1
|
Britz SM, Nelson S, Earhart KM, Pru JK, Schmitt EE. Circadian Disruption Impacts Fetal Development in Mice Using High-Frequency Ultrasound. J Circadian Rhythms 2024; 22:4. [PMID: 39712938 PMCID: PMC11661015 DOI: 10.5334/jcr.249] [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: 09/25/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024] Open
Abstract
The developmental origins of health and disease theory suggests that environmental exposures during early life, particularly during prenatal life, can greatly influence health status later in life. Irregular light-dark cycles, such as those experienced during shift work, result in the repeated disruption of circadian rhythms, which negatively impacts physiological and behavioral cycles. The purpose of our study was to assess parameters in the developing mouse embryo and fetus using high frequency ultrasound when exposed to circadian disruption. Pregnant female mice were subjected to a seven-hour advanced circadian disrupted protocol or remained on a normal 12/12 light-dark cycle throughout pregnancy. Significant differences were observed in placental length (p = 0.00016), placental thickness (p = 0.0332), and stomach diameter (p = 0.0186) at E14.5-18.5. These findings suggest that circadian disruption in pregnant dams, mimicking shift work, alters embryonic and fetal development in specific organs in utero.
Collapse
Affiliation(s)
- Samantha M. Britz
- WWAMI Medical Education, University of Washington School of Medicine, Seattle, WA, US
| | - Shay Nelson
- WWAMI Medical Education, University of Washington School of Medicine, Seattle, WA, US
| | - Kylie M. Earhart
- Division of Kinesiology & Health, University of Wyoming, Laramie, WY, US
| | - James K. Pru
- Program in Reproductive Biology, Department of Animal Science, University of Wyoming, Laramie, WY, US
| | - Emily E. Schmitt
- WWAMI Medical Education, University of Washington School of Medicine, Seattle, WA, US
- Division of Kinesiology & Health, University of Wyoming, Laramie, WY, US
| |
Collapse
|
2
|
Kloster AK, Adami LNG, Moysés-Oliveira M, Hachul H, Andersen ML, Tufik S. Exploring the Link between Premature Ovarian Insufficiency, Insomnia and Circadian Pathways. JBRA Assist Reprod 2024; 28:567-589. [PMID: 39254468 PMCID: PMC11622404 DOI: 10.5935/1518-0557.20240052] [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: 01/05/2024] [Accepted: 07/25/2024] [Indexed: 09/11/2024] Open
Abstract
OBJECTIVE To establish an interaction network for genes related to premature ovarian insufficiency (POI) and insomnia, and to identify biological processes that connect POI to the physiological clock. METHODS Previously reported lists of genes associated to POI and insomnia were contrasted and their intersection was used as input on protein-protein interaction analyses. POI-associated genes were contrasted with gene expression markers for neural circadian control and enriched pathways among their shared content were dissected. RESULTS The functional network generated from the intersection between POI and insomnia gene lists pointed to the central nervous system as the most relevant cellular context for this connection. After identifying POI-associated genes that play a role in neural circadian patterns, we observed the disruption of pathways related to the hypothalamic-pituitary-gonadal axis as the major genetic link between ovarian function and circadian neural circuits. CONCLUSIONS These findings highlight neurological mechanisms that support the POI-insomnia interplay.
Collapse
Affiliation(s)
- Anna K. Kloster
- Sleep Institute, Associação Fundo de Incentivo
à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São
Paulo, São Paulo, Brazil
| | - Luana N. G. Adami
- Sleep Institute, Associação Fundo de Incentivo
à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São
Paulo, São Paulo, Brazil
| | | | - Helena Hachul
- Sleep Institute, Associação Fundo de Incentivo
à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São
Paulo, São Paulo, Brazil
| | - Monica L. Andersen
- Sleep Institute, Associação Fundo de Incentivo
à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São
Paulo, São Paulo, Brazil
| | - Sergio Tufik
- Sleep Institute, Associação Fundo de Incentivo
à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São
Paulo, São Paulo, Brazil
| |
Collapse
|
3
|
Weng L, Hong H, Zhang Q, Xiao C, Zhang Q, Wang Q, Huang J, Lai D. Sleep Deprivation Triggers the Excessive Activation of Ovarian Primordial Follicles via β2 Adrenergic Receptor Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402393. [PMID: 39229959 PMCID: PMC11538700 DOI: 10.1002/advs.202402393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/23/2024] [Indexed: 09/05/2024]
Abstract
Sleep deprivation (SD) is observed to adversely affect the reproductive health of women. However, its precise physiological mechanisms remain largely elusive. In this study, using a mouse model of SD, it is demonstrated that SD induces the depletion of ovarian primordial follicles, a phenomenon not attributed to immune-mediated attacks or sympathetic nervous system activation. Rather, the excessive secretion of stress hormones, namely norepinephrine (NE) and epinephrine (E), by overactive adrenal glands, has emerged as a key mediator. The communication pathway mediated by the KIT ligand (KITL)-KIT between granulosa cells and oocytes plays a pivotal role in primordial follicle activation. SD heightened the levels of NE/E that stimulates the activation of the KITL-KIT/PI3K and mTOR signaling cascade in an β2 adrenergic receptor (ADRB2)-dependent manner, thereby promoting primordial follicle activation and consequent primordial follicle loss in vivo. In vitro experiments further corroborate these observations, revealing that ADRB2 upregulates KITL expression in granulosa cells via the activation of the downstream cAMP/PKA pathway. Together, these results reveal the significant involvement of ADRB2 signaling in the depletion of ovarian primordial follicles under sleep-deprived conditions. Additionally, ADRB2 antagonists are proposed for the treatment or prevention of excessive activation of primordial follicles induced by SD.
Collapse
Affiliation(s)
- Lichun Weng
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Hanqing Hong
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Qinyu Zhang
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Chengqi Xiao
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Qiuwan Zhang
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Qian Wang
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Ju Huang
- Songjiang Hospital and Songjiang Research InstituteShanghai Key Laboratory of Emotions and Affective DisordersShanghai Jiao Tong University School of MedicineShanghai201600China
| | - Dongmei Lai
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| |
Collapse
|
4
|
Le DC, Ngo MHT, Kuo YC, Chen SH, Lin CY, Ling TY, Pham QTT, Au HK, Myung J, Huang YH. Secretome from estrogen-responding human placenta-derived mesenchymal stem cells rescues ovarian function and circadian rhythm in mice with cyclophosphamide-induced primary ovarian insufficiency. J Biomed Sci 2024; 31:95. [PMID: 39390588 PMCID: PMC11468397 DOI: 10.1186/s12929-024-01085-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Primary ovarian insufficiency (POI) is an early decline in ovarian function that leads to ovarian failure. Conventional treatments for POI are inadequate, and treatments based on mesenchymal stem cells (MSCs) have emerged as an option. However, the lack of consideration of the estrogen niche in ovarian tissue significantly reduces the therapeutic efficacy, with an unclear mechanism in the MSCs in POI treatment. Furthermore, the disruption of circadian rhythm associated with POI has not been previously addressed. METHODS Conditioned medium (CM) and estradiol-conditioned medium (E2-CM) were generated from estrogen receptor positive MSCs (ER+pcMSCs). Chemotherapy-induced POI models were established using C57BL/6 mice (in vivo) and KGN cells (in vitro) treated with cyclophosphamide (CTX) or 4-hydroperoxycyclophosphamide (4-OOH-CP). Gene/protein expressions were detected using RT-qPCR, Western blotting, and immunohistochemistry assays. Locomotor activity was monitored for behavioral circadian rhythmicity. Cytokine arrays and miRNA analysis were conducted to analyze potential factors within CM/E2-CM. RESULTS The secretome of ER+pcMSCs (CM and E2-CM) significantly reduced the CTX-induced defects in ovarian folliculogenesis and circadian rhythm. CM/E2-CM also reduced granulosa cell apoptosis and rescued angiogenesis in POI ovarian tissues. E2-CM had a more favorable effect than the CM. Notably, ER+pcMSC secretome restored CTX-induced circadian rhythm defects, including the gene expressions associated with the ovarian circadian clock (e.g., Rora, E4bp4, Rev-erbα, Per2 and Dbp) and locomotor activity. Additionally, the cytokine array analysis revealed a significant increase in cytokines and growth factors associated with immunomodulation and angiogenesis, including angiogenin. Neutralizing the angiogenin in CM/E2-CM significantly reduced its ability to promote HUVEC tube formation in vitro. Exosomal miRNA analysis revealed the miRNAs involved in targeting the genes associated with POI rescue (PTEN and PDCD4), apoptosis (caspase-3, BIM), estrogen synthesis (CYP19A1), ovarian clock regulation (E4BP4, REV-ERBα) and fibrosis (COL1A1). CONCLUSION This study is the first to demonstrate that, in considering the estrogen niche in ovarian tissue, an estrogen-priming ER+pcMSC secretome achieved ovarian regeneration and restored the circadian rhythm in a CTX-induced POI mouse model. The potential factors involved include angiogenin and exosomal miRNAs in the ER+pcMSC secretome. These findings offer insights into potential stem cell therapies for chemotherapy-induced POI and circadian rhythm disruption.
Collapse
Affiliation(s)
- Duy-Cuong Le
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Laboratory, Vinmec International Hospital, Minh Khai Street, Hai Ba Trung, Hanoi, Vietnam
| | - Mai-Huong T Ngo
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Yung-Che Kuo
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Shu-Hwa Chen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chung-Yen Lin
- Institute of Information Science, Academia Sinica, Taipei, 11529, Taiwan
- Institute of Fishery Sciences, College of Life Science, National Taiwan University, Taipei, 10617, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, 10617, Taiwan
| | - Thai-Yen Ling
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Quoc Thao Trang Pham
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Heng-Kien Au
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, 11042, Taiwan.
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, 11042, Taiwan.
| | - Jihwan Myung
- Graduate Institute of Mind, Brain and Consciousness, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Brain and Consciousness Research Centre (BCRC), TMU-Shuang Ho Hospital, New Taipei City, 23561, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Wuxing Street, Taipei, 11031, Taiwan.
| | - Yen-Hua Huang
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, 11042, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Wuxing Street, Taipei, 11031, Taiwan.
| |
Collapse
|
5
|
Luo B, Song J, Zhang J, Han J, Zhou X, Chen L. The contribution of circadian clock to the biological processes. Front Mol Biosci 2024; 11:1387576. [PMID: 38903177 PMCID: PMC11187296 DOI: 10.3389/fmolb.2024.1387576] [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: 02/18/2024] [Accepted: 05/20/2024] [Indexed: 06/22/2024] Open
Abstract
All organisms have various circadian, behavioral, and physiological 24-h periodic rhythms, which are controlled by the circadian clock. The circadian clock controls various behavioral and physiological rhythms. In mammals, the primary circadian clock is present in the suprachiasmatic nucleus of the hypothalamus. The rhythm of the circadian clock is controlled by the interaction between negative and positive feedback loops, consisting of crucial clock regulators (including Bmal1 and Clock), three cycles (mPer1, mPer2, and mPer3), and two cryptochromes (Cry1 and Cry2). The development of early mammalian embryos is an ordered and complex biological process that includes stages from fertilized eggs to blastocysts and undergoes important morphological changes, such as blastocyst formation, cell multiplication, and compaction. The circadian clock affects the onset and timing of embryonic development. The circadian clock affects many biological processes, including eating time, immune function, sleep, energy metabolism, and endocrinology, therefore, it is also crucial for overall health, growth and development after birth. This review summarized the effects of the circadian clock in the body's physiological activities. A new strategy is proposed for the prevention of malformations or diseases by regulating the circadian clock or changing circadian rhythms.
Collapse
Affiliation(s)
- Beibei Luo
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Jiangyuan Song
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Jiaqi Zhang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Jun Han
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Xin Zhou
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| |
Collapse
|
6
|
Chen R, Qin Y, Du J, Liu J, Dai S, Lei M, Zhu H. Circadian clock gene BMAL1 regulates STAR expression in goose ovarian preovulatory granulosa cells. Poult Sci 2023; 102:103159. [PMID: 37871489 PMCID: PMC10598734 DOI: 10.1016/j.psj.2023.103159] [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: 07/10/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/25/2023] Open
Abstract
The ovarian circadian clock plays a regulatory role in the avian ovulation-oviposition cycle. However, little is known regarding the ovarian circadian clock of geese. In this study, we investigated rhythmic changes in clock genes over a 48-h period and identified potential clock-controlled genes involved in progesterone synthesis in goose ovarian preovulatory granulosa cells. The results showed that BMAL1, CRY1, and CRY2, as well as 4 genes (LHR, STAR, CYP11A1, and HSD3B) involved in progesterone synthesis exhibited rhythmic expression patterns in goose ovarian preovulatory granulosa cells over a 48-h period. Knockdown of BMAL1 decreased the progesterone concentration and downregulated STAR mRNA and protein levels in goose ovarian preovulatory granulosa cells. Overexpression of BMAL1 increased the progesterone concentration and upregulated the STAR mRNA level in goose ovarian preovulatory granulosa cells. Moreover, we demonstrated that the BMAL1/CLOCK complex activated the transcription of goose STAR gene by binding to an E-box motif. These results suggest that the circadian clock is involved in the regulation of progesterone synthesis in goose ovarian preovulatory granulosa cells by orchestrating the transcription of steroidogenesis-related genes.
