has clarified the involvement of a sophisticated network of non-coding RNAs, including microRNAs, circular RNAs, and long non-coding RNAs, in orchestrating these pivotal processes. Disconcerted expression of these molecules can disrupt the delicate equilibrium required for implantation, amplifying the risk of RIF. This comprehensive review presents an in-depth investigation of the complex role played by non-coding RNAs in the pathogenesis of RIF. Furthermore, it underscores the vast potential of non-coding RNAs as diagnostic biomarkers and therapeutic targets, with the ultimate goal of enhancing implantation success rates in IVF cycles. As ongoing research continues to unravel the intercalated web of molecular interactions, exploiting the power of non-coding RNAs may offer promising avenues for mitigating the challenges posed by RIF and improving the outcomes of assisted reproduction.
Collapse
Affiliation(s)
- Mazyar Zahir
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Tavakoli
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Biology, Maragheh University, Maragheh, Iran
| | - Majid Zaki-Dizaji
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Sedigheh Hantoushzadeh
- Vali-E-Asr Reproductive Health Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Masoumeh Majidi Zolbin
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Zhong J, Li J, Burton GJ, Koistinen H, Cheung KW, Ng EHY, Yao Y, Yeung WSB, Lee CL, Chiu PCN. The functional roles of protein glycosylation in human maternal-fetal crosstalk. Hum Reprod Update 2024; 30:81-108. [PMID: 37699855 DOI: 10.1093/humupd/dmad024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/20/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND The establishment of maternal-fetal crosstalk is vital to a successful pregnancy. Glycosylation is a post-translational modification in which glycans (monosaccharide chains) are attached to an organic molecule. Glycans are involved in many physiological and pathological processes. Human endometrial epithelium, endometrial gland secretions, decidual immune cells, and trophoblasts are highly enriched with glycoconjugates and glycan-binding molecules important for a healthy pregnancy. Aberrant glycosylation in the placenta and uterus has been linked to repeated implantation failure and various pregnancy complications, but there is no recent review summarizing the functional roles of glycosylation at the maternal-fetal interface and their associations with pathological processes. OBJECTIVE AND RATIONALE This review aims to summarize recent findings on glycosylation, glycosyltransferases, and glycan-binding receptors at the maternal-fetal interface, and their involvement in regulating the biology and pathological conditions associated with endometrial receptivity, placentation and maternal-fetal immunotolerance. Current knowledge limitations and future insights into the study of glycobiology in reproduction are discussed. SEARCH METHODS A comprehensive PubMed search was conducted using the following keywords: glycosylation, glycosyltransferases, glycan-binding proteins, endometrium, trophoblasts, maternal-fetal immunotolerance, siglec, selectin, galectin, repeated implantation failure, early pregnancy loss, recurrent pregnancy loss, preeclampsia, and fetal growth restriction. Relevant reports published between 1980 and 2023 and studies related to these reports were retrieved and reviewed. Only publications written in English were included. OUTCOMES The application of ultrasensitive mass spectrometry tools and lectin-based glycan profiling has enabled characterization of glycans present at the maternal-fetal interface and in maternal serum. The endometrial luminal epithelium is covered with highly glycosylated mucin that regulates blastocyst adhesion during implantation. In the placenta, fucose and sialic acid residues are abundantly presented on the villous membrane and are essential for proper placentation and establishment of maternal-fetal immunotolerance. Glycan-binding receptors, including selectins, sialic-acid-binding immunoglobulin-like lectins (siglecs) and galectins, also modulate implantation, trophoblast functions and maternal-fetal immunotolerance. Aberrant glycosylation is associated with repeated implantation failure, early pregnancy loss and various pregnancy complications. The current limitation in the field is that most glycobiological research relies on association studies, with few studies revealing the specific functions of glycans. Technological advancements in analytic, synthetic and functional glycobiology have laid the groundwork for further exploration of glycans in reproductive biology under both physiological and pathological conditions. WIDER IMPLICATIONS A deep understanding of the functions of glycan structures would provide insights into the molecular mechanisms underlying their involvement in the physiological and pathological regulation of early pregnancy. Glycans may also potentially serve as novel early predictive markers and therapeutic targets for repeated implantation failure, pregnancy loss, and other pregnancy complications.
Collapse
Affiliation(s)
- Jiangming Zhong
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R., China
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jianlin Li
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R., China
| | - Graham J Burton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Hannu Koistinen
- Department of Clinical Chemistry and Haematology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Ka Wang Cheung
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R., China
| | - Ernest H Y Ng
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R., China
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yuanqing Yao
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - William S B Yeung
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Cheuk-Lun Lee
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R., China
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Philip C N Chiu
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R., China
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
5
|
Lian S, Liu P, Li X, Lv G, Song J, Zhang H, Wu R, Wang D, Wang J. BLV-miR-B1-5p Promotes Staphylococcus aureus Adhesion to Mammary Epithelial Cells by Targeting MUC1. Animals (Basel) 2023; 13:3811. [PMID: 38136848 PMCID: PMC10741194 DOI: 10.3390/ani13243811] [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: 10/12/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Bovine leukemia virus (BLV) is widely prevalent worldwide and can persistently infect mammary epithelial cells in dairy cows, leading to reduced cellular antimicrobial capacity. BLV-encoded microRNAs (BLV-miRNAs) can modify host genes and promote BLV replication. We previously showed that BLV-miR-B1-5p significantly promoted Staphylococcus aureus (S. aureus) adhesion to bovine mammary epithelial (MAC-T) cells; however, the pathway responsible for this effect remained unclear. This study aims to examine how BLV-miR-B1-5p promotes S. aureus adhesion to MAC-T cells via miRNA target gene prediction and validation. Target site prediction showed that BLV-miR-B1-5p could target the mucin family gene mucin 1 (MUC1). Real-time polymerase chain reaction, immunofluorescence, and dual luciferase reporter assay further confirmed that BLV-miR-B1-5p could target and inhibit the expression of MUC1 in bovine MAC-T cells while interfering with the expression of MUC1 promoted S. aureus adhesion to MAC-T cells. These results indicate that BLV-miR-B1-5p promotes S. aureus adhesion to mammary epithelial cells by targeting MUC1.
Collapse
Affiliation(s)
- Shuai Lian
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Pengfei Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Xiao Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Guanxin Lv
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Jiahe Song
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Han Zhang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Rui Wu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
- College of Biology and Agriculture, Jiamusi University, Jiamusi 154007, China
| | - Di Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Jianfa Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| |
Collapse
|
6
|
Yang H, Chen X, Liu Y, Luo J, Huang R, Zhao Y, Li TC, Huang X. Does non-cavity distorting intramural fibroid affect endometrium around the time of embryo implantation? HUM FERTIL 2023; 26:712-719. [PMID: 37815345 DOI: 10.1080/14647273.2023.2264498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 06/17/2023] [Indexed: 10/11/2023]
Abstract
The effect of the intramural fibroids not distorting the cavity remains controversial on implantation and pregnancy. The aim of this study was to examine the impact of non-cavity distorting intramural fibroids on endometrium. Fifty-six women with non-cavity distorting intramural fibroid were recruited in this study. Paired endometrial specimens, one from beneath the fibroid (ipsilateral endometrium) and the other from the opposite side of uterine cavity, away from the fibroid (contralateral endometrium) were obtained 7-9 days after the luteinizing hormone surge in a natural cycle. Histological dating, Mucin1 and Glycodelin expression and uterine natural killer (uNK) cell density were compared between the paired samples. The median (IQR) H-score of Mucin1 staining in the ipsilateral luminal epithelium was 210% (142-230%), which was significantly (p < 0.05) higher than that of the contralateral luminal endometrium (157%, IQR 114-176%). There was no significant difference in Mucin1 expression in the glandular epithelium. There was no significant difference in Glycodelin expression in luminal and glandular epithelium, uNK cells density or histological dating results between the paired endometrial samples. In conclusion, it is uncertain whether the altered expression of Mucin1 in luminal epithelium alone may have impact on implantation when other markers are not changed.
Collapse
Affiliation(s)
- Haiyan Yang
- Hysteroscopic Center, Fu-Xing Hospital, Capital Medical University, Beijing, China
| | - Xiaoyan Chen
- Assisted Reproductive Center, Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yingyu Liu
- Assisted Reproductive Center, Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jing Luo
- Department of Pathology, Fu-Xing Hospital, Capital Medical University, Beijing, China
| | - Rui Huang
- Hysteroscopic Center, Fu-Xing Hospital, Capital Medical University, Beijing, China
| | - Yiwei Zhao
- Assisted Reproductive Center, Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Tin-Chiu Li
- Hysteroscopic Center, Fu-Xing Hospital, Capital Medical University, Beijing, China
- Assisted Reproductive Center, Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Xiaowu Huang
- Hysteroscopic Center, Fu-Xing Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Eivazi S, Tanhaye Kalate Sabz F, Amiri S, Zandieh Z, Bakhtiyari M, Rashidi M, Aflatoonian R, Mehraein F, Amjadi F. MiRNAs secreted by human blastocysts could be potential gene expression regulators during implantation. Mol Biol Rep 2023; 50:1375-1383. [PMID: 36469260 DOI: 10.1007/s11033-022-08121-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Micro RNAs (miRNAs) are small non-coding RNAs known as essential regulators of cell-cell communication. Recent studies have revealed that miRNAs are secreted by a blastocyst in culture media. We hypothesized that endometrial epithelial cells take up embryo-derived miRNAs as well as other soluble factors and regulate their receptivity-related gene expression. METHODS AND RESULTS Blastocyst culture media (BCM) were collected from the individually cultured embryos, while human endometrial epithelial cells (HEECs) were collected from healthy fertile volunteers. To evaluate the effect of BCM on the endometrial receptivity gene expression, HEECs were co-cultured with implanted BCM, non-implanted BCM, and a control culture medium. After determining altered gene expression in the HEECs, the miRNAs-related genes through bioinformatics databases were identified and evaluated in the BCM. Co-culture of primary HEECs with BCM significantly stimulated the expression levels of VEGFA, HBEGF, HOXA10, and LIF in the implanted group compared with non-implanted and control groups. The fold changes of miR-195 significantly diminished in the implanted BCM group compared with the non-implanted BCM group. Reduced fold changes of miR-29b, 145 and increased miR-223 were also observed in the implanted BCM group compared with the non-implanted ones. CONCLUSION miRNAs could function as potential gene expression regulators during implantation. These molecules are secreted by human blastocyst, taken up by endometrial epithelial cells, and cause a change in the endometrial function. We found that BCMs can be effective in implantation process by stimulating related receptivity gene expression.
Collapse
Affiliation(s)
- Sadegh Eivazi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1168743514, Iran
| | - Fatemeh Tanhaye Kalate Sabz
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1168743514, Iran
- Department of Anatomical Sciences and Pathology, School of Medicine, North Khorasan University of Medical Sciences, bojnurd, Iran
| | - Sadegh Amiri
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1168743514, Iran
| | - Zahra Zandieh
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1168743514, Iran
- Shahid Akbar Abadi Clinical Research Development Unit (SHACRDU), School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mehrdad Bakhtiyari
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1168743514, Iran
| | - Mandana Rashidi
- Shahid Akbar Abadi Clinical Research Development Unit (SHACRDU), School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Reza Aflatoonian
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Fereshteh Mehraein
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1168743514, Iran.
| | - Fatemehsadat Amjadi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1168743514, Iran.
- Shahid Akbar Abadi Clinical Research Development Unit (SHACRDU), School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
8
|
Siriwardena D, Boroviak TE. Evolutionary divergence of embryo implantation in primates. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210256. [PMID: 36252209 DOI: 10.1098/rstb.2021.0256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Implantation of the conceptus into the uterus is absolutely essential for successful embryo development. In humans, our understanding of this process has remained rudimentary owing to the inaccessibility of early implantation stages. Non-human primates recapitulate many aspects of human embryo development and provide crucial insights into trophoblast development, uterine receptivity and embryo invasion. Moreover, primate species exhibit a variety of implantation strategies and differ in embryo invasion depths. This review examines conservation and divergence of the key processes required for embryo implantation in different primates and in comparison with the canonical rodent model. We discuss trophectoderm compartmentalization, endometrial remodelling and embryo adhesion and invasion. Finally, we propose that studying the mechanism controlling invasion depth between different primate species may provide new insights and treatment strategies for placentation disorders in humans. This article is part of the theme issue 'Extraembryonic tissues: exploring concepts, definitions and functions across the animal kingdom'.
