1
|
Massarotti C, Cimadomo D, Spadoni V, Conforti A, Zacà C, Carosso AR, Vaiarelli A, Venturella R, Vitagliano A, Busnelli A, Cozzolino M, Borini A. Female fertility preservation for family planning: a position statement of the Italian Society of Fertility and Sterility and Reproductive Medicine (SIFES-MR). J Assist Reprod Genet 2024; 41:2521-2535. [PMID: 39030346 PMCID: PMC11405660 DOI: 10.1007/s10815-024-03197-4] [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: 04/09/2024] [Accepted: 07/03/2024] [Indexed: 07/21/2024] Open
Abstract
PURPOSE This position statement by the Italian Society of Fertility and Sterility and Reproductive Medicine (SIFES-MR) aims to establish an optimal framework for fertility preservation outside the standard before oncological therapies. Key topics include the role of fertility units in comprehensive fertility assessment, factors impacting ovarian potential, available preservation methods, and appropriate criteria for offering such interventions. METHODS The SIFES-MR writing group comprises Italian reproductive physicians, embryologists, and scientists. The consensus emerged after a six-month period of meetings, including extensive literature review, dialogue among authors and input from society members. Final approval was granted by the SIFES-MR governing council. RESULTS Fertility counselling transitions from urgent to long-term care, emphasizing family planning. Age, along with ovarian reserve markers, is the primary predictor of female fertility. Various factors, including gynecological conditions, autoimmune disorders, and prior gonadotoxic therapies, may impact ovarian reserve. Oocyte cryopreservation should be the preferred method. Women 30-34 years old and 35-39 years old, without known pathologies impacting the ovarian reserve, should cryopreserve at least 12-13 and 15-20 oocytes to achieve the same chance of a spontaneous live birth they would have if they tried to conceive at the age of cryopreservation (63% and 52%, respectively in the two age groups). CONCLUSIONS Optimal fertility counselling necessitates a long-term approach, that nurtures an understanding of fertility, facilitates timely evaluation of factors that may affect fertility, and explores fertility preservation choices at opportune intervals.
Collapse
Affiliation(s)
- Claudia Massarotti
- Physiopathology of Human Reproduction, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi, 10, 16132, Genova, Italy.
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health (DINOGMI Department), University of Genova, Genova, Italy.
| | - Danilo Cimadomo
- IVIRMA Global Reseach Alliance, Genera, Clinica Valle Giulia, Rome, Italy
| | | | - Alessandro Conforti
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Carlotta Zacà
- IVIRMA Global Research Alliance, 9.Baby, Bologna, Italy
| | - Andrea Roberto Carosso
- Obstetrics and Gynecology 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, Sant'Anna Hospital Città della Salute e della Scienza di Torino, University of Torino, Turin, Italy
| | - Alberto Vaiarelli
- IVIRMA Global Reseach Alliance, Genera, Clinica Valle Giulia, Rome, Italy
| | - Roberta Venturella
- Unit of Obstetrics and Gynecology, University of Catanzaro "Magna Grecia", Catanzaro, Italy
| | - Amerigo Vitagliano
- First Unit of Obstetrics and Gynecology, Department of Interdisciplinary Medicine (DIM), University of Bari, Bari, Italy
| | - Andrea Busnelli
- Department of Obstetrics and Gynecology, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Mauro Cozzolino
- IVIRMA Global Research Alliance, IVI Roma, Rome, Italy
- IVIRMA Global Research Alliance, Fundación IVI-IIS la Fe, Valencia, Spain
| | - Andrea Borini
- IVIRMA Global Research Alliance, 9.Baby, Bologna, Italy
| |
Collapse
|
2
|
Xu H, Mao X, Zhang S, Ren J, Jiang S, Cai L, Miao X, Tao Y, Peng C, Lv M, Li Y. Perfluorooctanoic acid triggers premature ovarian insufficiency by impairing NAD+ synthesis and mitochondrial function in adult zebrafish. Toxicol Sci 2024; 201:118-128. [PMID: 38830045 DOI: 10.1093/toxsci/kfae071] [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] [Indexed: 06/05/2024] Open
Abstract
High-dose perfluorooctanoic acid (PFOA) impairs oocyte maturation and offspring quality. However, the physiological concentrations of PFOA in follicular fluids of patients with premature ovarian insufficiency (POI) were detected at lower levels, thus the relationship between physiological PFOA and reproductive disorders remains elusive. Here, we investigated whether physiological PFOA exposure affects gonad function in adult zebrafish. Physiological PFOA exposure resulted in POI-like phenotypes in adult females, which exhibited decreased spawning frequency, reduced number of ovulated eggs, abnormal gonadal index, and aberrant embryonic mortality. Meanwhile, oocytes from PFOA-exposed zebrafish showed mitochondrial disintegration and diminished mitochondrial membrane potential. Unlike the high-dose treated oocytes exhibiting high reactive oxygen species (ROS) levels and excessive apoptosis, physiological PFOA reduced the ROS levels and did not trigger apoptosis. Interestingly, physiological PFOA exposure would not affect testis function, indicating specific toxicity in females. Mechanistically, PFOA suppressed the NAD+ biosynthesis and impaired mitochondrial function in oocytes, thus disrupting oocyte maturation and ovarian fertility. Nicotinamide mononucleotide (NMN), a precursor for NAD+ biosynthesis, alleviated the PFOA-induced toxic effects in oocytes and improved the oocyte maturation and fertility upon PFOA exposure. Our findings discover new insights into PFOA-induced reproductive toxicity and provide NMN as a potential drug for POI therapy.
Collapse
Affiliation(s)
- Hao Xu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, Southwest University, Chongqing 400715, China
| | - Xiaoyu Mao
- College of Language Intelligence, Sichuan International Studies University, Chongqing 400031, China
| | - Siling Zhang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China
| | - Jie Ren
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China
| | - Shanwen Jiang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China
| | - Lijuan Cai
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China
| | - Xiaomin Miao
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China
| | - Yixi Tao
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China
| | - Chao Peng
- Fisheries Development Department of Agriculture and Rural Committee of Nanchuan District, Chongqing 408400, China
| | - Mengzhu Lv
- Department of Immunology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Yun Li
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, Southwest University, Chongqing 400715, China
| |
Collapse
|
3
|
Benelli E, Marradi M, Sciarroni E, Di Cosmo C, Bagattini B, Del Ghianda S, Simoncini T, Fruzzetti F, Tonacchera M, Fiore E. Thyroid autoimmunity in different phenotypes of polycystic ovary syndrome: a single-center experience. J Endocrinol Invest 2024:10.1007/s40618-024-02404-4. [PMID: 38850509 DOI: 10.1007/s40618-024-02404-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 05/29/2024] [Indexed: 06/10/2024]
Abstract
PURPOSE Polycystic ovary syndrome (PCOS) has been associated with Hashimoto's thyroiditis (HT) and 4 phenotypes have been described in this syndrome. The aim of this work was to investigate the frequency of anti-thyroid antibodies (TAb) and thyroid function in the 4 phenotypes of PCOS. PATIENTS This study included 448 patients with PCOS: 260 (58.0%) with phenotype A, 119 (26.6%) with phenotype B, 38 (8.5%) with phenotype C and 31 (6.9%) with phenotype D. RESULTS TAb positivity was detected in 90/448 patients (20.1%) and was statistically significant higher (p = 0.03) in the grouped phenotypes A-B (83/379, 21.9%) than in phenotypes C-D (7/69, 10.1%). Positive anti-thyroglobulin antibodies (TgAb) were detected in 74/448 (16.5%) patients and positive anti-thyroperoxidase antibodies (TPOAb) in 66/448 (14.7%) patients. Both TgAb and TPOAb positivity was higher but not statistically significant in phenotype A-B than phenotype C-D. High titer TgAb (> 100 UI/ml) frequency was significantly higher (p = 0.005) in grouped phenotypes A-B (39/379, 10.3%) than in phenotypes C-D (0/69, 0.0%), while no significant difference was observed for low titer TgAb (≤ 100 UI/ml). According to a binary logistic regression analysis hypothyroidism was significantly associated with TAb positivity (OR 4.19; CI 2.25-7.79; p < 0.01) but not with PCOS phenotype. Androgen profile was not associated with TAb positivity. CONCLUSION A higher frequency of positive TAb and of high titer TgAb and TPOAb have been detected in PCOS women with phenotypes A and B, probably in relation to the greater imbalances between estrogen and progesterone levels present in these phenotypes.
Collapse
Affiliation(s)
- E Benelli
- Department of Clinical and Experimental Medicine, Section of Endocrinology, University Hospital of Pisa, Via Paradisa 2, 56124, Pisa, Pisa, Italy
| | - M Marradi
- Department of Clinical and Experimental Medicine, Section of Endocrinology, University Hospital of Pisa, Via Paradisa 2, 56124, Pisa, Pisa, Italy
| | - E Sciarroni
- Department of Clinical and Experimental Medicine, Section of Endocrinology, University Hospital of Pisa, Via Paradisa 2, 56124, Pisa, Pisa, Italy
| | - C Di Cosmo
- Department of Clinical and Experimental Medicine, Section of Endocrinology, University Hospital of Pisa, Via Paradisa 2, 56124, Pisa, Pisa, Italy
| | - B Bagattini
- Department of Clinical and Experimental Medicine, Section of Endocrinology, University Hospital of Pisa, Via Paradisa 2, 56124, Pisa, Pisa, Italy
| | - S Del Ghianda
- Department of Clinical and Experimental Medicine, Section of Endocrinology, University Hospital of Pisa, Via Paradisa 2, 56124, Pisa, Pisa, Italy
| | - T Simoncini
- Department of Obstetrics and Gynecology, University Hospital of Pisa, Pisa, Italy
| | - F Fruzzetti
- Department of Obstetrics and Gynecology, University Hospital of Pisa, Pisa, Italy
| | - M Tonacchera
- Department of Clinical and Experimental Medicine, Section of Endocrinology, University Hospital of Pisa, Via Paradisa 2, 56124, Pisa, Pisa, Italy
| | - E Fiore
- Department of Clinical and Experimental Medicine, Section of Endocrinology, University Hospital of Pisa, Via Paradisa 2, 56124, Pisa, Pisa, Italy.
| |
Collapse
|
4
|
Bafor EE, Erwin-Cohen RA, Martin T, Baker C, Kimmel AE, Duverger O, Fenimore JM, Ramba M, Spindel T, Hess MM, Sanford M, Lazarevic V, Benayoun BA, Young HA, Valencia JC. Aberrant CD8 +T cells drive reproductive dysfunction in female mice with elevated IFN-γ levels. Front Immunol 2024; 15:1368572. [PMID: 38698852 PMCID: PMC11064017 DOI: 10.3389/fimmu.2024.1368572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/22/2024] [Indexed: 05/05/2024] Open
Abstract
Introduction Interferon-gamma (IFN-γ) is pivotal in orchestrating immune responses during healthy pregnancy. However, its dysregulation, often due to autoimmunity, infections, or chronic inflammatory conditions, is implicated in adverse reproductive outcomes such as pregnancy failure or infertility. Additionally, the underlying immunological mechanisms remain elusive. Methods Here, we explore the impact of systemic IFN-γ elevation on cytotoxic T cell responses in female reproduction utilizing a systemic lupus-prone mouse model with impaired IFN-γ degradation. Results Our findings reveal that heightened IFN-γ levels triggered the infiltration of CD8+T cells in the pituitary gland and female reproductive tract (FRT), resulting in prolactin deficiency and subsequent infertility. Furthermore, we demonstrate that chronic IFN-γ elevation increases effector memory CD8+T cells in the murine ovary and uterus. Discussion These insights broaden our understanding of the role of elevated IFN-γ in female reproductive dysfunction and suggest CD8+T cells as potential immunotherapeutic targets in female reproductive disorders associated with chronic systemic IFN-γ elevation.
Collapse
Affiliation(s)
- Enitome E. Bafor
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Rebecca A. Erwin-Cohen
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Toni Martin
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Clayton Baker
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
- Molecular and Computational Biology Department, University of Southern California, Dornsife College of Letters, Arts and Sciences, Los Angeles, CA, United States
| | - Adrienne E. Kimmel
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Olivier Duverger
- Craniofacial Anomalies and Regeneration Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - John M. Fenimore
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Meredith Ramba
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Thea Spindel
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Megan M. Hess
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Michael Sanford
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Vanja Lazarevic
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Bérénice A. Benayoun
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
- Molecular and Computational Biology Department, University of Southern California, Dornsife College of Letters, Arts and Sciences, Los Angeles, CA, United States
| | - Howard A. Young
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Julio C. Valencia
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| |
Collapse
|
5
|
Moldenhauer LM, Foyle KL, Wilson JJ, Wong YY, Sharkey DJ, Green ES, Barry SC, Hull ML, Robertson SA. A disrupted FOXP3 transcriptional signature underpins systemic regulatory T cell insufficiency in early pregnancy failure. iScience 2024; 27:108994. [PMID: 38327801 PMCID: PMC10847744 DOI: 10.1016/j.isci.2024.108994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 12/22/2023] [Accepted: 01/18/2024] [Indexed: 02/09/2024] Open
Abstract
Regulatory T (Treg) cell defects are implicated in disorders of embryo implantation and placental development, but the origins of Treg cell dysfunction are unknown. Here, we comprehensively analyzed the phenotypes and transcriptional profile of peripheral blood Treg cells in individuals with early pregnancy failure (EPF). Compared to fertile subjects, EPF subjects had 32% fewer total Treg cells and 54% fewer CD45RA+CCR7+ naive Treg cells among CD4+ T cells, an altered Treg cell phenotype with reduced transcription factor FOXP3 and suppressive marker CTLA4 expression, and lower Treg:Th1 and Treg:Th17 ratios. RNA sequencing demonstrated an aberrant gene expression profile, with upregulation of pro-inflammatory genes including CSF2, IL4, IL17A, IL21, and IFNG in EPF Treg cells. In silico analysis revealed 25% of the Treg cell dysregulated genes are targets of FOXP3. We conclude that EPF is associated with systemic Treg cell defects arising due to disrupted FOXP3 transcriptional control and loss of lineage fidelity.
Collapse
Affiliation(s)
- Lachlan M. Moldenhauer
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Kerrie L. Foyle
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Jasmine J. Wilson
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Ying Y. Wong
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - David J. Sharkey
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Ella S. Green
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Simon C. Barry
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - M. Louise Hull
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Sarah A. Robertson
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
6
|
Qi L, Li Y, Zhang L, Li S, Zhang X, Li W, Qin J, Chen X, Ji Y, Xue Z, Lv B. Immune and oxidative stress disorder in ovulation-dysfunction women revealed by single-cell transcriptome. Front Immunol 2023; 14:1297484. [PMID: 38116006 PMCID: PMC10729704 DOI: 10.3389/fimmu.2023.1297484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/13/2023] [Indexed: 12/21/2023] Open
Abstract
Introduction Ovulation dysfunction is now a widespread cause of infertility around the world. Although the impact of immune cells in human reproduction has been widely investigated, systematic understanding of the changes of the immune atlas under female ovulation remain less understood. Methods Here, we generated single cell transcriptomic profiles of 80,689 PBMCs in three representative statuses of ovulation dysfunction, i.e., polycystic ovary syndrome (PCOS), primary ovarian insufficiency (POI) and menopause (MENO), and identified totally 7 major cell types and 25 subsets of cells. Results Our study revealed distinct cluster distributions of immune cells among individuals of ovulation disorders and health. In patients with ovulation dysfunction, we observed a significant reduction in populations of naïve CD8 T cells and effector memory CD4 T cells, whereas circulating NK cells and regulatory NK cells increased. Discussion Our results highlight the significant contribution of cDC-mediated signaling pathways to the overall inflammatory response within ovulation disorders. Furthermore, our data demonstrated a significant upregulation of oxidative stress in patients with ovulation disorder. Overall, our study gave a deeper insight into the mechanism of PCOS, POI, and menopause, which may contribute to the better diagnosis and treatments of these ovulatory disorder.