Collapse
Affiliation(s)
- Rong Chen
- Key Laboratory of Crop and Animal Integrated Farming, Ministry of Agriculture and Rural Affairs, Nanjing, China; Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Yifei Qin
- Key Laboratory of Crop and Animal Integrated Farming, Ministry of Agriculture and Rural Affairs, Nanjing, China; Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Jie Du
- Animal Husbandry and Veterinary College, Jiangsu Vocational College of Agriculture and Forestry, Jurong, Jiangsu, China
| | - Jie Liu
- Key Laboratory of Crop and Animal Integrated Farming, Ministry of Agriculture and Rural Affairs, Nanjing, China; Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Shudi Dai
- Key Laboratory of Crop and Animal Integrated Farming, Ministry of Agriculture and Rural Affairs, Nanjing, China; Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Mingming Lei
- Key Laboratory of Crop and Animal Integrated Farming, Ministry of Agriculture and Rural Affairs, Nanjing, China; Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Huanxi Zhu
- Key Laboratory of Crop and Animal Integrated Farming, Ministry of Agriculture and Rural Affairs, Nanjing, China; Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, China.
| |
Collapse
|
7
|
Bora G, Önel T, Yıldırım E, Yaba A. Circadian regulation of mTORC1 signaling via Per2 dependent mechanism disrupts folliculogenesis and oocyte maturation in female mice. J Mol Histol 2023:10.1007/s10735-023-10126-9. [PMID: 37162693 DOI: 10.1007/s10735-023-10126-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 05/04/2023] [Indexed: 05/11/2023]
Abstract
mTOR (mammalian target of Rapamycin) is an important signaling pathway involved in several crucial ovarian functions including folliculogenesis and oocyte maturation. The circadian rhythm regulates multiple physiological processes and PER2 is one of the core circadian rhythm components. mTOR is regulated by the circadian clock and in turn, the rhythmic mTOR activities strengthen the clock function. Our current study aims to investigate a possible interconnection between the circadian clock and the mTORC1 signaling pathway in folliculogenesis and oocyte maturation. Here we demonstrate that the circadian system regulates mTORC1 signaling via Per2 dependent mechanism in the mouse ovary. To investigate the effect of constant light on ovarian and oocyte morphology, animals were housed 12:12 h L:D group in standard lightening conditions and the 12:12 h L:L group in constant light for one week. Food intake and body weight changes were measured. Ovarian morphology, follicle counting, and oocyte aging were evaluated. Afterward, western blot for mTOR, p-mTOR, p70S6K, p-p70S6K, PER2, and Caspase-3 protein levels was performed. The study demonstrated that circadian rhythm disruption caused an alteration in their food intake and decrease in primordial follicle numbers and an increase in the number of atretic follicles. It caused an increase in oxidative stress and a decrease in ZP3 expression in oocytes. Decreased protein levels of mTOR, p-mTOR, p70S6K, and PER2 were shown. The results showed that the circadian clock regulates mTORC1 through PER2 dependent mechanism and that decreased mTORC1 activity can contribute to premature aging of mouse ovary. In conclusion, these results suggest that the circadian clock may control ovarian aging by regulating mTOR signaling pathway through Per2 expression.
Collapse
Affiliation(s)
- Gizem Bora
- Department of Histology and Embryology, Yeditepe University Faculty of Medicine, 34755, İstanbul, Turkey
| | - Tuğçe Önel
- Department of Histology and Embryology, Yeditepe University Faculty of Medicine, 34755, İstanbul, Turkey
| | - Ecem Yıldırım
- Department of Histology and Embryology, Yeditepe University Faculty of Medicine, 34755, İstanbul, Turkey
| | - Aylin Yaba
- Department of Histology and Embryology, Yeditepe University Faculty of Medicine, 34755, İstanbul, Turkey.
| |
Collapse
|
8
|
Freeman JR, Whitcomb BW, Bertone-Johnson ER, Balzer LB, O'Brien LM, Dunietz GL, Purdue-Smithe AC, Kim K, Silver RM, Schisterman EF, Mumford SL. Preconception sleep duration, sleep timing, and shift work in association with fecundability and live birth among women with a history of pregnancy loss. Fertil Steril 2023; 119:252-263. [PMID: 36586812 PMCID: PMC9899515 DOI: 10.1016/j.fertnstert.2022.10.026] [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: 02/14/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVE To evaluate the associations between preconception sleep characteristics and shift work with fecundability and live birth. DESIGN Secondary analysis of the Effects of Aspirin in Gestation and Reproduction study, a preconception cohort. SETTING Four US academic medical centers. PATIENT(S) Women aged 18-40 with a history of 1-2 pregnancy losses who were attempting to conceive again. INTERVENTION(S) Not applicable. MAIN OUTCOME MEASURES(S) We evaluated baseline, self-reported sleep duration, sleep midpoint, social jetlag, and shift work among 1,228 women who were observed for ≤6 cycles of pregnancy attempts to ascertain fecundability. We ascertained live birth at the end of follow up via chart abstraction. We estimated fecundability odds ratios (FORs) using discrete, Cox proportional hazards models and risk ratios (RRs) for live birth using log-Poisson models. RESULT(S) Sleep duration ≥9 vs. 7 to <8 hours (FOR: 0.81, 95% confidence interval [CI], 0.61; 1.08), later sleep midpoints (3rd tertile vs. 2nd tertile: FOR: 0.85; 95% CI, 0.69, 1.04) and social jetlag (continuous per hour; FOR: 0.93, 95% CI: 0.86, 1.00) were not associated with reduced fecundability. In sensitivity analyses, excluding shift workers, sleep duration ≥9 vs. 7 to <8 hours (FOR: 0.62; 95% CI, 0.42; 0.93) was associated with low fecundability. Night shift work was not associated with fecundability (vs. non-night shift work FOR: 1.17, 95% CI, 0.96; 1.42). Preconception sleep was not associated with live birth. CONCLUSION(S) Overall, there does not appear to be a strong association between sleep characteristics, fecundability, and live birth. Although these findings may suggest weak and imprecise associations with some sleep characteristics, our findings should be evaluated in larger cohorts of women with extremes of sleep characteristics. CLINICAL TRIAL REGISTRATION NUMBER Clinicaltrials.gov NCT00467363.
Collapse
Affiliation(s)
- Joshua R Freeman
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, Massachusetts; Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Brian W Whitcomb
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, Massachusetts
| | - Elizabeth R Bertone-Johnson
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, Massachusetts; Department of Health Promotion and Policy, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, Massachusetts
| | - Laura B Balzer
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, Massachusetts
| | - Louise M O'Brien
- Division of Sleep Medicine, Department of Neurology, University of Michigan, Ann Arbor, Michigan; Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
| | - Galit L Dunietz
- Division of Sleep Medicine, Department of Neurology, University of Michigan, Ann Arbor, Michigan
| | - Alexandra C Purdue-Smithe
- Division of Women's Health, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Keewan Kim
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Robert M Silver
- Department of Obstetrics and Gynecology, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Enrique F Schisterman
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sunni L Mumford
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
9
|
Gong X, Zhang Y, Ai J, Li K. Application of Single-Cell RNA Sequencing in Ovarian Development. Biomolecules 2022; 13:47. [PMID: 36671432 PMCID: PMC9855652 DOI: 10.3390/biom13010047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 12/28/2022] Open
Abstract
The ovary is a female reproductive organ that plays a key role in fertility and the maintenance of endocrine homeostasis, which is of great importance to women's health. It is characterized by a high heterogeneity, with different cellular subpopulations primarily containing oocytes, granulosa cells, stromal cells, endothelial cells, vascular smooth muscle cells, and diverse immune cell types. Each has unique and important functions. From the fetal period to old age, the ovary experiences continuous structural and functional changes, with the gene expression of each cell type undergoing dramatic changes. In addition, ovarian development strongly relies on the communication between germ and somatic cells. Compared to traditional bulk RNA sequencing techniques, the single-cell RNA sequencing (scRNA-seq) approach has substantial advantages in analyzing individual cells within an ever-changing and complicated tissue, classifying them into cell types, characterizing single cells, delineating the cellular developmental trajectory, and studying cell-to-cell interactions. In this review, we present single-cell transcriptome mapping of the ovary, summarize the characteristics of the important constituent cells of the ovary and the critical cellular developmental processes, and describe key signaling pathways for cell-to-cell communication in the ovary, as revealed by scRNA-seq. This review will undoubtedly improve our understanding of the characteristics of ovarian cells and development, thus enabling the identification of novel therapeutic targets for ovarian-related diseases.
Collapse
Affiliation(s)
| | | | - Jihui Ai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kezhen Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
10
|
Xu LN, Li HT, Liu S, Jiang J, Liu YQ, Cheng HYM, Yu Y, Cao JM, Zhang P. Constitutional delay of growth and puberty in female mice is induced by circadian rhythm disruption in utero. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 241:113723. [PMID: 35679725 DOI: 10.1016/j.ecoenv.2022.113723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 05/27/2022] [Accepted: 05/29/2022] [Indexed: 06/15/2023]
Abstract
Constitutional delay of growth and puberty (CDGP) refers to the late onset of puberty. CDGP is associated with poor psychosocial outcomes and elevated risk of cardiovascular and osteoporotic diseases, especially in women. The environmental factors that contribute to CDGP are poorly understood. Here, we investigated the effects of chronic circadian disturbance (CCD) during the fetal stage on the pubertal development of female mice. Compared to non-stressed female (NS-F) mice that were not exposed to CCD in utero, adolescent CCD female (CCD-F) mice exhibited phenotypes that were consistent with CDGP, including lower body weight, reduced levels of circulating gonadal hormones, decreased expression of gonadal hormones and steroid synthesis-related enzymes in the ovary and hypothalamus, irregular estrus cycles, and tardive vaginal introitus initial opening (VO) days (equivalent to the menarche). Phenotypic differences in the above-noted parameters were not observed in CCD-F mice once they had reached adulthood. The expression of genes involved in fatty acid metabolism was perturbed in the ovary and hypothalamus of CCD-F mice. In addition, the ovaries of these animals exhibited altered diurnal expression profiles of circadian clock genes. Together, our findings not only suggest that CCD during fetal development may result in delayed puberty in female mice, they also offer insights on potential mechanisms that underlie CDGP.
Collapse
Affiliation(s)
- Lin-Na Xu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Hui-Ting Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Shuang Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Jie Jiang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Ya-Qin Liu
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hai-Ying Mary Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| | - Yang Yu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China; Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China.
| | - Ji-Min Cao
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China.