Collapse
Affiliation(s)
- Dylan Siriwardena
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK.,Centre for Trophoblast Research, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK.,Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Thorsten E Boroviak
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK.,Centre for Trophoblast Research, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK.,Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| |
Collapse
|
9
|
Qu H, Khalil RA. Role of ADAM and ADAMTS Disintegrin and Metalloproteinases in Normal Pregnancy and Preeclampsia. Biochem Pharmacol 2022; 206:115266. [PMID: 36191626 DOI: 10.1016/j.bcp.2022.115266] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022]
Abstract
Normal pregnancy (NP) involves intricate processes starting with egg fertilization, proceeding to embryo implantation, placentation and gestation, and culminating in parturition. These pregnancy-related processes require marked uteroplacental and vascular remodeling by proteolytic enzymes and metalloproteinases. A disintegrin and metalloproteinase (ADAM) and ADAM with thrombospondin motifs (ADAMTS) are members of the zinc-dependent family of proteinases with highly conserved protein structure and sequence homology, which include a pro-domain, and a metalloproteinase, disintegrin and cysteine-rich domain. In NP, ADAMs and ADAMTS regulate sperm-egg fusion, embryo implantation, trophoblast invasion, placental angiogenesis and spiral arteries remodeling through their ectodomain proteolysis of cell surface cytokines, cadherins and growth factors as well as their adhesion with integrins and cell-cell junction proteins. Preeclampsia (PE) is a serious complication of pregnancy characterized by new-onset hypertension (HTN) in pregnancy (HTN-Preg) at or after 20 weeks of gestation, with or without proteinuria. Insufficient trophoblast invasion of the uterine wall, inadequate expansive remodeling of the spiral arteries, reduced uteroplacental perfusion pressure, and placental ischemia/hypoxia are major initiating events in the pathogenesis of PE. Placental ischemia/hypoxia increase the release of reactive oxygen species (ROS), which lead to aberrant expression/activity of certain ADAMs and ADAMTS. In PE, abnormal expression/activity of specific ADAMs and ADAMTS that function as proteolytic sheddases could alter proangiogenic and growth factors, and promote the release of antiangiogenic factors and inflammatory cytokines into the placenta and maternal circulation leading to generalized inflammation, endothelial cell injury and HTN-Preg, renal injury and proteinuria, and further decreases in uteroplacental blood flow, exaggeration of placental ischemia, and consequently fetal growth restriction. Identifying the role of ADAMs and ADAMTS in NP and PE has led to a better understanding of the underlying molecular and vascular pathways, and advanced the potential for novel biomarkers for prediction and early detection, and new approaches for the management of PE.
Collapse
Affiliation(s)
- Hongmei Qu
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA.
| |
Collapse
|
10
|
Gauster M, Moser G, Wernitznig S, Kupper N, Huppertz B. Early human trophoblast development: from morphology to function. Cell Mol Life Sci 2022; 79:345. [PMID: 35661923 PMCID: PMC9167809 DOI: 10.1007/s00018-022-04377-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/13/2022] [Accepted: 05/13/2022] [Indexed: 12/18/2022]
Abstract
Human pregnancy depends on the proper development of the embryo prior to implantation and the implantation of the embryo into the uterine wall. During the pre-implantation phase, formation of the morula is followed by internalization of blastomeres that differentiate into the pluripotent inner cell mass lineage, while the cells on the surface undergo polarization and differentiate into the trophectoderm of the blastocyst. The trophectoderm mediates apposition and adhesion of the blastocyst to the uterine epithelium. These processes lead to a stable contact between embryonic and maternal tissues, resulting in the formation of a new organ, the placenta. During implantation, the trophectoderm cells start to differentiate and form the basis for multiple specialized trophoblast subpopulations, all of which fulfilling specific key functions in placentation. They either differentiate into polar cells serving typical epithelial functions, or into apolar invasive cells that adapt the uterine wall to progressing pregnancy. The composition of these trophoblast subpopulations is crucial for human placenta development and alterations are suggested to result in placenta-associated pregnancy pathologies. This review article focuses on what is known about very early processes in human reproduction and emphasizes on morphological and functional aspects of early trophoblast differentiation and subpopulations.
Collapse
Affiliation(s)
- Martin Gauster
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Gerit Moser
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Stefan Wernitznig
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Nadja Kupper
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Berthold Huppertz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria.
| |
Collapse
|
11
|
Lillehoj EP, Luzina IG, Atamas SP. Mammalian Neuraminidases in Immune-Mediated Diseases: Mucins and Beyond. Front Immunol 2022; 13:883079. [PMID: 35479093 PMCID: PMC9035539 DOI: 10.3389/fimmu.2022.883079] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/21/2022] [Indexed: 12/28/2022] Open
Abstract
Mammalian neuraminidases (NEUs), also known as sialidases, are enzymes that cleave off the terminal neuraminic, or sialic, acid resides from the carbohydrate moieties of glycolipids and glycoproteins. A rapidly growing body of literature indicates that in addition to their metabolic functions, NEUs also regulate the activity of their glycoprotein targets. The simple post-translational modification of NEU protein targets-removal of the highly electronegative sialic acid-affects protein folding, alters protein interactions with their ligands, and exposes or covers proteolytic sites. Through such effects, NEUs regulate the downstream processes in which their glycoprotein targets participate. A major target of desialylation by NEUs are mucins (MUCs), and such post-translational modification contributes to regulation of disease processes. In this review, we focus on the regulatory roles of NEU-modified MUCs as coordinators of disease pathogenesis in fibrotic, inflammatory, infectious, and autoimmune diseases. Special attention is placed on the most abundant and best studied NEU1, and its recently discovered important target, mucin-1 (MUC1). The role of the NEU1 - MUC1 axis in disease pathogenesis is discussed, along with regulatory contributions from other MUCs and other pathophysiologically important NEU targets.
Collapse
Affiliation(s)
- Erik P. Lillehoj
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Irina G. Luzina
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Research Service, Baltimore Veterans Affairs (VA) Medical Center, Baltimore, MD, United States
| | - Sergei P. Atamas
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
12
|
Kubota K, Miwa M, Hayashi KG, Hosoe M, Sakatani M. Steroidal but not embryonic regulation of mucin 1 expression in bovine endometrium. J Reprod Dev 2021; 67:386-391. [PMID: 34645736 PMCID: PMC8668378 DOI: 10.1262/jrd.2021-087] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In cow herd management, inadequate embryo implantation leads to pregnancy loss and causes severe economic losses. Thus, it is crucial to understand the molecular mechanisms underlying
endometrial receptivity and subsequent embryo implantation. Transmembrane glycocalyx mucin 1 (MUC1) has a large and highly glycosylated extracellular domain known to inhibit embryo
implantation via steric hindrance. The role of MUC1 in the bovine endometrium remains to be explored. Herein, we used simple but reliable in vivo and in
vitro experiments to investigate the expression and regulation of MUC1 in the bovine endometrium. MUC1 gene expression was analyzed in endometrial epithelial
cells collected by the cytobrush technique using reverse transcription-quantitative polymerase chain reaction. MUC1 protein expression was evaluated by immunohistochemical analysis of
endometrial samples collected from slaughtered cows. We used an in vitro cell culture model to study the regulation of MUC1 expression by treating cells with sex steroidal
hormones or co-culturing cells with a blastocyst. The results revealed that MUC1 was expressed and localized to the apical surface of luminal epithelial cells in the bovine endometrium. MUC1
expression disappeared during the luteal phase of the estrous cycle and during pregnancy. 17β-estradiol induced MUC1 expression, whereas progesterone inhibited its increase
and co-culturing with blastocysts did not affect the expression. A long postpartum interval is a known risk factor for reduced fertility, and MUC1 expression was higher in this compromised
condition. Our results demonstrated the MUC1 regulation by steroid hormones in bovine endometrium for embryo implantation, and we observed a negative correlation between MUC1 expression and
fertility.
Collapse
Affiliation(s)
- Kaiyu Kubota
- Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO), Tochigi 329-2793, Japan
| | - Masafumi Miwa
- Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO), Tochigi 329-2793, Japan
| | - Ken-Go Hayashi
- Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO), Ibaraki 305-0901, Japan
| | - Misa Hosoe
- Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO), Ibaraki 305-0901, Japan
| | - Miki Sakatani
- Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO), Tochigi 329-2793, Japan
| |
Collapse
|
13
|
Abstract
Morphological transitions are typically attributed to the actions of proteins and lipids. Largely overlooked in membrane shape regulation is the glycocalyx, a pericellular membrane coat that resides on all cells in the human body. Comprised of complex sugar polymers known as glycans as well as glycosylated lipids and proteins, the glycocalyx is ideally positioned to impart forces on the plasma membrane. Large, unstructured polysaccharides and glycoproteins in the glycocalyx can generate crowding pressures strong enough to induce membrane curvature. Stress may also originate from glycan chains that convey curvature preference on asymmetrically distributed lipids, which are exploited by binding factors and infectious agents to induce morphological changes. Through such forces, the glycocalyx can have profound effects on the biogenesis of functional cell surface structures as well as the secretion of extracellular vesicles. In this review, we discuss recent evidence and examples of these mechanisms in normal health and disease.
Collapse
Affiliation(s)
- Joe Chin-Hun Kuo
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, USA; ,
| | - Matthew J Paszek
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, USA; , .,Field of Biomedical Engineering and Field of Biophysics, Cornell University, Ithaca, New York 14853, USA.,Kavli Institute at Cornell for Nanoscale Science, Ithaca, New York 14853, USA
| |
Collapse
|
14
|
Al-Bataineh MM, Kinlough CL, Mi Z, Jackson EK, Mutchler SM, Emlet DR, Kellum JA, Hughey RP. KIM-1-mediated anti-inflammatory activity is preserved by MUC1 induction in the proximal tubule during ischemia-reperfusion injury. Am J Physiol Renal Physiol 2021; 321:F135-F148. [PMID: 34151589 PMCID: PMC8424662 DOI: 10.1152/ajprenal.00127.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 11/22/2022] Open
Abstract
Cell-associated kidney injury molecule-1 (KIM-1) exerts an anti-inflammatory role following kidney injury by mediating efferocytosis and downregulating the NF-κB pathway. KIM-1 cleavage blunts its anti-inflammatory activities. We reported that mucin 1 (MUC1) is protective in a mouse model of ischemia-reperfusion injury (IRI). As both KIM-1 and MUC1 are induced in the proximal tubule (PT) during IRI and are a disintegrin and metalloprotease 17 (ADAM17) substrates, we tested the hypothesis that MUC1 protects KIM-1 activity. Muc1 knockout (KO) mice and wild-type (WT) littermates were subjected to IRI. KIM-1, MUC1, and ADAM17 levels (and signaling pathways) were assessed by immunoblot analysis. PT localization was assessed by confocal microscopy and an in situ proximity ligation assay. Findings were extended using human kidneys and urine as well as KIM-1-mediated efferocytosis assays in mouse PT cultures. In response to tubular injury in mouse and human kidneys, we observed induction and coexpression of KIM-1 and MUC1 in the PT. Compared with WT mice, Muc1 KO mice had higher urinary KIM-1 and lower kidney KIM-1. KIM-1 was apical in the PT of WT kidneys but predominately with luminal debris in Muc1 KO mice. Efferocytosis was reduced in Muc1 KO PT cultures compared with WT cultures, whereas inflammation was increased in Muc1 KO kidneys compared with WT kidneys. MUC1 was cleaved by ADAM17 in PT cultures and blocked KIM-1 shedding in Madin-Darby canine kidney cells. We conclude that KIM-1-mediated efferocytosis and thus anti-inflammatory activity during IRI is preserved in the injured kidney by MUC1 inhibition of KIM-1 shedding.NEW & NOTEWORTHY KIM-1 plays a key role in the recovery of the tubule epithelium during renal IRI by mediating efferocytosis and associated signaling that suppresses inflammation. Excessive cleavage of KIM-1 by ADAM17 provides a decoy receptor that aggravates efferocytosis and subsequent signaling. Our data from experiments in mice, patients, and cultured cells show that MUC1 is also induced during IRI and competes with KIM-1 for cleavage by ADAM17. Consequently, MUC1 protects KIM-1 anti-inflammatory activity in the damaged kidney.