Collapse
Affiliation(s)
- Lingbin Qi
- Reproductive Medical Center, Department of Gynecology and Obstetrics, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yumei Li
- Department of Assisted Reproduction, Xiangya Hospital, Central South University, Changsha, China
| | - Lina Zhang
- Reproductive Medical Center, Department of Gynecology and Obstetrics, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shuyue Li
- Reproductive Medical Center, Department of Gynecology and Obstetrics, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xunyi Zhang
- Reproductive Medical Center, Department of Gynecology and Obstetrics, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wanqiong Li
- Reproductive Medical Center, Department of Gynecology and Obstetrics, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiaying Qin
- Reproductive Medical Center, Department of Gynecology and Obstetrics, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xian Chen
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
| | - Yazhong Ji
- Reproductive Medical Center, Department of Gynecology and Obstetrics, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhigang Xue
- Reproductive Medical Center, Department of Gynecology and Obstetrics, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bo Lv
- Reproductive Medical Center, Department of Gynecology and Obstetrics, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Liu D, Guan X, Liu W, Jia Y, Zhou H, Xi C, Zhao M, Fang Y, Wu L, Li K. Identification of transcriptome characteristics of granulosa cells and the possible role of UBE2C in the pathogenesis of premature ovarian insufficiency. J Ovarian Res 2023; 16:203. [PMID: 37848988 PMCID: PMC10580542 DOI: 10.1186/s13048-023-01266-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 08/17/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is an important cause of infertility characterized by the functional decline of the ovary. Granulosa cells (GCs) around oocytes are critical for folliculogenesis, and GC dysfunction is one of the important etiologies of POI. The aim of this study was to explore the potential biomarkers of POI by identifying hub genes and analyze the correlation of biomarkers with immune infiltration in POI using RNA profiling and bioinformatics analysis. METHODS RNA sequencing was performed on GCs from biochemical POI (bPOI) patients and controls. Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were used to explore the candidate genes. qRT‒PCR was performed to verify the expression of hub genes. Western blot, Cell Counting Kit-8, 5-ethynyl-2'-deoxyuridine (EdU) assays, TUNEL (TdT-mediated dUTP Nick-End Labeling) and flow cytometry analysis were used to validate the possible role of ubiquitin-conjugating enzyme 2C (UBE2C) in POI. CIBERSORT was adopted to explore immune cell infiltration and the correlation between UBE2C and immune cells in bPOI. RESULTS Through analysis of differentially expressed genes (DEGs) and WGCNA, we obtained 143 candidate genes. After construction of the protein‒protein interaction (PPI) network and analysis with Cytoscape, 10 hub genes, including UBE2C, PBK, BUB1, CDC20, NUSAP1, CENPA, CCNB2, TOP2A, AURKB, and FOXM1, were identified and verified by qRT‒PCR. Subsequently, UBE2C was chosen as a possible biomarker of POI because knockdown of UBE2C could inhibit the proliferation and promote the apoptosis of GCs. Immune infiltration analysis indicated that monocytes and M1 macrophages may be associated with the pathogenesis of POI. In addition, UBE2C was negatively correlated with monocytes and M1 macrophages in POI. CONCLUSIONS This study identified a hub gene in GCs that might be important in the pathogenesis of POI and revealed the key role of UBE2C in driving POI. Immune infiltration may be highly related with the onset and etiology of POI.
Collapse
Affiliation(s)
- Dan Liu
- Centre for Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, 2699 Gaoke West Road, Pudong District, Shanghai, 201204, China
| | - Xiaohong Guan
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Wenqiang Liu
- Centre for Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, 2699 Gaoke West Road, Pudong District, Shanghai, 201204, China
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yanping Jia
- Centre for Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, 2699 Gaoke West Road, Pudong District, Shanghai, 201204, China
| | - Hong Zhou
- Centre for Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, 2699 Gaoke West Road, Pudong District, Shanghai, 201204, China
| | - Chenxiang Xi
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Mei Zhao
- Centre for Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, 2699 Gaoke West Road, Pudong District, Shanghai, 201204, China
| | - Yuan Fang
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Li Wu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Kunming Li
- Centre for Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, 2699 Gaoke West Road, Pudong District, Shanghai, 201204, China.
| |
Collapse
|
8
|
Kong D, Cho H, Hwang S, Choi E, Lee AY, Choi EK, Kim YB, Kim HJ, Hong S. Bioinformatics and integrated pharmacology network to identify the therapeutic targets and potential molecular mechanism of alpha-lipoic acid on primary ovarian insufficiency. J Cell Biochem 2023; 124:1557-1572. [PMID: 37660319 DOI: 10.1002/jcb.30464] [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: 10/15/2022] [Revised: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 09/05/2023]
Abstract
Women experiencing primary ovarian insufficiency (POI) are more likely to experience infertility, and its incidence is increasing worldwide annually. Recently, the role of alpha-lipoic acid (ALA) in the treatment of POI has been reported. However, details of the potential pharmacological targets and related molecular pathways of ALA remain unclear and need to be elucidated. Thus, this study aims to elucidate the potential therapeutic target and related molecular mechanism of ALA on POI. First, the potential targets of POI and ALA-related targets were downloaded from online public databases. Subsequently, the overlapped target genes between POI and ALA were acquired, and gene ontology (GO), Kyoto encyclopedia of genes and genomes (KEGG) analysis, protein-protein interaction (PPI) networks were performed and constructed. Finally, molecular docking was performed to verify protein-to-protein effect. A total of 152 potential therapeutic targets were identified. The biological processes of the intersecting targets were mainly involved in the cellular response to peptides, response to xenobiotic stimuli, and response to peptide hormones. The highly enriched pathways were the cAMP, PI3K/AKT, estrogen, progesterone mediated oocyte maturation, and apoptosis signaling pathways. The top 10 hub targets for ALA in the treatment of POI were STAT3, STAT1, CASP3, MTOR, PTGS2, CASP8, HSP90AA1, PIK3CA, MAPK1, and ESR1. The binding between ALA and all top hub targets were verified using the molecular docking analysis. In summary, using the systematic integrated pharmacology network and bioinformatics analysis, this study illustrated that ALA participates in the treatment of POI via multiple targets and multiple pathways mechanisms.
Collapse
Affiliation(s)
- Deqi Kong
- Department of Biomedical Science, Graduate School of Medicine, Korea University, Seoul, Korea
| | - Heeryun Cho
- Department of Biomedical Science, Graduate School of Medicine, Korea University, Seoul, Korea
| | - Soowon Hwang
- Department of Biomedical Science, Graduate School of Medicine, Korea University, Seoul, Korea
| | - Eunsaem Choi
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Korea
| | - Ah-Young Lee
- Central Research Institute, Designed Cells Co., Ltd., Cheongju, Korea
| | - Ehn-Kyoung Choi
- Central Research Institute, Designed Cells Co., Ltd., Cheongju, Korea
| | - Yun-Bae Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju, Korea
| | - Hai-Joong Kim
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Korea
| | - Sooncheol Hong
- Department of Biomedical Science, Graduate School of Medicine, Korea University, Seoul, Korea
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Korea
- Institute of Stem Cell Research, Korea University, Seoul, Korea
| |
Collapse
|
9
|
Geng X, He Z, Bao Z, Di W, Gu Z. Aberrant HPO Axis Alterations and Autoimmune Abnormalities in PCOS Patients with DOR: A Retrospective Analysis. J Clin Med 2023; 12:5212. [PMID: 37629254 PMCID: PMC10455465 DOI: 10.3390/jcm12165212] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/20/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND There is a group of polycystic ovary syndrome (PCOS) patients in clinic who have diminished ovarian reserve (DOR) in combination. This study was designed to evaluate the differences in glucolipid metabolism, hypothalamic-pituitary-ovarian (HPO) axis-related parameters, and autoimmune antibodies in PCOS patients with and without DOR. METHODS A total of 2307 PCOS patients, including 1757 patients with PCOS alone and 550 patients who have both PCOS and DOR, were enrolled in this retrospective study. Parameters of glucolipid metabolism, HPO axis-related parameters, and autoimmune antibodies were measured and analyzed. RESULTS The prevalence of DOR among all patients with PCOS was 23.84%. Many HPO axis-related parameters, such as follicle-stimulating hormone (FSH), luteinizing hormone (LH), estradiol (E2), and prolactin (PRL) were significantly different in PCOS with DOR compared with PCOS without DOR. The FSH levels were positively correlated with LH, testosterone (T), and androstenedione (AD) levels, but had no association with glucolipid metabolism after adjusting for body mass index (BMI). Moreover, anti-ovarian antibody (AOAb) and anti-21-OH antibody (21-OHAb) levels were significantly elevated in PCOS patients with DOR. CONCLUSIONS PCOS patients with DOR showed more chaotic HPO axis hormone levels and elevated autoimmune antibodies, suggesting that autoimmune factors may be the cause of DOR in women with PCOS.
Collapse
Affiliation(s)
- Xueying Geng
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Zhihong He
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Zhouzhou Bao
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Wen Di
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
- Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Zhuowei Gu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
- Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| |
Collapse
|
10
|
Wang J, Yin T, Liu S. Dysregulation of immune response in PCOS organ system. Front Immunol 2023; 14:1169232. [PMID: 37215125 PMCID: PMC10196194 DOI: 10.3389/fimmu.2023.1169232] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common reproductive endocrine disorder affecting women, which can lead to infertility. Infertility, obesity, hirsutism, acne, and irregular menstruation are just a few of the issues that PCOS can be linked to. PCOS has a complicated pathophysiology and a range of clinical symptoms. Chronic low-grade inflammation is one of the features of PCOS. The inflammatory environment involves immune and metabolic disturbances. Numerous organ systems across the body, in addition to the female reproductive system, have been affected by the pathogenic role of immunological dysregulation in PCOS in recent years. Insulin resistance and hyperandrogenism are associated with immune cell dysfunction and cytokine imbalance. More importantly, obesity is also involved in immune dysfunction in PCOS, leading to an inflammatory environment in women with PCOS. Hormone, obesity, and metabolic interactions contribute to the pathogenesis of PCOS. Hormone imbalance may also contribute to the development of autoimmune diseases. The aim of this review is to summarize the pathophysiological role of immune dysregulation in various organ systems of PCOS patients and provide new ideas for systemic treatment of PCOS in the future.
Collapse
Affiliation(s)
- Jingxuan Wang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tailang Yin
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Su Liu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| |
Collapse
|
11
|
Stem Cell-Based Therapeutic Strategies for Premature Ovarian Insufficiency and Infertility: A Focus on Aging. Cells 2022; 11:cells11233713. [PMID: 36496972 PMCID: PMC9738202 DOI: 10.3390/cells11233713] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/14/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
Reproductive aging is on the rise globally and inseparable from the entire aging process. An extreme form of reproductive aging is premature ovarian insufficiency (POI), which to date has mostly been of idiopathic etiology, thus hampering further clinical applications and associated with enormous socioeconomic and personal costs. In the field of reproduction, the important functional role of inflammation-induced ovarian deterioration and therapeutic strategies to prevent ovarian aging and increase its function are current research hotspots. This review discusses the general pathophysiology and relative causes of POI and comprehensively describes the association between the aging features of POI and infertility. Next, various preclinical studies of stem cell therapies with potential for POI treatment and their molecular mechanisms are described, with particular emphasis on the use of human induced pluripotent stem cell (hiPSC) technology in the current scenario. Finally, the progress made in the development of hiPSC technology as a POI research tool for engineering more mature and functional organoids suitable as an alternative therapy to restore infertility provides new insights into therapeutic vulnerability, and perspectives on this exciting research on stem cells and the derived exosomes towards more effective POI diagnosis and treatment are also discussed.
Collapse
|
12
|
Wang Y, Liu L, Tan C, Meng G, Meng L, Nie H, Du J, Lu GX, Lin G, He WB, Tan YQ. Novel MEIOB variants cause primary ovarian insufficiency and non-obstructive azoospermia. Front Genet 2022; 13:936264. [PMID: 35991565 PMCID: PMC9388730 DOI: 10.3389/fgene.2022.936264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Infertility is a global health concern. MEIOB has been found to be associated with premature ovarian insufficiency (POI) and non-obstructive azoospermia (NOA), but its variants have not been reported in Chinese patients. The aim of this study was to identify the genetic aetiology of POI or NOA in three Han Chinese families. Methods: Whole-exome sequencing (WES) was used to identify candidate pathogenic variants in three consanguineous Chinese infertile families with POI or NOA. Sanger sequencing was performed to validate these variants in the proband of family I and her affected family members. In vitro functional analyses were performed to confirm the effects of these variants. Results: Two novel homozygous frameshift variants (c.258_259del and c.1072_1073del) and one novel homozygous nonsense variant (c.814C > T) in the MEIOB gene were identified in three consanguineous Han Chinese families. In vitro functional analyses revealed that these variants produced truncated proteins and affected their function. Conclusion: We identified three novel MEIOB loss-of-function variants in local Chinese patients for the first time and confirmed their pathogenicity using in vitro functional analyses. These results extend the mutation spectrum of the MEIOB gene and have important significance for genetic counselling in these families.
Collapse
Affiliation(s)
- Yurong Wang
- Hunan Guangxiu Hospital, Hunan Normal University, Changsha, China
| | - Ling Liu
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
| | - Chen Tan
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
| | - Guiquan Meng
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
| | - Lanlan Meng
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- National Engineering and Research Center of Human Stem Cells, Changsha, China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, China
| | - Hongchuan Nie
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Juan Du
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, China
| | - Guang-Xiu Lu
- Hunan Guangxiu Hospital, Hunan Normal University, Changsha, China
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- National Engineering and Research Center of Human Stem Cells, Changsha, China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, China
| | - Wen-Bin He
- Hunan Guangxiu Hospital, Hunan Normal University, Changsha, China
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- National Engineering and Research Center of Human Stem Cells, Changsha, China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, China
| | - Yue-Qiu Tan
- Hunan Guangxiu Hospital, Hunan Normal University, Changsha, China
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- National Engineering and Research Center of Human Stem Cells, Changsha, China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, China
| |
Collapse
|
13
|
Garzia E, Galiano V, Guarnaccia L, Marfia G, Murru G, Guermandi E, Riparini J, Sulpizio P, Marconi AM. Basal serum level of Δ4-androstenedione reflects the ovaries' ability to respond to stimulation in IVF cycles: setting up a new reliable index of both ovarian reserve and response. J Assist Reprod Genet 2022; 39:1917-1926. [PMID: 35759063 PMCID: PMC9428103 DOI: 10.1007/s10815-022-02546-5] [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: 01/24/2022] [Accepted: 06/09/2022] [Indexed: 01/19/2023] Open
Abstract
PURPOSE Adequate androgen levels are necessary for regular follicular growth, progression beyond the pre-antral stage, and prevention of follicular atresia. The main purpose of this study was to investigate whether baseline androgen levels had a predictive value on stimulation outcomes in IVF cycles. The secondary purpose was to compare the possible predictive value of androgens with that of already known markers. METHODS The study included 91 infertile patients aged 30-45 years awaiting the first IVF cycle. All women underwent the same stimulation protocol and the same starting dose of recombinant FSH. As stimulation outcomes, the number of follicles recruited, estradiol and progesterone levels on the day of trigger, the total dose of gonadotropins administered, and the number of oocytes collected were recorded. Multiple linear regression and multivariate logistic regression were used to evaluate the significant predictive value of the variables for response to controlled ovarian stimulation (COS). By studying the reliability of different markers, an attempt was made to develop a single index with the highest predictive value. RESULTS Pearson's correlation revealed a statistically significant inverse correlation between oocytes collected and age (r = - 0.333, p < 0.001) and a positive correlation with AMH (anti-müllerian hormone) (r = 0.360, p < 0.001), antral follicle count (AFC) (r = 0.639, p < 0.001), and androstenedione (Δ4-A) (r = 0.359, p < 0.001). No significant correlation was reported with FSH (r = - 0.133, p = 0.207) and total testosterone (r = 0.180, p = 0.088). In COS good responders, the G-index (= AMH ng/mL*AFC/Δ4-A ng/dL) revealed a significantly higher level (p < 0.001) than AMH, AFC, and Δ4-A alone. CONCLUSION Baseline serum Δ4-A, presumably crucial for ensuring a regular follicular growth, is a reliable marker of ovarian response to stimulation. Since the ovarian capacity to respond to gonadotropins does not depend exclusively on the presence of follicles, we suggest a new index, the G-index, able to contemplate both the ovarian reserve and the Δ4-A level.