| | - Peng Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
11
|
Johnson BS, Krishna MB, Padmanabhan RA, Pillai SM, Jayakrishnan K, Laloraya M. Derailed peripheral circadian genes in polycystic ovary syndrome patients alters peripheral conversion of androgens synthesis. Hum Reprod 2022; 37:1835-1855. [PMID: 35728080 DOI: 10.1093/humrep/deac139] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION Do circadian genes exhibit an altered profile in peripheral blood mononuclear cells (PBMCs) of polycystic ovary syndrome (PCOS) patients and do they have a potential role in androgen excess? SUMMARY ANSWER Our findings revealed that an impaired circadian clock could hamper the regulation of peripheral steroid metabolism in PCOS women. WHAT IS KNOWN ALREADY PCOS patients exhibit features of metabolic syndrome. Circadian rhythm disruption is involved in the development of metabolic diseases and subfertility. An association between shift work and the incidence of PCOS in females was recently reported. STUDY DESIGN, SIZE, DURATION This is a retrospective case-referent study in which peripheral blood samples were obtained from 101 control and 101 PCOS subjects. PCOS diagnoses were based on Rotterdam Consensus criteria. PARTICIPANTS/MATERIALS, SETTING, METHODS This study comprised 101 women with PCOS and 101 control volunteers, as well as Swiss albino mice treated with dehydroepiandrosterone (DHEA) to induce PCOS development. Gene expression analyses of circadian and steroidogenesis genes in human PBMC and mice ovaries and blood were executed by quantitative real-time PCR. MAIN RESULTS AND THE ROLE OF CHANCE We observed aberrant expression of peripheral circadian clock genes in PCOS, with a significant reduction in the core clock genes, circadian locomotor output cycles kaput (CLOCK) (P ≤ 0.00001), brain and muscle ARNT-like 1 (BMAL1) (P ≤ 0.00001) and NPAS2 (P ≤ 0.001), and upregulation of their negative feedback loop genes, CRY1 (P ≤ 0.00003), CRY2 (P ≤ 0.00006), PER1 (P ≤ 0.003), PER2 (P ≤ 0.002), DEC1 (P ≤ 0.0001) and DEC2 (P ≤ 0.00005). Transcript levels of an additional feedback loop regulating BMAL1 showed varied expression, with reduced RORA (P ≤ 0.008) and increased NR1D1 (P ≤ 0.02) in PCOS patients in comparison with the control group. We also demonstrated the expression pattern of clock genes in PBMCs of PCOS women at three different time points. PCOS patients also exhibited increased mRNA levels of steroidogenic enzymes like StAR (P ≤ 0.0005), CYP17A1 (P ≤ 0.005), SRD5A1 (P ≤ 0.00006) and SRD5A2 (P ≤ 0.009). Knockdown of CLOCK/BMAL1 in PBMCs resulted in a significant reduction in estradiol production, by reducing CYP19A1 and a significant increase in dihydrotestosterone production, by upregulating SRD5A1 and SRD5A2 in PBMCs. Our data also showed that CYP17A1 as a direct CLOCK-BMAL1 target in PBMCs. Phenotypic classification of PCOS subgroups showed a higher variation in expression of clock genes and steroidogenesis genes with phenotype A of PCOS. In alignment with the above results, altered expression of ovarian core clock genes (Clock, Bmal1 and Per2) was found in DHEA-treated PCOS mice. The expression of peripheral blood core clock genes in DHEA-induced PCOS mice was less robust and showed a loss of periodicity in comparison with that of control mice. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION We could not evaluate the circadian oscillation of clock genes and clock-controlled genes over a 24-h period in the peripheral blood of control versus PCOS subjects. Additionally, circadian genes in the ovaries of PCOS women could not be evaluated due to limitations in sample availability, hence we employed the androgen excess mouse model of PCOS for ovarian circadian assessment. Clock genes were assessed in the whole ovary of the androgen excess mouse model of PCOS rather than in granulosa cells, which is another limitation of the present work. WIDER IMPLICATIONS OF THE FINDINGS Our observations suggest that the biological clock is one of the contributing factors in androgen excess in PCOS, owing to its potential role in modulating peripheral androgen metabolism. Considering the increasing prevalence of PCOS and the rising frequency of delayed circadian rhythms and insufficient sleep among women, our study emphasizes the potential in modulating circadian rhythm as an important strategy in PCOS management, and further research on this aspect is highly warranted. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the RGCB-DBT Core Funds and a grant (#BT/PR29996/MED/97/472/2020) from the Department of Biotechnology (DBT), India, to M.L. B.S.J. was supported by a DST/INSPIRE Fellowship/2015/IF150361 and M.B.K. was supported by the Research Fellowship from Council of Scientific & Industrial Research (CSIR) (10.2(5)/2007(ii).E.U.II). The authors declare no competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Betcy Susan Johnson
- Female Reproduction and Metabolic Syndromes Laboratory, Division of Molecular Reproduction, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Research Scholar, Research Centre, University of Kerala, Thiruvananthapuram, Kerala, India
| | - Meera B Krishna
- Female Reproduction and Metabolic Syndromes Laboratory, Division of Molecular Reproduction, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Renjini A Padmanabhan
- Female Reproduction and Metabolic Syndromes Laboratory, Division of Molecular Reproduction, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | | | - K Jayakrishnan
- KJK Hospital and Fertility Research Centre, Thiruvananthapuram, Kerala, India
| | - Malini Laloraya
- Female Reproduction and Metabolic Syndromes Laboratory, Division of Molecular Reproduction, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
12
|
Roenneberg T, Foster RG, Klerman EB. The circadian system, sleep, and the health/disease balance: a conceptual review. J Sleep Res 2022; 31:e13621. [PMID: 35670313 PMCID: PMC9352354 DOI: 10.1111/jsr.13621] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 11/30/2022]
Abstract
The field of "circadian medicine" is a recent addition to chronobiology and sleep research efforts. It represents a logical step arising from the increasing insights into the circadian system and its interactions with life in urbanised societies; applying these insights to the health/disease balance at home and in the medical practice (outpatient) and clinic (inpatient). Despite its fast expansion and proliferating research efforts, circadian medicine lacks a formal framework to categorise the many observations describing interactions among the circadian system, sleep, and the health/disease balance. A good framework allows us to categorise observations and then assign them to one or more components with hypothesised interactions. Such assignments can lead to experiments that document causal (rather than correlational) relationships and move from describing observations to discovering mechanisms. This review details such a proposed formal framework for circadian medicine and will hopefully trigger discussion among our colleagues, so that the framework can be improved and expanded. As the basis of the framework for circadian medicine, we define "circadian health" and how it links to general health. We then define interactions among the circadian system, sleep, and the health/disease balance and put the framework into the context of the literature with examples from six domains of health/disease balance: fertility, cancer, immune system, mental health, cardiovascular, and metabolism.
Collapse
Affiliation(s)
- Till Roenneberg
- Institute of Medical Psychology and Institute for Occupational, Social and Environmental Medicine, Munich, Germany
| | - Russell G Foster
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, New Biochemistry Building, University of Oxford, Oxford, UK
| | - Elizabeth B Klerman
- Department of Neurology, Massachusetts General Hospital, Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Suyama A, Iwata N, Soejima Y, Nakano Y, Yamamoto K, Nada T, Otsuka F. Roles of NR5A1 and NR5A2 in the regulation of steroidogenesis by Clock gene and bone morphogenetic proteins by human granulosa cells. Endocr J 2021; 68:1283-1291. [PMID: 34176817 DOI: 10.1507/endocrj.ej21-0223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The functional role of the transcription factors NR5A1 and NR5A2 and their interaction with Clock gene and bone morphogenetic proteins (BMPs) were investigated in human granulosa KGN cells. Treatment with BMP-15 and GDF-9 suppressed forskolin (FSK)-induced steroidogenesis as shown by the mRNA expression levels of StAR and P450scc but not the mRNA expression level of P450arom. Of interest, treatment with BMP-15 and GDF-9 also suppressed FSK-induced NR5A2 mRNA expression. Treatment with BMP-15 suppressed NR5A2 mRNA and protein expression but increased Clock mRNA and protein expression levels by granulosa cells. The mRNA expression levels of NR5A1, but not those of NR5A2, were positively correlated with the levels of Clock mRNA, while the mRNA levels of Id-1, the target gene of BMP signaling, were positively correlated with those of NR5A1 but not with those of NR5A2. It was also demonstrated that the mRNA expression levels of NR5A1 were positively correlated with those of P450arom and 3βHSD, whereas the mRNA expression level of NR5A2 was correlated with those of StAR and P450scc. Furthermore, inhibition of Clock gene expression by siRNA attenuated the expression of NR5A1, and the mRNA levels of Clock gene were significantly correlated with those of NR5A1. Collectively, the results suggested a novel mechanism by which Clock gene expression induced by BMP-15 is functionally linked to the expression of NR5A1, whereas NR5A2 expression is suppressed by BMP-15 in granulosa cells. The interaction between Clock NR5A1/NR5A2 and BMP-15 is likely to be involved in the fine-tuning of steroidogenesis by ovarian granulosa cells.
Collapse
Affiliation(s)
- Atsuhito Suyama
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Nahoko Iwata
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Yoshiaki Soejima
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Yasuhiro Nakano
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Koichiro Yamamoto
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Takahiro Nada
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Fumio Otsuka
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
14
|
Androulakis IP. Circadian rhythms and the HPA axis: A systems view. WIREs Mech Dis 2021; 13:e1518. [PMID: 33438348 DOI: 10.1002/wsbm.1518] [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: 07/10/2020] [Revised: 11/20/2020] [Accepted: 11/30/2020] [Indexed: 12/26/2022]
Abstract
The circadian timing system comprises a network of time-keeping clocks distributed across a living host whose responsibility is to allocate resources and distribute functions temporally to optimize fitness. The molecular structures generating these rhythms have evolved to accommodate the rotation of the earth in an attempt to primarily match the light/dark periods during the 24-hr day. To maintain synchrony of timing across and within tissues, information from the central clock, located in the suprachiasmatic nucleus, is conveyed using systemic signals. Leading among those signals are endocrine hormones, and while the hypothalamic-pituitary-adrenal axis through the release of glucocorticoids is a major pacesetter. Interestingly, the fundamental units at the molecular and physiological scales that generate local and systemic signals share critical structural properties. These properties enable time-keeping systems to generate rhythmic signals and allow them to adopt specific properties as they interact with each other and the external environment. The purpose of this review is to provide a broad overview of these structures, discuss their functional characteristics, and describe some of their fundamental properties as these related to health and disease. This article is categorized under: Immune System Diseases > Computational Models Immune System Diseases > Biomedical Engineering.
Collapse
Affiliation(s)
- Ioannis P Androulakis
- Biomedical Engineering Department, Chemical & Biochemical Engineering Department, Rutgers University, New Brunswick, New Jersey.,Department of Surgery, Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| |
Collapse
|
15
|
Wang F, Xie N, Wu Y, Zhang Q, Zhu Y, Dai M, Zhou J, Pan J, Tang M, Cheng Q, Shi B, Guo Q, Li X, Xie L, Wang B, Yang D, Weng Q, Guo L, Ye J, Pan M, Zhang S, Zhou H, Zhen C, Liu P, Ning K, Brackenridge L, Hardiman PJ, Qu F. Association between circadian rhythm disruption and polycystic ovary syndrome. Fertil Steril 2020; 115:771-781. [PMID: 33358334 DOI: 10.1016/j.fertnstert.2020.08.1425] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To explore the association of circadian rhythm disruption with polycystic ovary syndrome (PCOS) and the potential underlying mechanism in ovarian granulosa cells (GCs). DESIGN Multicenter questionnaire-based survey, in vivo and ex vivo studies. SETTING Twelve hospitals in China, animal research center, and research laboratory of a women's hospital. PATIENTS/ANIMALS A total of 436 PCOS case subjects and 715 control subjects were recruited for the survey. In vivo and ex vivo studies were conducted in PCOS-model rats and on ovarian GCs collected from women with PCOS and control subjects. INTERVENTION(S) The PCOS rat model was established with the use of testosterone propionate. MAIN OUTCOME MEASURE(S) Assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq), RNA sequencing, rhythmicity analysis, functional enrichment analysis. RESULT(S) There was a significant correlation between night shift work and PCOS. PCOS-model rats presented distinct differences in the circadian variation of corticotropin-releasing hormone, adrenocorticotropic hormone, prolactin, and a 4-h phase delay in thyrotropic hormone levels. The motif enrichment analysis of ATAC-seq revealed the absence of clock-related transcription factors in specific peaks of PCOS group, and RNA sequencing ex vivo at various time points over 24 hours demonstrated the differential rhythmic expression patterns of women with PCOS. Kyoto Encyclopedia of Genes and Genomes analysis further highlighted metabolic dysfunction, including both carbohydrate and amino acid metabolism and the tricarboxylic acid cycle. CONCLUSION(S) There is a significant association of night shift work with PCOS, and genome-wide chronodisruption exists in ovarian GCs.
Collapse
Affiliation(s)
- Fangfang Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Institute for Women's Health, University College London, London, United Kingdom
| | - Ningning Xie
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yan Wu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Qing Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yuhang Zhu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Minchen Dai
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jue Zhou
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China; Institute for Women's Health, University College London, London, United Kingdom
| | - Jiexue Pan
- First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Mengling Tang
- School of Public Health, Zhejiang University, Hangzhou, China
| | - Qi Cheng
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Biwei Shi
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Qinyuan Guo
- Maternal and Child Healthcare Hospital of Liuzhou, Liuzhou, China
| | - Xinling Li
- Maternal and Child Healthcare Hospital of Liuzhou, Liuzhou, China
| | - Lifeng Xie
- Maternal and Child Healthcare Hospital of Liuzhou, Liuzhou, China
| | - Bing Wang
- Second Hospital of Jiaxing, Jiaxing, China
| | - Dongxia Yang
- Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qing Weng
- First People's Hospital of Yuhang District of Hangzhou, Hangzhou, China
| | - Lanzhong Guo
- Dongyang Women's and Children's Hospital, Dongyang, China
| | - Jisheng Ye
- Dongyang Women's and Children's Hospital, Dongyang, China
| | - Mingwo Pan
- Guangdong Women and Children Hospital, Guangzhou, China
| | - Shuyi Zhang
- Baiyin City Maternity and Childcare Hospital, Baiyin, China
| | - Hua Zhou
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cailan Zhen
- People's Hospital of Lucheng, Lucheng, China
| | - Ping Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second Hospital, Chengdu, People's Republic of China
| | - Ke Ning
- Department of Social Science, Institute of Education, University College London, London, United Kingdom
| | - Lisa Brackenridge
- Institute for Women's Health, University College London, London, United Kingdom
| | - Paul J Hardiman
- Institute for Women's Health, University College London, London, United Kingdom
| | - Fan Qu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Institute for Women's Health, University College London, London, United Kingdom.
| |
Collapse
|
16
|
Xu Y, Wang L, Cao S, Hu R, Liu R, Hua K, Guo Z, Di HJ, Hu Z. Genipin improves reproductive health problems caused by circadian disruption in male mice. Reprod Biol Endocrinol 2020; 18:122. [PMID: 33308222 PMCID: PMC7731552 DOI: 10.1186/s12958-020-00679-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 12/06/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Circadian rhythm disruption impacts a wide range of physiological processes, including fertility. However, the effect of circadian disruption on male spermatogenesis and fertility, and treatments for these effects have been largely unexplored at the molecular level. METHODS In this study, we examined the effects of genipin on improving the reproductive health problems caused by circadian disruption. Three groups of animals were fed under different conditions: control group (normal T cycle with saline), group of shortened T cycles (Light/Dark = 4 hours/4 hours) with saline, and a group of shortened T cycles with genipin by oral gavage. The male fertility was evaluated by fertility study and pups parameters analysis after successful sexual behavior and mating with female mice. We sacrificed the treated animals after 5 or 10 weeks and collected the testis, sperm and serum for histological analysis, sperm motility assay, and serum hormone detection, respectively. Furthermore, the effect of genipin was assessed by detection of progesterone secretion and steroidogenic key proteins expression, including StAR and CYP11A1, in mouse Leydig tumor MLTC-1 cells. RESULTS Male mice exposed to shortened light-dark cycles, much shorter than 24 hours, had reduced fertility with decreased sperm concentrations and sperm motility. Male mice under circadian disruption have reduced testis size and abnormal morphology, leading to lower fertility rates, reduced litter size and pup body weight. Treatment with exogenous genipin, a natural plant-derived compound, alleviated circadian disruption-induced damage to fertility and spermatogenesis and normalized testosterone, dihydrotestosterone (DHT), and androstenedione (ASD) levels in the male mice. The levels of key proteins involved in steroidogenesis, StAR and CYP11A1, were reduced in mouse testes after the circadian disruption, but genipin treatment restored the reduction. The mRNA expression of SRD5A1, which encodes an androgen synthesis enzyme, was also upregulated by genipin treatment. Furthermore, genipin treatment showed a positive effect on steroidogenesis in MLTC-1 cells, resulting in an increase in hormone secretion and the upregulation of StAR and CYP11A1. CONCLUSIONS Our results showed an association between circadian disruption and reproductive health problems in male mice and indicated that treatments with genipin have positive effects on the reproductive health of male mice with circadian rhythm disorders.