Collapse
Affiliation(s)
- Mohammad M Al-Bataineh
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Carol L Kinlough
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Zaichuan Mi
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Stephanie M Mutchler
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - David R Emlet
- Department of Critical Care Medicine, Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - John A Kellum
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Critical Care Medicine, Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Rebecca P Hughey
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
15
|
Sekulovski N, Whorton AE, Shi M, Hayashi K, MacLean JA. Insulin signaling is an essential regulator of endometrial proliferation and implantation in mice. FASEB J 2021; 35:e21440. [PMID: 33749878 DOI: 10.1096/fj.202002448r] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 01/04/2023]
Abstract
Insulin signaling is critical for the development of preovulatory follicles and progression through the antral stage. Using a conditional knockout model that escapes this blockage, we recently described the role of insulin signaling in granulosa cells during the periovulatory window in mice lacking Insr and Igf1r driven by Pgr-Cre. These mice were infertile, exhibiting defects in ovulation, luteinization, steroidogenesis, and early embryo development. Herein, we demonstrate that while these mice exhibit normal uterine receptivity, uterine cell proliferation and decidualization are compromised resulting in complete absence of embryo implantation in uteri lacking both receptors. While the histological organization of double knockout mice appeared normal, the thickness of their endometrium was significantly reduced. This was supported by the reduced proliferation of both epithelial and stromal cells during the preimplantation stages of pregnancy. Expression and localization of the main drivers of uterine proliferation, ESR1 and PGR, was normal in knockouts, suggesting that insulin signaling acts downstream of these two receptors. While AKT/PI3K signaling was unaffected by insulin receptor ablation, activation of p44/42 MAPK was significantly reduced in both single and double knockout uteri at 3.5 dpc. Overall, we conclude that both INSR and IGF1R are necessary for optimal endometrial proliferation and implantation.
Collapse
Affiliation(s)
- Nikola Sekulovski
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL, USA
| | - Allison E Whorton
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL, USA
| | - Mingxin Shi
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL, USA
| | - Kanako Hayashi
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL, USA.,Center for Reproductive Biology, Washington State University, Pullman, WA, USA
| | - James A MacLean
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL, USA.,Center for Reproductive Biology, Washington State University, Pullman, WA, USA.,School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| |
Collapse
|
16
|
Evaluation of uterine receptivity after gonadotropin releasing hormone agonist administration as an oocyte maturation trigger: a rodent model. Sci Rep 2019; 9:12519. [PMID: 31467307 PMCID: PMC6715633 DOI: 10.1038/s41598-019-48918-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 08/12/2019] [Indexed: 01/16/2023] Open
Abstract
In natural cycle or minimal stimulation cycle IVF, buserelin acetate (buserelin), a gonadotropin-releasing hormone agonist, is often used as a maturation trigger; however, its effect on pregnancy outcomes remains unclear. Therefore, in the present study, we compared uterine receptivity in buserelin-administered mice with that in human chorionic gonadotropin (hCG)-administered mice during the peri-implantation period. Implantation, decidualisation, and term-pregnancy were impaired following hCG, but not buserelin administration. hCG stimulated the synthesis and secretion of progesterone and oestradiol, whereas ovarian steroidogenesis in the buserelin-treated group was comparable with that in the control group. Furthermore, similar to the observation in controls, the buserelin-treated group exhibited activation of progesterone receptor signalling and inhibition of oestrogen receptor signalling in the endometrial epithelium on the day of implantation. However, epithelial progesterone signalling was not detected, and a high expression of genes downstream to oestrogen was observed on day 4 following hCG administration. These results suggest that buserelin administration does not impact uterine receptivity as it did not affect ovarian steroidogenesis and endometrial steroid signalling. Therefore, buserelin is preferred as an oocyte maturation trigger to optimise uterine receptivity during treatments involving timed intercourse, intrauterine insemination, or fresh embryo transfer following in vitro fertilisation.
Collapse
|
17
|
Heydarian M, Yang T, Schweinlin M, Steinke M, Walles H, Rudel T, Kozjak-Pavlovic V. Biomimetic Human Tissue Model for Long-Term Study of Neisseria gonorrhoeae Infection. Front Microbiol 2019; 10:1740. [PMID: 31417529 PMCID: PMC6685398 DOI: 10.3389/fmicb.2019.01740] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 07/15/2019] [Indexed: 11/18/2022] Open
Abstract
Gonorrhea is the second most common sexually transmitted infection in the world and is caused by Gram-negative diplococcus Neisseria gonorrhoeae. Since N. gonorrhoeae is a human-specific pathogen, animal infection models are only of limited use. Therefore, a suitable in vitro cell culture model for studying the complete infection including adhesion, transmigration and transport to deeper tissue layers is required. In the present study, we generated three independent 3D tissue models based on porcine small intestinal submucosa (SIS) scaffold by co-culturing human dermal fibroblasts with human colorectal carcinoma, endometrial epithelial, and male uroepithelial cells. Functional analyses such as transepithelial electrical resistance (TEER) and FITC-dextran assay indicated the high barrier integrity of the created monolayer. The histological, immunohistochemical, and ultra-structural analyses showed that the 3D SIS scaffold-based models closely mimic the main characteristics of the site of gonococcal infection in human host including the epithelial monolayer, the underlying connective tissue, mucus production, tight junction, and microvilli formation. We infected the established 3D tissue models with different N. gonorrhoeae strains and derivatives presenting various phenotypes regarding adhesion and invasion. The results indicated that the disruption of tight junctions and increase in interleukin production in response to the infection is strain and cell type-dependent. In addition, the models supported bacterial survival and proved to be better suitable for studying infection over the course of several days in comparison to commonly used Transwell® models. This was primarily due to increased resilience of the SIS scaffold models to infection in terms of changes in permeability, cell destruction and bacterial transmigration. In summary, the SIS scaffold-based 3D tissue models of human mucosal tissues represent promising tools for investigating N. gonorrhoeae infections under close-to-natural conditions.
Collapse
Affiliation(s)
| | - Tao Yang
- Biocenter, Chair of Microbiology, University of Würzburg, Würzburg, Germany
| | - Matthias Schweinlin
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Maria Steinke
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Heike Walles
- Research Center "Dynamic Systems: Systems Engineering" (CDS), Otto von-Guericke-University, Magdeburg, Germany
| | - Thomas Rudel
- Biocenter, Chair of Microbiology, University of Würzburg, Würzburg, Germany
| | | |
Collapse
|
18
|
Shariati MBH, Niknafs B, Seghinsara AM, Shokrzadeh N, Alivand MR. Administration of dexamethasone disrupts endometrial receptivity by alteration of expression of miRNA 223, 200a, LIF, Muc1, SGK1, and ENaC via the ERK1/2-mTOR pathway. J Cell Physiol 2019; 234:19629-19639. [PMID: 30993706 DOI: 10.1002/jcp.28562] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 02/28/2019] [Accepted: 03/06/2019] [Indexed: 12/30/2022]
Abstract
Successful implantation of embryos requires endometrial receptivity. Glucocorticoids are one of the factors influencing the implantation window. In this study, 40 female BALB/c mice were used to study the impacts of dexamethasone administration on endometrial receptivity markers during implantation window. The mice mated and were randomly divided into four groups: control (vehicle), dexamethasone (100 μg/kg, IP), PP242 (30 mg/kg, IP), and dexamethasone + PP242 (Dex + PP242). On the Day 4th and 5th of gestation, mice received their respective treatments and were killed on the 5th day. To assess the expression of Muc1, leukemia inflammatory inhibitor (LIF), serum/glucocorticoid-inducible kinase 1 (SGK1), epithelial Na+ channel (ENaC), miRNA 200a, and miRNA 223-3p in the endometrium real-time polymerase chain reaction was performed. Furthermore, using Western blot analysis protein expressions of extracellular signal-regulated kinase 1/2 (ERK1/2), mammalian target of rapamycin (mTOR), and eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) were evaluated. Periodic Acid-Schiff staining was used to examine the histomorphological changes of the uterus. According to the results dexamethasone declined the expression of LIF, whereas upregulated expression of Muc1, SGK1, ENaC mRNA, miRNA 200a, and miRNA 223-3p in the endometrium. In addition, PP242, an mTOR inhibitor, induced mRNA expression of Muc1, miRNA200a, and miRNa223-3p whereas it declined the expression of LIF. Moreover, activity of the ERK1/2-mTOR pathway in the endometrial cells was deterred by dexamethasone and PP242. Nonstop epithelium proliferation and elevated surface glycoproteins layer on epithelium of dexamethasone and/or PP242-received groups were divulged through histochemical analysis. According to the above mentioned results, uterine receptivity during implantation period was declined by dexamethasone, at least in part, through modulation of involved genes in endometrial receptivity and inhibition of the ERK1/2-mTOR pathway.
Collapse
Affiliation(s)
| | - Behrooz Niknafs
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Science, Tabriz, Iran
| | - Abbas Majdi Seghinsara
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Naser Shokrzadeh
- Infertility and Reproductive Health Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mohammad Reza Alivand
- Department of Genetic, Facualty of Medcine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
19
|
Hesam Shariati MB, Seghinsara AM, Shokrzadeh N, Niknafs B. The effect of fludrocortisone on the uterine receptivity partially mediated by ERK1/2-mTOR pathway. J Cell Physiol 2019; 234:20098-20110. [PMID: 30968418 DOI: 10.1002/jcp.28609] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 03/10/2019] [Accepted: 03/19/2019] [Indexed: 12/13/2022]
Abstract
Implantation of embryos needs endometrial receptivity. Mineralocorticoids is one of the causes influencing the implantation window. This study targeted to evaluation fludrocortisone different properties on endometrial receptivity. The objective of this study was to assess whether treatment with fludrocortisone could impact the expression of diverse genes and proteins that are involved in uterine receptivity in mice. In this study, 40 female adult BALB/c mice were used. The samples were allocated to four groups of ten. Control group (C) received: vehicle; fludrocortisone group (FCA): received 1.5 mg/kg fludrocortisone; PP242 group (PP242): received 30 mg/kg PP242; fludrocortisone+PP242 group (FCA+PP242): received fludrocortisone and PP242. Mice were killed on window implantation day after mating and confirmed pregnancy. The endometrial epithelium of mouse was collected to assess mRNA expression of leukemia inhibitory factor (LIF), mucin-1 (MUC1), heparin-binding epidermal growth factor (HB-EGF), (Msx.1), miRNA Let-7a, and miRNA 223-3p as well as protein expression of extracellular signal-regulated kinase 1/2 (ERK1/2), mammalian target of rapamycin (mTOR), and eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) in the uterine using real-time PCR and western blot, respectively. In comparison with the control group, fludrocortisone administration upregulated the expression of LIF, HB-EGF, Msx.1, miRNA Let-7a, ERK1/2, and mTOR in the epithelial endometrium. The PP242-treated group demonstrated a significant rise in the expression of MUC1, miRNA 223-3p and a remarkable decline in ERK1/2 and p-4E-BP1 levels in comparison with the control group. Combination therapy of (FCA+PP242) resulted in a remarkable rise in LIF, Msx-1, HB-EGF, ERK1/2, and mTOR levels, in comparison with the PP242 group. Furthermore, combination therapy of (FCA+PP242) downregulated the expression of MUC1 in comparison with the PP242-treated group. According to the results, fludrocortisone affected uterine receptivity possibly by means of modulating the expression of genes involved in the uterine receptivity and activation of the ERK1/2-mTOR pathway.
Collapse
Affiliation(s)
| | - Abbas Majdi Seghinsara
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Naser Shokrzadeh
- Infertility and Reproductive Health research center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Behrooz Niknafs
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Science, Tabriz, Iran
| |
Collapse
|
20
|
El Dairi R, Huuskonen P, Pasanen M, Rysä J. Peroxisome proliferator activated receptor gamma (PPAR-γ) ligand pioglitazone regulated gene networks in term human primary trophoblast cells. Reprod Toxicol 2018; 81:99-107. [DOI: 10.1016/j.reprotox.2018.07.077] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/13/2018] [Accepted: 07/12/2018] [Indexed: 01/02/2023]
|
21
|
Kho HS. Oral epithelial MUC1 and oral health. Oral Dis 2018; 24:19-21. [PMID: 29480594 DOI: 10.1111/odi.12713] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 07/12/2017] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To provide information about MUC1, epithelial membrane-bound mucin, in terms of its role in oral health. DESIGN The expression and functional roles of MUC1 in the oral mucosa and salivary glands were reviewed. Information on the modulation of oral mucosal epithelial MUC1 expression compared with that of endometrial epithelial MUC1 expression was also reviewed. In addition, the possible associations between oral mucosal epithelial MUC1 and oral diseases were explored. RESULTS MUC1 is expressed in the oral mucosa and major and minor salivary glands. Protection of oral mucosal surfaces is believed to be the main function of oral mucosal epithelial MUC1. Its role in the salivary glands is thought to facilitate salivary flow through the ductal system. Information on the role of MUC1 in signal transduction and modulation of immune function in the oral cavity is sparse. However, the possible roles of MUC1 in dry mouth conditions, burning mouth syndrome, dental caries, and oral candidiasis have been suggested. CONCLUSIONS Despite limited information, it is clear that oral epithelial MUC1 plays an important role in oral health. Further research evaluating the physiological and pathological roles of MUC1 in the oral cavity is warranted.
