1
|
Dai Y, Ono M, Suzuki T, Hayashi S, Kojima J, Sasaki T, Fujiwara T, Daikoku T, Terakawa J, Maida Y, Ando H, Fujiwara H, Kuji N, Nishi H. BMAL1 Regulates Collagen Production in the Myometrium and Leiomyomas. Reprod Sci 2025:10.1007/s43032-025-01812-y. [PMID: 39966227 DOI: 10.1007/s43032-025-01812-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/30/2025] [Indexed: 02/20/2025]
Abstract
Infertility and reproductive issues are commonly observed in animals with clock abnormalities. Substantial rodent data is available; however, relatively few studies have investigated the connection between fertility and clock abnormalities in humans. Therefore, this study aimed to analyze the expression of circadian clock genes and their impact on genes involved in collagen production in the human myometrium and leiomyomas (LM). The relationship between the expression of brain and muscle aryl-hydrocarbon-receptor-nuclear-translocator (Arnt)-like protein-1 (BMAL1) and the genes responsible for collagen synthesis in the human MM and LMs were investigated. Human MM and LM tissues were collected for analysis from patients who underwent hysterectomy analysis. Immunohistochemical analysis, cell culturing, immunofluorescence, small interfering RNA transfection, reverse transcription quantitative real-time polymerase chain reaction, scratch wound assays, and transwell assays were employed to gain a comprehensive understanding of the cellular and molecular processes. A correlation was found between BMAL1 expression and genes regulating collagen synthesis in primary cultures of human MM and LM cells. Moreover, the inhibition of BMAL1 differentially increased the migration and invasion of MM and LM cells. This work discloses the role of BMAL1 in collagen production in primary cultures of human MM and LM cells, offering insight into clock gene involvement in both normal and pathological uterine conditions. Furthermore, this study highlights the crucial role of BMAL1 in collagen synthesis in human MM and LM cells, underscoring the significance of BMAL1 in the regulation of reproductive physiology. These results suggest that BMAL1 might be a useful target molecule for anti-LM therapy.
Collapse
Affiliation(s)
- Yidan Dai
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo, 160-0023, Japan
| | - Masanori Ono
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo, 160-0023, Japan.
| | - Tomoo Suzuki
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo, 160-0023, Japan
| | - Shigehiro Hayashi
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo, 160-0023, Japan
| | - Junya Kojima
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo, 160-0023, Japan
| | - Toru Sasaki
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo, 160-0023, Japan
| | - Tomoko Fujiwara
- Department of Human Life Environments, Kyoto Notre Dame University, Kyoto, 606-0848, Japan
| | - Takiko Daikoku
- Division of Animal Disease Model, Research Center for Experimental Modeling of Human Disease, Graduate School of Medical Science, Kanazawa University, Kanazawa, 920-8641, Japan
| | - Jumpei Terakawa
- School of Veterinary Medicine, Azabu University, Kanagawa, 252-0206, Japan
| | - Yoshiko Maida
- Department of Nursing, College of Medical, Pharmaceutical, and Health Sciences, Kanazawa University, Kanazawa, 920-8641, Japan
| | - Hitoshi Ando
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Science, Kanazawa University, Kanazawa, 920-8641, Japan
| | - Hiroshi Fujiwara
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, 920-8641, Japan
| | - Naoaki Kuji
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo, 160-0023, Japan
| | - Hirotaka Nishi
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo, 160-0023, Japan
| |
Collapse
|
2
|
Xu P, Morishige JI, Jing Z, Nagata N, Shi Y, Iba T, Daikoku T, Ono M, Maida Y, Fujiwara T, Fujiwara H, Ando H. Exenatide administration time-dependently affects the hepatic circadian clock through glucagon-like peptide-1 receptors in the central nervous system. Biochem Pharmacol 2024; 230:116567. [PMID: 39369911 DOI: 10.1016/j.bcp.2024.116567] [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: 05/29/2024] [Revised: 08/23/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Accumulating evidence indicates that disruption of the circadian clock contributes to the development of lifestyle-related diseases. We have previously shown that exenatide, a glucagon-like peptide-1 (GLP-1) receptor agonist, can strongly affect the molecular clocks in the peripheral tissues. This study aimed to investigate the effects of its dosing time and the central nervous system-specific GLP-1 receptor knockdown (GLP1RKD) on the hepatic clock in mice treated with exenatide. Male C57BL/6J and GLP1RKD mice were housed under a 12-h/12-h light/dark cycle, and feeding was restricted to either the light period (L-TRF) or the first 4 h in the dark period (D-TRF). In parallel, exenatide was administered 4-5 times, once daily either at the beginning of the dark (ZT 12) or light period (ZT 0), and we assessed the mRNA expression rhythms of clock genes in the liver thereafter. Exenatide administration at ZT 12 counteracted the phase shift effect of the L-TRF on the hepatic clock of wild-type mice, whereas the dosing at ZT 0 enhanced its effect. However, exenatide did not influence the phase of the hepatic clock under D-TRF regardless of the dosing time. The effect of exenatide in wild-type mice weakened in GLP1RKD mice. These results showed that exenatide dosing time-dependently affects the hepatic circadian clock through the central GLP-1 system. Exenatide administration at the beginning of the active period (i.e., in the morning for humans) might prevent disruption of the peripheral clocks caused by irregular eating habits.