Collapse
Affiliation(s)
- Yihua Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, 210023, Nanjing, China
| | - Li Wang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, 210023, Nanjing, China
| | - Siyuan Cao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, 210023, Nanjing, China
| | - Ruihua Hu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, 210023, Nanjing, China
| | - Rui Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, 210023, Nanjing, China
| | - Ke Hua
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, 210023, Nanjing, China
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, 210023, Nanjing, China
| | - Hong-Jie Di
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, 210017, Nanjing, China.
| | - Zhigang Hu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, 210023, Nanjing, China.
| |
Collapse
|
17
|
Bedont JL, Iascone DM, Sehgal A. The Lineage Before Time: Circadian and Nonclassical Clock Influences on Development. Annu Rev Cell Dev Biol 2020; 36:469-509. [PMID: 33021821 PMCID: PMC10826104 DOI: 10.1146/annurev-cellbio-100818-125454] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Diverse factors including metabolism, chromatin remodeling, and mitotic kinetics influence development at the cellular level. These factors are well known to interact with the circadian transcriptional-translational feedback loop (TTFL) after its emergence. What is only recently becoming clear, however, is how metabolism, mitosis, and epigenetics may become organized in a coordinated cyclical precursor signaling module in pluripotent cells prior to the onset of TTFL cycling. We propose that both the precursor module and the TTFL module constrain cellular identity when they are active during development, and that the emergence of these modules themselves is a key lineage marker. Here we review the component pathways underlying these ideas; how proliferation, specification, and differentiation decisions in both developmental and adult stem cell populations are or are not regulated by the classical TTFL; and emerging evidence that we propose implies a primordial clock that precedes the classical TTFL and influences early developmental decisions.
Collapse
Affiliation(s)
- Joseph Lewis Bedont
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| | - Daniel Maxim Iascone
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| | - Amita Sehgal
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
- The Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
18
|
Yaw AM, Duong TV, Nguyen D, Hoffmann HM. Circadian rhythms in the mouse reproductive axis during the estrous cycle and pregnancy. J Neurosci Res 2020; 99:294-308. [PMID: 32128870 DOI: 10.1002/jnr.24606] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/17/2020] [Accepted: 02/12/2020] [Indexed: 12/26/2022]
Abstract
Molecular and behavioral timekeeping is regulated by the circadian system which includes the brain's suprachiasmatic nucleus (SCN) that translates environmental light information into neuronal and endocrine signals aligning peripheral tissue rhythms to the time of day. Despite the critical role of circadian rhythms in fertility, it remains unexplored how circadian rhythms change within reproductive tissues during pregnancy. To determine how estrous cycle and pregnancy impact phase relationships of reproductive tissues, we used PER2::Luciferase (PER2::LUC) circadian reporter mice and determined the time of day of PER2::LUC peak (phase) in the SCN, pituitary, uterus, and ovary. The relationships between reproductive tissue PER2::LUC phases changed throughout the estrous cycle and late pregnancy and were accompanied by changes to PER2::LUC period in the SCN, uterus, and ovary. To determine if the phase relationship adaptations were driven by sex steroids, we asked if progesterone, a hormone involved in estrous cyclicity and pregnancy, could regulate Per2-luciferase expression. Using an in vitro transfection assay, we found that progesterone increased Per2-luciferase expression in immortalized SCN (SCN2.2) and arcuate nucleus (KTAR) cells. In addition, progesterone shortened PER2::LUC period in ex vivo uterine tissue recordings collected during pregnancy. As progesterone dramatically increases during pregnancy, we evaluated wheel-running patterns in PER2::LUC mice. We confirmed that activity levels decrease during pregnancy and found that activity onset was delayed. Although SCN, but not arcuate nucleus, PER2::LUC period changed during late pregnancy, onset of locomotor activity did not correlate with SCN or arcuate nucleus PER2::LUC period.
Collapse
Affiliation(s)
- Alexandra M Yaw
- Department of Animal Science and the Reproductive and Developmental Science Program, Michigan State University, East Lansing, MI, USA
| | - Thu V Duong
- Department of Animal Science and the Reproductive and Developmental Science Program, Michigan State University, East Lansing, MI, USA
| | - Duong Nguyen
- Department of Animal Science and the Reproductive and Developmental Science Program, Michigan State University, East Lansing, MI, USA
| | - Hanne M Hoffmann
- Department of Animal Science and the Reproductive and Developmental Science Program, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
19
|
Hohos NM, Elliott EM, Cho KJ, Lin IS, Rudolph MC, Skaznik-Wikiel ME. High-fat diet-induced dysregulation of ovarian gene expression is restored with chronic omega-3 fatty acid supplementation. Mol Cell Endocrinol 2020; 499:110615. [PMID: 31628964 PMCID: PMC6878773 DOI: 10.1016/j.mce.2019.110615] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/09/2019] [Accepted: 10/09/2019] [Indexed: 12/26/2022]
Abstract
Chronic high-fat diet (HFD) consumption causes ovarian dysfunction in rodents. Acute dietary treatment with docosahexaenoic acid (DHA) increases oocyte quality and ovarian reserve at advanced reproductive age. We hypothesized that DHA supplementation after HFD exposure reverses HFD-induced ovarian defects. We conducted a dietary intervention with reversal to chow, DHA-supplemented chow, or DHA-supplemented HFD after HFD consumption. After 10 weeks, HFD-fed mice had impaired estrous cyclicity, decreased primordial follicles, and altered ovarian expression of 24 genes compared to chow controls. Diet reversal to either chow or chow + DHA restored estrous cyclicity, however only chow + DHA appeared to mitigated the impact of HFD on ovarian reserve. All dietary interventions restored HFD-dysregulated gene expression to chow levels. We found no association between follicular fluid DHA levels and ovarian reserve. In conclusion our data suggest some benefit of DHA supplementation after HFD, particularly in regards to ovarian gene expression, however complete restoration of ovarian function was not achieved.
Collapse
Affiliation(s)
- Natalie M Hohos
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Emily M Elliott
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kirstin J Cho
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ivy S Lin
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Michael C Rudolph
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, 12700 East 19th Ave, Aurora, CO, 80045, USA
| | | |
Collapse
|
20
|
Mills J, Kuohung W. Impact of circadian rhythms on female reproduction and infertility treatment success. Curr Opin Endocrinol Diabetes Obes 2019; 26:317-321. [PMID: 31644470 DOI: 10.1097/med.0000000000000511] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW In recent years, new studies have investigated the role and influence of sleep on female fertility and early pregnancy outcomes, providing a growing body of knowledge demonstrating how regulation by sleep of hormones are important to reproduction, and how disruptions in sleep, circadian rhythms, and genes regulating circadian rhythmicity can negatively impact fertility and early pregnancy outcomes. This review aims to summarize the most recent research on the relationship among circadian rhythms, fertility, and early pregnancy outcomes in women, and to explore possible fertility interventions. RECENT FINDINGS Recent studies have found altered levels of FSH, LH, and prolactin with sleep disturbance or circadian dysrhythmia. Disruption of circadian rhythms in the form of shift work, jet lag, and daylight savings time changes have been associated with poorer fertility and early pregnancy outcomes. Alterations in the expression of circadian rhythm-regulating circadian locomotor output cycles kaput (CLOCK) genes have been associated with decreased fertility and increased rates of miscarriage. SUMMARY Overall, undisrupted sleep and circadian rhythmicity appear to optimize fertility and early pregnancy outcomes and may play an important role in the success of fertility treatment.
Collapse
Affiliation(s)
- Jacqueline Mills
- Department of Obstetrics and Gynecology, Boston Medical Center, Boston, Massachusetts, USA
| | | |
Collapse
|
21
|
Day JR, David A, Barbosa MGDM, Brunette MA, Cascalho M, Shikanov A. Encapsulation of ovarian allograft precludes immune rejection and promotes restoration of endocrine function in immune-competent ovariectomized mice. Sci Rep 2019; 9:16614. [PMID: 31719632 PMCID: PMC6851353 DOI: 10.1038/s41598-019-53075-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 10/15/2019] [Indexed: 12/22/2022] Open
Abstract
Premature ovarian insufficiency (POI) is a significant complication of cytotoxic treatments due to extreme ovarian sensitivity to chemotherapy and radiation. POI is particularly devastating for young girls reaching puberty, because it irreversibly affects their physical and cognitive development. Changes occurring during puberty determine their height, bone health, insulin responsiveness, lipid metabolism, cardiovascular health and cognition. The only available treatment for POI during puberty is hormone replacement therapy (HRT), which delivers non-physiological levels of estrogen, lacks other ovarian hormones and pulsatility, and is not responsive to feedback regulation. Here we report that ovarian allografts encapsulated in a hydrogel-based capsule and implanted in ovariectomized mice restore ovarian endocrine function in immune competent mice. Ovarian tissue from BALB/c mice was encapsulated in poly(ethylene-glycol) (PEG) hydrogels, with a proteolytically degradable core and a non-degradable shell. The dual capsules were implanted subcutaneously in immune competent ovariectomized C57BL/6 mice for a period of 60 days. As expected, non-encapsulated ovarian allografts implanted in a control group sensitized the recipients as confirmed with donor-specific IgG in the serum, which increased 26-fold in the 3 weeks following transplantation (p = 0.02) and infiltration of the graft with CD8 T cells consistent with allo-immunity. In contrast, encapsulation in the Dual PEG capsules prevented sensitization to the allograft in all the recipients with no evidence of lymphocytic infiltration. In summary, the approach of hydrogel-based immunoisolation presents a minimally invasive and robust cell-therapy to restore hormonal balance in ovarian insufficiency. This report is the first to demonstrate the application of a tunable PEG-based hydrogel as an immunoisolator of allogeneic ovarian tissue to restore endocrine function in ovariectomized mice and prevent cell-mediated immune rejection in immune competent mice.
Collapse
Affiliation(s)
- James Ronald Day
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA
| | - Anu David
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA
| | - Mayara Garcia de Mattos Barbosa
- Department of Surgery, University of Michigan, Ann Arbor, USA
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, USA
| | | | - Marilia Cascalho
- Department of Surgery, University of Michigan, Ann Arbor, USA
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, USA
| | - Ariella Shikanov
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA.
- Department of Macromolecular Science & Engineering, University of Michigan, Ann Arbor, USA.
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, USA.
| |
Collapse
|
22
|
Laine VN, Verhagen I, Mateman AC, Pijl A, Williams TD, Gienapp P, van Oers K, Visser ME. Exploration of tissue-specific gene expression patterns underlying timing of breeding in contrasting temperature environments in a song bird. BMC Genomics 2019; 20:693. [PMID: 31477015 PMCID: PMC6720064 DOI: 10.1186/s12864-019-6043-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/19/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Seasonal timing of breeding is a life history trait with major fitness consequences but the genetic basis of the physiological mechanism underlying it, and how gene expression is affected by date and temperature, is not well known. In order to study this, we measured patterns of gene expression over different time points in three different tissues of the hypothalamic-pituitary-gonadal-liver axis, and investigated specifically how temperature affects this axis during breeding. We studied female great tits (Parus major) from lines artificially selected for early and late timing of breeding that were housed in two contrasting temperature environments in climate-controlled aviaries. We collected hypothalamus, liver and ovary samples at three different time points (before and after onset of egg-laying). For each tissue, we sequenced whole transcriptomes of 12 pools (n = 3 females) to analyse gene expression. RESULTS Birds from the selection lines differed in expression especially for one gene with clear reproductive functions, zona pellucida glycoprotein 4 (ZP4), which has also been shown to be under selection in these lines. Genes were differentially expressed at different time points in all tissues and most of the differentially expressed genes between the two temperature treatments were found in the liver. We identified a set of hub genes from all the tissues which showed high association to hormonal functions, suggesting that they have a core function in timing of breeding. We also found ample differentially expressed genes with largely unknown functions in birds. CONCLUSIONS We found differentially expressed genes associated with selection line and temperature treatment. Interestingly, the latter mainly in the liver suggesting that temperature effects on egg-laying date may happen down-stream in the physiological pathway. These findings, as well as our datasets, will further the knowledge of the mechanisms of tissue-specific avian seasonality in the future.