Collapse
Affiliation(s)
- H-S Kho
- Department of Oral Medicine and Oral Diagnosis, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea.,Institute on Aging Seoul National University, Seoul, Korea
| |
Collapse
|
22
|
Benkhalifa M, Zayani Y, Bach V, Copin H, Feki M, Benkhalifa M, Allal-Elasmi M. Does the dysregulation of matrix metalloproteinases contribute to recurrent implantation failure? Expert Rev Proteomics 2018; 15:311-323. [PMID: 29648896 DOI: 10.1080/14789450.2018.1464915] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
INTRODUCTION The progress in in vitro fertilization (IVF) techniques for infertility management has led to the investigation of embryo implantation site proteins such as Matrix metalloproteinases (MMPs), which may have a key role in embryo-endometrium crosstalk and in the molecular mechanisms of the embryo implantation. Areas covered: Numerous studies have generated much information concerning the relation between the different proteins at the site of implantation such as cytokines, growth factors, adhesion molecules and MMPs. However, the exact role of the MMPs in embryo implantation and the impact of their dysregulation in recurrent implantation failure have yet to be characterized. Expert commentary: The proteomic investigation of the MMPs and their molecular pathways may enable scientists and clinicians to correct this dysregulation (via appropriate means of prevention and treatment), better manage embryo transfer during IVF cycles, and thus increase the ongoing pregnancy rate.
Collapse
Affiliation(s)
- Mustapha Benkhalifa
- a Department of Biochemistry , University of Tunis El Manar , Tunis , Tunisia.,b Faculty of sciences of Bizerte , Carthage University , Jarzouna Bizerte , Tunisia
| | - Yosra Zayani
- a Department of Biochemistry , University of Tunis El Manar , Tunis , Tunisia
| | - Véronique Bach
- c PERITOX-INERIS laboratory, CURS , Picardie University Jules Verne , Amiens , France
| | - Henri Copin
- d Reproductive Medicine and developmental Biology , University Hospital and School of Medicine Picardie University Jules Verne , Amiens , France
| | - Moncef Feki
- a Department of Biochemistry , University of Tunis El Manar , Tunis , Tunisia
| | - Moncef Benkhalifa
- c PERITOX-INERIS laboratory, CURS , Picardie University Jules Verne , Amiens , France.,d Reproductive Medicine and developmental Biology , University Hospital and School of Medicine Picardie University Jules Verne , Amiens , France
| | - Monia Allal-Elasmi
- a Department of Biochemistry , University of Tunis El Manar , Tunis , Tunisia
| |
Collapse
|
23
|
Dietary Supplementation of Leucine in Premating Diet Improves the Within-Litter Birth Weight Uniformity, Antioxidative Capability, and Immune Function of Primiparous SD Rats. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1523147. [PMID: 29850484 PMCID: PMC5932505 DOI: 10.1155/2018/1523147] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 03/13/2018] [Indexed: 11/18/2022]
Abstract
The high within-litter birth weight variation has become a big issue in multiparous animals. The present study was conducted to investigate the effects of leucine supplementation in premating diet on the reproductive performance, maternal antioxidative capability, and immune function in primiparous rats. Six-week-old female SD rats were assigned to basal diet or 0.6% leucine supplemented diet for two weeks. After mating during the eighth week of age, the rats were fed with regular gestation diet. Maternal blood samples were collected on the day before mating (day −1) and day 7 and day 20 of pregnancy, while ovaries and uteruses were obtained on day −1 and on day 7, respectively. The results indicate that, compared with control group, within-litter birth weight variation was significantly decreased, while birth weights were significantly increased in the leucine group (P < 0.01). Also, leucine improved the embryo distribution uniformity and the number of implantation sites in uterine. The ovarian gene expressions of LHR, CYP19A1, and VEGFA were upregulated, while Mucin-1 was decreased significantly (P < 0.05). Leucine also increased the maternal antioxidant capacity and immune function. Conclusively, leucine supplementation in premating diet could improve the reproductive performance, which could be attributed to the improved oxidative and immune status.
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Recent studies in the kidney have revealed that the well characterized tumor antigen mucin 1 (MUC1/Muc1) also has numerous functions in the normal and injured kidney. RECENT FINDINGS Mucin 1 is a transmembrane mucin with a robust glycan-dependent apical targeting signal and efficient recycling from endosomes. It was recently reported that the TRPV5 calcium channel is stabilized on the cell surface by galectin-dependent cross-linking to mucin 1, providing a novel mechanism for regulation of ion channels and normal electrolyte balance.Our recent studies in mice show that Muc 1 is induced after ischemia, stabilizing hypoxia-inducible factor 1 (HIF-1)α and β-catenin levels, and transactivating the HIF-1 and β-catenin protective pathways. However, prolonged induction of either pathway in the injured kidney can proceed from apparent full recovery to chronic kidney disease. A very recent report indicates that aberrant activation of mucin 1 signaling after ischemic injury in mice and humans is associated with development of chronic kidney disease and fibrosis. A frameshift mutation in MUC1 was recently identified as the genetic lesion causing medullary cystic kidney disease type 1, now appropriately renamed MUC1 Kidney Disease. SUMMARY Studies of mucin 1 in the kidney now reveal significant functions for the extracellular mucin-like domain and signaling through the cytoplasmic tail.
Collapse
|
25
|
Placental development during early pregnancy in sheep: Progesterone and estrogen receptor protein expression. Theriogenology 2018; 114:273-284. [PMID: 29665573 DOI: 10.1016/j.theriogenology.2018.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/02/2018] [Accepted: 04/04/2018] [Indexed: 02/08/2023]
Abstract
The aim of this study was to evaluate the pattern of protein expression of the steroid receptor isoforms of nuclear progesterone receptors (PGR) A and B, and estrogen receptors (ESR1 and 2) in utero-placental compartments during early pregnancy. Utero-placental tissues were collected from days 14-30 (n = 4 ewes/day), and uterine tissues were collected from non-pregnant ewes on day 10 after estrus (n = 4). Cross sections of formalin-fixed and paraffin embedded tissues were immunofluorescently stained to detect PGRAB, PGRB, ESR1 and ESR2, followed by image generation of entire cross-sections of uterine and utero-placental tissues, confocal imaging of individual uterine and utero-placental compartments, and image and statistical analyses. PGRAB, PGRB, ESR1 and ESR2 were detected in several compartments of uterine and utero-placental tissues. Quantitative image analysis of staining intensity demonstrated that compared to non-pregnant controls 1) expression of PGRAB and PGRB was less in luminal epithelium and endometrial glands from day 14-16 till 30; 2) PGRAB expression tended to be greater in endometrial and myometrial blood vessels on days 28 and/or 30; 3) PGRB expression in myometrum was lower on days 16 and 28; 4) ESR1 in endometrial stroma was lower in all days of pregnancy; 5) ESR2 expression was similar in all compartments and not affected by pregnancy stage; and 6) in FM, expression of steroid receptors was similar. Thus, we have demonstrated spatial and temporal expression of nuclear PGR and ESR isoforms in utero-placental compartments during early pregnancy.
Collapse
|
26
|
Straßburger D, Glaffig M, Stergiou N, Bialas S, Besenius P, Schmitt E, Kunz H. Synthetic MUC1 Antitumor Vaccine with Incorporated 2,3-Sialyl-T Carbohydrate Antigen Inducing Strong Immune Responses with Isotype Specificity. Chembiochem 2018; 19:1142-1146. [PMID: 29633523 DOI: 10.1002/cbic.201800148] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Indexed: 11/10/2022]
Abstract
The endothelial glycoprotein MUC1 is known to underlie alterations in cancer by means of aberrant glycosylation accompanied by changes in morphology. The heavily shortened glycans induce a collapse of the peptide backbone and enable accessibility of the latter to immune cells, rendering it a tumor-associated antigen. Synthetic vaccines based on MUC1 tandem repeat motifs, comprising tumor-associated 2,3-sialyl-T antigen, conjugated to the immunostimulating tetanus toxoid, are reported herein. Immunization with these vaccines in a simple water/oil emulsion produced a strong immune response in mice to which stimulation with complete Freund's adjuvant (CFA) was not superior. In both cases, high levels of IgG1 and IgG2a/b were induced in C57BL/6 mice. Additional glycosylation in the immunodominant PDTRP domain led to improved binding of the induced antisera to MCF-7 breast tumor cells, compared with that of the monoglycosylated peptide vaccine.
Collapse
Affiliation(s)
- David Straßburger
- Institut für Organische Chemie, Johannes Gutenberg-Universität Mainz, Duesbergweg 10-14, 55128, Mainz, Germany
| | - Markus Glaffig
- Institut für Organische Chemie, Johannes Gutenberg-Universität Mainz, Duesbergweg 10-14, 55128, Mainz, Germany
| | - Natascha Stergiou
- Institut für Immunologie, Johannes Gutenberg-Universität Mainz, Obere Zahlbacher Strasse 67, 55101, Mainz, Germany
| | - Sabrina Bialas
- Institut für Organische Chemie, Johannes Gutenberg-Universität Mainz, Duesbergweg 10-14, 55128, Mainz, Germany
| | - Pol Besenius
- Institut für Organische Chemie, Johannes Gutenberg-Universität Mainz, Duesbergweg 10-14, 55128, Mainz, Germany
| | - Edgar Schmitt
- Institut für Immunologie, Johannes Gutenberg-Universität Mainz, Obere Zahlbacher Strasse 67, 55101, Mainz, Germany
| | - Horst Kunz
- Institut für Organische Chemie, Johannes Gutenberg-Universität Mainz, Duesbergweg 10-14, 55128, Mainz, Germany
| |
Collapse
|
27
|
Wang X, Zhu B, Xiong S, Sheng X, Qi X, Huang Q, Chen C, Guo Y, Ni H. Expression and function of MUC1 in uterine tissues during early pregnancy in sheep after natural oestrous or artificially-induced oestrous. Theriogenology 2018; 108:339-347. [PMID: 29288978 DOI: 10.1016/j.theriogenology.2017.12.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 12/08/2017] [Accepted: 12/13/2017] [Indexed: 10/18/2022]
Abstract
Mucin 1 (MUC1), a cell surface glycoprotein, is expressed mainly in the endometrial luminal epithelium (LE) and glandular epithelium (GE) of the endometrium in many mammalian species including mice, rats, pigs, sheep, horses and humans during various stages of a menstrual or oestrous cycle, where it plays an important role in embryo implantation and placentation. However, the expression and function of MUC1 in uterine tissues during early pregnancy in sheep after artificially-induced oestrous is not known. Therefore, we investigated the expression and function of MUC1 in the early pregnant and non-pregnant uterine tissues of sheep with natural oestrous or artificially-induced oestrous on days 10, 12, 14, 16 and 18 of the cycle by in situ hybridization, quantitative real-time polymerase chain reaction, immunohistochemical staining and western blotting methods. According to our results, MUC1 mRNA and protein expression increased initially but then decreased from days 10-18, peaking on day 14 in the uterine tissues of non-pregnant ewes after both natural and artificially-induced oestrous. MUC1 protein localisation was observed in the LE on days 10, 12 and 14 and in the GE on days 16 and 18. In contrast, MUC1 mRNA and protein expression increased on days 10 and 12, decreased on day 14, but increased again on days 16 and 18 in the uterine tissues of pregnant ewes both in natural oestrous and in artificially-induced oestrous. Additionally, the MUC1 mRNA and protein expression levels in the uterine tissues of the early pregnant sheep were significantly lower than those in the non-pregnant sheep on days 10, 14, and 18, except on day 16(P < 0.01). Enhancing MUC1 protein expression with oestrogen or/and progesterone decreased the blastocyst adhesion rate when blastocysts were co-cultured with endometrial epithelial cells (EECs), while inhibiting MUC1 protein expression with IFN-τ increased the blastocyst adhesion rate when the blastocysts were co-cultured with EECs. Compared with the ewes undergoing natural oestrus, the expression trend and regulation of MUC1 did not change, and the MUC1 expression levels only increased under artificial oestrus conditions. Our data provide important information for improving the conception rate in sheep undergoing artificially-induced oestrus and offer some reference points relating to embryo transfer, oestrus synchronisation and superovulation.