Collapse
Affiliation(s)
- Emanuele Garzia
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 via di Rudinì, 8, Milan, Italy
- Istituto Di Medicina Aerospaziale “A. Mosso,” Aeronautica Militare, Milan, Italy
| | - Valentina Galiano
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 via di Rudinì, 8, Milan, Italy
- Gynecology and Obstetrics Unit, San Carlo Borromeo Hospital, ASST Santi Paolo e Carlo, Milan, Italy
| | - Laura Guarnaccia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Aldo Ravelli Research Center, Milan, Italy
| | - Giovanni Marfia
- Istituto Di Medicina Aerospaziale “A. Mosso,” Aeronautica Militare, Milan, Italy
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Aldo Ravelli Research Center, Milan, Italy
| | - Giulia Murru
- Gynecology and Obstetrics Unit, ASST Rhodense, Ospedale G. Salvini, Garbagnate Milanese, Italy
| | - Ellade Guermandi
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 via di Rudinì, 8, Milan, Italy
| | - Jennifer Riparini
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 via di Rudinì, 8, Milan, Italy
| | - Patrizia Sulpizio
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 via di Rudinì, 8, Milan, Italy
| | - Anna Maria Marconi
- Gynecology and Obstetrics Unit, Department of Mother and Child, Department of Health Sciences, San Paolo Hospital Medical School, University of Milan, Milan, Italy
| |
Collapse
|
14
|
Sun IH, Gillis-Buck E, Mackenzie TC, Gardner JM. Thymic and extrathymic Aire-expressing cells in maternal-fetal tolerance. Immunol Rev 2022; 308:93-104. [PMID: 35535447 DOI: 10.1111/imr.13082] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/04/2022] [Indexed: 12/16/2022]
Abstract
Healthy pregnancy requires maternal immune tolerance to both fetal and placental tissues which contain a range of self- and non-self-antigens. While many of the components and mechanisms of maternal-fetal tolerance have been investigated in detail and previously and thoroughly reviewed (Erlebacher A. Annu Rev Immunol. 2013;31:387-411), the role of autoimmune regulator (Aire), a critical regulator of central tolerance expressed by medullary thymic epithelial cells (mTECs), has been less explored. Aire is known to facilitate the expression of a range of otherwise tissue-specific antigens (TSAs) in mTECs, and here we highlight recent work showing a role for mTEC-mediated thymic selection in maintaining maternal-fetal tolerance. Recently, however, our group and others have identified additional populations of extrathymic Aire-expressing cells (eTACs) in the secondary lymphoid organs. These hematopoietic antigen-presenting cells possess the ability to induce functional inactivation and/or deletion of cognate T cells, and deletion of maternal eTACs during pregnancy increases T-cell activation in the lymph nodes and lymphocytic infiltration of the uterus, leading to pregnancy complications including intrauterine growth restriction (IUGR) and fetal resorption. In this review, we briefly summarize findings related to essential Aire biology, discuss the known roles of Aire-deficiency related to pregnancy complications and infertility, review the newly discovered role for eTACs in the maintenance of maternal-fetal tolerance-as well as recent work defining eTACs at the single-cell level-and postulate potential mechanisms by which eTACs may regulate this process.
Collapse
Affiliation(s)
- Im-Hong Sun
- Department of Surgery, University of California, San Francisco, California, USA.,Diabetes Center, University of California, San Francisco, California, USA
| | - Eva Gillis-Buck
- Department of Surgery, University of California, San Francisco, California, USA
| | - Tippi C Mackenzie
- Department of Surgery, University of California, San Francisco, California, USA.,Center for Maternal-Fetal Precision Medicine, University of California, San Francisco, California, USA
| | - James M Gardner
- Department of Surgery, University of California, San Francisco, California, USA.,Diabetes Center, University of California, San Francisco, California, USA
| |
Collapse
|
15
|
Altuntaş SÇ, Güneş M. Investigation of the Relationship Between Autoimmune and Nodular Goiter in Patients with Euthyroid Polycystic Ovary Syndrome and Their Phenotypes. Horm Metab Res 2022; 54:396-406. [PMID: 35419775 DOI: 10.1055/a-1825-0316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine disorder that frequently affects women of reproductive age. In PCOS, the incidence of thyroid diseases has increased in addition to reproductive and metabolic problems. To compare thyroid nodule, volume, autoimmunity, and thyroid function tests of euthyroid PCOS and its phenotypes. The files of 178 patients with PCOS aged 18-45 years and 92 patients with no disease who were matched for body mass index were retrospectively scanned. Women with PCOS were divided into four phenotypes, ABCD. Anti-TPO titer and prevalence, fT3, and thyroid volume were higher in the PCOS group compared with the control group in terms of anti-Tg levels, presence of nodules, and the number of nodules. There was no statistical difference between the PCOS group and the healthy controls. The number of nodules of 1 cm and above was found to be higher only in patients with PCOS compared with the control group. When the phenotypes were examined, thyroid dysfunction features were found in phenotype A, which was the most prominent. Thyroid autoimmunity, thyroid volume, and the number of nodules larger than 1 cm increased in patients with PCOS compared with controls. This situation is thought to be caused by the reproductive and metabolic properties of PCOS because thyroid dysfunction was detected more in phenotype A, which is called the full phenotype. Therefore, all patients with PCOS, especially phenotype A, should be evaluated for the presence of nodules with autoimmunity using USG, even if there are no symptoms, and thyroid functions.
Collapse
Affiliation(s)
- Seher Çetinkaya Altuntaş
- Division of Endocrinology, Bursa Yuksek Ihtisas Education and Training Hospital, University of Health Sciences, Bursa, Turkey
| | - Mutlu Güneş
- Department of Endocrinology and Metabolism, Bursa Yuksek Ihtisas Education and Training Hospital, University of Health Sciences, Bursa, Turkey
| |
Collapse
|
16
|
P450 Side-Chain Cleavage Enzyme (P450-SCC) Is an Ovarian Autoantigen in a Mouse Model for Autoimmune Oophoritis. Reprod Sci 2022; 29:2391-2400. [PMID: 35585293 DOI: 10.1007/s43032-022-00970-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/06/2022] [Indexed: 10/18/2022]
Abstract
Steroid-producing cells contain key cytochrome P450 enzymes, such as side-chain cleavage (P450-SCC) and 17α-hydroxylase (17α-OH). They are required for steroid hormone synthesis and considered antigens associated with Addison's disease and autoimmune primary ovarian insufficiency (POI). We studied an animal model for human autoimmune POI in mice with autoimmune oophoritis induced by neonatal thymectomy performed at day 3 (TX3). We previously identified an oocyte-specific protein as a major antigen inciting autoimmune oophoritis in mice. In this study, we characterized ovarian steroid-producing cell antigens. Using indirect immunofluorescence staining, we tested immune reactions in mouse ovarian and adrenal tissue sections with sera from TX3 female mice. More than half of the TX3 mice (8 of 15) produced antibodies reacting with both ovarian and adrenal steroid-producing cells, including some that reacted to oocytes as well. We produced recombinant proteins for the three key steroidogenic enzymes 17α-OH, P450-SSC, and 3β-hydroxysteroid dehydrogenase (3β-HSD) and tested their immune reactions with individual mouse sera. By immunoblotting, all mouse sera that reacted with the steroid-producing cells (n = 8) were shown to react with the P450-SCC, but not with the 17α-OH or 3β-HSD recombinant proteins. The sham-operated mouse sera and TX3 mouse sera negative for steroid-producing cells did not react with the P450-SCC recombinant protein. Our findings indicate that the P450-SCC is a specific and unique major antigen within the ovarian steroid-producing cells. Given their similarity of predicted antigenicity, we assume that P450-SCC acts in human autoimmune POI as it does in mouse autoimmune oophoritis.
Collapse
|
17
|
Personalized Nutrition in the Management of Female Infertility: New Insights on Chronic Low-Grade Inflammation. Nutrients 2022; 14:nu14091918. [PMID: 35565885 PMCID: PMC9105997 DOI: 10.3390/nu14091918] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/19/2022] [Accepted: 04/29/2022] [Indexed: 02/01/2023] Open
Abstract
Increasing evidence on the significance of nutrition in reproduction is emerging from both animal and human studies, suggesting a mutual association between nutrition and female fertility. Different “fertile” dietary patterns have been studied; however, in humans, conflicting results or weak correlations are often reported, probably because of the individual variations in genome, proteome, metabolome, and microbiome and the extent of exposure to different environmental conditions. In this scenario, “precision nutrition”, namely personalized dietary patterns based on deep phenotyping and on metabolomics, microbiome, and nutrigenetics of each case, might be more efficient for infertile patients than applying a generic nutritional approach. In this review, we report on new insights into the nutritional management of infertile patients, discussing the main nutrigenetic, nutrigenomic, and microbiomic aspects that should be investigated to achieve effective personalized nutritional interventions. Specifically, we will focus on the management of low-grade chronic inflammation, which is associated with several infertility-related diseases.
Collapse
|
18
|
Zammit NW, McDowell J, Warren J, Muskovic W, Gamble J, Shi YC, Kaczorowski D, Chan CL, Powell J, Ormandy C, Brown D, Oakes SR, Grey ST. TNFAIP3 Reduction-of-Function Drives Female Infertility and CNS Inflammation. Front Immunol 2022; 13:811525. [PMID: 35464428 PMCID: PMC9027572 DOI: 10.3389/fimmu.2022.811525] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/21/2022] [Indexed: 11/17/2022] Open
Abstract
Women with autoimmune and inflammatory aetiologies can exhibit reduced fecundity. TNFAIP3 is a master negative regulator of inflammation, and has been linked to many inflammatory conditions by genome wide associations studies, however its role in fertility remains unknown. Here we show that mice harbouring a mild Tnfaip3 reduction-of-function coding variant (Tnfaip3I325N) that reduces the threshold for inflammatory NF-κB activation, exhibit reduced fecundity. Sub-fertility in Tnfaip3I325N mice is associated with irregular estrous cycling, low numbers of ovarian secondary follicles, impaired mammary gland development and insulin resistance. These pathological features are associated with infertility in human subjects. Transplantation of Tnfaip3I325N ovaries, mammary glands or pancreatic islets into wild-type recipients rescued estrous cycling, mammary branching and hyperinsulinemia respectively, pointing towards a cell-extrinsic hormonal mechanism. Examination of hypothalamic brain sections revealed increased levels of microglial activation with reduced levels of luteinizing hormone. TNFAIP3 coding variants may offer one contributing mechanism for the cause of sub-fertility observed across otherwise healthy populations as well as for the wide variety of auto-inflammatory conditions to which TNFAIP3 is associated. Further, TNFAIP3 represents a molecular mechanism that links heightened immunity with neuronal inflammatory homeostasis. These data also highlight that tuning-up immunity with TNFAIP3 comes with the potentially evolutionary significant trade-off of reduced fertility.
Collapse
Affiliation(s)
- Nathan W. Zammit
- Immunity and Inflammation Theme, Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Joseph McDowell
- Immunity and Inflammation Theme, Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Joanna Warren
- Immunity and Inflammation Theme, Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Walter Muskovic
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Joanne Gamble
- Centre for NSW Health Pathology, Institute of Clinical Pathology And Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - Yan-Chuan Shi
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Dominik Kaczorowski
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Chia-Ling Chan
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Joseph Powell
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Chris Ormandy
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Translation Science Pillar, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - David Brown
- Centre for NSW Health Pathology, Institute of Clinical Pathology And Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - Samantha R. Oakes
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Translation Science Pillar, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Shane T. Grey
- Immunity and Inflammation Theme, Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Translation Science Pillar, Garvan Institute of Medical Research, Sydney, NSW, Australia
| |
Collapse
|
19
|
Zhou P, Yao Q, Zhao Q, Yang L, Yu Y, Xie J, Feng C, Zhou L, Jin M. IVF/ICSI outcomes of euthyroid infertile women with thyroid autoimmunity: does treatment with aspirin plus prednisone matter? BMC Pregnancy Childbirth 2022; 22:263. [PMID: 35351031 PMCID: PMC8966173 DOI: 10.1186/s12884-022-04532-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/02/2022] [Indexed: 11/23/2022] Open
Abstract
Background Thyroid autoimmunity (TAI) has been demonstrated to be associated with adverse pregnancy including recurrent miscarriage, unexplained infertility, and implantation failure. To settle with the fertility problem, prescribing aspirin combined with prednisone (P + A) to women positive for anti-thyroid antibodies is frequent in clinical practice, but the underlying effect remains controversial. Methods A multicenter, retrospective study was conducted in three reproductive centers from 2017 to 2020. A total of 494 euthyroid infertile women were recruited who were positive for anti-thyroperoxidase and/or thyroglobulin antibodies (TPOAb and TgAb, respectively) with thyroid-stimulating hormone (TSH) levels ranging 0.35-4.0mIU/L and underwent their first in vitro fertilization and embryo transfer (IVF-ET) cycle. Ultimately, 346 women were included of which 150 women were treated with prednisone (10 mg/d) and aspirin (100 mg/d). The remaining 196 women were untreated (control group). Treatment started on the day of embryo transfer and continued until clinical pregnancy was determined. Results The clinical pregnancy rate was 57.5% vs. 63.5% in the control and treated groups (P = 0.414) for first fresh embryo transfer cycles and 57.8% vs. 61.8% for frozen-thawed embryo transfer cycles (P = 0.606). In addition, the live birth rate for the fresh embryo transfer was 49.6% vs. 47.3% in the control and treated groups (P = 0.762). Logistic regression revealed that aspirin plus prednisone did not improve the clinical pregnancy rate or miscarriage rate. Furthermore, it was observed that low free triiodothyronine (FT3) was associated with high miscarriage rates. Conclusions Utilizing an adjuvant treatment of P + A after the embryo transfer may not be necessary in euthyroid women with thyroid autoimmunity undergoing their first IVF-ET, regardless of the embryo type (fresh or frozen). Supplementary Information The online version contains supplementary material available at 10.1186/s12884-022-04532-2.