Collapse
Affiliation(s)
- Veronika N. Laine
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), P.O. Box 50, 6700 AB Wageningen, The Netherlands
| | - Irene Verhagen
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), P.O. Box 50, 6700 AB Wageningen, The Netherlands
| | - A. Christa Mateman
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), P.O. Box 50, 6700 AB Wageningen, The Netherlands
| | - Agata Pijl
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), P.O. Box 50, 6700 AB Wageningen, The Netherlands
| | - Tony D. Williams
- Department of Biological Sciences, Simon Fraser University, Burnaby, Canada
| | - Phillip Gienapp
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), P.O. Box 50, 6700 AB Wageningen, The Netherlands
| | - Kees van Oers
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), P.O. Box 50, 6700 AB Wageningen, The Netherlands
| | - Marcel E. Visser
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), P.O. Box 50, 6700 AB Wageningen, The Netherlands
| |
Collapse
|
23
|
Ye P, Li M, Liao W, Ge K, Jin S, Zhang C, Chen X, Geng Z. Hypothalamic transcriptome analysis reveals the neuroendocrine mechanisms in controlling broodiness of Muscovy duck (Cairina moschata). PLoS One 2019; 14:e0207050. [PMID: 31071089 PMCID: PMC6508920 DOI: 10.1371/journal.pone.0207050] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 04/22/2019] [Indexed: 02/07/2023] Open
Abstract
Broodiness, one of the maternal behaviors and instincts for natural breeding in birds, is an interesting topic in reproductive biology. Broodiness in poultry is characterized by persistent nesting, usually associated with cessation of egg laying. The study of avian broodiness is essential for bird conservation breeding and commercial poultry industry. In this study, we examined the hypothalamus transcriptome of Muscovy duck in three reproductive stages, including egg-laying anaphase (LA), brooding prophase (BP) and brooding metaphase (BM). Differences in gene expression during the transition from egg-laying to broodiness were examined, and 155, 379, 292 differently expressed genes (DEGs) were obtained by pairwise comparisons of LA-vs-BP, LA-vs-BM and BP-vs-BM, respectively (fold change≥1.5, P < 0.05). Gene Ontology Term (GO) enrichment analysis suggested a possible role of oxidative stress in the hypothalamus might invoke reproductive costs that potentially change genes expression. KEGG analysis revealed glutamatergic synapse, dopaminergic synapse, serotonergic synapse and GABAergic synapse pathway were significantly enriched, and regulator genes were identified. Eight gene expression patterns were illustrated by trend analysis and further clustered into three clusters. Additional six hub genes were identified through combining trend analysis and protein-protein interaction (PPI) analysis. Our results suggested that the cyclical mechanisms of reproductive function conversion include effects of oxidative stress, biosynthesis of neurotransmitters or their receptors, and interactions between glucocorticoids and thyroid hormones and regulatory genes. These candidate genes and biological pathways may be used as targets for artificial manipulation and marker-assisted breeding in the reproductive behavior.
Collapse
Affiliation(s)
- Pengfei Ye
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, P.R. China
| | - Min Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, P.R. China
| | - Wang Liao
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, P.R. China
| | - Kai Ge
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, P.R. China
- College of Biological and Pharmaceutical Engineering, West Anhui University, Liuan, China
| | - Sihua Jin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, P.R. China
| | - Cheng Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, P.R. China
| | - Xingyong Chen
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, P.R. China
| | - Zhaoyu Geng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, P.R. China
| |
Collapse
|
24
|
Daily rhythms of expression in reproductive genes along the brain-pituitary-gonad axis and liver of zebrafish. Comp Biochem Physiol A Mol Integr Physiol 2019; 231:158-169. [DOI: 10.1016/j.cbpa.2019.02.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/19/2019] [Accepted: 02/16/2019] [Indexed: 12/26/2022]
|
25
|
Nagao S, Iwata N, Soejima Y, Takiguchi T, Aokage T, Kozato Y, Nakano Y, Nada T, Hasegawa T, Otsuka F. Interaction of ovarian steroidogenesis and clock gene expression modulated by bone morphogenetic protein-7 in human granulosa cells. Endocr J 2019; 66:157-164. [PMID: 30518737 DOI: 10.1507/endocrj.ej18-0423] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
A functional link between clock gene expression and ovarian steroidogenesis was studied using human granulosa KGN cells. Similarities between changes in the mRNA and protein expression levels of Bmal1 and Clock and those of Per2 and Cry1 were found in KGN cells after treatment with forskolin. Among the interrelationships between the expression levels of clock and steroidogenic factors, Clock mRNA had a strongly positive correlation with P450arom and a negative correlation with 3βHSD. Knockdown of Clock gene by siRNA resulted in a significant reduction of estradiol production by inhibiting P450arom expression, while it induced a significant increase of progesterone production by upregulating 3βHSD in KGN cells treated with forskolin. Moreover, BMP-7 had an enhancing effect on the expression of Clock mRNA and protein in KGN cells. Thus, the expression levels of Clock, being upregulated by forskolin and BMP-7, were functionally linked to estradiol production and progesterone suppression by human granulosa cells.
Collapse
Affiliation(s)
- Satoko Nagao
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Kitaku, Okayama 700-8558, Japan
| | - Nahoko Iwata
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Kitaku, Okayama 700-8558, Japan
| | - Yoshiaki Soejima
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Kitaku, Okayama 700-8558, Japan
| | - Takaaki Takiguchi
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Kitaku, Okayama 700-8558, Japan
| | - Tamami Aokage
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Kitaku, Okayama 700-8558, Japan
| | - Yuka Kozato
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Kitaku, Okayama 700-8558, Japan
| | - Yasuhiro Nakano
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Kitaku, Okayama 700-8558, Japan
| | - Takahiro Nada
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Kitaku, Okayama 700-8558, Japan
| | - Toru Hasegawa
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Kitaku, Okayama 700-8558, Japan
| | - Fumio Otsuka
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Kitaku, Okayama 700-8558, Japan
| |
Collapse
|
26
|
Kobayashi M, Watanabe K, Matsumura R, Anayama N, Miyamoto A, Miyazaki H, Miyazaki K, Shimizu T, Akashi M. Involvement of the luteinizing hormone surge in the regulation of ovary and oviduct clock gene expression in mice. Genes Cells 2018; 23:649-657. [PMID: 29920869 DOI: 10.1111/gtc.12605] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 12/15/2022]
Abstract
Circadian dysfunction perturbs the female reproductive cycle. In particular, mice lacking the clock gene Bmal1 show severe infertility, implying that BMAL1 plays roles in ovulation and luteinization. Here, we examined temporal changes in clock gene expression in the ovary and oviduct before and during gonadotropin-induced follicular growth, ovulation, and luteinization in sexually immature mice. While the oviduct did not show a drastic change in clock gene expression, Bmal1 expression in the ovary was higher than that in control mice during the period from 4 to 16 hr after human chorionic gonadotropin (hCG) administration. Bmal1 expression reached a maximum at 16 hr after hCG administration, when follicle luteinization occurred. In an interesting manner, administration of hCG to ex vivo-cultured oviduct triggered a shorter circadian period and inevitably resulted in phase advance. Together, our present data suggest that LH surge induces continuous expression of BMAL1 in the mouse ovary and modulates circadian phase in the mouse oviduct.
Collapse
Affiliation(s)
- Momoko Kobayashi
- Graduate School of Animal and Food Hygiene, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Kaya Watanabe
- Graduate School of Animal and Food Hygiene, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Ritsuko Matsumura
- The Research Institute for Time Studies, Yamaguchi University, Yamaguchi, Yamaguchi, Japan
| | - Nozomi Anayama
- Graduate School of Animal and Food Hygiene, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Akio Miyamoto
- Graduate School of Animal and Food Hygiene, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Hitoshi Miyazaki
- Gene Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Koyomi Miyazaki
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Takashi Shimizu
- Graduate School of Animal and Food Hygiene, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Makoto Akashi
- The Research Institute for Time Studies, Yamaguchi University, Yamaguchi, Yamaguchi, Japan
| |
Collapse
|
27
|
Katandukila JV, Ngalameno MK, Mgode GF, Bastos AD, Bennett NC. Pattern of ovulation in an ancient, solitary mole-rat lineage: Heliophobius argenteocinereus emini from Tanzania. CAN J ZOOL 2017. [DOI: 10.1139/cjz-2016-0292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The genus Heliophobius Peters, 1846 is an ancient subterranean rodent lineage within the family Bathyergidae that displays seasonal breeding over a broad geographical range. In East Africa, reproduction in these solitary mole-rats has been shown to coincide with the long rains, but it is not clear whether the silvery mole-rat subspecies Heliophobius argenteocinereus emini Noack, 1894 from Tanzania is an induced or spontaneous ovulator. To address this, urinary progesterone was measured every second day over a period of 132 days in six wild-caught females subjected to three sequential experimental treatments. In the first, control (C) treatment, females were housed singly for a period of 44 days, following which nonphysical contact (NPC) with a vasectomised male and then physical contact (PC) with the same vasectomised male occurred in each of the subsequent 44-day treatments. Noninvasive monitoring of ovarian cyclicity confirmed that mean urinary progesterone concentration was higher during PC than during either NPC or C, despite the fact that the males were vasectomised and incapable of fertilising the females. Examination of penile morphology revealed that males possess epidermal projections on the glans penis, which probably bring about cervical stimulation during coitus. These findings together with the female progesterone profiles imply the species is an induced ovulator stimulated by penile intromission.
Collapse
Affiliation(s)
- Jestina V. Katandukila
- South African Research Chair for Mammal Behavioural Ecology and Physiology, Department of Zoology and Entomology, University of Pretoria, Pretoria 0002, Republic of South Africa
- Department of Zoology and Wildlife Conservation, College of Natural and Applied Sciences, University of Dar es Salaam, P.O. Box 35064, Dar es Salaam, Tanzania
| | - Mungo K. Ngalameno
- South African Research Chair for Mammal Behavioural Ecology and Physiology, Department of Zoology and Entomology, University of Pretoria, Pretoria 0002, Republic of South Africa
| | - Georgies F. Mgode
- Pest Management Centre, Sokoine University, P.O. Box 3110, Morogoro, Tanzania
| | - Armanda D.S. Bastos
- Mammal Research Institute, Department of Zoology and Entomology, University of Pretoria, Pretoria 0002, Republic of South Africa
| | - Nigel C. Bennett
- South African Research Chair for Mammal Behavioural Ecology and Physiology, Department of Zoology and Entomology, University of Pretoria, Pretoria 0002, Republic of South Africa
| |
Collapse
|
28
|
Lin YJ, Tsai CC, Huang LT, Sheen JM, Tiao MM, Yu HR, Chen CC, Tain YL. Detrimental effect of maternal and post-weaning high-fat diet on the reproductive function in the adult female offspring rat: roles of insulin-like growth factor 2 and the ovarian circadian clock. J Assist Reprod Genet 2017; 34:817-826. [PMID: 28417351 PMCID: PMC5445052 DOI: 10.1007/s10815-017-0915-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 03/29/2017] [Indexed: 11/24/2022] Open
Abstract
PURPOSE We evaluate the impact of maternal and post-weaning high-fat (HF) diet on ovarian follicular population, steroidogenesis, and gene expression with a focus on the circadian clock system and insulin-like growth factor 2 (Igf2) in adult offspring ovaries, and to elucidate whether a maternal and post-weaning diet confers similar risks. METHODS Virgin Sprague-Dawley rats were fed with normal chow (C) diet or HF diet for 5 weeks before mating, during gestation, and lactation. Female offspring were fed with the C or HF diet from weaning to 6 months of age, resulting in four study groups (n = 6 per group): C/C, C/HF, HF/C, and HF/HF. RESULTS Ovaries from offspring exposed to post-weaning HF diet (i.e., the C/HF and HF/HF groups) had a decrease in small follicle numbers, but with similar numbers of antral follicles and corpora lutea. Offspring from HF-fed dams (i.e., the HF/C and HF/HF groups) had increased plasma estradiol concentrations and decreased luteinizing hormone levels at 6 months of age. In addition, Igf2 and each of the circadian rhythm core genes Clock, Per1, Per2, and Per3 were increased in the ovaries of offspring exposed to maternal HF diet (both HF/C and HF/HF groups). CONCLUSIONS Maternal and post-weaning HF diet programs the reproductive profile of the female offspring in adult life through different manners. Post-weaning HF intake resulted in the reduction of small follicles in adulthood, whereas maternal HF diet had long-term deleterious consequences on female offspring steroidogenesis and coincided with alteration of the upregulation of the imprinted gene Igf2 and changes in ovarian circadian rhythms.
Collapse
Affiliation(s)
- Yu-Ju Lin
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung, Taiwan
| | - Ching-Chou Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung, Taiwan
| | - Li-Tung Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, 123 Dabi Road, Niausung, Kaohsiung, 833, Taiwan
| | - Jiunn-Ming Sheen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, 123 Dabi Road, Niausung, Kaohsiung, 833, Taiwan
| | - Mao-Meng Tiao
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, 123 Dabi Road, Niausung, Kaohsiung, 833, Taiwan
| | - Hong-Ren Yu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, 123 Dabi Road, Niausung, Kaohsiung, 833, Taiwan
| | - Chih-Cheng Chen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, 123 Dabi Road, Niausung, Kaohsiung, 833, Taiwan
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, 123 Dabi Road, Niausung, Kaohsiung, 833, Taiwan. .,Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|
29
|
Hernández-Díaz N, Torres R, Ramírez-Pinilla MP. Proteomic Profile of Mabuya sp. (Squamata: Scincidae) Ovary and Placenta During Gestation. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2017; 328:371-389. [PMID: 28397398 DOI: 10.1002/jez.b.22739] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/28/2017] [Accepted: 03/02/2017] [Indexed: 02/07/2023]
Abstract
Reptiles are one of the most diverse groups of vertebrates, providing an integrated system for comparative studies on metabolic, animal physiology, and developmental biology. However, the molecular data available are limited and only recently have started to call attention in the "omics" sciences. Mabuya sp. is a viviparous placentrotrophic skink with particular reproductive features, including microlecithal eggs, early luteolysis, prolonged gestation, and development of a highly specialized placenta. This placenta is responsible for respiratory exchange and the transference of all nutrients necessary for embryonic development. Our aim was to identify differentially expressed proteins in the ovary and placenta of Mabuya sp. during early, mid, and late gestation; their possible metabolic pathways; and biological processes. We carried out a comparative proteomic analysis during gestation in both tissues by sodium dodecyl sulfate polyacrylamide gel electrophoresis, two-dimensional gel electrophoresis, and matrix-assisted laser desorption/ionization. Differential protein expression in both tissues (Student's t-test P < 0.05) was related to several processes such as cell structure, cell movement, and energy. Proteins found in ovary are mainly associated with follicular development and its regulation. In the placenta, particularly during mid and late gestation, protein expression is involved in nutrient metabolism, transport, protein synthesis, and embryonic development. This work provides new insights about the proteins expressed and their physiological mechanisms in Mabuya sp. placenta and ovary during gestation.