Collapse
Affiliation(s)
- Xiangguo Wang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China.
| | - Boyang Zhu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China.
| | - Shanhui Xiong
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China.
| | - Xihui Sheng
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China.
| | - Xiaolong Qi
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China.
| | - Qizhen Huang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China.
| | - Chaolei Chen
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China.
| | - Yong Guo
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China.
| | - Hemin Ni
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China.
| |
Collapse
|
28
|
Černá M, Kuntová B, Talacko P, Stopková R, Stopka P. Differential regulation of vaginal lipocalins (OBP, MUP) during the estrous cycle of the house mouse. Sci Rep 2017; 7:11674. [PMID: 28916783 PMCID: PMC5601457 DOI: 10.1038/s41598-017-12021-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 09/01/2017] [Indexed: 01/08/2023] Open
Abstract
Female house mice produce pheromone-carrying major urinary proteins (MUPs) in a cycling manner, thus reaching the maximum urinary production just before ovulation. This is thought to occur to advertise the time of ovulation via deposited urine marks. This study aimed to characterize the protein content from the house mouse vaginal flushes to detect putative vaginal-advertising molecules for a direct identification of reproductive states. Here we show that the mouse vaginal discharge contains lipocalins including those from the odorant binding (OBP) and major urinary (MUP) protein families. OBPs were highly expressed but only slightly varied throughout the cycle, whilst several MUPs were differentially abundant. MUP20 or 'darcin', was thought to be expressed only by males. However, in females it was significantly up-regulated during estrus similarly as the recently duplicated central/group-B MUPs (sMUP17 and highly expressed sMUP9), which in the mouse urine are male biased. MUPs rise between proestrus and estrus, remain steady throughout metestrus, and are co-expressed with antimicrobial proteins. Thus, we suggest that MUPs and potentially also OBPs are important components of female vaginal advertising of the house mouse.
Collapse
Affiliation(s)
- Martina Černá
- BIOCEV group, Department of Zoology, Faculty of Science, Charles University, Viničná 7, Prague, CZ 12844, Czech Republic
| | - Barbora Kuntová
- BIOCEV group, Department of Zoology, Faculty of Science, Charles University, Viničná 7, Prague, CZ 12844, Czech Republic
| | - Pavel Talacko
- BIOCEV group, Department of Zoology, Faculty of Science, Charles University, Viničná 7, Prague, CZ 12844, Czech Republic
| | - Romana Stopková
- BIOCEV group, Department of Zoology, Faculty of Science, Charles University, Viničná 7, Prague, CZ 12844, Czech Republic
| | - Pavel Stopka
- BIOCEV group, Department of Zoology, Faculty of Science, Charles University, Viničná 7, Prague, CZ 12844, Czech Republic.
| |
Collapse
|
29
|
Fullerton PT, Monsivais D, Kommagani R, Matzuk MM. Follistatin is critical for mouse uterine receptivity and decidualization. Proc Natl Acad Sci U S A 2017; 114:E4772-E4781. [PMID: 28559342 PMCID: PMC5474784 DOI: 10.1073/pnas.1620903114] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Embryo implantation remains a significant challenge for assisted reproductive technology, with implantation failure occurring in ∼50% of in vitro fertilization attempts. Understanding the molecular mechanisms underlying uterine receptivity will enable the development of new interventions and biomarkers. TGFβ family signaling in the uterus is critical for establishing and maintaining pregnancy. Follistatin (FST) regulates TGFβ family signaling by selectively binding TGFβ family ligands and sequestering them. In humans, FST is up-regulated in the decidua during early pregnancy, and women with recurrent miscarriage have lower endometrial expression of FST during the luteal phase. Because global knockout of Fst is perinatal lethal in mice, we generated a conditional knockout (cKO) of Fst in the uterus using progesterone receptor-cre to study the roles of uterine Fst during pregnancy. Uterine Fst-cKO mice demonstrate severe fertility defects and deliver only 2% of the number of pups delivered by control females. In Fst-cKO mice, the uterine luminal epithelium does not respond properly to estrogen and progesterone signals and remains unreceptive to embryo attachment by continuing to proliferate and failing to differentiate. The uterine stroma of Fst-cKO mice also responds poorly to artificial decidualization, with lower levels of proliferation and differentiation. In the absence of uterine FST, activin B expression and signaling are up-regulated, and bone morphogenetic protein (BMP) signals are impaired. Our findings support a model in which repression of activin signaling by FST enables uterine receptivity by preserving critical BMP signaling.
Collapse
Affiliation(s)
- Paul T Fullerton
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030
| | - Diana Monsivais
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030
| | - Ramakrishna Kommagani
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Martin M Matzuk
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030;
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Department of Pharmacology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
30
|
Batistel F, Osorio JS, Tariq MR, Li C, Caputo J, Socha MT, Loor JJ. Peripheral leukocyte and endometrium molecular biomarkers of inflammation and oxidative stress are altered in peripartal dairy cows supplemented with Zn, Mn, and Cu from amino acid complexes and Co from Co glucoheptonate. J Anim Sci Biotechnol 2017; 8:33. [PMID: 28469842 PMCID: PMC5410708 DOI: 10.1186/s40104-017-0163-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 03/23/2017] [Indexed: 12/31/2022] Open
Abstract
Background Immune dysfunction and a higher risk of uterine infections are characteristics of the transition into lactation in dairy cows. The supply of complexed trace minerals, which are more bioavailable, could help overcome the greater needs of these nutrients in tissues around parturition and early lactation. Results Twenty Holstein cows received an oral bolus with a mix of inorganic trace minerals (INO) or complexed trace minerals (AAC) to achieve 75, 65, 11, and 1 ppm supplemental Zn, Mn, Cu, and Co, respectively, in the total diet dry matter from -30 d through +30 d relative to parturition. Blood for polymorphonuclear leukocyte (PMNL) isolation was collected at -30, -15, +10, and + 30 d relative to parturition, whereas endometrium biopsies were performed at +14 and +30 d. Feeding AAC led to greater PMNL expression of genes related with inflammation response (DDX58), oxidative stress response (MPO), eicosanoid metabolism (PLA2G4A and ALOX5AP), transcription regulation (PPARG), and cellular adhesion (TLN1). The upregulation by AAC in endometrium of genes related with inflammation response (TLR2, TLR4, NFKB1, TNF, IL6, IL1B, IL10, IL8), prostaglandin synthesis (PTGS2, PTGES), and antioxidant responses (NFE2L2, SOD1) indicated a faster remodeling of uterine tissue and potentially greater capacity to control a local bacterial invasion. Conclusions Data indicate that trace mineral supplementation from amino acid complexes improves PMNL activity and allows the prompt recovery of uterine tissue during early lactation. As such, the benefits of complexed trace minerals extend beyond an improvement of liver function and productive performance. Electronic supplementary material The online version of this article (doi:10.1186/s40104-017-0163-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fernanda Batistel
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, 1207 West Gregory Drive, Urbana, IL 61801 USA
| | - Johan S Osorio
- Department of Dairy Science, South Dakota State University, Brookings, SD USA
| | - Muhammad Rizwan Tariq
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, 1207 West Gregory Drive, Urbana, IL 61801 USA.,Department of Food Science and Technology, University College of Agriculture & Environmental Sciences, The Islamia University of Bahawalpur, Bahawalpur, Punjab Pakistan
| | - Cong Li
- College of Animal Science and Technology, Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, 100193 China
| | - Jessica Caputo
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, 1207 West Gregory Drive, Urbana, IL 61801 USA
| | | | - Juan J Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, 1207 West Gregory Drive, Urbana, IL 61801 USA
| |
Collapse
|
31
|
Li X, Wang L, Nunes DP, Troxler RF, Offner GD. Pro-inflammatory Cytokines Up-regulate MUC1 Gene Expression in Oral Epithelial Cells. J Dent Res 2016; 82:883-7. [PMID: 14578499 DOI: 10.1177/154405910308201107] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The membrane-bound mucin MUC1 is expressed ubiquitously on epithelial surfaces and is thought to provide protection from bacterial and chemical injury. The present study was undertaken to determine whether MUC1 was expressed in cultured oral epithelial cells and whether expression is modulated by pro-inflammatory mediators released as part of the host response to infection by oral pathogens. Northern and Western blotting experiments showed that KB cells express MUC1 mRNA and protein. When cells were treated with interleukins (IL-1β, IL-6), tumor necrosis factor-alpha (TNF-α), or interferon-gamma (IFN-γ), or combinations of these, real-time PCR demonstrated that MUC1 mRNA increased 1.4- to 3.2-fold. Interestingly, a significant increase in levels of MUC1 protein was also observed. While no effect was observed when KB cells were incubated with LPS from Porphyromonas gingivalis, infection of KB monolayers with this oral pathogen caused a 2.85-fold increase in MUC1 transcript levels. These results suggest that increased MUC1 synthesis may be a key element in the host response to infection with oral pathogens.
Collapse
Affiliation(s)
- X Li
- Department of Periodontology and Oral Biology, Boston University Medical Center, MA 02118, USA
| | | | | | | | | |
Collapse
|
32
|
Uterine glands impact uterine receptivity, luminal fluid homeostasis and blastocyst implantation. Sci Rep 2016; 6:38078. [PMID: 27905495 PMCID: PMC5131473 DOI: 10.1038/srep38078] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 11/03/2016] [Indexed: 01/10/2023] Open
Abstract
Uterine glands are essential for pregnancy in mice and likely humans, because they secrete or transport bioactive substances that regulate uterine receptivity for blastocyst implantation. In mice, the uterus becomes receptive to blastocyst implantation on day 4, but is refractory by day 5. Here, blastocysts could be recovered from progesterone-induced uterine gland (PUGKO) but not wildtype (WT) mice on day 5 post-mating. Anti-adhesive Muc1 protein and microvilli were present on the luminal epithelium of PUGKO but not WT uteri. A number of known uterine receptivity genes and gland-specific genes were altered in the PUGKO uterus. Next, the uterus and uterine luminal fluid (ULF) were obtained from WT and PUGKO mice on day 3, 4 and 5. Transcriptome analysis revealed that 580 genes were decreased in the PUGKO uterus, however ULF secrotome analysis revealed that many proteins and several amino acids were increased in the PUGKO ULF. Of note, many proteins encoded by many gland-specific genes were not identified in the ULF of WT mice. These results support the ideas that uterine glands secrete factors that regulate ULF homeostasis and interact with other cell types in the uterus to influence uterine receptivity and blastocyst implantation for the establishment of pregnancy.
Collapse
|
33
|
Superovulation at a specific stage of the estrous cycle determines the reproductive performance in mice. Reprod Biol 2016; 16:279-286. [PMID: 27843090 DOI: 10.1016/j.repbio.2016.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 10/07/2016] [Accepted: 10/16/2016] [Indexed: 11/22/2022]
Abstract
Inconsistent reproductive performance has been reported in superovulated mice. Hence, the aim of this study was to analyze the effect and possible mechanism of superovulation timing on mouse reproductive performance. The results showed that mice superovulated at the metestrous (23.08±6.08%) and diestrous stages (33.33±11.45%) presented significantly lower pregnancy rates compared with those superovulated at the estrous stage (66.67±9.20%). After superovulation at the proestrous and estrous stages, mucin 1 (MUC1) and let-7a/let-7b microRNA (miRNA) expression levels were significantly attenuated and enhanced on embryonic day 3.5 (E3.5), respectively, whereas no significant differences in the expression level were found in mice superovulated at the other two stages. A higher number of developing and Graafian follicles was observed in the ovarian sections 48h after the administration of pregnant mare serum gonadotropin (PMSG) at the proestrous and estrous stages. The sections from mice treated at the metestrous and diestrous stages, however, presented more corpora lutea. Therefore, mice superovulated at the proestrous and estrous stages exhibited the best pregnancy rates. Furthermore, the disordered expression of MUC1 and let-7a/let-7b miRNA in mice superovulated at the metestrous and diestrous stages may impair reproduction performance.
Collapse
|
34
|
Geary TW, Burns GW, Moraes JGN, Moss JI, Denicol AC, Dobbs KB, Ortega MS, Hansen PJ, Wehrman ME, Neibergs H, O'Neil E, Behura S, Spencer TE. Identification of Beef Heifers with Superior Uterine Capacity for Pregnancy. Biol Reprod 2016; 95:47. [PMID: 27417907 PMCID: PMC5029478 DOI: 10.1095/biolreprod.116.141390] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 07/06/2016] [Indexed: 11/16/2022] Open
Abstract
Infertility and subfertility represent major problems in domestic animals and humans, and the majority of embryonic loss occurs during the first month of gestation that involves pregnancy recognition and conceptus implantation. The critical genes and physiological pathways in the endometrium that mediate pregnancy establishment and success are not well understood. In study one, predominantly Angus heifers were classified based on fertility using serial embryo transfer to select animals with intrinsic differences in pregnancy loss. In each of the four rounds, a single in vitro-produced, high-quality embryo was transferred into heifers on Day 7 postestrus and pregnancy was determined on Days 28 and 42 by ultrasound and then terminated. Heifers were classified based on pregnancy success as high fertile (HF), subfertile (SF), or infertile (IF). In study two, fertility-classified heifers were resynchronized and bred with semen from a single high-fertility bull. Blood samples were collected every other day from Days 0 to 36 postmating. Pregnancy rate was determined on Day 28 by ultrasound and was higher in HF (70.4%) than in heifers with low fertility (36.8%; SF and IF). Progesterone concentrations in serum during the first 20 days postestrus were not different in nonpregnant heifers and also not different in pregnant heifers among fertility groups. In study three, a single in vivo-produced embryo was transferred into fertility-classified heifers on Day 7 postestrus. The uteri were flushed on Day 14 to recover embryos, and endometrial biopsies were obtained from the ipsilateral uterine horn. Embryo recovery rate and conceptus length and area were not different among the heifer groups. RNA was sequenced from the Day 14 endometrial biopsies of pregnant HF, SF, and IF heifers (n = 5 per group) and analyzed by edgeR-robust analysis. There were 26 differentially expressed genes (DEGs) in the HF compared to SF endometrium, 12 DEGs for SF compared to IF endometrium, and three DEGs between the HF and IF endometrium. Several of the DEG-encoded proteins are involved in immune responses and are expressed in B cells. Results indicate that preimplantation conceptus survival and growth to Day 14 is not compromised in SF and IF heifers. Thus, the observed difference in capacity for pregnancy success in these fertility-classified heifers is manifest between Days 14 and 28 when pregnancy recognition signaling and conceptus elongation and implantation must occur for the establishment of pregnancy.