Collapse
Affiliation(s)
- Ping Zhou
- Second Affiliated Hospital, School of Medicine, Zhejiang University, 310052, Hangzhou, Zhejiang Province, P.R. China
| | - Qiuping Yao
- Second Affiliated Hospital, School of Medicine, Zhejiang University, 310052, Hangzhou, Zhejiang Province, P.R. China.,Jiaxing Maternity and Child Health Care Hospital, Zhejiang Province, 314051, Jiaxing, P.R. China
| | - Qiaohang Zhao
- Second Affiliated Hospital, School of Medicine, Zhejiang University, 310052, Hangzhou, Zhejiang Province, P.R. China
| | - Lihua Yang
- Jinhua People's Hospital, 321000, Jinhua, Zhejiang Province, P.R. China
| | - Ya Yu
- Second Affiliated Hospital, School of Medicine, Zhejiang University, 310052, Hangzhou, Zhejiang Province, P.R. China
| | - Jilai Xie
- Second Affiliated Hospital, School of Medicine, Zhejiang University, 310052, Hangzhou, Zhejiang Province, P.R. China
| | - Chun Feng
- Second Affiliated Hospital, School of Medicine, Zhejiang University, 310052, Hangzhou, Zhejiang Province, P.R. China
| | - Liming Zhou
- Ningbo Women and Children's Hospital, 315000, Ningbo, Zhejiang Province, P.R. China.
| | - Min Jin
- Second Affiliated Hospital, School of Medicine, Zhejiang University, 310052, Hangzhou, Zhejiang Province, P.R. China.
| |
Collapse
|
20
|
Venkatesh SS, Ferreira T, Benonisdottir S, Rahmioglu N, Becker CM, Granne I, Zondervan KT, Holmes MV, Lindgren CM, Wittemans LBL. Obesity and risk of female reproductive conditions: A Mendelian randomisation study. PLoS Med 2022; 19:e1003679. [PMID: 35104295 PMCID: PMC8806071 DOI: 10.1371/journal.pmed.1003679] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 12/07/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Obesity is observationally associated with altered risk of many female reproductive conditions. These include polycystic ovary syndrome (PCOS), abnormal uterine bleeding, endometriosis, infertility, and pregnancy-related disorders. However, the roles and mechanisms of obesity in the aetiology of reproductive disorders remain unclear. Thus, we aimed to estimate observational and genetically predicted causal associations between obesity, metabolic hormones, and female reproductive disorders. METHODS AND FINDINGS Logistic regression, generalised additive models, and Mendelian randomisation (MR) (2-sample, non-linear, and multivariable) were applied to obesity and reproductive disease data on up to 257,193 women of European ancestry in UK Biobank and publicly available genome-wide association studies (GWASs). Body mass index (BMI), waist-to-hip ratio (WHR), and WHR adjusted for BMI were observationally (odds ratios [ORs] = 1.02-1.87 per 1-SD increase in obesity trait) and genetically (ORs = 1.06-2.09) associated with uterine fibroids (UF), PCOS, heavy menstrual bleeding (HMB), and pre-eclampsia. Genetically predicted visceral adipose tissue (VAT) mass was associated with the development of HMB (OR [95% CI] per 1-kg increase in predicted VAT mass = 1.32 [1.06-1.64], P = 0.0130), PCOS (OR [95% CI] = 1.15 [1.08-1.23], P = 3.24 × 10-05), and pre-eclampsia (OR [95% CI] = 3.08 [1.98-4.79], P = 6.65 × 10-07). Increased waist circumference posed a higher genetic risk (ORs = 1.16-1.93) for the development of these disorders and UF than did increased hip circumference (ORs = 1.06-1.10). Leptin, fasting insulin, and insulin resistance each mediated between 20% and 50% of the total genetically predicted association of obesity with pre-eclampsia. Reproductive conditions clustered based on shared genetic components of their aetiological relationships with obesity. This study was limited in power by the low prevalence of female reproductive conditions among women in the UK Biobank, with little information on pre-diagnostic anthropometric traits, and by the susceptibility of MR estimates to genetic pleiotropy. CONCLUSIONS We found that common indices of overall and central obesity were associated with increased risks of reproductive disorders to heterogenous extents in a systematic, large-scale genetics-based analysis of the aetiological relationships between obesity and female reproductive conditions. Our results suggest the utility of exploring the mechanisms mediating the causal associations of overweight and obesity with gynaecological health to identify targets for disease prevention and treatment.
Collapse
Affiliation(s)
- Samvida S. Venkatesh
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Teresa Ferreira
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Women’s and Reproductive Health, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Stefania Benonisdottir
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| | - Nilufer Rahmioglu
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Women’s and Reproductive Health, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Christian M. Becker
- Nuffield Department of Women’s and Reproductive Health, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Ingrid Granne
- Nuffield Department of Women’s and Reproductive Health, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Krina T. Zondervan
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Women’s and Reproductive Health, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Michael V. Holmes
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, United Kingdom
| | - Cecilia M. Lindgren
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Women’s and Reproductive Health, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Laura B. L. Wittemans
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Women’s and Reproductive Health, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
21
|
|
22
|
Liu M, Wang D, Zhu L, Yin J, Ji X, Zhong Y, Gao Y, Zhang J, Liu Y, Zhang R, Chen H. Association of thyroid peroxidase antibodies with the rate of first-trimester miscarriage in euthyroid women with unexplained recurrent spontaneous abortion. Front Endocrinol (Lausanne) 2022; 13:966565. [PMID: 36120428 PMCID: PMC9471195 DOI: 10.3389/fendo.2022.966565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 08/12/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Unexplained recurrent spontaneous abortion is a serious reproductive problem of unknown etiology. Thyroid peroxidase antibodies (TPO-Ab) may be associated with pregnancy outcomes in unexplained recurrent spontaneous abortion with normal thyroid function. OBJECTIVE This study aimed to investigate the relationship between TPO-Ab and the first trimester miscarriage rate/live birth rate in women of unexplained recurrent spontaneous abortion with normal thyroid function. METHODS We retrospectively analyzed the clinical data of 297 women who met our strict inclusion criteria, comparing the first trimester miscarriage rate/live birth rate between the TPO-Ab positive and TPO-Ab negative groups. For the same purpose, we also performed subgroup analysis. RESULTS Of the included women, 76 (25.6%) were TPO-Ab positive, and 221 (74.4%) were negative. First trimester miscarriage rate differed between the two groups (36.8% vs 24.0%, RR = 1.54, 95% CI: 1.05-2.24, P = 0.030). In the younger subgroup (<35 years) and the primary RSA subgroup, First trimester miscarriage rate was also higher in the TPO-Ab positive group (33.3% vs 19.0%, RR = 1.75, 95% CI: 1.07-2.87, P = 0.030; 36.5% vs 21.7%, RR = 1.69, 95% CI: 1.10-2.58, P = 0.020). While the live birth rate was lower in women with TPO-Ab positive, the difference did not reach statistical significance, even in the subgroup analysis. CONCLUSION Our results suggest that TPO-Ab is associated with first trimester miscarriage rate in euthyroid women with unexplained recurrent spontaneous abortion.
Collapse
Affiliation(s)
- Meilan Liu
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dongyan Wang
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Center for Reproductive Genetics and Reproductive Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liqiong Zhu
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianlan Yin
- Department of Obstetrics and Gynecology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiaohui Ji
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yilei Zhong
- Department of Obstetrics and Gynecology, YueBei People’s Hospital, Shaoguan, China
| | - Yuan Gao
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianping Zhang
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yukun Liu
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Hui Chen, ; Rui Zhang, ; Yukun Liu,
| | - Rui Zhang
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Hui Chen, ; Rui Zhang, ; Yukun Liu,
| | - Hui Chen
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Hui Chen, ; Rui Zhang, ; Yukun Liu,
| |
Collapse
|
23
|
Medenica S, Abazovic D, Ljubić A, Vukovic J, Begovic A, Cucinella G, Zaami S, Gullo G. The Role of Cell and Gene Therapies in the Treatment of Infertility in Patients with Thyroid Autoimmunity. Int J Endocrinol 2022; 2022:4842316. [PMID: 36081621 PMCID: PMC9448571 DOI: 10.1155/2022/4842316] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 08/03/2022] [Indexed: 12/02/2022] Open
Abstract
There is a rising incidence of infertility worldwide, and many couples experience difficulties conceiving nowadays. Thyroid autoimmunity (TAI) is recognized as one of the major female infertility causes related to a diminished ovarian reserve and potentially impaired oocyte maturation and embryo development, causing adverse pregnancy outcomes. Growing evidence has highlighted its impact on spontaneously achieved pregnancy and pregnancy achieved by in vitro fertilization. Despite the influence of thyroid hormones on the male reproductive system, there is insufficient data on the association between TAI and male infertility. In past years, significant progress has been achieved in cell and gene therapies as emerging treatment options for infertility. Cell therapies utilize living cells to restore healthy tissue microenvironment and homeostasis and usually involve platelet-rich plasma and various stem cells. Using stem cells as therapeutic agents has many advantages, including simple sampling, abundant sources, poor immunogenicity, and elimination of ethical concerns. Mesenchymal Stem Cells (MSCs) represent a heterogeneous fraction of self-renewal, multipotent non-hematopoietic stem cells that display profound immunomodulatory and immunosuppressive features and promising therapeutic effects. Infertility has a genetic component in about half of all cases, although most of its genetic causes are still unknown. Hence, it is essential to identify genes involved in meiosis, DNA repair, ovarian development, steroidogenesis, and folliculogenesis, as well as those involved in spermatogenesis in order to develop potential gene therapies for infertility. Despite advances in therapy approaches such as biological agents, autoimmune disorders remain impossible to cure. Recent research demonstrates the remarkable therapeutic effectiveness of MSCs in a wide array of autoimmune diseases. TAI is one of many autoimmune disorders that can benefit from the use of MSCs, which can be derived from bone marrow and adipose tissue. Cell and gene therapies hold great potential for treating autoimmune conditions, although further research is still needed.
Collapse
Affiliation(s)
- Sanja Medenica
- Department of Endocrinology, Internal Medicine Clinic, Clinical Center of Montenegro, School of Medicine, University of Montenegro, Podgorica, Montenegro
| | | | - Aleksandar Ljubić
- Biocell Hospital, Belgrade, Serbia
- Special Gynecology Hospital with Maternity Ward Jevremova, Belgrade, Serbia
- Libertas International University, Dubrovnik, Croatia
| | | | | | - Gaspare Cucinella
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, IVF UNIT, University of Palermo, Palermo, Italy
| | - Simona Zaami
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, “Sapienza” University of Rome, Rome, Italy
| | - Giuseppe Gullo
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, IVF UNIT, University of Palermo, Palermo, Italy
| |
Collapse
|
24
|
Ma H, Ishida K, Xu C, Takahashi K, Li Y, Zhang C, Kang Q, Jia Y, Hu W, Matsumaru D, Nakanishi T, Hu J. Triphenyl phosphate delayed pubertal timing and induced decline of ovarian reserve in mice as an estrogen receptor antagonist. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 290:118096. [PMID: 34488164 DOI: 10.1016/j.envpol.2021.118096] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/29/2021] [Accepted: 09/01/2021] [Indexed: 06/13/2023]
Abstract
Although concerns have been raised about the adverse effects of triphenyl phosphate (TPhP) on female fertility, its risk to ovarian functioning remains unknown. In this study, female C57BL/6 mice at postnatal day 21 were exposed on a daily basis to TPhP dose of 2, 10, and 50 mg/kg for 40 days. A significant delay in pubertal timing was observed in the mice exposed to 50 mg/kg of TPhP. An estrogen-responsive reporter transgenic mice assay demonstrated that TPhP significantly downregulated the estrogen receptor (ER) signaling by 45.1% in the whole body in the 50 mg/kg group, and by 14.7-43.7% in the uterus for all exposure groups compared with the control. This strong antagonistic activity of TPhP toward ER explained the delay in pubertal timing. A significant reduction in the number of follicles in all stages was observed in mice after being exposed to TPhP for 40 days at concentrations of 10 and 50 mg/kg, resulting in a decline of the ovarian reserve. The elevation of the follicle-stimulating hormone concentration may have contributed to this phenomenon, as controlled by the antagonistic activity of TPhP toward ER in the brain. The toxic effects of TPhP on ovarian functioning highlight this chemical as a potential risk factor for female fertility.
Collapse
Affiliation(s)
- Haojia Ma
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Keishi Ishida
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Chenke Xu
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Kyosuke Takahashi
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Yu Li
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Chenhao Zhang
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Qiyue Kang
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Yingting Jia
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Wenxin Hu
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Daisuke Matsumaru
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Tsuyoshi Nakanishi
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Jianying Hu
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
25
|
D'Ippolito S, Capozzi A, Scambia G, Sorge R, Lello S, Simone ND. Glucose/insulin metabolism and vitamin D in women with recurrent pregnancy loss. Am J Reprod Immunol 2021; 87:e13505. [PMID: 34687115 DOI: 10.1111/aji.13505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 09/02/2021] [Accepted: 10/18/2021] [Indexed: 10/20/2022] Open
Abstract
OBJECTIVE Glucose/insulin metabolism has been related to recurrent pregnancy loss (RPL) through mechanisms not really clarified. Also, vitamin D deficiency seems to be associated to RPL. The purpose of our study was to evaluate the correlation between glucose/insulin metabolism parameters and vitamin D levels in women with history of RPL. STUDY DESIGN Observational retrospective study on RPL women. The correlation among vitamin D levels and fasting glucose (FG), fasting insulin (FI), Homeostatic model assessment of insulin resistance (HOMA-IR) index, area under glucose curve (AUC-Glyc) and area under insulin curve (AUC-Ins), was evaluated. RESULTS One-hundred and twenty-seven RPL women were classified into three subgroups (0-1-2) according to the levels of FI. We found a statistically significant linear Pearson correlation between FI and HOMA-IR (r = .840; P = .001). An, inverse, but non-significant correlation both between vitamin D and FI (R = -.202, ns) and vitamin D levels and AUC-Ins (R = -.288, ns) was observed. The variables vitamin D, HOMA-IR and AUC-Ins were statistically significant in the considered subgroups (Vitamin D: ANOVA + Bonferroni test: 0 vs. 1; P = .001; 0 vs. 2; P = .010; 1 vs. 2; P = .657; HOMA-IR: ANOVA + Bonferroni test: 0 vs. 1; P = .014; 0 vs. 2; P = .001; 1 vs. 2; P = .001; AUC-Ins: ANOVA + Bonferroni test: 0 vs. 1; P = .010; 0 vs. 2; P = .206; 1 vs. 2; P = .980). CONCLUSIONS Vitamin D might play additional roles in the pathogenesis of RPL, beyond its well known immunomodulatory role.
Collapse
Affiliation(s)
- Silvia D'Ippolito
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.), Roma, Italia
| | - Anna Capozzi
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.), Roma, Italia
| | - Giovanni Scambia
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.), Roma, Italia.,Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Roberto Sorge
- Laboratory of Biometry, University of Rome, Tor Vergata, Rome, Italy
| | - Stefano Lello
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.), Roma, Italia
| | - Nicoletta Di Simone
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, 20072, Italy.,IRCCS Humanitas Research Hospital, Rozzano, Milan, 20089, Italy
| |
Collapse
|
26
|
Han L, Ma Y, Liang Z, Chen D. Laboratory characteristics analysis of the efficacy of levothyroxine on subclinical hypothyroidism during pregnancy: a single-center retrospective study. Bioengineered 2021; 12:4183-4190. [PMID: 34288808 PMCID: PMC8806776 DOI: 10.1080/21655979.2021.1955589] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
To reassess the efficacy of levothyroxine on subclinical hypothyroidism (SCH, 4.0 mIU/L ≤ TSH (thyroid stimulating hormone) <10 mIU/L with normal free T4) during pregnancy. 165 levothyroxine-treated pregnant women experiencing SCH were screened. And controls were randomly selected using euthyroidism (EU) women, matched by age, gravidity, and parity in the EU group (n = 660). We evaluated laboratory characteristics and pregnancy outcomes during follow-ups. Compared with the EU group, the SCH group displayed higher inadequate maternal gestational weight gain, premature delivery, low birth weight offspring and infant offspring small for their gestational age. After levothyroxine treatment, the SCH group displayed lower total cholesterol, low-density lipoprotein levels, and higher serum homocysteine levels before delivery. Pregnant women with SCH still exhibit adverse pregnancy outcomes after levothyroxine treatment. Taken together, we believe that besides levothyroxine, vitamin B12 and folic acid could be added to the treatment of pregnant women with SCH. In addition, regular monitoring of blood sugar levels, lipid and homocysteine levels, and intervention gestational weight gain could alleviate the adverse effects of SCH on pregnancy outcomes.