Collapse
Affiliation(s)
- Nathaly Hernández-Díaz
- Laboratorio de Biología Reproductiva de Vertebrados, Escuela de Biología, Facultad de Ciencias, Universidad Industrial de Santander, Bucaramanga, Santander, Colombia.,Grupo de Investigación en Bioquímica y Microbiología, GIBIM, Escuela de Química, Universidad Industrial de Santander, Bucaramanga, Santander, Colombia
| | - Rodrigo Torres
- Grupo de Investigación en Bioquímica y Microbiología, GIBIM, Escuela de Química, Universidad Industrial de Santander, Bucaramanga, Santander, Colombia.,Laboratorio de Biotecnología-CEO, Instituto Colombiano del Petróleo, ECOPETROL, Piedecuesta, Santander, Colombia
| | - Martha Patricia Ramírez-Pinilla
- Laboratorio de Biología Reproductiva de Vertebrados, Escuela de Biología, Facultad de Ciencias, Universidad Industrial de Santander, Bucaramanga, Santander, Colombia
| |
Collapse
|
30
|
Chen S, Qiao H, Fu H, Sun S, Zhang W, Jin S, Gong Y, Jiang S, Xiong W, YanWu. Molecular cloning, characterization, and temporal expression of the clock genes period and timeless in the oriental river prawn Macrobrachium nipponense during female reproductive development. Comp Biochem Physiol A Mol Integr Physiol 2017; 207:43-51. [PMID: 28192242 DOI: 10.1016/j.cbpa.2017.02.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 02/08/2017] [Accepted: 02/08/2017] [Indexed: 10/20/2022]
Abstract
The circadian clock is crucial for sustaining rhythmic biochemical, physiological, and behavioral processes in living creatures. In this study, we isolated and characterized two circadian clock genes in Macrobrachium nipponense, period (Mnper) and timeless (Mntim). The complete Mnper cDNA measures 4283bp in length with an open reading frame encoding 1292 amino acids, including functional domains such as PER-ARNT-SIM (PAS), cytoplasmic localization domain (CLD), TIM interaction site (TIS), and nuclear localization signal (NLS). The deduced Mntim protein comprises1540 amino acids with functional domains such as PER interaction site (PIS), NLS, and CLD. Tissue distribution analyses showed that the two genes were highly expressed in the eyestalk and brain in both males and females, as well as being expressed in the ovary. The expression profiles of Mnper and Mntim were determined in the eyestalk, brain, and ovary under simulated breeding season and non-breeding season conditions. The expression profiles of both Mnper and Mntim appeared to be unaffected in the eyestalk. However, the expression of both genes exhibited significant seasonal variations in the brain, and thus we assumed the brain to be their functional location. The expression profiles under different simulated seasons and the variations during different ovarian stages indicate that both genes might be involved with female reproduction. Especially the mRNA levels in the brain varied greatly during these stages indicating that the clock function in the brain is closely related to ovarian development and female reproduction. And the reproductive roles of clock genes need to be elucidated.
Collapse
Affiliation(s)
- SuHua Chen
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, PR China
| | - Hui Qiao
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, PR China
| | - HongTuo Fu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, PR China; Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, PR China.
| | - Shengming Sun
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, PR China
| | - WenYi Zhang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, PR China
| | - ShuBo Jin
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, PR China
| | - Yongsheng Gong
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, PR China
| | - Sufei Jiang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, PR China
| | - Weiyi Xiong
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, PR China
| | - YanWu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, PR China
| |
Collapse
|
31
|
Chavan R, Preitner N, Okabe T, Strittmatter LM, Xu C, Ripperger JA, Pitteloud N, Albrecht U. REV-ERBα regulates Fgf21 expression in the liver via hepatic nuclear factor 6. Biol Open 2017; 6:1-7. [PMID: 27875243 PMCID: PMC5278426 DOI: 10.1242/bio.021519] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The circadian clock contributes to the timing of many body functions including metabolism and reproduction. The hepatokine fibroblast growth factor 21 (FGF21) is a critical metabolic regulator involved in modulation of fertility. Here we show that lack of the clock component REV-ERBα elevates FGF21 levels in liver and plasma. At the molecular level, REV-ERBα modulates the expression of FGF21 via the liver-specific hepatic nuclear factor 6 (HNF6). We conclude that REV-ERBα regulates metabolism and reproduction, at least in part, via regulation of Fgf21. Summary: The hepatokine Fgf21 is transcriptionally regulated by the nuclear receptor REV-ERBα with the hepatocyte-specific factor HNF6 to regulate metabolism and fertility.
Collapse
Affiliation(s)
- Rohit Chavan
- Department of Biology, Unit of Biochemistry, University of Fribourg, Fribourg CH1700, Switzerland
| | - Nadia Preitner
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne CH1011, Switzerland
| | - Takashi Okabe
- Department of Biology, Unit of Biochemistry, University of Fribourg, Fribourg CH1700, Switzerland
| | | | - Cheng Xu
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne CH1011, Switzerland
| | - Jürgen A Ripperger
- Department of Biology, Unit of Biochemistry, University of Fribourg, Fribourg CH1700, Switzerland
| | - Nelly Pitteloud
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne CH1011, Switzerland
| | - Urs Albrecht
- Department of Biology, Unit of Biochemistry, University of Fribourg, Fribourg CH1700, Switzerland
| |
Collapse
|
32
|
Xu J, Li Y, Wang Y, Xu Y, Zhou C. Loss of Bmal1 decreases oocyte fertilization, early embryo development and implantation potential in female mice. ZYGOTE 2016; 24:760-7. [PMID: 27140828 DOI: 10.1017/s0967199416000083] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Biological clock genes expressed in reproductive tissues play important roles in maintaining the normal functions of reproductive system. However, disruption of female circadian rhythm on oocyte fertilization, preimplantation embryo development and blastocyst implantation potential is still unclear. In this study, ovulation, in vivo and in vitro oocyte fertilization, embryo development, implantation and intracellular reactive oxygen species (ROS) levels in ovary and oviduct were studied in female Bmal1+/+ and Bmal1-/- mice. The number of naturally ovulated oocyte in Bmal1-/- mice decreased (5.2 ± 0.8 vs 7.8 ± 0.8, P < 0.001), with an increasing abnormal oocyte ratio (20.4 ± 3.5 vs 11.7 ± 2.0%, P = 0.001) after superovulation. Significantly lower fertilization rate and obtained blastocyst number were observed in Bmal1-/- female mice either mated with wild-type in vivo or fertilized by sperm from wild-type male mice in vitro (all P < 0.05). Interestingly, in vitro fertilization rate of oocytes derived from Bmal1-/- increased significantly compared with in vivo study (P < 0.01). After transferring blastocysts derived from Bmal1+/+ and Bmal1-/- female mice to pseudopregnant mice, the implantation sites of the latter decreased 5 days later (8.0 ± 0.8 vs 5.3 ± 1.0, P = 0.005). The intracellular ROS levels in the ovary on proestrus day and in the oviduct on metestrus day increased significantly in Bmal1-/- mice compared with that of Bmal1+/+ mice. Deletion of the core biological clock gene Bmal1 significantly decreases oocyte fertilization rate, early embryo development and implantation potential in female mice, and these may be possibly caused by excess ROS levels generated in ovary and oviduct.
Collapse
Affiliation(s)
- Jian Xu
- Reproductive Medicine Center,Guangzhou Women and Children's Medical Center,Guangzhou,Guangzhou Medical University,China
| | - Yan Li
- Reproductive Medicine Center,Henan Provincial People's Hospital,Zhengzhou,Henan,China
| | - Yizi Wang
- Reproductive Medicine Center,First Affiliated Hospital of Sun Yat-sen University,Guangzhou,China;Guangdong Provincial Key Laboratory of Reproductive Medicine,Guangdong,China
| | - Yanwen Xu
- Reproductive Medicine Center,First Affiliated Hospital of Sun Yat-sen University,58 Zhongshan Road II,Guangzhou,China
| | - Canquan Zhou
- Reproductive Medicine Center,First Affiliated Hospital of Sun Yat-sen University,58 Zhongshan Road II,Guangzhou,China
| |
Collapse
|
33
|
Nakamura TJ, Takasu NN, Nakamura W. The suprachiasmatic nucleus: age-related decline in biological rhythms. J Physiol Sci 2016; 66:367-74. [PMID: 26915078 PMCID: PMC10717791 DOI: 10.1007/s12576-016-0439-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/08/2016] [Indexed: 10/22/2022]
Abstract
Aging is associated with changes in sleep duration and quality, as well as increased rates of pathologic/disordered sleep. While several factors contribute to these changes, emerging research suggests that age-related changes in the mammalian central circadian clock within the suprachiasmatic nucleus (SCN) may be a key factor. Prior work from our group suggests that circadian output from the SCN declines because of aging. Furthermore, we have previously observed age-related infertility in female mice, caused by a mismatch between environmental light-dark cycles and the intrinsic, internal biological clocks. In this review, we address regulatory mechanisms underlying circadian rhythms in mammals and summarize recent literature describing the effects of aging on the circadian system.
Collapse
Affiliation(s)
- Takahiro J Nakamura
- Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Kanagawa, 214-8571, Japan
| | - Nana N Takasu
- Laboratory of Oral Chronobiology, Graduate School of Dentistry, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Wataru Nakamura
- Laboratory of Oral Chronobiology, Graduate School of Dentistry, Osaka University, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
34
|
Khan ZA, Yumnamcha T, Rajiv C, Sanjita Devi H, Mondal G, Devi SD, Bharali R, Chattoraj A. Melatonin biosynthesizing enzyme genes and clock genes in ovary and whole brain of zebrafish (Danio rerio): Differential expression and a possible interplay. Gen Comp Endocrinol 2016; 233:16-31. [PMID: 27179881 DOI: 10.1016/j.ygcen.2016.05.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 05/05/2016] [Accepted: 05/11/2016] [Indexed: 12/27/2022]
Abstract
The present study on zebrafish (Danio rerio) is the first attempt to demonstrate the circadian mRNA expression of melatonin biosynthesizing enzyme genes (Tph1a, Aanat1, Aanat2 and Hiomt) and clock associated genes (Bmal1a, Clock1a, Per1b, Per2 and Cry2a) in the ovary with a comparison to whole brain in normal (LD=12h L:12h D) and altered photic conditions (continuous dark, DD; continuous light, LL). Moreover, the present study also confirmed the ability of zebrafish ovary to biosynthesize melatonin both in vivo and in vitro with a significant difference at day and night. qRT-PCR analysis of genes revealed a dark acrophase of Aanat2 in both organs while Tph1 is in whole brain in LD condition. On the contrary, Bmal1a and Clock1a giving their peak in light, thereby showing a negative correlation with Tph1a and Aanat2. In LD-ovary, the acrophase of Tph1a, Bmal1a and Clock1a is in light and thus display a positive correlation. This trend of relationship in respect to Tph1a is not changing in altered photic conditions in both organs (except in DD-ovary). On the other hand this association for Aanat2 is varying in ovary under altered photic conditions but only in DD-whole brain. Both in LD and LL the expression of Aanat2 in brain presenting an opposite acrophase with both Bmal1a and Clock1a of ovary and consequently displaying a strong negative correlation among them. Interestingly, all ovarian clock associated genes become totally arrhythmic in DD, representing a loss of correlation between the melatonin synthesizing genes in brain and clock associated genes in ovary. The result is also indicating the formation of two heterodimers namely Clock1a:Bmal1a and Per2:Cry2a in the functioning of clock genes in both organs, irrespective of photic conditions, as they are exhibiting a strong significant positive correlation. Collectively, our data suggest that ovary of zebrafish is working as peripheral oscillator having its own melatonin biosynthesizing machinery and signifying a possible correlation with central oscillating system in various photic conditions.