Collapse
Affiliation(s)
- Thomas W Geary
- USDA-ARS, Fort Keogh Livestock and Range Research Laboratory, Miles City, Montana
| | - Gregory W Burns
- Division of Animal Sciences, University of Missouri, Columbia, Missouri
| | - Joao G N Moraes
- Division of Animal Sciences, University of Missouri, Columbia, Missouri
| | - James I Moss
- Department of Animal Sciences, University of Florida, Gainesville, Florida
| | - Anna C Denicol
- Department of Animal Sciences, University of Florida, Gainesville, Florida
| | - Kyle B Dobbs
- Department of Animal Sciences, University of Florida, Gainesville, Florida
| | - M Sofia Ortega
- Department of Animal Sciences, University of Florida, Gainesville, Florida
| | - Peter J Hansen
- Department of Animal Sciences, University of Florida, Gainesville, Florida
| | | | - Holly Neibergs
- Department of Animal Sciences, Washington State University, Pullman, Washington
| | - Eleanore O'Neil
- Division of Animal Sciences, University of Missouri, Columbia, Missouri
| | - Susanta Behura
- Division of Animal Sciences, University of Missouri, Columbia, Missouri
| | - Thomas E Spencer
- Division of Animal Sciences, University of Missouri, Columbia, Missouri
| |
Collapse
|
35
|
Chapela PJ, Broaddus RR, Hawkins SM, Lessey BA, Carson DD. Cytokine stimulation of MUC4 expression in human female reproductive tissue carcinoma cell lines and endometrial cancer. J Cell Biochem 2016; 116:2649-57. [PMID: 25923310 DOI: 10.1002/jcb.25213] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 04/22/2015] [Indexed: 12/27/2022]
Abstract
MUC4, a transmembrane glycoprotein, interferes with cell adhesion, and promotes EGFR signaling in cancer. Studies in rat models have demonstrated steroid hormonal regulation of endometrial MUC4 expression. In this study, qRT-PCR screening of mouse tissues determined that Muc4 mRNA also was robustly expressed in mouse uteri. Previous studies from our labs have demonstrated MUC4 mRNA was expressed at levels <1% of MUC1 mRNA in human endometrium and endometriotic tissue. Multiple human endometrial adenocarcinoma cell lines were assayed for MUC4 mRNA expression revealing extremely low basal expression in the Ishikawa, RL-95-2, AN3CA, and KLE lines. Moderate to high expression was observed in HEC50 and HEC-1A cells. MUC4 mRNA expression was not affected by progesterone and/or estrogen treatment, but was greatly stimulated at both mRNA and protein levels by proinflammatory cytokines (IFN-γ and TNF-α), particularly when used in combination. In endometrial tissue, MUC4 mRNA levels did not change significantly between normal or cancerous samples; although, a subset of patients with grade 1 and 2 tumors displayed substantially higher expression. Likewise, immunostaining of human endometrial adenocarcinoma tissues revealed little to no staining in many patients (low MUC4), but strong staining in some patients (high MUC4) independent of cancer grade. In cases where staining was observed, it was heterogeneous with some cells displaying robust MUC4 expression and others displaying little or no staining. Collectively, these observations demonstrate that while MUC4 is highly expressed in the mouse uterus, it is not a major mucin in normal human endometrium. Rather, MUC4 is a potential marker of endometrial adenocarcinoma in a subset of patients.
Collapse
Affiliation(s)
| | - Russell R Broaddus
- Department of Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030
| | - Shannon M Hawkins
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas, 77030
| | - Bruce A Lessey
- Department of Obstetrics and Gynecology, Greenville Health System, Greenville, South Carolina, 29605
| | - Daniel D Carson
- Department of Biosciences, Rice University, Houston, Texas, 77251.,Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030
| |
Collapse
|
36
|
Apostolopoulos V, Stojanovska L, Gargosky SE. MUC1 (CD227): a multi-tasked molecule. Cell Mol Life Sci 2015; 72:4475-500. [PMID: 26294353 PMCID: PMC11113675 DOI: 10.1007/s00018-015-2014-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 07/23/2015] [Accepted: 08/06/2015] [Indexed: 12/16/2022]
Abstract
Mucin 1 (MUC1 [CD227]) is a high-molecular weight (>400 kDa), type I membrane-tethered glycoprotein that is expressed on epithelial cells and extends far above the glycocalyx. MUC1 is overexpressed and aberrantly glycosylated in adenocarcinomas and in hematological malignancies. As a result, MUC1 has been a target for tumor immunotherapeutic studies in mice and in humans. MUC1 has been shown to have anti-adhesive and immunosuppressive properties, protects against infections, and is involved in the oncogenic process as well as in cell signaling. In addition, MUC1 plays a key role in the reproductive tract, in the immune system (affecting dendritic cells, monocytes, T cells, and B cells), and in chronic inflammatory diseases. Evidence for all of these roles for MUC1 is discussed herein and demonstrates that MUC1 is truly a multitasked molecule.
Collapse
Affiliation(s)
- Vasso Apostolopoulos
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia.
| | - Lily Stojanovska
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | | |
Collapse
|
37
|
Bastu E, Mutlu MF, Yasa C, Dural O, Nehir Aytan A, Celik C, Buyru F, Yeh J. Role of Mucin 1 and Glycodelin A in recurrent implantation failure. Fertil Steril 2015; 103:1059-1064.e2. [PMID: 25747132 DOI: 10.1016/j.fertnstert.2015.01.025] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 01/14/2015] [Accepted: 01/15/2015] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To evaluate and compare the levels of Mucin 1 (MUC-1) and Glycodelin A (GdA) in precisely timed endometrial biopsies and blood samples taken from women with recurrent implantation failure, and women with proven fertility, in a control group. DESIGN Molecular studies in human blood and tissue. SETTING University hospital. PATIENT(S) Women with recurrent implantation failure and women with proven fertility. INTERVENTION(S) Primary endometrial cells and blood samples during the implantation "window" (between day 7 and day 9 after the surge in luteinizing hormone). MAIN OUTCOME MEASURE(S) Expression of MUC-1 and GdA in the human endometrium and in blood during the implantation window were analyzed by enzyme-linked immunosorbent assay. Additionally, MUC-1 and GdA levels in tissue were analyzed by western blot during the same period. RESULT(S) Both blood and tissue measurements of MUC-1 and GdA were significantly lower in women with recurrent implantation failure than in fertile women during the implantation window. In addition, we found a highly significant correlation between blood vs. tissue measurements of both MUC-1 and GdA. CONCLUSION(S) The present study reveals that blood and tissue levels of MUC-1 and GdA are much lower in women with RIF, compared with those in fertile women. Receptivity can be evaluated with noninvasive blood sampling, rather than more-invasive endometrium sampling, as the blood and tissue measurements of MUC-1 and GdA are correlated.
Collapse
Affiliation(s)
- Ercan Bastu
- Department of Obstetrics and Gynecology, Istanbul University School of Medicine, Istanbul, Turkey.
| | | | - Cenk Yasa
- Department of Obstetrics and Gynecology, Istanbul University School of Medicine, Istanbul, Turkey
| | - Ozlem Dural
- Department of Obstetrics and Gynecology, Istanbul University School of Medicine, Istanbul, Turkey
| | - Asli Nehir Aytan
- Department of Obstetrics and Gynecology, Istanbul University School of Medicine, Istanbul, Turkey
| | - Cem Celik
- Bahceci Health Group, Istanbul, Turkey
| | - Faruk Buyru
- Department of Obstetrics and Gynecology, Istanbul University School of Medicine, Istanbul, Turkey
| | - John Yeh
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
38
|
Inyawilert W, Fu TY, Lin CT, Tang PC. MicroRNA-199a mediates mucin 1 expression in mouse uterus during implantation. Reprod Fertil Dev 2015; 26:653-64. [PMID: 23759257 DOI: 10.1071/rd12097] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 04/16/2013] [Indexed: 12/19/2022] Open
Abstract
Embryo implantation is a complicated process involving interactions between the blastocyst and the luminal epithelium of the receptive uterus. Mucin 1 (MUC1) is an integral membrane glycoprotein expressed apically by secretory epithelial cells and the glandular epithelium in different organs, including the uterus. It is believed that loss of MUC1 on the surface of uterine epithelial cells is necessary for embryo implantation. The endogenous non-protein coding microRNAs (miRNAs) of 21-24 nucleotides are found in diverse organisms. It has been shown that miRNAs participate in a range of cellular processes by regulating gene expression at the post-transcriptional level. In the present study, the regulatory role of miRNA-199a on the expression of MUC1 in mouse uterus during implantation was investigated for its effect on embryo implantation. Western blotting and immunohistochemistry results showed high MUC1 expression on Day 0.5 and low expression by Day 4.5 of pregnancy. In contrast with MUC1 expression, increased miRNA-199a expression was evident at Day 4.5 of pregnancy, as measured by real-time reverse transcription-polymerase chain reaction. In addition, we demonstrated direct binding of miRNA-199a to the 3'-untranslated region of MUC1. Transfection of miRNA-199a into mouse uterine epithelial cells isolated from Day 0.5 of pregnancy also downregulated expression of MUC1. Therefore, the present study provides evidence that MUC1 is a direct target of miRNA-199a and suggests that development of novel strategies to facilitate a successful pregnancy and repair implantation failure humans may include miRNA.
Collapse
Affiliation(s)
- Wilasinee Inyawilert
- Department of Animal Science, National Chung Hsing University, 250 Kuo Kuang Road, Taichung 40227, Taiwan
| | - Tzu-Yen Fu
- Department of Animal Science, National Chung Hsing University, 250 Kuo Kuang Road, Taichung 40227, Taiwan
| | - Chun-Ting Lin
- Department of Animal Science, National Chung Hsing University, 250 Kuo Kuang Road, Taichung 40227, Taiwan
| | - Pin-Chi Tang
- Department of Animal Science, National Chung Hsing University, 250 Kuo Kuang Road, Taichung 40227, Taiwan
| |
Collapse
|
39
|
Inyawilert W, Fu TY, Lin CT, Tang PC. Let-7-mediated suppression of mucin 1 expression in the mouse uterus during embryo implantation. J Reprod Dev 2015; 61:138-44. [PMID: 25739861 PMCID: PMC4410312 DOI: 10.1262/jrd.2014-106] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Mucin 1 (Muc1) is an integral transmembrane mucin glycoprotein expressed on the apical surface of most epithelia. It is considered to be a barrier to the regulation of embryo implantation by inhibiting attachment of the embryo to the endometrium. Therefore, loss of Muc1 on the surface of uterine epithelial cells is necessary for embryo implantation. Studies have demonstrated that microRNAs (miRNAs) play a key role in enhancing embryo implantation in mammals. In this study, we investigated the regulatory role of two miRNAs (let-7a and let-7b) on the expression of Muc1 in mouse uteri during implantation. Western blotting indicated that Muc1 expression was highest on day1 of pregnancy and constantly decreased thereafter until day 4. In contrast to Muc1 expression, increased expression of let-7a and let-7b was evident on day 4 of pregnancy as measured by real-time reverse transcription-polymerase chain reaction
(real-time RT-PCR). We demonstrated direct binding of let-7a and let-7b to the 3’untranslated region of muc1. Furthermore, Muc1 expression was suppressed after transfection of mouse uterine epithelial cells isolated from day 1 of pregnancy with let-7a and let-7b. In summary, the present study provides evidence that Muc1 is a direct target of let-7a and let-7b. Additionally, the current study suggests that miRNAs are novel targets which can be used to facilitate a successful pregnancy and repair implantation failure.