Collapse
Affiliation(s)
- Luyang Han
- Department of Obstetrics, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Yan Ma
- Department of Obstetrics, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Zhaoxia Liang
- Department of Obstetrics, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Danqing Chen
- Department of Obstetrics, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| |
Collapse
|
27
|
Gillis-Buck E, Miller H, Sirota M, Sanders SJ, Ntranos V, Anderson MS, Gardner JM, MacKenzie TC. Extrathymic Aire-expressing cells support maternal-fetal tolerance. Sci Immunol 2021; 6:eabf1968. [PMID: 34272228 PMCID: PMC9363019 DOI: 10.1126/sciimmunol.abf1968] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 06/17/2021] [Indexed: 12/21/2022]
Abstract
Healthy pregnancy requires tolerance to fetal alloantigens as well as syngeneic embryonic and placental antigens. Given the importance of the autoimmune regulator (Aire) gene in self-tolerance, we investigated the role of Aire-expressing cells in maternal-fetal tolerance. We report that maternal ablation of Aire-expressing (Aire +) cells during early mouse pregnancy caused intrauterine growth restriction (IUGR) in both allogeneic and syngeneic pregnancies. This phenotype is immune mediated, as IUGR was rescued in Rag1-deficient mice, and involved a memory response, demonstrated by recurrence of severe IUGR in second pregnancies. Single-cell RNA sequencing demonstrated that Aire + cell depletion in pregnancy results in expansion of activated T cells, particularly T follicular helper cells. Unexpectedly, selective ablation of either Aire-expressing medullary thymic epithelial cells or extrathymic Aire-expressing cells (eTACs) mapped the IUGR phenotype exclusively to eTACs. Thus, we report a previously undescribed mechanism for the maintenance of maternal-fetal immune homeostasis and demonstrate that eTACs protect the conceptus from immune-mediated IUGR.
Collapse
Affiliation(s)
- Eva Gillis-Buck
- Department of Surgery, University of California, San Francisco, CA, USA
| | - Haleigh Miller
- Department of Epidemiology and Biostatistics University of California, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
- Diabetes Center University of California, San Francisco, CA, USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
- Department of Pediatrics University of California, San Francisco, CA, USA
| | - Stephan J Sanders
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- Center for Maternal-Fetal Precision Medicine, University of California, San Francisco, CA, USA
| | - Vasilis Ntranos
- Department of Epidemiology and Biostatistics University of California, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
- Diabetes Center University of California, San Francisco, CA, USA
| | - Mark S Anderson
- Diabetes Center University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - James M Gardner
- Department of Surgery, University of California, San Francisco, CA, USA.
- Diabetes Center University of California, San Francisco, CA, USA
| | - Tippi C MacKenzie
- Department of Surgery, University of California, San Francisco, CA, USA.
- Department of Pediatrics University of California, San Francisco, CA, USA
- Center for Maternal-Fetal Precision Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
28
|
Jiao X, Zhang X, Li N, Zhang D, Zhao S, Dang Y, Zanvit P, Jin W, Chen Z, Chen W, Qin Y. T reg deficiency-mediated T H 1 response causes human premature ovarian insufficiency through apoptosis and steroidogenesis dysfunction of granulosa cells. Clin Transl Med 2021; 11:e448. [PMID: 34185428 PMCID: PMC8214854 DOI: 10.1002/ctm2.448] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 12/22/2022] Open
Abstract
Immune dysregulation has long been proposed as a component of premature ovarian insufficiency (POI), but the underlying mediators and mechanisms remain largely unknown. Here we showed that patients with POI had augmented T helper 1 (TH 1) responses and regulatory T (Treg ) cell deficiency in both the periphery and the ovary compared to the control women. The increased ratio of TH 1:Treg cells was strongly correlated with the severity of POI. In mouse models of POI, the increased infiltration of TH 1 cells in the ovary resulted in follicle atresia and ovarian insufficiency, which could be prevented and reversed by Treg cells. Importantly, interferon (IFN) -γ and tumor necrosis factor (TNF) -α cooperatively promoted the apoptosis of granulosa cells and suppressed their steroidogenesis by modulating CTGF and CYP19A1. We have thus revealed a previously unrecognized Treg cell deficiency-mediated TH 1 response in the pathogenesis of POI, which should have implications for therapeutic interventions in patients with POI.
Collapse
Affiliation(s)
- Xue Jiao
- Center for Reproductive MedicineCheeloo College of MedicineShandong UniversityJinanShandongChina
- Mucosal Immunology SectionNIDCRNational Institutes of HealthBethesdaMarylandUSA
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsShandong UniversityJinanShandongChina
- Key laboratory of Reproductive Endocrinology of Ministry of EducationShandong UniversityJinanShandongChina
| | - Xiruo Zhang
- Center for Reproductive MedicineCheeloo College of MedicineShandong UniversityJinanShandongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsShandong UniversityJinanShandongChina
- Key laboratory of Reproductive Endocrinology of Ministry of EducationShandong UniversityJinanShandongChina
| | - Nianyu Li
- Center for Reproductive MedicineCheeloo College of MedicineShandong UniversityJinanShandongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsShandong UniversityJinanShandongChina
- Key laboratory of Reproductive Endocrinology of Ministry of EducationShandong UniversityJinanShandongChina
| | - Dunfang Zhang
- Mucosal Immunology SectionNIDCRNational Institutes of HealthBethesdaMarylandUSA
| | - Shidou Zhao
- Center for Reproductive MedicineCheeloo College of MedicineShandong UniversityJinanShandongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsShandong UniversityJinanShandongChina
- Key laboratory of Reproductive Endocrinology of Ministry of EducationShandong UniversityJinanShandongChina
| | - Yujie Dang
- Center for Reproductive MedicineCheeloo College of MedicineShandong UniversityJinanShandongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsShandong UniversityJinanShandongChina
- Key laboratory of Reproductive Endocrinology of Ministry of EducationShandong UniversityJinanShandongChina
| | - Peter Zanvit
- Mucosal Immunology SectionNIDCRNational Institutes of HealthBethesdaMarylandUSA
| | - Wenwen Jin
- Mucosal Immunology SectionNIDCRNational Institutes of HealthBethesdaMarylandUSA
| | - Zi‐Jiang Chen
- Center for Reproductive MedicineCheeloo College of MedicineShandong UniversityJinanShandongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsShandong UniversityJinanShandongChina
- Key laboratory of Reproductive Endocrinology of Ministry of EducationShandong UniversityJinanShandongChina
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghaiChina
- Center for Reproductive MedicineRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Wanjun Chen
- Mucosal Immunology SectionNIDCRNational Institutes of HealthBethesdaMarylandUSA
| | - Yingying Qin
- Center for Reproductive MedicineCheeloo College of MedicineShandong UniversityJinanShandongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsShandong UniversityJinanShandongChina
- Key laboratory of Reproductive Endocrinology of Ministry of EducationShandong UniversityJinanShandongChina
| |
Collapse
|
29
|
Chalan P, Thomas N, Caturegli P. Th17 Cells Contribute to the Pathology of Autoimmune Hypophysitis. THE JOURNAL OF IMMUNOLOGY 2021; 206:2536-2543. [PMID: 34011522 DOI: 10.4049/jimmunol.2001073] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 03/26/2021] [Indexed: 12/11/2022]
Abstract
Autoimmune hypophysitis is classified as primary if its origin is idiopathic and secondary if it develops as a consequence of treatment with immune checkpoint inhibitors. Expanding use of immunotherapy has been paralleled by the increasing hypophysitis prevalence. However, understanding of the immune responses driving the disease remains limited. Using a mouse model of primary hypophysitis, we have identified CD4+ T lymphocytes to be the main pituitary-infiltrating immune cell population. Functional analysis showed that they display a Th17 and Th1/Th17 phenotype. To examine involvement of proinflammatory Th1, Th17, and Th1/17 subsets in hypophysitis, we have isolated RNA from the formalin-fixed paraffin-embedded pituitary specimens from 16 hypophysitis patients (three of whom had hypophysitis secondary to immune checkpoint inhibitors), 10 patients with adenoma, and 23 normal pituitaries obtained at autopsy. Transcript levels of IFN-γ, IL-17A, IL-4, IL-10, TGF-β, CD4, CD8α, and class II MHC transactivator were analyzed by the reverse transcription-quantitative PCR (RT-qPCR). Pituitary glands of patients with hypophysitis showed significantly higher IL-17A, CD4, and class II MHC transactivator mRNA levels compared with adenoma and normal pituitaries. All three secondary hypophysitis patients showed detectable IL-17A levels, but other cytokines were not detected in their pituitaries. Levels of IFN-γ, IL-4, IL-10, and TGF-β did not differ between the groups. TGF-β transcript was found in significantly fewer hypophysitis pituitaries (2 out of 16) compared with adenoma (7 out of 10) and normal pituitaries (11 out of 23). Presence of TGF-β in two hypophysitis patients was associated with significantly lower IL-17A mRNA levels compared with hypophysitis patients with no detectable TGF-β (p = 0.03).
Collapse
Affiliation(s)
- Paulina Chalan
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD
| | - Nithya Thomas
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD
| | - Patrizio Caturegli
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD
| |
Collapse
|
30
|
Gleicher N, Darmon S, Molinari E, Zhang L, Hu J, Albertini DF, Barad DH. A form of secondary ovarian insufficiency (SOI) due to adrenal hypoandrogenism as new infertility diagnosis. Endocrine 2021; 72:260-267. [PMID: 33009651 DOI: 10.1007/s12020-020-02512-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/23/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Mediated via the androgen receptor on granulosa cells, models of small growing follicle stages demonstrate dependence on testosterone. Androgen deficiency reduces ovarian response to follicle stimulation hormone (FSH), granulosa cell mass and estradiol (E2) production falls and FSH, therefore, rises. Though potentially of adrenal and/or ovarian origin, androgen deficiency in association with female infertility is almost universally primarily of adrenal origin, raising the possibility that women with presumptive diagnosis of primary ovarian insufficiency (POI), also called primary ovarian failure (POF) may actually suffer from secondary ovarian insufficiency (SOI) due to adrenal hypoandrogenism that leads to follicular arrest at small-growing follicle stages. METHODS This retrospective cohort study was performed in a private, academically affiliated infertility center in New York City. We searched the center's anonymized electronic research data bank for consecutive patients who presented with a diagnosis of POI, defined by age <41 year, FSH > 40.0 mIU/mL, amenorrhea for at least 6 month, and low testosterone (T), defined as total T (TT) in the lowest age-specific quartile of normal range. This study did not include patients with oophoritis. Since dehydroepiandrosterone sulfate (DHEAS) is the only androgen almost exclusively produce by adrenals, adrenal hypoandrogenism was defined by DHEAS < 100ug/dL. Thirteen of 78 presumed POI women (16.67%) qualified and represented the original study population. POI patients are usually treated with third-party egg donation; 6/13, however, rejected egg donation for personal or religious reasons and insisted on undergoing at least one last IVF cycle attempt (final study population). In preparation, they were supplemented with DHEA 25 mg TID and CoQ10 333 mg TID for at least 6 weeks prior to ovarian stimulation for IVF with FSH and human menopausal gonadotropins (hMG). Since POI patients are expected to be resistant to ovarian stimulation, primary outcome for the study was ovarian response, while secondary outcome was pregnancy/delivery. RESULTS Though POI/POF patients usually are completely unresponsive to ovarian stimulation, to our surprise, 5/6 (83.3%) patients demonstrated an objective follicle response. In addition, 2/6 (33.3%) conceived spontaneously between IVF cycles, while on DHEA and CoQ10 supplementation and delivered healthy offspring. One of those is currently in treatment for a second child. CONCLUSIONS This preliminary report suggests that a surprising portion of young women below age 41, tagged with a diagnosis of POI/POF, due to adrenal hypoandrogenism actually suffer from a form of SOI, at least in some cases amenable to treatment by androgen supplementation. Since true POI/POF usually requires third-party egg donation, correct differentiation between POI and SOI in such women appears of great importance and may warrant a trial stimulation after androgen pre-supplementation for at least 6 weeks.
Collapse
Affiliation(s)
- Norbert Gleicher
- The Center for Human Reproduction, New York, NY, 10021, USA.
- The Foundation for Reproductive Medicine, New York, NY, 10022, USA.
- Stem Cell Biology and Molecular Embryology Laboratory, Rockefeller University, New York, NY, 10016, USA.
- Department of Obstetrics and Gynecology, Vienna University School of Medicine, 1009, Vienna, Austria.
| | - Sarah Darmon
- The Center for Human Reproduction, New York, NY, 10021, USA
| | | | - Lin Zhang
- The Center for Human Reproduction, New York, NY, 10021, USA
| | - Jianjun Hu
- The Center for Human Reproduction, New York, NY, 10021, USA
| | - David F Albertini
- The Center for Human Reproduction, New York, NY, 10021, USA
- Stem Cell Biology and Molecular Embryology Laboratory, Rockefeller University, New York, NY, 10016, USA
| | - David H Barad
- The Center for Human Reproduction, New York, NY, 10021, USA
- The Foundation for Reproductive Medicine, New York, NY, 10022, USA
| |
Collapse
|
31
|
Naim N, Amrit FRG, McClendon TB, Yanowitz JL, Ghazi A. The molecular tug of war between immunity and fertility: Emergence of conserved signaling pathways and regulatory mechanisms. Bioessays 2020; 42:e2000103. [PMID: 33169418 DOI: 10.1002/bies.202000103] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 12/21/2022]
Abstract
Reproduction and immunity are energy intensive, intimately linked processes in most organisms. In women, pregnancy is associated with widespread immunological adaptations that alter immunity to many diseases, whereas, immune dysfunction has emerged as a major cause for infertility in both men and women. Deciphering the molecular bases of this dynamic association is inherently challenging in mammals. This relationship has been traditionally studied in fast-living, invertebrate species, often in the context of resource allocation between life history traits. More recently, these studies have advanced our understanding of the mechanistic underpinnings of the immunity-fertility dialogue. Here, we review the molecular connections between reproduction and immunity from the perspective of human pregnancy to mechanistic discoveries in laboratory organisms. We focus particularly on recent invertebrate studies identifying conserved signaling pathways and transcription factors that regulate resource allocation and shape the balance between reproductive status and immune health.
Collapse
Affiliation(s)
- Nikki Naim
- Departments of Pediatrics, Developmental Biology and Cell Biology and Physiology, John, G. Rangos Sr. Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Francis R G Amrit
- Departments of Pediatrics, Developmental Biology and Cell Biology and Physiology, John, G. Rangos Sr. Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - T Brooke McClendon
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Judith L Yanowitz
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Arjumand Ghazi
- Departments of Pediatrics, Developmental Biology and Cell Biology and Physiology, John, G. Rangos Sr. Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
32
|
Yalçın Bahat P, Kadiroğulları P, Topbas Selcuki NF, Yücel B, Çakmak K, Üreyen Özdemir E. Ovarian reserve in patients with ankylosing spondylitis. Arch Gynecol Obstet 2020; 303:189-193. [PMID: 33030584 DOI: 10.1007/s00404-020-05824-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/25/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE The aim of this study was to determine the autoimmune effects of ankylosing spondylitis (AS) on the fertility potential of women by evaluating ovarian reserves of AS patients. METHODS A total of 104 patients, 52 in the AS group (study group) and 52 in the control group were included in the study. Ovarian reserve was evaluated by serum anti-Müllerian hormone (AMH) levels, antral follicle count (AFC) and baseline serum follicle-stimulating hormone (FSH) levels. RESULTS The mean serum AMH levels were significantly lower in the study group when compared to the controls (2.203 ± 1.110 vs. 1.188 ± 0.891, p < 0.001). In addition, the mean AFC was also significantly lower in the study group. (10.67 ± 1.81 vs. 9.54 ± 2.50, p = 0.009). Mean FSH levels were calculated to be 6.72 ± 1.14 in the study group and 7.21 ± 1.22 in the control group. The difference was not statistically significant (p = 0.781). CONCLUSION This study shows that AS like several other autoimmune conditions has an adverse effect on the female fertility potential. Therefore, an early start and long-term management of AS patients who have fertility desire is recommended. Serum AMH levels can be used in monitoring ovarian reserve and in early detection of reproductive decline of AS patients. CLINICALTRIAL NUMBER NCT04209881.