Collapse
Affiliation(s)
- Zeeshan Ahmad Khan
- Biological Rhythm Laboratory, Animal Resources Programme, Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Takyelpat, Imphal 795 001, Manipur, India
| | - Thangal Yumnamcha
- Biological Rhythm Laboratory, Animal Resources Programme, Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Takyelpat, Imphal 795 001, Manipur, India
| | - Chongtham Rajiv
- Biological Rhythm Laboratory, Animal Resources Programme, Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Takyelpat, Imphal 795 001, Manipur, India
| | - Haobijam Sanjita Devi
- Biological Rhythm Laboratory, Animal Resources Programme, Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Takyelpat, Imphal 795 001, Manipur, India
| | - Gopinath Mondal
- Biological Rhythm Laboratory, Animal Resources Programme, Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Takyelpat, Imphal 795 001, Manipur, India
| | - Sh Dharmajyoti Devi
- Biological Rhythm Laboratory, Animal Resources Programme, Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Takyelpat, Imphal 795 001, Manipur, India
| | - Rupjyoti Bharali
- Department of Biotechnology, Gauhati University, Guwahati 781 014, Assam, India
| | - Asamanja Chattoraj
- Biological Rhythm Laboratory, Animal Resources Programme, Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Takyelpat, Imphal 795 001, Manipur, India.
| |
Collapse
|
35
|
Yan L, Silver R. Neuroendocrine underpinnings of sex differences in circadian timing systems. J Steroid Biochem Mol Biol 2016; 160:118-26. [PMID: 26472554 PMCID: PMC4841755 DOI: 10.1016/j.jsbmb.2015.10.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 10/04/2015] [Accepted: 10/08/2015] [Indexed: 01/05/2023]
Abstract
There are compelling reasons to study the role of steroids and sex differences in the circadian timing system. A solid history of research demonstrates the ubiquity of circadian changes that impact virtually all behavioral and biological responses. Furthermore, steroid hormones can modulate every attribute of circadian responses including the period, amplitude and phase. Finally, desynchronization of circadian rhythmicity, and either enhancing or damping amplitude of various circadian responses can produce different effects in the sexes. Studies of the neuroendocrine underpinnings of circadian timing systems and underlying sex differences have paralleled the overall development of the field as a whole. Early experimental studies established the ubiquity of circadian rhythms by cataloging daily and seasonal changes in whole organism responses. The next generation of experiments demonstrated that daily changes are not a result of environmental synchronizing cues, and are internally orchestrated, and that these differ in the sexes. This work was followed by the revelation of molecular circadian rhythms within individual cells. At present, there is a proliferation of work on the consequences of these daily oscillations in health and in disease, and awareness that these may differ in the sexes. In the present discourse we describe the paradigms used to examine circadian oscillation, to characterize how these internal timing signals are synchronized to local environmental conditions, and how hormones of gonadal and/or adrenal origin modulate circadian responses. Evidence pointing to endocrinologically and genetically mediated sex differences in circadian timing systems can be seen at many levels of the neuroendocrine and endocrine systems, from the cell, the gland and organ, and to whole animal behavior, including sleep/wake or rest/activity cycles, responses to external stimuli, and responses to drugs. We review evidence indicating that the analysis of the circadian timing system is amenable to experimental analysis at many levels of the neuraxis, and on several different time scales, rendering it especially useful for the exploration of mechanisms associated with sex differences.
Collapse
Affiliation(s)
- Lily Yan
- Department of Psychology, Michigan State University, East Lansing, MI 48824, USA; Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA.
| | - Rae Silver
- Psychology Department, Barnard College, New York, NY 10027, USA; Department of Psychology, Columbia University, New York, NY 10027, USA; Department of Pathology and Cell Biology, Columbia University Health Sciences, New York, NY 10032, USA
| |
Collapse
|
36
|
Wang Y, Gu F, Deng M, Guo L, Lu C, Zhou C, Chen S, Xu Y. Rotating shift work and menstrual characteristics in a cohort of Chinese nurses. BMC WOMENS HEALTH 2016; 16:24. [PMID: 27145834 PMCID: PMC4857333 DOI: 10.1186/s12905-016-0301-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 04/20/2016] [Indexed: 11/25/2022]
Abstract
Background Shift work disrupts the circadian rhythm and may cause menstruation disorders. This study assessed the impact of shift work on menstrual cycle in a population of Chinese nurses. Methods Questionnaires on menstrual characteristics and shift schedules were sent to female nurses of the First Affiliated Hospital of Sun Yat-sen University (FAHSYSU) and Guanghua Hospital of Stomatology (GHHS), affiliated to Sun Yat-sen University. Part I was a cross-sectional study and included 139 nurses in GHHS who had regular 8:00–17:30 working (non-shift group), and 334 nurses from FAHSYSU who worked shifts, a response rate of 67.5 % and 59.6 %, respectively (age ≤ 50 years). Menstrual patterns were compared and age-adjusted relative risks of shift work were analyzed. Part II was a nested case–control study. Cases were nurses in Part I who had regular cycle with mean cycle length (MCL) of 25–31 days and but at least 3 days variation in MCL after starting shift work (n = 45). Controls consisted of 67 nurses with matching shift patterns and age, but no MCL changes. A control non-shift age-matched group consisted of 30 GHHS nurses with no MCL changes. A follow-up second questionnaire was sent 2 years later. Results In Part I, the shift group had a significantly higher proportion of nurses with menstrual cycle irregularity. The proportion of nurses with a cycle of 25–31 days decreased from 81.7 to 67.8 % after changing to shift work. Logistic regression analysis showed that night shift frequency was the only risk factor associated with cycle shortening. After adjusting for age, MCL was shorter when night work was performed > 7 times per month. In Part II, the mean change in MCL in the case group, including prolongation or shortening, was 4.115 ± 2.084 days after shift working. In the 2 years’ follow-up, the MCL of the study group did not recover to the original length. Conclusions Rotating shift work can increase the prevalence of menstrual cycle irregularity. Night shift frequency was the only risk factor associated with cycle reduced. Changes in MCL did not show recovery over a follow-up period of 2 years. Electronic supplementary material The online version of this article (doi:10.1186/s12905-016-0301-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yizi Wang
- Reproductive Center, the 6th Building, the First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2 Road No. 58, YueXiu District, Guangzhou, 510080, Guangdong Province, China
| | - Fang Gu
- Reproductive Center, the 6th Building, the First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2 Road No. 58, YueXiu District, Guangzhou, 510080, Guangdong Province, China
| | - Mingfen Deng
- Reproductive Center, the 6th Building, the First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2 Road No. 58, YueXiu District, Guangzhou, 510080, Guangdong Province, China
| | - Lan Guo
- Department of Biostatistics and Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Ciyong Lu
- Department of Biostatistics and Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Canquan Zhou
- Reproductive Center, the 6th Building, the First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2 Road No. 58, YueXiu District, Guangzhou, 510080, Guangdong Province, China
| | - Shouzhen Chen
- Nursing Department, the First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2 Road No.58, YueXiu District, Guangzhou, Guangdong Province, 510080, China.
| | - Yanwen Xu
- Reproductive Center, the 6th Building, the First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2 Road No. 58, YueXiu District, Guangzhou, 510080, Guangdong Province, China.
| |
Collapse
|
37
|
Reinberg A, Smolensky MH, Touitou Y. The full moon as a synchronizer of circa-monthly biological rhythms: Chronobiologic perspectives based on multidisciplinary naturalistic research. Chronobiol Int 2016; 33:465-79. [DOI: 10.3109/07420528.2016.1157083] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Alain Reinberg
- Unité de Chronobiologie, Fondation A de Rothschild, Paris cedex 19, France
| | - Michael H. Smolensky
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Yvan Touitou
- Unité de Chronobiologie, Fondation A de Rothschild, Paris cedex 19, France
| |
Collapse
|
38
|
Temperature compensation and temperature sensation in the circadian clock. Proc Natl Acad Sci U S A 2015; 112:E6284-92. [PMID: 26578788 DOI: 10.1073/pnas.1511215112] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
All known circadian clocks have an endogenous period that is remarkably insensitive to temperature, a property known as temperature compensation, while at the same time being readily entrained by a diurnal temperature oscillation. Although temperature compensation and entrainment are defining features of circadian clocks, their mechanisms remain poorly understood. Most models presume that multiple steps in the circadian cycle are temperature-dependent, thus facilitating temperature entrainment, but then insist that the effect of changes around the cycle sums to zero to enforce temperature compensation. An alternative theory proposes that the circadian oscillator evolved from an adaptive temperature sensor: a gene circuit that responds only to temperature changes. This theory implies that temperature changes should linearly rescale the amplitudes of clock component oscillations but leave phase relationships and shapes unchanged. We show using timeless luciferase reporter measurements and Western blots against TIMELESS protein that this prediction is satisfied by the Drosophila circadian clock. We also review evidence for pathways that couple temperature to the circadian clock, and show previously unidentified evidence for coupling between the Drosophila clock and the heat-shock pathway.
Collapse
|
39
|
Takasu N, Nakamura T, Tokuda I, Todo T, Block G, Nakamura W. Recovery from Age-Related Infertility under Environmental Light-Dark Cycles Adjusted to the Intrinsic Circadian Period. Cell Rep 2015; 12:1407-13. [DOI: 10.1016/j.celrep.2015.07.049] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 06/29/2015] [Accepted: 07/23/2015] [Indexed: 11/25/2022] Open
|
40
|
Han W, Zou J, Wang K, Su Y, Zhu Y, Song C, Li G, Qu L, Zhang H, Liu H. High-Throughput Sequencing Reveals Hypothalamic MicroRNAs as Novel Partners Involved in Timing the Rapid Development of Chicken (Gallus gallus) Gonads. PLoS One 2015; 10:e0129738. [PMID: 26061962 PMCID: PMC4465036 DOI: 10.1371/journal.pone.0129738] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 05/12/2015] [Indexed: 12/21/2022] Open
Abstract
Onset of the rapid gonad growth is a milestone in sexual development that comprises many genes and regulatory factors. The observations in model organisms and mammals including humans have shown a potential link between miRNAs and development timing. To determine whether miRNAs play roles in this process in the chicken (Gallus gallus), the Solexa deep sequencing was performed to analyze the profiles of miRNA expression in the hypothalamus of hens from two different pubertal stages, before onset of the rapid gonad development (BO) and after onset of the rapid gonad development (AO). 374 conserved and 46 novel miRNAs were identified as hypothalamus-expressed miRNAs in the chicken. 144 conserved miRNAs were showed to be differentially expressed (reads > 10, P < 0.05) during the transition from BO to AO. Five differentially expressed miRNAs were validated by real-time quantitative RT-PCR (qRT-PCR) method. 2013 putative genes were predicted as the targets of the 15 most differentially expressed miRNAs (fold-change > 4.0, P < 0.01). Of these genes, 7 putative circadian clock genes, Per2, Bmal1/2, Clock, Cry1/2, and Star were found to be targeted multiple times by the miRNAs. qRT-PCR revealed the basic transcription levels of these clock genes were much higher (P < 0.01) in AO than in BO. Further functional analysis suggested that these 15 miRNAs play important roles in transcriptional regulation and signal transduction pathways. The results provide new insights into miRNAs functions in timing the rapid development of chicken gonads. Considering the characteristics of miRNA functional conservation, the results will contribute to the research on puberty onset in humans.
Collapse
Affiliation(s)
- Wei Han
- College of Animal Science & Technology, Nanjing Agricultural University, Nanjing, PR China
- National Chickens Genetic Resources, Poultry institute, Chinese Academy of Agricultural Science, Yangzhou, PR China
| | - Jianmin Zou
- National Chickens Genetic Resources, Poultry institute, Chinese Academy of Agricultural Science, Yangzhou, PR China
| | - Kehua Wang
- National Chickens Genetic Resources, Poultry institute, Chinese Academy of Agricultural Science, Yangzhou, PR China
| | - Yijun Su
- National Chickens Genetic Resources, Poultry institute, Chinese Academy of Agricultural Science, Yangzhou, PR China
| | - Yunfen Zhu
- National Chickens Genetic Resources, Poultry institute, Chinese Academy of Agricultural Science, Yangzhou, PR China
| | - Chi Song
- National Chickens Genetic Resources, Poultry institute, Chinese Academy of Agricultural Science, Yangzhou, PR China
| | - Guohui Li
- National Chickens Genetic Resources, Poultry institute, Chinese Academy of Agricultural Science, Yangzhou, PR China
| | - Liang Qu
- National Chickens Genetic Resources, Poultry institute, Chinese Academy of Agricultural Science, Yangzhou, PR China
| | - Huiyong Zhang
- National Chickens Genetic Resources, Poultry institute, Chinese Academy of Agricultural Science, Yangzhou, PR China
| | - Honglin Liu
- College of Animal Science & Technology, Nanjing Agricultural University, Nanjing, PR China
- * E-mail:
| |
Collapse
|
41
|
Moore CJ, DeLong NE, Chan KA, Holloway AC, Petrik JJ, Sloboda DM. Perinatal Administration of a Selective Serotonin Reuptake Inhibitor Induces Impairments in Reproductive Function and Follicular Dynamics in Female Rat Offspring. Reprod Sci 2015; 22:1297-311. [DOI: 10.1177/1933719115578925] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- C. J. Moore
- Department of Biochemistry and Biomedical Sciences, Pediatrics McMaster University, Hamilton, Canada
- Department of Obstetrics and Gynaecology, Pediatrics McMaster University, Hamilton, Canada
| | - N. E. DeLong
- Department of Obstetrics and Gynaecology, Pediatrics McMaster University, Hamilton, Canada
| | - K. A. Chan
- Department of Biochemistry and Biomedical Sciences, Pediatrics McMaster University, Hamilton, Canada
| | - A. C. Holloway
- Department of Obstetrics and Gynaecology, Pediatrics McMaster University, Hamilton, Canada
| | - J. J. Petrik
- Department of Obstetrics and Gynaecology, Pediatrics McMaster University, Hamilton, Canada
- Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - D. M. Sloboda
- Department of Biochemistry and Biomedical Sciences, Pediatrics McMaster University, Hamilton, Canada
- Department of Obstetrics and Gynaecology, Pediatrics McMaster University, Hamilton, Canada
- Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| |
Collapse
|
42
|
Chan KA, Bernal AB, Vickers MH, Gohir W, Petrik JJ, Sloboda DM. Early life exposure to undernutrition induces ER stress, apoptosis, and reduced vascularization in ovaries of adult rat offspring. Biol Reprod 2015; 92:110. [PMID: 25810471 DOI: 10.1095/biolreprod.114.124149] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 03/16/2015] [Indexed: 12/15/2022] Open
Abstract
Maternal nutritional restriction has been shown to induce impairments in a number of organ systems including the ovary. We have previously shown that maternal undernutrition induces fetal growth restriction and low birth weight, and results in an offspring ovarian phenotype characteristic of premature ovarian aging with reduced ovarian reserve. In the present study, we set out to investigate the underlying mechanisms that lead offspring of undernourished mothers to premature ovarian aging. Pregnant dams were randomized to 1) a standard diet throughout pregnancy and lactation (control), 2) a calorie-restricted (50% of control) diet during pregnancy, 3) a calorie-restricted (50% of control) diet during pregnancy and lactation, or 4) a calorie-restricted (50% of control) diet during lactation alone. The present study shows that early life undernutrition-induced reduction of adult ovarian follicles may be mediated by increased ovarian endoplasmic reticulum stress in a manner that increased follicular apoptosis but not autophagy. These changes were associated with a loss of ovarian vessel density and are consistent with an accelerated ovarian aging phenotype. Whether these changes are mediated specifically by a reduction in the local antioxidant environment is unclear, although our data suggest the possibility that ovarian melatonin may play a part in early life nutritional undernutrition and impaired offspring folliculogenesis.