Collapse
Affiliation(s)
- Wilasinee Inyawilert
- Department of Animal Science, National Chung Hsing University, Taichung 40227, Taiwan
| | | | | | | |
Collapse
|
40
|
Sheldon IM. Genes and environmental factors that influence disease resistance to microbes in the female reproductive tract of dairy cattle. Reprod Fertil Dev 2015; 27:72-81. [DOI: 10.1071/rd14305] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Microbes commonly infect the female reproductive tract of cattle, causing infertility, abortion and post partum uterine diseases. When organisms reach the uterus, the resistance to disease depends on the balance between the classic triad of the virulence of the microbes, the host defence systems and the environment. The present review considers each aspect of this triad, using postpartum uterine disease as an exemplar for understanding disease resistance. The bacteria that cause postpartum uterine disease are adapted to the endometrium, and their microbial toxins cause tissue damage and inflammation. However, non-specific defence systems counter ascending infections of the female reproductive tract, and inflammatory responses in the endometrium are driven by innate immunity. Disease resistance to bacterial infection involves many genes involved in the maintenance or restoration of tissue homeostasis in the endometrium, including antimicrobial peptides, complement, cytokines, chemokines and Toll-like receptors. The most important environmental factors facilitating the development of postpartum uterine disease are related to trauma of the reproductive tract and to the metabolic stress of lactation in dairy cows. Long-term solutions for uterine disease will include genetic selection for disease resistance and optimising the care of the animal before, during and after parturition.
Collapse
|
41
|
Transmembrane Mucin Expression and Function in Embryo Implantation and Placentation. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2015; 216:51-68. [PMID: 26450494 DOI: 10.1007/978-3-319-15856-3_4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Transmembrane mucins (TMs) are extremely large, complex glycoproteins that line the apical surfaces of simple epithelia including those of the female reproductive tract. TMs provide a physical barrier consistent with their role as part of the innate immune system. This barrier function must be overcome in the context of embryo implantation to permit blastocyst attachment. Three major TMs have been identified in uterine epithelia of multiple species: MUC1, MUC4, and MUC16. MUC1 has been found in all species studied to date, whereas expression of MUC4 and MUC16 have been less well studied and may be species specific. The strategies for removing mucins to permit embryo attachment also vary in a species-specific way and include both hormonal suppression of TM gene expression and membrane clearance via cell surface proteases. Studies emerging from the cancer literature indicate that TMs can modulate a surprisingly wide variety of signal transduction processes. Furthermore, various cell surface proteins have been identified that bind either the oligosaccharide or protein motifs of TMs suggesting that these molecules may support cell attachment in some contexts, including trophoblast interactions with cells of the immune system. The intimate association of TMs at sites of embryo-maternal interaction and the varied functions these complex molecules can play make them key players in embryo implantation and placentation processes.
Collapse
|
42
|
Lei W, Herington J, Galindo CL, Ding T, Brown N, Reese J, Paria BC. Cross-species transcriptomic approach reveals genes in hamster implantation sites. Reproduction 2014; 148:607-21. [PMID: 25252651 DOI: 10.1530/rep-14-0388] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The mouse model has greatly contributed to understanding molecular mechanisms involved in the regulation of progesterone (P4) plus estrogen (E)-dependent blastocyst implantation process. However, little is known about contributory molecular mechanisms of the P4-only-dependent blastocyst implantation process that occurs in species such as hamsters, guineapigs, rabbits, pigs, rhesus monkeys, and perhaps humans. We used the hamster as a model of P4-only-dependent blastocyst implantation and carried out cross-species microarray (CSM) analyses to reveal differentially expressed genes at the blastocyst implantation site (BIS), in order to advance the understanding of molecular mechanisms of implantation. Upregulation of 112 genes and downregulation of 77 genes at the BIS were identified using a mouse microarray platform, while use of the human microarray revealed 62 up- and 38 down-regulated genes at the BIS. Excitingly, a sizable number of genes (30 up- and 11 down-regulated genes) were identified as a shared pool by both CSMs. Real-time RT-PCR and in situ hybridization validated the expression patterns of several up- and down-regulated genes identified by both CSMs at the hamster and mouse BIS to demonstrate the merit of CSM findings across species, in addition to revealing genes specific to hamsters. Functional annotation analysis found that genes involved in the spliceosome, proteasome, and ubiquination pathways are enriched at the hamster BIS, while genes associated with tight junction, SAPK/JNK signaling, and PPARα/RXRα signalings are repressed at the BIS. Overall, this study provides a pool of genes and evidence of their participation in up- and down-regulated cellular functions/pathways at the hamster BIS.
Collapse
Affiliation(s)
- Wei Lei
- Division of NeonatologyDepartment of PediatricsDivision of Cardiovascular MedicineDepartment of Obstetrics and GynecologyVanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Jennifer Herington
- Division of NeonatologyDepartment of PediatricsDivision of Cardiovascular MedicineDepartment of Obstetrics and GynecologyVanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Cristi L Galindo
- Division of NeonatologyDepartment of PediatricsDivision of Cardiovascular MedicineDepartment of Obstetrics and GynecologyVanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Tianbing Ding
- Division of NeonatologyDepartment of PediatricsDivision of Cardiovascular MedicineDepartment of Obstetrics and GynecologyVanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Naoko Brown
- Division of NeonatologyDepartment of PediatricsDivision of Cardiovascular MedicineDepartment of Obstetrics and GynecologyVanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Jeff Reese
- Division of NeonatologyDepartment of PediatricsDivision of Cardiovascular MedicineDepartment of Obstetrics and GynecologyVanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Bibhash C Paria
- Division of NeonatologyDepartment of PediatricsDivision of Cardiovascular MedicineDepartment of Obstetrics and GynecologyVanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| |
Collapse
|
43
|
Ezoe K, Daikoku T, Yabuuchi A, Murata N, Kawano H, Abe T, Okuno T, Kobayashi T, Kato K. Ovarian stimulation using human chorionic gonadotrophin impairs blastocyst implantation and decidualization by altering ovarian hormone levels and downstream signaling in mice. Mol Hum Reprod 2014; 20:1101-16. [PMID: 25122188 DOI: 10.1093/molehr/gau065] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Ovarian stimulation induced by follicle-stimulating hormone and human chorionic gonadotrophin (hCG) is commonly used in assisted reproductive technology to increase embryo production. However, recent clinical and animal studies have shown that ovarian stimulation disrupts endometrial function and embryo development and adversely affects pregnancy outcomes. How ovarian stimulation impairs pregnancy establishment and the precise mechanisms by which this stimulation reduces the chances of conception remain unclear. In this study, we first demonstrated that ovarian stimulation using hCG alone impairs implantation, decidualization and fetal development of mice by generating abnormal ovarian hormone levels. We also showed that ovarian hormone levels were altered because of changes in the levels of the enzymes involved in their synthesis in the follicles and corpora lutea. Furthermore, we determined that anomalous ovarian hormone secretion induced by ovarian stimulation alters the spatiotemporal expression of progesterone receptors and their downstream genes, especially in the uterine epithelium. Epithelial estrogenic signaling and cell proliferation were promoted on the day of implantation in stimulated mice and these changes led to the failure of uterine transition from the prereceptive to the receptive state. Collectively, our findings indicate that ovarian stimulation using hCG induces an imbalance in steroid hormone secretion, which causes a failure of the development of uterine receptivity and subsequent implantation and decidualization by altering the expression of steroid receptors and their downstream signaling associated with embryo implantation.
Collapse
Affiliation(s)
- Kenji Ezoe
- Kato Ladies Clinic, 7-20-3 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Takiko Daikoku
- Division of Reproductive Sciences, Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Akiko Yabuuchi
- Kato Ladies Clinic, 7-20-3 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Nana Murata
- Kato Ladies Clinic, 7-20-3 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Hiroomi Kawano
- Kato Ladies Clinic, 7-20-3 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Takashi Abe
- Kato Ladies Clinic, 7-20-3 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Takashi Okuno
- Kato Ladies Clinic, 7-20-3 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Tamotsu Kobayashi
- Kato Ladies Clinic, 7-20-3 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Keiichi Kato
- Kato Ladies Clinic, 7-20-3 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| |
Collapse
|
44
|
Sheldon IM, Cronin JG, Healey GD, Gabler C, Heuwieser W, Streyl D, Bromfield JJ, Miyamoto A, Fergani C, Dobson H. Innate immunity and inflammation of the bovine female reproductive tract in health and disease. Reproduction 2014; 148:R41-51. [PMID: 24890752 DOI: 10.1530/rep-14-0163] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mammalian reproductive physiology and the development of viviparity co-evolved with inflammation and immunity over millennia. Many inflammatory mediators contribute to paracrine and endocrine signalling, and the maintenance of tissue homeostasis in the female reproductive tract. However, inflammation is also a feature of microbial infections of the reproductive tract. Bacteria and viruses commonly cause endometritis, perturb ovarian follicle development and suppress the endocrine activity of the hypothalamus and pituitary in cattle. Innate immunity is an evolutionary ancient system that orchestrates host cell inflammatory responses aimed at eliminating pathogens and repairing damaged tissue. Pattern recognition receptors on host cells bind pathogen-associated molecular patterns and damage-associated molecular patterns, leading to the activation of intracellular MAPK and NFκB signalling pathways and the release of inflammatory mediators. Inflammatory mediators typically include the interleukin cytokines IL1β and IL6, chemokines such as IL8, interferons and prostaglandins. This review outlines the mechanisms of inflammation and innate immunity in the bovine female reproductive tract during health and disease condition.