Collapse
Affiliation(s)
- Pınar Yalçın Bahat
- Department of Obstetrics and Gynecology, Kanuni Sultan Suleyman Training and Research Hospital, Health Sciences University, 34000, Istanbul, Turkey.
| | - Pınar Kadiroğulları
- Department of Obstetrics and Gynecology, Acibadem University Atakent Hospital, Istanbul, Turkey
| | - Nura Fitnat Topbas Selcuki
- Department of Obstetrics and Gynecology, Istanbul Sisli Hamidiye Etfal Training and Research Hospital, Health Sciences University, Istanbul, Turkey
| | - Burak Yücel
- Department of Obstetrics and Gynecology, Kanuni Sultan Suleyman Training and Research Hospital, Health Sciences University, 34000, Istanbul, Turkey
| | - Kübra Çakmak
- Obstetrics and Gynecology, Esenler Maternity and Children's Hospital, Istanbul, Turkey
| | - Eda Üreyen Özdemir
- Department of Obstetrics and Gynecology Ankara, Çubuk Halil Şıvgın Hospital, Çubuk, Turkey
| |
Collapse
|
33
|
He R, Zhao Z, Yang Y, Liang X. Using bioinformatics and metabolomics to identify altered granulosa cells in patients with diminished ovarian reserve. PeerJ 2020; 8:e9812. [PMID: 32923184 PMCID: PMC7457930 DOI: 10.7717/peerj.9812] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 08/04/2020] [Indexed: 11/20/2022] Open
Abstract
Background During fertility treatment, diminished ovarian reserve (DOR) is a challenge that can seriously affect a patient's reproductive potential. However, the pathogenesis of DOR is still unclear and its treatment options are limited. This study aimed to explore DOR's molecular mechanisms. Methods We used R software to analyze the mRNA microarray dataset E-MTAB-391 downloaded from ArrayExpress, screen for differentially expressed genes (DEGs), and perform functional enrichment analyses. We also constructed the protein-protein interaction (PPI) and miRNA-mRNA networks. Ovarian granulosa cells (GCs) from women with DOR and the control group were collected to perform untargeted metabolomics analyses. Additionally, small molecule drugs were identified using the Connectivity Map database. Results We ultimately identified 138 DEGs. Our gene ontology (GO) analysis indicated that DEGs were mainly enriched in cytokine and steroid biosynthetic processes. According to the Kyoto Encyclopedia of Genes and Genomes (KEGG), the DEGs were mainly enriched in the AGE (advanced glycation end-product)-RAGE (receptor for AGE) signaling pathway in diabetic complications and steroid biosynthesis. In the PPI network, we determined that JUN, EGR1, HMGCR, ATF3, and SQLE were hub genes that may be involved in steroid biosynthesis and inflammation. miRNAs also played a role in DOR development by regulating target genes. We validated the differences in steroid metabolism across GCs using liquid chromatography-tandem mass spectrometry (LC-MS/MS). We selected 31 small molecules with potentially positive or negative influences on DOR development. Conclusion We found that steroidogenesis and inflammation played critical roles in DOR development, and our results provide promising insights for predicting and treating DOR.
Collapse
Affiliation(s)
- Ruifen He
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Zhongying Zhao
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yongxiu Yang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Key Laboratory for Gynecologic Oncology Gansu Province, Lanzhou, China
| | - Xiaolei Liang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Key Laboratory for Gynecologic Oncology Gansu Province, Lanzhou, China
| |
Collapse
|
34
|
Ho CW, Chen HH, Hsieh MC, Chen CC, Hsu SP, Yip HT, Kao CH. Hashimoto's thyroiditis might increase polycystic ovary syndrome and associated comorbidities risks in Asia. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:684. [PMID: 32617304 PMCID: PMC7327368 DOI: 10.21037/atm-19-4763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background To investigate whether increased the comorbidities such as coronary artery disease (CAD) and risks between Hashimoto’s thyroiditis (HT) and polycystic ovary syndrome (PCOS) in Taiwanese women. Methods Patients newly diagnosed as having HT during 2000–2012 were assigned to the case group. Cases and controls were matched for age and comorbidities at a 1:2 ratio using propensity score matching. Incidence was calculated in the unit of 1000 person-year. Univariate and multivariate Cox proportional hazard regression, multivariate Cox, logistic regression, and Kaplan-Meier analyses were performed. Results Among 3,996 participants, 2,664 constituted the control group and 1,332 constituted the case group. The PCOS risk in patients with HT increased by 2.37 times [95% confidence interval (CI): 1.22–4.62] compared with the controls. Hypertension (HTN) [adjusted odds ratio (OR): 1.31, 95% CI: 1.03–1.66] and hyperlipidemia (adjusted OR: 1.55, 95% CI: 1.2–1.9) were more common in HT patients without PCOS than in other patients. The adjusted OR for CAD in patients with HT was 1.51 (95% CI: 1.11–2.06), whereas that in patients with HT and PCOS was 5.92 (95% CI: 1.32–26.53). Conclusions In our study, the PCOS risk in patients with HT increased by 2.37 times, which is lower than the increase in HT risk in Asian patients with PCOS (4.56 times). The proportion of CAD increased significantly by 5.92 times in patients with HT and PCOS compared with patients with HT only.
Collapse
Affiliation(s)
- Chun-Wei Ho
- Intelligent Diabetes Metabolism and Exercise Center, China Medical University Hospital, Taichung
| | - Hsin-Hung Chen
- Intelligent Diabetes Metabolism and Exercise Center, China Medical University Hospital, Taichung.,School of Medicine, Institute of Medicine and Public Health, Chung Shan, Medical University, Taichung
| | - Ming-Chia Hsieh
- Intelligent Diabetes Metabolism and Exercise Center, China Medical University Hospital, Taichung.,Graduate Institute of Integrative Medicine, China Medical University, Taichung.,Division of Clinical Nutrition, Department of Internal Medicine, China Medical University Hospital, Taichung
| | - Ching-Chu Chen
- Division of Endocrinology and Metabolism, Department of Internal Medicine, China Medical University Hospital, Taichung.,School of Chinese Medicine, China Medical University, Taichung
| | - Sheng-Pang Hsu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, China Medical University Hospital, Taichung
| | - Hei-Tung Yip
- Management Office for Health Data, China Medical University Hospital, Taichung.,School of Medicine, College of Medicine China Medical University, Taichung
| | - Chia-Hung Kao
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung.,Department of Nuclear Medicine and PET Center, China Medical University Hospital, Taichung.,Department of Bioinformatics and Medical Engineering, Asia University, Taichung.,Center of Augmented Intelligence in Healthcare, China Medical University Hospital, Taichung
| |
Collapse
|
35
|
Chen HH, Chen CC, Hsieh MC, Ho CW, Hsu SP, Yip HT, Kao CH. Graves' disease could increase polycystic ovary syndrome and comorbidities in Taiwan. Curr Med Res Opin 2020; 36:1063-1067. [PMID: 32297812 DOI: 10.1080/03007995.2020.1756235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Background: To investigate the association between Graves' disease (GD) and polycystic ovary syndrome (PCOS) and its comorbidities.Methods: Logistic regression was performed to determine the association between the two conditions. Cumulative incidence curves were obtained using the Kaplan-Meier method and log-rank test. Hazard ratios were determined using the Cox proportional hazards regression model.Results: We included 5399 patients with GD as the study group and 10,798 patients without GD as the control group. The cumulative incidence curve of PCOS in patients with GD was significantly higher than that in patients without GD (p = .02). The adjusted hazard ratio for PCOS in patients with GD compared with patients without GD was 1.47 (95%CI = 1.09-1.98). The adjusted odds ratio of hyperlipidemia in patients with GD and PCOS was 2.18 (95%CI = 1.14-4.17) higher than that in patients with GD only.Conclusion: Our study demonstrated that women with GD could be at risk of developing PCOS; additionally, a higher incidence of comorbidities, including hyperlipidemia, was noted in women with GD and PCOS.
Collapse
Affiliation(s)
- Hsin-Hung Chen
- Intelligent Diabetes Metabolism and Exercise Center, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, Institute of Medicine and Public Health, Chung Shan Medical University, Taichung, Taiwan
| | - Ching-Chu Chen
- Division of Endocrinology and Metabolism, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Ming-Chia Hsieh
- Intelligent Diabetes Metabolism and Exercise Center, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Integrative Medicine, China Medical University, Taichung, Taiwan
- Division of Clinical Nutrition, China Medical University Hospital, Taichung, Taiwan
| | - Chun-Wei Ho
- Intelligent Diabetes Metabolism and Exercise Center, China Medical University Hospital, Taichung, Taiwan
| | - Sheng-Pang Hsu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Hei-Tung Yip
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Chia-Hung Kao
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Nuclear Medicine and PET Center, China Medical University Hospital, Taichung, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
- Center of Augmented Intelligence in Healthcare, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
36
|
Rao M, Zeng Z, Zhou F, Wang H, Liu J, Wang R, Wen Y, Yang Z, Su C, Su Z, Zhao S, Tang L. Effect of levothyroxine supplementation on pregnancy loss and preterm birth in women with subclinical hypothyroidism and thyroid autoimmunity: a systematic review and meta-analysis. Hum Reprod Update 2020; 25:344-361. [PMID: 30951172 DOI: 10.1093/humupd/dmz003] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 12/15/2018] [Accepted: 01/16/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Subclinical hypothyroidism (SCH) and thyroid autoimmunity (TAI) are associated with adverse pregnancy outcomes such as pregnancy loss and preterm birth. However, the ability of levothyroxine (LT4) supplementation to attenuate the risks of these outcomes remains controversial. OBJECTIVE AND RATIONALE This systematic review and meta-analysis was conducted to determine the effect of LT4 supplementation on pregnancy loss rate (PLR) and preterm birth rate (PBR) among pregnant women with SCH and TAI. SEARCH METHODS A systematic literature search of the PubMed, EMBASE, Web of Science and Cochrane Controlled Trials Register databases and Clinicaltrials.gov was performed to identify all relevant English studies published up to April 2018. The following terms were used for the search: [subclinical hypothyroidism OR thyroid autoimmunity OR thyroperoxidase antibody (TPO-Ab) OR thyroglobulin antibodies (Tg-Ab)] AND (levothyroxine OR euthyrox) AND [pregnancy outcome OR miscarriage OR abortion OR pregnancy loss OR preterm birth OR premature delivery OR early labo(u)r]. The reference lists of the relevant publications were also manually searched for related studies. Published manuscripts were included if they reported data on pregnancy loss, preterm birth or both. We separately analysed the pooled effects of LT4 supplementation on PLR and PBR in women with SCH and TAI. OUTCOMES Overall, 13 eligible studies including 7970 women were included in the meta-analysis. Eight and five of these studies were randomized controlled trials (RCTs) and retrospective studies, respectively. The pooled results indicated that LT4 supplementation significantly decreased the PLR [relative risk (RR) = 0.56, 95% confidence interval (CI): 0.42-0.75, I2 = 1%, 12 studies] and PBR (RR = 0.68, 95% CI: 0.51-0.91, I2 = 21%, eight studies) in women with SCH and/or TAI. We further found that LT4 supplementation significantly decreased the risk of pregnancy loss (RR = 0.43, 95% CI: 0.26-0.72, P = 0.001, I2 = 0%) but not of preterm birth (RR = 0.67, 95% CI: 0.41-1.12, P = 0.13, I2 = 0%) in women with SCH. Furthermore, LT4 supplementation significantly decreased the risks of both pregnancy loss (RR = 0.63, 95% CI: 0.45-0.89, P = 0.009, I2 = 0%) and preterm birth (RR = 0.68 95% CI: 0.48-0.98, P = 0.04, I2 = 46%) in women with TAI. These results were consistent when only RCTs were included in the analysis. Further, in women with SCH, LT4 supplementation reduced the risk of pregnancy loss in pregnancies achieved by assisted reproduction (RR = 0.27, 95% CI: 0.14-0.52, P < 0.001, I2 = 14%) but not in naturally conceived pregnancies (RR = 0.60, 95% CI: 0.28-1.30, P = 0.13, I2 = 0%). By contrast, in women with TAI, LT4 supplementation reduced the risks of both pregnancy loss (RR = 0.61, 95% CI: 0.39-0.96, P = 0.03, I2 = 0%) and preterm birth (RR = 0.49, 95% CI: 0.30-0.79, P = 0.003, I2 = 0%) in naturally conceived pregnancies but not in pregnancies achieved by assisted reproduction (RR = 0.68, 95% CI: 0.40-1.15, P = 0.15, I2 = 0% for pregnancy loss and RR = 1.20, 95% CI: 0.68-2.13, P = 0.53, I2 not applicable for preterm birth). WIDER IMPLICATIONS This meta-analysis confirmed the beneficial effects of LT4 supplementation, namely the reduced risks of pregnancy loss and preterm birth, among pregnant women with SCH and/or TAI. The different effects of LT4 supplementation on naturally conceived pregnancies and pregnancies achieved by assisted reproduction in women with SCH and/or TAI suggest that these women should be managed separately. Due to the limited number of studies included in this meta-analysis, especially in the subgroup analysis, further large RCTs and fundamental studies are warranted to confirm the conclusions and better clarify the molecular mechanism underlying these associations.