Collapse
Affiliation(s)
- Kaitlyn A Chan
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Angelica B Bernal
- Liggins Institute and Gravida: National Centre for Growth and Development, University of Auckland, New Zealand
| | - Mark H Vickers
- Liggins Institute and Gravida: National Centre for Growth and Development, University of Auckland, New Zealand
| | - Wajiha Gohir
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Jim J Petrik
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Deborah M Sloboda
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
43
|
Mereness AL, Murphy ZC, Sellix MT. Developmental programming by androgen affects the circadian timing system in female mice. Biol Reprod 2015; 92:88. [PMID: 25695720 DOI: 10.1095/biolreprod.114.126409] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 02/11/2015] [Indexed: 12/16/2022] Open
Abstract
Circadian clocks play essential roles in the timing of events in the mammalian hypothalamo-pituitary-ovarian (HPO) axis. The molecular oscillator driving these rhythms has been localized to tissues of the HPO axis. It has been suggested that synchrony among these oscillators is a feature of normal reproductive function. The impact of fertility disorders on clock function and the role of the clock in the etiology of endocrine pathology remain unknown. Polycystic ovarian syndrome (PCOS) is a particularly devastating fertility disorder, affecting 5%-10% of women at childbearing age with features including a polycystic ovary, anovulation, and elevated serum androgen. Approximately 40% of these women have metabolic syndrome, marked by hyperinsulinemia, dyslipidemia, and insulin resistance. It has been suggested that developmental exposure to excess androgen contributes to the etiology of fertility disorders, including PCOS. To better define the role of the timing system in these disorders, we determined the effects of androgen-dependent developmental programming on clock gene expression in tissues of the metabolic and HPO axes. Female PERIOD2::luciferase (PER2::LUC) mice were exposed to androgen (dihydrotestosterone [DHT]) in utero (Days 16-18 of gestation) or for 9-10 wk (DHT pellet) beginning at weaning (pubertal androgen excess [PAE]). As expected, both groups of androgen-treated mice had disrupted estrous cycles. Analysis of PER2::LUC expression in tissue explants revealed that excess androgen produced circadian misalignment via tissue-dependent effects on phase distribution. In vitro treatment with DHT differentially affected the period of PER2::LUC expression in tissue explants and granulosa cells, indicating that androgen has direct and tissue-specific effects on clock gene expression that may account for the effects of developmental programming on the timing system.
Collapse
Affiliation(s)
- Amanda L Mereness
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Zachary C Murphy
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Michael T Sellix
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, New York
| |
Collapse
|
44
|
Liu J, Cheng KM, Silversides FG. Recovery of fertility from adult ovarian tissue transplanted into week-old Japanese quail chicks. Reprod Fertil Dev 2015; 27:281-4. [DOI: 10.1071/rd13256] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Accepted: 09/30/2013] [Indexed: 01/21/2023] Open
Abstract
Fertility of cryopreserved ovarian tissue from immature chickens and Japanese quail has been recovered by transplantation. This is of special importance for non-mammalian vertebrates in which cryopreservation and in vitro maturation of oocytes are challenging because their oogenesis is characterised by vitellogenesis. This study tested whether fertility of adult quail ovarian tissue could be recovered by transplantation. Ovaries were isolated from mature Japanese quail hens, trimmed, cut into 3- to 4-mm2 pieces and transplanted into ovariectomised, week-old chicks. Recipients were administered an immunosuppressant for two weeks. Ten of 12 recipients survived until sexual maturity and seven laid eggs, but all stopped laying by 17 weeks of age. The age at first egg of recipients laying eggs (75.7 ± 4.2 days) was greater than that of untreated hens (51.8 ± 1.7 days) and egg production of recipients during the laying period (21.7 ± 5.7) was less than that of untreated hens (60.8 ± 3.5). Recipients were paired with males from the WB line for test mating. Only two hens laid eggs during the test period but both produced 100% donor-derived offspring. This research demonstrated that the reproductive potential of ovarian tissue from adult quail hens can be restored by transplantation.
Collapse
|
45
|
Abstract
Rhythmic events in the female reproductive system depend on the coordinated and synchronized activity of multiple neuroendocrine and endocrine tissues. This coordination is facilitated by the timing of gene expression and cellular physiology at each level of the hypothalamo-pituitary-ovarian (HPO) axis, including the basal hypothalamus and forebrain, the pituitary gland, and the ovary. Central to this pathway is the primary circadian pacemaker in the suprachiasmatic nucleus (SCN) that, through its myriad outputs, provides a temporal framework for gonadotropin release and ovulation. The heart of the timing system, a transcription-based oscillator, imparts SCN pacemaker cells and a company of peripheral tissues with the capacity for daily oscillations of gene expression and cellular physiology. Although the SCN sits comfortably at the helm, peripheral oscillators (such as the ovary) have undefined but potentially critical roles. Each cell type of the ovary, including theca cells, granulosa cells, and oocytes, harbor a molecular clock implicated in the processes of follicular growth, steroid hormone synthesis, and ovulation. The ovarian clock is influenced by the reproductive cycle and diseases that perturb the cycle and/or follicular growth can disrupt the timing of clock gene expression in the ovary. Chronodisruption is known to negatively affect reproductive function and fertility in both rodent models and women exposed to shiftwork schedules. Thus, influencing clock function in the HPO axis with chronobiotics may represent a novel avenue for the treatment of common fertility disorders, particularly those resulting from chronic circadian disruption.
Collapse
Affiliation(s)
- Michael T. Sellix
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
46
|
Yeh CM, Shay J, Zeng TC, Chou JL, Huang THM, Lai HC, Chan MWY. Epigenetic silencing of ARNTL, a circadian gene and potential tumor suppressor in ovarian cancer. Int J Oncol 2014; 45:2101-7. [PMID: 25175925 DOI: 10.3892/ijo.2014.2627] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 08/13/2014] [Indexed: 11/06/2022] Open
Abstract
Ovarian cancer is the fifth leading cause of cancer death and the most deadly gynecological malignancy in women. Epigenetic modifications play an important role in regulating gene transcription. Specifically, aberrant promoter hypermethylation has been implicated as a hallmark of cancer. In order to identify genes that are differentially methylated in ovarian cancer, we performed meDIP-chip in various ovarian cancer cell lines using Agilent 244K CpG island microarray. One of the targets, ARNTL which is a core component of the circadian clock is methylated in a sub-set of ovarian cancer cell lines. Combined bisulfite restriction analysis (COBRA) confirmed the results of the microarray. Additional analysis using ChIP-PCR revealed that promoter of ARNTL is enriched with the repressive histone mark H3K27me3 in CP70 and MCP2 ovarian cancer cells. Treatment with the EZH2 inhibitor (GSK126) significantly restored ARNTL expression in these cells (CP70 and MCP2). Further functional analysis demonstrated that overexpression of ARNTL inhibited cell growth and enhanced chemosensitivity of cisplatin in ovarian cancer cells. Finally, overexpression of ARNTL restored the rhythmic activity of c-MYC in ovarian cancer cells. These results suggested that ARNTL may be a tumor suppressor and is epigenetically silenced in ovarian cancer.
Collapse
Affiliation(s)
- Chia-Ming Yeh
- Department of Life Science and, National Chung Cheng University, Chia-Yi, Taiwan, R.O.C
| | - Jacqueline Shay
- Department of Life Science and, National Chung Cheng University, Chia-Yi, Taiwan, R.O.C
| | - Ting-Chuan Zeng
- Department of Life Science and, National Chung Cheng University, Chia-Yi, Taiwan, R.O.C
| | - Jian-Liang Chou
- Department of Life Science and, National Chung Cheng University, Chia-Yi, Taiwan, R.O.C
| | - Tim H-M Huang
- Department of Molecular Medicine, Institute of Biotechnology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Hung-Cheng Lai
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, R.O.C
| | - Michael W Y Chan
- Department of Life Science and, National Chung Cheng University, Chia-Yi, Taiwan, R.O.C
| |
Collapse
|
47
|
Shah S, Meisenberg G. Chronobiology awareness concerning the menstrual cycle. BIOL RHYTHM RES 2014. [DOI: 10.1080/09291016.2014.882095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
48
|
Beltramo M, Dardente H, Cayla X, Caraty A. Cellular mechanisms and integrative timing of neuroendocrine control of GnRH secretion by kisspeptin. Mol Cell Endocrinol 2014; 382:387-399. [PMID: 24145132 DOI: 10.1016/j.mce.2013.10.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 10/08/2013] [Accepted: 10/11/2013] [Indexed: 01/11/2023]
Abstract
The hypothalamus integrates endogenous and exogenous inputs to control the pituitary-gonadal axis. The ultimate hypothalamic influence on reproductive activity is mediated through timely secretion of GnRH in the portal blood, which modulates the release of gonadotropins from the pituitary. In this context neurons expressing the RF-amide neuropeptide kisspeptin present required features to fulfill the role of the long sought-after hypothalamic integrative centre governing the stimulation of GnRH neurons. Here we focus on the intracellular signaling pathways triggered by kisspeptin through its cognate receptor KISS1R and on the potential role of proteins interacting with this receptor. We then review evidence implicating both kisspeptin and RFRP3--another RF-amide neuropeptide--in the temporal orchestration of both the pre-ovulatory LH surge in female rodents and the organization of seasonal breeding in photoperiodic species.
Collapse
Affiliation(s)
- Massimiliano Beltramo
- UMR Physiologie de la Reproduction et des Comportements (INRA, UMR85, CNRS, UMR7247, Université François Rabelais Tours, IFCE), F-37380 Nouzilly, France.
| | - Hugues Dardente
- UMR Physiologie de la Reproduction et des Comportements (INRA, UMR85, CNRS, UMR7247, Université François Rabelais Tours, IFCE), F-37380 Nouzilly, France
| | - Xavier Cayla
- UMR Physiologie de la Reproduction et des Comportements (INRA, UMR85, CNRS, UMR7247, Université François Rabelais Tours, IFCE), F-37380 Nouzilly, France
| | - Alain Caraty
- UMR Physiologie de la Reproduction et des Comportements (INRA, UMR85, CNRS, UMR7247, Université François Rabelais Tours, IFCE), F-37380 Nouzilly, France
| |
Collapse
|
49
|
Bailey M, Silver R. Sex differences in circadian timing systems: implications for disease. Front Neuroendocrinol 2014; 35:111-39. [PMID: 24287074 PMCID: PMC4041593 DOI: 10.1016/j.yfrne.2013.11.003] [Citation(s) in RCA: 229] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 11/13/2013] [Accepted: 11/17/2013] [Indexed: 12/22/2022]
Abstract
Virtually every eukaryotic cell has an endogenous circadian clock and a biological sex. These cell-based clocks have been conceptualized as oscillators whose phase can be reset by internal signals such as hormones, and external cues such as light. The present review highlights the inter-relationship between circadian clocks and sex differences. In mammals, the suprachiasmatic nucleus (SCN) serves as a master clock synchronizing the phase of clocks throughout the body. Gonadal steroid receptors are expressed in almost every site that receives direct SCN input. Here we review sex differences in the circadian timing system in the hypothalamic-pituitary-gonadal axis (HPG), the hypothalamic-adrenal-pituitary (HPA) axis, and sleep-arousal systems. We also point to ways in which disruption of circadian rhythms within these systems differs in the sexes and is associated with dysfunction and disease. Understanding sex differentiated circadian timing systems can lead to improved treatment strategies for these conditions.
Collapse
Affiliation(s)
- Matthew Bailey
- Department of Psychology, Columbia University, United States.
| | - Rae Silver
- Department of Psychology, Columbia University, United States; Department of Psychology, Barnard College, United States; Department of Pathology and Cell Biology, Columbia University Medical Center, United States.
| |
Collapse
|
50
|
Hou W, Xiong L, Li S, Wang Y, Jiang Z, Cheng S, Liu Y, Xiao J, Guo H, Wang Z. A continuous electromagnetic radiation exposure affected the expressions ofClockandfviigenes in mice. BIOL RHYTHM RES 2013. [DOI: 10.1080/09291016.2013.770295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|