Collapse
Affiliation(s)
- I Martin Sheldon
- College of MedicineInstitute of Life Science, Swansea University, Singleton Park, Swansea SA2 8PP, UKInstitute of Veterinary BiochemistryClinic of Animal ReproductionFreie Universitaet Berlin, Berlin, GermanyClinic for Ruminants with Ambulatory and Herd Health ServicesCentre for Clinical Veterinary Medicine, Ludwig Maximilian University of Munich, Oberschleißheim, GermanyDepartment of Animal SciencesUniversity of Florida, Gainesville, Florida 32608, USAGraduate School for Animal and Food HygieneObihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, JapanSchool of Veterinary ScienceUniversity of Liverpool, Leahurst, Neston CH64 7TE, UK
| | - James G Cronin
- College of MedicineInstitute of Life Science, Swansea University, Singleton Park, Swansea SA2 8PP, UKInstitute of Veterinary BiochemistryClinic of Animal ReproductionFreie Universitaet Berlin, Berlin, GermanyClinic for Ruminants with Ambulatory and Herd Health ServicesCentre for Clinical Veterinary Medicine, Ludwig Maximilian University of Munich, Oberschleißheim, GermanyDepartment of Animal SciencesUniversity of Florida, Gainesville, Florida 32608, USAGraduate School for Animal and Food HygieneObihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, JapanSchool of Veterinary ScienceUniversity of Liverpool, Leahurst, Neston CH64 7TE, UK
| | - Gareth D Healey
- College of MedicineInstitute of Life Science, Swansea University, Singleton Park, Swansea SA2 8PP, UKInstitute of Veterinary BiochemistryClinic of Animal ReproductionFreie Universitaet Berlin, Berlin, GermanyClinic for Ruminants with Ambulatory and Herd Health ServicesCentre for Clinical Veterinary Medicine, Ludwig Maximilian University of Munich, Oberschleißheim, GermanyDepartment of Animal SciencesUniversity of Florida, Gainesville, Florida 32608, USAGraduate School for Animal and Food HygieneObihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, JapanSchool of Veterinary ScienceUniversity of Liverpool, Leahurst, Neston CH64 7TE, UK
| | - Christoph Gabler
- College of MedicineInstitute of Life Science, Swansea University, Singleton Park, Swansea SA2 8PP, UKInstitute of Veterinary BiochemistryClinic of Animal ReproductionFreie Universitaet Berlin, Berlin, GermanyClinic for Ruminants with Ambulatory and Herd Health ServicesCentre for Clinical Veterinary Medicine, Ludwig Maximilian University of Munich, Oberschleißheim, GermanyDepartment of Animal SciencesUniversity of Florida, Gainesville, Florida 32608, USAGraduate School for Animal and Food HygieneObihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, JapanSchool of Veterinary ScienceUniversity of Liverpool, Leahurst, Neston CH64 7TE, UK
| | - Wolfgang Heuwieser
- College of MedicineInstitute of Life Science, Swansea University, Singleton Park, Swansea SA2 8PP, UKInstitute of Veterinary BiochemistryClinic of Animal ReproductionFreie Universitaet Berlin, Berlin, GermanyClinic for Ruminants with Ambulatory and Herd Health ServicesCentre for Clinical Veterinary Medicine, Ludwig Maximilian University of Munich, Oberschleißheim, GermanyDepartment of Animal SciencesUniversity of Florida, Gainesville, Florida 32608, USAGraduate School for Animal and Food HygieneObihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, JapanSchool of Veterinary ScienceUniversity of Liverpool, Leahurst, Neston CH64 7TE, UK
| | - Dominik Streyl
- College of MedicineInstitute of Life Science, Swansea University, Singleton Park, Swansea SA2 8PP, UKInstitute of Veterinary BiochemistryClinic of Animal ReproductionFreie Universitaet Berlin, Berlin, GermanyClinic for Ruminants with Ambulatory and Herd Health ServicesCentre for Clinical Veterinary Medicine, Ludwig Maximilian University of Munich, Oberschleißheim, GermanyDepartment of Animal SciencesUniversity of Florida, Gainesville, Florida 32608, USAGraduate School for Animal and Food HygieneObihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, JapanSchool of Veterinary ScienceUniversity of Liverpool, Leahurst, Neston CH64 7TE, UK
| | - John J Bromfield
- College of MedicineInstitute of Life Science, Swansea University, Singleton Park, Swansea SA2 8PP, UKInstitute of Veterinary BiochemistryClinic of Animal ReproductionFreie Universitaet Berlin, Berlin, GermanyClinic for Ruminants with Ambulatory and Herd Health ServicesCentre for Clinical Veterinary Medicine, Ludwig Maximilian University of Munich, Oberschleißheim, GermanyDepartment of Animal SciencesUniversity of Florida, Gainesville, Florida 32608, USAGraduate School for Animal and Food HygieneObihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, JapanSchool of Veterinary ScienceUniversity of Liverpool, Leahurst, Neston CH64 7TE, UK
| | - Akio Miyamoto
- College of MedicineInstitute of Life Science, Swansea University, Singleton Park, Swansea SA2 8PP, UKInstitute of Veterinary BiochemistryClinic of Animal ReproductionFreie Universitaet Berlin, Berlin, GermanyClinic for Ruminants with Ambulatory and Herd Health ServicesCentre for Clinical Veterinary Medicine, Ludwig Maximilian University of Munich, Oberschleißheim, GermanyDepartment of Animal SciencesUniversity of Florida, Gainesville, Florida 32608, USAGraduate School for Animal and Food HygieneObihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, JapanSchool of Veterinary ScienceUniversity of Liverpool, Leahurst, Neston CH64 7TE, UK
| | - Chrys Fergani
- College of MedicineInstitute of Life Science, Swansea University, Singleton Park, Swansea SA2 8PP, UKInstitute of Veterinary BiochemistryClinic of Animal ReproductionFreie Universitaet Berlin, Berlin, GermanyClinic for Ruminants with Ambulatory and Herd Health ServicesCentre for Clinical Veterinary Medicine, Ludwig Maximilian University of Munich, Oberschleißheim, GermanyDepartment of Animal SciencesUniversity of Florida, Gainesville, Florida 32608, USAGraduate School for Animal and Food HygieneObihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, JapanSchool of Veterinary ScienceUniversity of Liverpool, Leahurst, Neston CH64 7TE, UK
| | - Hilary Dobson
- College of MedicineInstitute of Life Science, Swansea University, Singleton Park, Swansea SA2 8PP, UKInstitute of Veterinary BiochemistryClinic of Animal ReproductionFreie Universitaet Berlin, Berlin, GermanyClinic for Ruminants with Ambulatory and Herd Health ServicesCentre for Clinical Veterinary Medicine, Ludwig Maximilian University of Munich, Oberschleißheim, GermanyDepartment of Animal SciencesUniversity of Florida, Gainesville, Florida 32608, USAGraduate School for Animal and Food HygieneObihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, JapanSchool of Veterinary ScienceUniversity of Liverpool, Leahurst, Neston CH64 7TE, UK
| |
Collapse
|
45
|
Bojić-Trbojević Ž, Jovanović Krivokuća M, Kolundžić N, Petronijević M, Vrzić-Petronijević S, Golubović S, Vićovac L. Galectin-1 binds mucin in human trophoblast. Histochem Cell Biol 2014; 142:541-53. [DOI: 10.1007/s00418-014-1229-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2014] [Indexed: 12/11/2022]
|
46
|
Gabryel-Porowska H, Gornowicz A, Bielawska A, Wójcicka A, Maciorkowska E, Grabowska SZ, Bielawski K. Mucin levels in saliva of adolescents with dental caries. Med Sci Monit 2014; 20:72-7. [PMID: 24441930 PMCID: PMC3907531 DOI: 10.12659/msm.889718] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background Human saliva, a complex secretion that contains a mixture of inorganic and organic molecules, plays an essential role in the maintenance of oral health. Mucins are the major macromolecular component of the secretion and are considered the first line of defense for epithelial tissues. The aim of this study was to compare levels of mucins (MUC5B, MUC7, and MUC1) in saliva of young subjects with dental caries. Material/Methods All patients had DMF (decay/missing/filled) higher than value 0. Eight subjects with DMF=3 (control group) and 27 adolescents with DMF >11 (research group) were recruited for this study. Clinical evaluation procedures were oral examination, including tooth, periodontal, oral mucosal status, and collection of saliva samples. Saliva was collected for mucin assay. Enzyme-linked immunosorbent assay was used to quantitate MUC5B, MUC7, and MUC1. Results Our results indicate that adolescents with very high intensity of dental caries disease had increased levels of MUC1 and MUC5B. The membrane mucin MUC1 protein levels in the group with DMF>11 (research group) were higher compared to the group with DMF=3 (control group), and the increase was statistically significant (p=0.011). Similarly, secreted mucin MUC5B protein levels were higher (p=0.06) in the group with DMF>11 (research group). Although MUC7 protein levels were slightly reduced in symptomatic subjects, the decrease was statistically insignificant (p=0.918). Conclusions Our data suggest links between the production of mucins, especially MUC1 and MUC5B in saliva, and dental caries disease.
Collapse
Affiliation(s)
| | - Agnieszka Gornowicz
- Department of Biotechnology, Medical University of Białystok, Białystok, Poland
| | - Anna Bielawska
- Department of Biotechnology, Medical University of Białystok, Białystok, Poland
| | - Anna Wójcicka
- Maxillofacial and Plastic Surgery Clinic, Białystok, Poland
| | - Elżbieta Maciorkowska
- Department of Developmental Period Medicine and Pediatric Nursing, Medical University of Białystok, Białystok, Poland
| | | | - Krzysztof Bielawski
- Department of Synthesis and Technology of Drugs, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
47
|
Check JH, Cohen R. The role of progesterone and the progesterone receptor in human reproduction and cancer. Expert Rev Endocrinol Metab 2013; 8:469-484. [PMID: 30754194 DOI: 10.1586/17446651.2013.827380] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Insufficient progesterone, effect possibly more on immune factors rather than adequate endometrial development, can be an easy remedial cause of infertility by simply supplementing the luteal phase with either vaginal or intramuscular or oral (dydrogesterone) progesterone. Progesterone will also help to reduce miscarriage rates when follicle maturing drugs are used for those with regular menses but follicular maturation defects, or women with recurrent miscarriages. One mechanism of action seems to be related to production of an immunomodulatory protein, the progesterone-induced blocking factor either in the cytoplasm or in the circulation. PIBF inhibits cytotoxicity of natural killer cells. Cancer cells may 'borrow' the same mechanism to escape NK cell immunosurveillance.
Collapse
Affiliation(s)
- Jerome H Check
- a Department of Obstetrics and Gynecology, Cooper Medical School of Rowan University, Division of Reproductive Endocrinology & Infertility, Camden, NJ, USA
| | - Rachael Cohen
- b Department of Obstetrics and Gynecology, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| |
Collapse
|
48
|
Sun X, Bartos A, Whitsett JA, Dey SK. Uterine deletion of Gp130 or Stat3 shows implantation failure with increased estrogenic responses. Mol Endocrinol 2013; 27:1492-501. [PMID: 23885093 DOI: 10.1210/me.2013-1086] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Leukemia inhibitory factor (LIF), a downstream target of estrogen, is essential for implantation in mice. LIF function is thought to be mediated by its binding to LIF receptor (LIFR) and recruitment of coreceptor GP130 (glycoprotein 130), and this receptor complex then activates signal transducer and activator of transcription (STAT)1/3. However, the importance of LIFR and GP130 acting via STAT3 in implantation remains uncertain, because constitutive inactivation of Lifr, Gp130, or Stat3 shows embryonic lethality in mice. To address this issue, we generated mice with conditional deletion of uterine Gp130 or Stat3 and show that both GP130 and STAT3 are critical for uterine receptivity and implantation. Implantation failure in these deleted mice is associated with higher uterine estrogenic responses prior to the time of implantation. These heightened estrogenic responses are not due to changes in ovarian hormone levels or expression of their nuclear receptors. In the deleted mice, estrogen-responsive gene, Lactoferrin (Ltf), and Mucin 1 protein, were up-regulated in the uterus. In addition, progesterone-responsive genes, Hoxa10 and Indian hedgehog (Ihh), were markedly down-regulated in STAT3-inactivated uteri. These changes in uteri of deleted mice were reflected by the failure of differentiation of the luminal epithelium, which is essential for blastocyst attachment.
Collapse
Affiliation(s)
- Xiaofei Sun
- Division of Reproductive Sciences,Perinatal Institute, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA
| | | | | | | |
Collapse
|
49
|
Lillehoj EP, Kato K, Lu W, Kim KC. Cellular and molecular biology of airway mucins. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 303:139-202. [PMID: 23445810 PMCID: PMC5593132 DOI: 10.1016/b978-0-12-407697-6.00004-0] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Airway mucus constitutes a thin layer of airway surface liquid with component macromolecules that covers the luminal surface of the respiratory tract. The major function of mucus is to protect the lungs through mucociliary clearance of inhaled foreign particles and noxious chemicals. Mucus is comprised of water, ions, mucin glycoproteins, and a variety of other macromolecules, some of which possess anti-microbial, anti-protease, and anti-oxidant activities. Mucins comprise the major protein component of mucus and exist as secreted and cell-associated glycoproteins. Secreted, gel-forming mucins are mainly responsible for the viscoelastic property of mucus, which is crucial for effective mucociliary clearance. Cell-associated mucins shield the epithelial surface from pathogens through their extracellular domains and regulate intracellular signaling through their cytoplasmic regions. However, neither the exact structures of mucin glycoproteins, nor the manner through which their expression is regulated, are completely understood. This chapter reviews what is currently known about the cellular and molecular properties of airway mucins.
Collapse
Affiliation(s)
- Erik P. Lillehoj
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kosuke Kato
- Center for Inflammation, Translational and Clinical Lung Research and Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA
| | - Wenju Lu
- Guangzhou Institute of Respiratory Diseases, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, PR China
| | - Kwang C. Kim
- Center for Inflammation, Translational and Clinical Lung Research and Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
50
|
Menon BB, Govindarajan B. Identification of an atypical zinc metalloproteinase, ZmpC, from an epidemic conjunctivitis-causing strain of Streptococcus pneumoniae. Microb Pathog 2012; 56:40-6. [PMID: 23168398 DOI: 10.1016/j.micpath.2012.11.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 11/08/2012] [Indexed: 11/26/2022]
Abstract
Streptococcus pneumoniae is a pathogen associated with a range of invasive and noninvasive infections. Despite the identification of the majority of virulence factors expressed by S. pneumoniae, knowledge of the strategies used by this bacterium to trigger infections, especially those originating at wet-surfaced epithelia, remains limited. In this regard, we recently reported a mechanism used by a nonencapsulated, epidemic conjunctivitis-causing strain of S. pneumoniae (strain SP168) to gain access into ocular surface epithelial cells. Mechanistically, strain SP168 secretes a zinc metalloproteinase, encoded by a truncated zmpC gene, to cleave off the ectodomain of a vital defense component - the membrane mucin MUC16 - from the apical glycocalyx barrier of ocular surface epithelial cells and, thereby invades underlying epithelial cells. Here, we compare the truncated SP168 ZmpC to its highly conserved archetype from S. pneumoniae serotype 4 (TIGR4), which has been linked to pneumococcal virulence in previous studies. Comparative nucleotide sequence analyses revealed that the zmpC gene corresponding to strain SP168 has two stretches of DNA deleted near its 5' end. A third 3 bp in-frame deletion, resulting in the elimination of an alanine residue, was found towards the middle segment of the SP168 zmpC. Closer examination of the primary structure revealed that the SP168 ZmpC lacks the canonical LPXTG motif - a signature typical of several surface proteins of gram-positive bacteria and of other pneumococcal zinc metalloproteinases. Surprisingly, in vitro assays performed using recombinant forms of ZmpC indicated that the truncated SP168 ZmpC induces more cleavage of the MUC16 ectodomain than its TIGR4 counterpart. This feature may help explain, in part, why S. pneumoniae strain SP168 is better equipped at abrogating the MUC16 glycocalyx barrier en route to causing epidemic conjunctivitis.
Collapse
Affiliation(s)
- Balaraj B Menon
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford St., Boston, MA 02114, USA.
| | | |
Collapse
|