Collapse
Affiliation(s)
- Meng Rao
- Department of Reproduction and Genetics, the First Affiliated Hospital of Kunming Medical University, No. 295 Xi Chang road, Kunming, China
| | - Zhengyan Zeng
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, No. 295 Xi Chang road, Kunming, China
| | - Fang Zhou
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hang Kong road, Wuhan, China
| | - Huawei Wang
- Department of Reproduction and Genetics, the First Affiliated Hospital of Kunming Medical University, No. 295 Xi Chang road, Kunming, China
| | - Jiang Liu
- Department of Reproduction and Genetics, the First Affiliated Hospital of Kunming Medical University, No. 295 Xi Chang road, Kunming, China
| | - Rui Wang
- Department of Reproduction and Genetics, the First Affiliated Hospital of Kunming Medical University, No. 295 Xi Chang road, Kunming, China
| | - Ya Wen
- Department of Reproduction and Genetics, the First Affiliated Hospital of Kunming Medical University, No. 295 Xi Chang road, Kunming, China
| | - Zexing Yang
- Department of Reproduction and Genetics, the First Affiliated Hospital of Kunming Medical University, No. 295 Xi Chang road, Kunming, China
| | - Cunmei Su
- Department of Reproduction and Genetics, the First Affiliated Hospital of Kunming Medical University, No. 295 Xi Chang road, Kunming, China
| | - Zhenfang Su
- Department of Reproduction and Genetics, the First Affiliated Hospital of Kunming Medical University, No. 295 Xi Chang road, Kunming, China
| | - Shuhua Zhao
- Department of Reproduction and Genetics, the First Affiliated Hospital of Kunming Medical University, No. 295 Xi Chang road, Kunming, China
| | - Li Tang
- Department of Reproduction and Genetics, the First Affiliated Hospital of Kunming Medical University, No. 295 Xi Chang road, Kunming, China
| |
Collapse
|
37
|
Demir B, Sarıdaş Demir S, Özkan Karacaer K, Paşa S, Sılan F. Evaluation inflammatory markers of hemogram parameters in primary ovarian insufficiency. Turk J Obstet Gynecol 2020; 17:9-14. [PMID: 32341824 PMCID: PMC7171537 DOI: 10.4274/tjod.galenos.2019.09476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 12/28/2019] [Indexed: 01/04/2023] Open
Abstract
Objective: In most of primary ovarian insufficiency (POI) cases, etiologic factors have not been fully elucidated. Recent studies have revealed that inflammatory agents play an important role in the etiopathogenesis of POI. Therefore, the aim of this study was to investigate the role of inflammatory markers of hemogram parameters in POI. Materials and Methods: The study compared 47 healthy women and 47 women diagnosed as having POI retrospectively by scanning electronic and written recording systems. Complete blood counts, day-3 hormone profiles levels of all subjects were analyzed. The neutrophil-lymphocyte ratio (NLR), red cell distribution width (RDW), platelet ratio (RPR), platelet lymphocyte ratio (PLR), and mean platelet volume (MPV) mean platelet lymphocyte ratio (MPLR) were calculated from the complete blood count parameters. Results: White blood cell and MPV values, platelet, and lymphocyte counts were significantly higher in the POI patients (p<0.001, p=0.042, p=0.038, p=0.049, respectively), RPR was significantly lower than the control group (p=0.011), but there were no significant differences in hemoglobin, RDW, NLR, PLR, and MPLR (p=0.454, p=0.057, p=0.635, p=0.780, p=0.126, respectively). The neutrophil count of the study group was higher than in the control group (p=0.057). Bivariate correlation analyses showed no correlations between blood parameters and hormone levels. The area under the receiver operating characteristic curve for RPR in POI was 0.652, with a threshold value 0.053, sensitivity=63% and specificity=63. Conclusion: Inflammatory markers of hemogram detected higher in patients with POI then control subjects.
Collapse
Affiliation(s)
- Bülent Demir
- Çanakkale Onsekiz Mart University Faculty of Medicine, Department of Gynecology and Obstetrics, Çanakkale, Turkey
| | - Süreyya Sarıdaş Demir
- Dokuz Eylül University Faculty of Medicine, Department of Gynecology and Obstetrics, İzmir, Turkey
| | - Kübra Özkan Karacaer
- Çanakkale Onsekiz Mart University Faculty of Medicine, Department of Gynecology and Obstetrics, Çanakkale, Turkey
| | - Semir Paşa
- Mardin Artuklu University Faculty of Health Sciences, Mardin, Turkey
| | - Fatma Sılan
- Çanakkale Onsekiz Mart University Faculty of Medicine, Department of Medical Genetic, Çanakkale, Turkey
| |
Collapse
|
38
|
Xie J, Jiang L, Sadhukhan A, Yang S, Yao Q, Zhou P, Rao J, Jin M. Effect of antithyroid antibodies on women with recurrent miscarriage: A meta-analysis. Am J Reprod Immunol 2020; 83:e13238. [PMID: 32198952 PMCID: PMC7317526 DOI: 10.1111/aji.13238] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/15/2020] [Accepted: 03/16/2020] [Indexed: 02/06/2023] Open
Abstract
Problem The effect of thyroid autoimmunity (TAI) on the prevalence of recurrent miscarriage (RM) is highly debatable. No meta‐analysis has been published in the past decade to investigate the impact of TAI on women with RM. Method of Study Systemic literature search was conducted on PubMed, Embase, Cochrane, and Web of Science databases. English language literatures published between 1993 and 2019 were selected. We assessed the relationship between the prevalence of RM and thyroid peroxidase antibodies (TPO‐Ab) or antithyroid antibodies (ATA) and evaluated the thyroid‐stimulating hormone (TSH) level in TPO‐Ab‐positive women with RM. We also observed the treatment effect with levothyroxine (LT4) for RM. Review Manager 5.3 software was used to obtain the pooled odds ratios (OR). Results Analysis of 22 eligible studies revealed significant association between TPO‐Ab and the prevalence of RM (OR = 1.85; 95% CI, 1.38 to 2.49; P < .001)(n ≥ 3), (OR = 1.82; 95% CI, 1.13 to 2.92; P = .01) (n ≥ 3). Women with ATA + had higher risk of RM (OR = 2.36; 95% CI, 1.71 to 3.25; P < .00001)(n ≥ 3), (OR = 2.34; 95% CI, 1.70 to 3.22; P < .00001)(n ≥ 2). RM women with TPO‐Ab had higher TSH level when compared with those negative for TPO‐Ab (random‐effect SMD = 0.60; 95% CI, 0.31 to 0.90; P < .0001). We also found beneficial effects of LT4 supplementation on the outcome of live birth rate (LBR) among pregnant women with TPO‐Ab (OR = 3.04; 95% CI, 0.69 to 13.36; P = .14). Conclusion The presence of serum antithyroid antibodies does harms to women and can even lead to recurrent miscarriage; LT4 treatment may have beneficial to RM women.
Collapse
Affiliation(s)
- Jilai Xie
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lihong Jiang
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Taizhou Women and Children's Hospital Affiliated to Wenzhou Medical University, Taizhou, China
| | - Annapurna Sadhukhan
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Songqing Yang
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qiuping Yao
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ping Zhou
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jinpeng Rao
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Min Jin
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
39
|
Chen J, Zhao X, Ao L, Yin T, Yang J. Transcriptomic changes and potential regulatory mechanism of intrauterine human chorionic gonadotropin co-cultured with peripheral blood mononuclear cells infusion in mice with embryonic implantation dysfunction. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:99. [PMID: 32175392 DOI: 10.21037/atm.2019.12.109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background This study aimed to explore whether intrauterine infusion of peripheral blood mononuclear cells (PBMCs) could induce favorable transcriptomic changes in the endometrium for embryo implantation and the potential mechanism. Methods Twenty-one mice were randomly divided to five groups, including a normal pregnancy (NP) group, an embryo implantation dysfunction (EID) group, an EID with human chorionic gonadotropin (hCG) group, an EID with PBMCs group, and an EID with hCG co-cultured with PBMCs group. The endometrium in the implantation window from mice were collected and determined by RNA sequencing (RNA-Seq), and the expression of significantly different genes with high degree of coincidence was recommended and validated by quantitative real-time polymerase chain reaction (qRT-PCR). Results There were totally 1,366 up-regulated and 1,374 down-regulated genes in the EID mice compared with the normal pregnant mice. We selected (fold change ≥2, P<0.05) and verified the candidate genes associated with embryo implantation, immune response and other reproductive processes in previous reports by qRT-PCR. Leukemia inhibitory factor (LIF), solute carrier family 15 member 2 (SLC15A2), retinoic acid receptor responder 1 (RARRES1), vascular cell adhesion molecule 1 (VCAM1) were down-regulated and musculin (MSC), chemokine (C-X-C motif) ligand 14 (CXCL14) were up-regulated significantly in EID group (P<0.05), and the synergistic effects of hCG were seen. In addition, the expression of glucocorticoid receptor (GR)-β in PBMCs of NP mice was higher than that of EID mice, and up-regulated GR-β in EID mice could significantly increase the expression of LIF, SLC15A2, RARRES1 and VCAM1, and decrease the expression of CXCL14 and MSC, which indicated GR-β might be a transcriptional factor of the six genes above. Conclusions Intrauterine PBMCs perfusion might improve the performance of impaired endometrial receptivity by regulating LIF, SLC15A2, RARRES1, VCAM1, MSC as well as CXCL14, and hCG could enhance the effect of PBMCs. In addition, GR-β, as a transcriptional factor, could regulate the six genes in PBMCs.
Collapse
Affiliation(s)
- Jiao Chen
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Hubei Clinical Research Center for Assisted Reproductive and Embryonic Development, Wuhan 430060, China
| | - Xuehan Zhao
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Hubei Clinical Research Center for Assisted Reproductive and Embryonic Development, Wuhan 430060, China
| | - Liangfei Ao
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Hubei Clinical Research Center for Assisted Reproductive and Embryonic Development, Wuhan 430060, China
| | - Tailang Yin
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Hubei Clinical Research Center for Assisted Reproductive and Embryonic Development, Wuhan 430060, China
| | - Jing Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Hubei Clinical Research Center for Assisted Reproductive and Embryonic Development, Wuhan 430060, China
| |
Collapse
|
40
|
Ghafoori F, Dehghan-Nayeri N, Khakbazan Z, Hedayatnejad M, Nabavi SM. Pregnancy and Motherhood Concerns Surrounding Women with Multiple Sclerosis: A Qualitative Content Analysis. INTERNATIONAL JOURNAL OF COMMUNITY BASED NURSING AND MIDWIFERY 2020; 8:2-11. [PMID: 32039275 PMCID: PMC6969949 DOI: 10.30476/ijcbnm.2019.73900.0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Background There is a high prevalence of multiple sclerosis (MS) among Iranian women of reproductive age. However, very few studies have been conducted in Iran to assess the experiences with and concerns about pregnancy and motherhood of such patients. The present study was conducted to better understand the experiences and concerns of women with MS about pregnancy and motherhood. Methods The present qualitative study was conducted from August 2016 to January 2017 among Iranian women with MS visiting the Iran MS Society in Tehran, Iran. The purposive sampling method was used to recruit the participants and the sampling was continued until data saturation. Based on the inclusion criteria, a total of 25 women with MS were recruited in the study. The data were collected by in-depth semi-structured face-to-face interviews and analyzed using the MAXQDA 10 software. Results The analysis of the interview data resulted in four main categories, namely "Pregnancy concerns", "Fear of failing as a parent", "Feeling of threatened fertility", and "Lack of social support". The results showed that Iranian women with MS avoided pregnancy due to the negative effects of the disease on their physical abilities and on life in general. They were also concerned about possible infertility, the effect of MS medications on their menstrual cycle, and the limitations of infertility treatments due to the presence of MS. These concerns led them to postpone pregnancy and lose time or opt for voluntary childlessness and consequently miss out on the experience of motherhood. Conclusion MS poses a serious challenge to women who consider getting pregnant and wish to experience motherhood. Health care professionals should support such patients to overcome their concerns and indecisiveness by providing appropriate information and counseling.
Collapse
Affiliation(s)
- Faezeh Ghafoori
- Department of Reproductive Health, School of Nursing and Midwifery, Tehran University of Medical Sciences, Tehran, Iran.,Ministery of Health and Medical Education, Tehran, Iran
| | - Nahid Dehghan-Nayeri
- Department of Nursing Management, School of Nursing and Midwifery, Tehran University of Medical Sciences, Tehran, Iran
| | - Zohreh Khakbazan
- Department of Reproductive Health, School of Nursing and Midwifery, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Seyed Massood Nabavi
- Department of Brain and Cognitive Sciences, Royan Institute for Stem Cell Biology and Technology, Royan Institute, Tehran, Iran
| |
Collapse
|
41
|
Hosotani M, Ichii O, Nakamura T, Masum MA, Otani Y, Elewa YHA, Kon Y. Altered ciliary morphofunction in the oviductal infundibulum of systemic autoimmune disease-prone MRL/MpJ-Faslpr/lpr mice. Cell Tissue Res 2020; 380:627-641. [DOI: 10.1007/s00441-020-03175-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/20/2020] [Indexed: 12/11/2022]
|
42
|
Adamska A, Łebkowska A, Krentowska A, Hryniewicka J, Adamski M, Leśniewska M, Polak AM, Kowalska I. Ovarian Reserve and Serum Concentration of Thyroid Peroxidase Antibodies in Euthyroid Women With Different Polycystic Ovary Syndrome Phenotypes. Front Endocrinol (Lausanne) 2020; 11:440. [PMID: 32849259 PMCID: PMC7399035 DOI: 10.3389/fendo.2020.00440] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 06/04/2020] [Indexed: 11/25/2022] Open
Abstract
Objective: It has been shown that women with polycystic ovary syndrome (PCOS), as well as Hashimoto's thyroiditis (HT), are characterized by increased incidence of infertility. Serum anti-Müllerian hormone (AMH), which reflects ovarian reserve, is elevated in PCOS women and is decreased in women with HT. The Rotterdam criteria recognize four clinical PCOS phenotypes, i.e., phenotypes A, B, C, and D. The aim of the present study was to investigate the relation between serum concentrations of thyroid peroxidase antibodies (TPOAbs) and ovarian reserve in different PCOS phenotypes. Patients and methods: We examined 141 women with PCOS [phenotype A was diagnosed in 67 (47.5%) women, phenotype B in 30 (21.3%), phenotype C in 28 (19.9%), and phenotype D in 16 (11.3%)] and 88 control subjects of similar age; all women were euthyroid. Serum concentrations of AMH, thyroid-stimulating hormone (TSH), thyroid hormones, and TPOAbs were assessed. Results: We observed positive serum TPOAbs in 21.9% women with PCOS and in 23.9% controls (p = 0.07). We did not find differences in the frequency of detection of positive serum TPOAbs between phenotypes A, B, and C and the control group (p > 0.05). We did not observe a difference in AMH levels between TPOAbs-positive and TPOAbs-negative women, both in the control group and the PCOS women (all p > 0.05). However, serum AMH concentration was markedly higher in the whole PCOS group (p < 0.01) and in phenotype A (p < 0.01) vs. controls when the serum concentration of TPOAbs was negative. In the groups with positive serum levels of TPOAbs, serum concentration of AMH did not differ between PCOS phenotypes and controls (p = 0.23). Additionally, we observed that serum AMH concentration was related to the level of TPOAbs in the PCOS group (r = -0.4, p = 0.02). Conclusions: The frequency of serum detection of positive TPOAbs did not differ between PCOS phenotypes with clinical/biochemical hyperandrogenism and the control group. The observation of the difference in serum AMH between the PCOS and control groups only in TPOAbs negative women together with the inverse relation of TPOAbs with serum AMH only in the PCOS group might suggest that ovarian reserve is influenced by TPOAbs in PCOS.
Collapse
Affiliation(s)
- Agnieszka Adamska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Białystok, Białystok, Poland
- *Correspondence: Agnieszka Adamska
| | - Agnieszka Łebkowska
- Department of Internal Medicine and Metabolic Diseases, Medical University of Białystok, Białystok, Poland
| | - Anna Krentowska
- Department of Internal Medicine and Metabolic Diseases, Medical University of Białystok, Białystok, Poland
| | - Justyna Hryniewicka
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Białystok, Białystok, Poland
| | - Marcin Adamski
- Faculty of Computer Science, Bialystok University of Technology, Białystok, Poland
| | - Monika Leśniewska
- Department of Reproduction and Gynecological Endocrinology, Medical University of Białystok, Białystok, Poland
| | - Aleksandra Maria Polak
- Department of Internal Medicine and Metabolic Diseases, Medical University of Białystok, Białystok, Poland
| | - Irina Kowalska
- Department of Internal Medicine and Metabolic Diseases, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
43
|
Elevated blood mercury level has a non-linear association with infertility in U.S. women: Data from the NHANES 2013-2016. Reprod Toxicol 2019; 91:53-58. [PMID: 31756438 DOI: 10.1016/j.reprotox.2019.11.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 12/18/2022]
Abstract
Mercury is a ubiquitous toxic heavy metal associated with an increased risk of female infertility; however, the evidence supporting this is limited and controversial. We aimed to explore the relationship between the total blood mercury and infertility in 1796 selected participants from the National Health and Nutrition Examination Survey (NHANES) (2013-2016). We found no significant association between mercury and infertility based on a fully-adjusted model (OR 1.04; 95 % CI 0.91, 1.19), and the results remained robust in a series of sensitive analysis. However, a non-linear relationship was detected. By a two-piecewise linear regression model and recursive algorithm, we identified an inflection point of 5.278 μg/L, when blood mercury was >5.278 μg/L, a 1-unit increase in mercury (log2) was associated with 157 % greater adjusted odds of infertility (OR 2.57; 95 % CI 1.12, 5.87). Our findings provide new insights to advance the research of the link between mercury and infertility.
Collapse
|
44
|
Saei Ghare Naz M, Ramezani Tehrani F, Ahmadi F, Alavi Majd H, Ozgoli G. Threats to Feminine Identity as the Main Concern of Iranian Adolescents with Polycystic Ovary Syndrome: A Qualitative Study. J Pediatr Nurs 2019; 49:e42-e47. [PMID: 31645273 DOI: 10.1016/j.pedn.2019.08.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 08/13/2019] [Accepted: 08/13/2019] [Indexed: 10/25/2022]
Abstract
PURPOSE Polycystic Ovary Syndrome (PCOS) is a prevalent hormonal disorder in adolescents; this study seeks to elaborate the main concerns of adolescents with PCOS. DESIGN AND METHODS This qualitative content analysis was conducted on 15 adolescents with PCOS using purposive sampling in 2018. Semi-structured in-depth interviews were held for data collection, and data saturation occurred when no new data were being obtained. Data were analyzed concurrently with data collection. RESULTS Fifteen adolescents with PCOS aged 13-19 years were enrolled into the study. Thirteen were single and two were married. The data analysis led to the extraction of "threats to feminine identity" as the main theme of this research, which was then divided into two categories: 1) concerns about attractiveness, with subcategories including fashion-related stress and the loss of physical beauty 2) concerns about femininity, with subcategories including the underlying issues of worrying about future marriage prospects, anxiety about infertility in the future and stress about menstrual abnormalities. CONCLUSION According to the results of this study, attractiveness and threat to femininity is the main concern of adolescents with PCOS that should be further considered in the management of their treatment. IMPLICATIONS FOR PRACTICE Understanding the main concerns of adolescents with PCOS can help healthcare professionals better manage this disease in this population subgroup.
Collapse
Affiliation(s)
- Marzieh Saei Ghare Naz
- Student Research Committee, Department of Midwifery and Reproductive Health, School of Nursing and Midwifery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fahimeh Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fazlollah Ahmadi
- Department of Nursing, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hamid Alavi Majd
- Department of Biostatistics, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Giti Ozgoli
- Department of Midwifery and Reproductive Health, School of Nursing and Midwifery, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
45
|
Che D, Yang Y, Xu Y, Fang Z, Pi L, Fu L, Zhou H, Tan Y, Lu Z, Li L, Liang Q, Xuan Q, Gu X. The lncRNA MALAT1 rs619586 G Variant Confers Decreased Susceptibility to Recurrent Miscarriage. Front Physiol 2019; 10:385. [PMID: 31024342 PMCID: PMC6465954 DOI: 10.3389/fphys.2019.00385] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 03/21/2019] [Indexed: 12/20/2022] Open
Abstract
Cardiovascula disease and recurrent miscarriage have shared risk factors, and some cardiovascular disease-related candidate genes have been confirmed to be associated with recurrent miscarriage. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a long non-coding RNA (lncRNA) that is considered to be associated with susceptibility to cardiovascular disease. However, whether lncRNA MALAT1 polymorphisms are related to recurrent miscarriage susceptibility is unclear. We genotyped three lncRNA MALAT1 polymorphisms (rs591291, rs619586, and rs3200401) in 284 patients and 392 controls using TaqMan methods. Logistic regression was used to evaluate the odds ratios (ORs) and 95% confidence intervals (CIs) adjusted for age. Our results showed that the rs619586 G variant had protective effects against recurrent miscarriage (AG vs. AA: adjusted OR = 0.670, 95% CI = 0.457–0.982, p = 0.040; GG vs. AA: adjusted OR = 0.278, 95% CI = 0.079–0.975, p = 0.046; GG/AG vs. AA adjusted OR = 0.621, 95% CI = 0.429–0.900, p = 0.012). In a combined analyses of protective genotypes, with regard to the three single nucleotide polymorphisms (SNPs), we found that individuals with two or three protective genotypes exhibited a significantly lower risk of recurrent miscarriage than those with no or only one protective genotype (adjusted OR = 0.369, 95% CI = 0.199–0.684, p = 0.002). Moreover, the decrease in recurrent miscarriage risk with two or three protective genotypes was most pronounced in women less than 35 years of age (OR = 0.290, 95% CI = 0.142–0.589, p < 0.001) and in women with 2–3 miscarriages (adjusted OR = 0.270, 95% CI = 0.126–0.580, p < 0.001). In conclusion, our study suggests that the rs619586 G variant may have potential protective effects conferring a decreased risk of recurrent miscarriage in the southern Chinese population.
Collapse
Affiliation(s)
- Di Che
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yanfang Yang
- Department of Prenatal Diagnosis, Maoming People's Hospital, Maoming, China
| | - Yufen Xu
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Zhenzhen Fang
- Program of Molecular Medicine, Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lei Pi
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - LanYan Fu
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Huazhong Zhou
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yaqian Tan
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Zhaoliang Lu
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Li Li
- Department of Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Qihua Liang
- Department of Clinical Lab, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Qingshan Xuan
- Department of Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiaoqiong Gu
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Department of Clinical Lab, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Department of Blood Transfusion, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
46
|
Decreased Expression of Cannabinoid Receptors in the Eutopic and Ectopic Endometrium of Patients with Adenomyosis. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5468954. [PMID: 30800671 PMCID: PMC6360557 DOI: 10.1155/2019/5468954] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 01/08/2019] [Indexed: 01/12/2023]
Abstract
Objective Adenomyosis is a common gynecologic benign disease that may have a life-long negative impact on women. Previous studies have indicated that the endocannabinoid system may participate in the progress of endometriosis. Our research aims to analyze the expression patterns of the typical cannabinoid receptors (CB1 and CB2), the main constituents of the endocannabinoid system, in endometrial samples derived from patients diagnosed as adenomyosis or not. Methods Eutopic and corresponding ectopic endometrium from 45 premenopausal women diagnosed as adenomyosis and normal endometrium from 34 age-matched women lacking evidence of adenomyosis were examined by immunohistochemistry and quantitative real-time polymerase chain reaction (qRT-PCR) to determine the CB1 and CB2 expression levels. Results In either the proliferative or the secretory phase, CB1 and CB2 protein and mRNA levels were both significantly lower in the eutopic and ectopic endometrium of adenomyosis when compared with normal endometrium. For women with adenomyosis, CB1 and CB2 protein and mRNA levels were much lower in the ectopic endometrium than the eutopic in both phases of the cycle. Both CB1 and CB2 protein and mRNA levels were increased during the secretory phase in normal endometrium, while CB1 lost its cyclic variation in the eutopic and ectopic endometrium from patients diagnosed as adenomyosis. Conclusion The decreased expression of CB1 and CB2 in the eutopic and ectopic endometrium from patients diagnosed as adenomyosis suggests that cannabinoid receptors may participate in the pathogenesis of adenomyosis.
Collapse
|
47
|
Inflamm-Aging: A New Mechanism Affecting Premature Ovarian Insufficiency. J Immunol Res 2019; 2019:8069898. [PMID: 30719458 PMCID: PMC6334348 DOI: 10.1155/2019/8069898] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 11/15/2018] [Indexed: 11/18/2022] Open
Abstract
The normal function of ovaries, along with the secretion of sex hormones, is among the most important endocrine factors that maintain the female sexual characteristics and promote follicular development and ovulation. Premature ovarian insufficiency (POI) is a common cause in the etiology of female infertility. It is defined as the loss of ovarian function before the age of 40. The characteristics of POI are menstrual disorders, including amenorrhea and delayed menstruation, accompanied by a raised gonadotrophin level and decreased estradiol level. Inflammatory aging is a new concept in the research field of aging. It refers to a chronic and low-degree proinflammatory state which occurs with increasing age. Inflammatory aging is closely associated with multiple diseases, as excessive inflammation can induce the inflammatory lesions in certain organs of the body. In recent years, studies have shown that inflammatory aging plays a significant role in the pathogenesis of POI. This paper begins with the pathogenesis of inflammatory aging and summarizes the relationship between inflammatory aging and premature ovarian insufficiency in a comprehensive way, as well as discussing the new diagnostic and therapeutic methods of POI.
Collapse
|
48
|
Romitti M, Fabris VC, Ziegelmann PK, Maia AL, Spritzer PM. Association between PCOS and autoimmune thyroid disease: a systematic review and meta-analysis. Endocr Connect 2018; 7:1158-1167. [PMID: 30352422 PMCID: PMC6215798 DOI: 10.1530/ec-18-0309] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/10/2018] [Indexed: 12/11/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most prevalent endocrine disorder affecting women of reproductive age. PCOS has been associated with distinct metabolic and cardiovascular diseases and with autoimmune conditions, predominantly autoimmune thyroid disease (AITD). AITD has been reported in 18-40% of PCOS women, depending on PCOS diagnostic criteria and ethnicity. The aim of this systematic review and meta-analysis was to summarize the available evidence regarding the likelihood of women with PCOS also having AITD in comparison to a reference group of non-PCOS women. We systematically searched EMBASE and MEDLINE for non-interventional case control, cross-sectional, or cohort studies published until August 2017. The Ottawa-Newcastle Scale was used to assess the methodological quality of studies. Statistical meta-analysis was performed with R. Thirteen studies were selected for the present analysis, including 1,210 women diagnosed with PCOS and 987 healthy controls. AITD was observed in 26.03% and 9.72% of PCOS and control groups respectively. A significant association was detected between PCOS and chance of AITD (OR= 3.27, 95%CI 2.32-4.63). Notably, after geographical stratification, the higher risk of AITD in PCOS women persisted for Asians (OR= 4.56, 95%CI 2.47-8.43), Europeans (OR= 3.27, 95%CI 2.07-5.15), and South Americans (OR= 1.86, 95 %CI 1.05-3.29). AIDT is a frequent condition in PCOS patients, and might affect thyroid function. Thus, screening for thyroid function and thyroid-specific autoantibodies should be considered in patients with PCOS even in the absence of overt symptoms. This systematic review and meta-analysis is registered in PROSPERO under number CRD42017079676.
Collapse
Affiliation(s)
- Mírian Romitti
- Thyroid UnitEndocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Vitor C Fabris
- Gynecological Endocrinology UnitEndocrine Division, Hospital de Clínicas de Porto Alegre, and Department of Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Patricia K Ziegelmann
- Postgraduate Program in Epidemiology and Department of StatisticsInstitute of Mathematics, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Ana Luiza Maia
- Thyroid UnitEndocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Poli Mara Spritzer
- Gynecological Endocrinology UnitEndocrine Division, Hospital de Clínicas de Porto Alegre, and Department of Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Correspondence should be addressed to P M Spritzer:
| |
Collapse
|
49
|
Rao M, Zeng Z, Zhao S, Tang L. Effect of levothyroxine supplementation on pregnancy outcomes in women with subclinical hypothyroidism and thyroid autoimmuneity undergoing in vitro fertilization/intracytoplasmic sperm injection: an updated meta-analysis of randomized controlled trials. Reprod Biol Endocrinol 2018; 16:92. [PMID: 30249251 PMCID: PMC6154908 DOI: 10.1186/s12958-018-0410-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 09/14/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Evidence suggests that subclinical hypothyroidism (SCH) and thyroid autoimmunity (TAI) are associated with adverse pregnancy outcomes. This systematic review and meta-analysis was conducted to determine whether levothyroxine (LT4) supplementation would improve pregnancy outcomes among infertile women with SCH and/or TAI who underwent in vitro fertilization (IVF) or intracytoplastic sperm injection (ICSI). METHODS We searched databases of PubMed, EMBASE, Web of Science, Cochrane Controlled Trials Register databases, and Clinicaltrials.gov up to April 2018 to identify eligible studies. Studies that focused on the treatment effect of LT4 on pregnancy outcomes of women with SCH and/or TAI who underwent IVF/ICSI were included in the data synthesis. We only included randomized controlled trials (RCTs). Relative risks (RR) and 95% confidence intervals (CI) were calculated using a random-effects model to assess the results of pregnancy outcomes, including clinical pregnancy rate, miscarriage rate, live birth rate and preterm birth rate. RESULTS Four published RCTs including 787 infertile couples undergoing IVF/ICSI were included in this meta-analysis. Notably, the study observed no significant associations of LT4 treatment with the clinical pregnancy rate (RR = 1.46, 95% CI: 0.86-2.48), live birth rate (RR = 2.05, 95% CI: 0.96-4.36), or preterm birth rate (RR = 1.13, 95% CI: 0.65-1.96). However, patients receiving LT4 supplementation had a significantly decreased miscarriage rate relative to those receiving a placebo or no treatment (RR = 0.51, 95% CI: 0.32-0.82). A further sub-group analysis showed that LT4 supplementation did not improve the miscarriage rates among patients with SCH (RR = 0.67, 95% CI: 0.39-1.15) or TAI (RR = 0.28, 95% CI: 0.07-1.06). CONCLUSIONS Given its potential to reduce the miscarriage rate, LT4 supplementation is recommended for infertile women with SCH and/or TAI who are undergoing IVF/ICSI. However, additional population-based RCTs are needed to confirm this recommendation.
Collapse
Affiliation(s)
- Meng Rao
- Department of Reproduction and Genetics, the First Affiliated Hospital of Kunming Medical University, No. 295 Xi Chang road, Kunming, 650032, China
| | - Zhengyan Zeng
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Shuhua Zhao
- Department of Reproduction and Genetics, the First Affiliated Hospital of Kunming Medical University, No. 295 Xi Chang road, Kunming, 650032, China
| | - Li Tang
- Department of Reproduction and Genetics, the First Affiliated Hospital of Kunming Medical University, No. 295 Xi Chang road, Kunming, 650032, China.
| |
Collapse
|
50
|
Ding C, Zou Q, Ding J, Ling M, Wang W, Li H, Huang B. Increased N6-methyladenosine causes infertility is associated with FTO expression. J Cell Physiol 2018; 233:7055-7066. [PMID: 29384212 DOI: 10.1002/jcp.26507] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/24/2018] [Indexed: 12/25/2022]
Abstract
The N6-methyladenosine (m6A) modification plays a central role in epigenetic regulation of the mammalian transcriptome. m6A can be demethylated by the fat mass- and obesity-associated (FTO) protein and the α-ketoglutarate-dependent dioxygenase alkB homolog 5 (ALKBH5) protein. Much less is known about that whether m6A content is involved in POI (premature ovarian insufficiency) disease. In this case-controlled study, 69 POI and 53 tubal occlusion patients were recruited from the reproduction centers in our hospital. For the POI animal model experiment, ovarian tissue was obtained from ten POI and nine healthy mice. An m6A test kit was developed to determine the m6A content in the RNA, and qPCR and western blot were used to examine the mRNA and protein expression levels of FTO and ALKBH5. FACS was used to measure the levels of proliferation and apoptosis, and siRNA was used to establish FTO and ALKBH5 knockdown cell lines. Our results showed that the m6A content in the RNA from POI patients and POI mice was significantly higher than control groups and that POI was characterized by the content of m6A. The mRNA and protein expression levels of FTO were significantly lower in the POI patients than control group and were associated with a risk of POI. These data suggest that the decreased mRNA and protein expression levels of FTO may be responsible for the increase in m6A in POI, which may further increase the risk of complications of POI. High m6A should be investigated further as a novel potential biomarker of POI.
Collapse
Affiliation(s)
- Chenyue Ding
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Qinyan Zou
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Jie Ding
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Mingfa Ling
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Wei Wang
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Hong Li
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Boxian Huang
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China.,Central Laboratory, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|