Collapse
Affiliation(s)
- Pingping Xu
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Jun-Ichi Morishige
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Zheng Jing
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Naoto Nagata
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yifan Shi
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Tomohiro Iba
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Takiko Daikoku
- Division of Animal Disease Model, Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa, Japan
| | - Masanori Ono
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo, Japan
| | - Yoshiko Maida
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Tomoko Fujiwara
- Department of Human Life Environments, Kyoto Notre Dame University, Kyoto, Japan
| | - Hiroshi Fujiwara
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan; Ochi Yume Clinic Nagoya, Nagoya, Japan
| | - Hitoshi Ando
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
3
|
Timpani CA, Debrincat D, Kourakis S, Boyer R, Formosa LE, Steele JR, Zhang H, Schittenhelm RB, Russell AP, Rybalka E, Lindsay A. Loss of endogenous estrogen alters mitochondrial metabolism and muscle clock-related protein Rbm20 in female mdx mice. FASEB J 2024; 38:e23718. [PMID: 38847487 DOI: 10.1096/fj.202400329r] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 05/08/2024] [Accepted: 05/19/2024] [Indexed: 11/01/2024]
Abstract
Female carriers of a Duchenne muscular dystrophy (DMD) gene mutation manifest exercise intolerance and metabolic anomalies that may be exacerbated following menopause due to the loss of estrogen, a known regulator of skeletal muscle function and metabolism. Here, we studied the impact of estrogen depletion (via ovariectomy) on exercise tolerance and muscle mitochondrial metabolism in female mdx mice and the potential of estrogen replacement therapy (using estradiol) to protect against functional and metabolic perturbations. We also investigated the effect of estrogen depletion, and replacement, on the skeletal muscle proteome through an untargeted proteomic approach with TMT-labelling. Our study confirms that loss of estrogen in female mdx mice reduces exercise capacity, tricarboxylic acid cycle intermediates, and citrate synthase activity but that these deficits are offset through estrogen replacement therapy. Furthermore, ovariectomy downregulated protein expression of RNA-binding motif factor 20 (Rbm20), a critical regulator of sarcomeric and muscle homeostasis gene splicing, which impacted pathways involving ribosomal and mitochondrial translation. Estrogen replacement modulated Rbm20 protein expression and promoted metabolic processes and the upregulation of proteins involved in mitochondrial dynamics and metabolism. Our data suggest that estrogen mitigates dystrophinopathic features in female mdx mice and that estrogen replacement may be a potential therapy for post-menopausal DMD carriers.
Collapse
Affiliation(s)
- Cara A Timpani
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Inherited and Acquired Myopathies Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
- Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, St Albans, Victoria, Australia
| | - Didier Debrincat
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Stephanie Kourakis
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Inherited and Acquired Myopathies Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
| | - Rebecca Boyer
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Luke E Formosa
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Joel R Steele
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Haijian Zhang
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Aaron P Russell
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Emma Rybalka
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Inherited and Acquired Myopathies Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
- Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, St Albans, Victoria, Australia
- Division of Neuropaediatrics and Developmental Medicine, University Children's Hospital of Basel (UKBB), Basel, Switzerland
| | - Angus Lindsay
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
- Department of Medicine, University of Otago, Christchurch, New Zealand
| |
Collapse
|
4
|
Zhang Z, Cheng J, Yang L, Li X, Li Q. Period circadian regulator 2-mediated steroid hormone synthesis by regulating transcription of steroidogenic acute regulatory protein in porcine granulosa cells. J Anim Sci 2024; 102:skae185. [PMID: 38982717 PMCID: PMC11303873 DOI: 10.1093/jas/skae185] [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: 03/20/2024] [Accepted: 07/09/2024] [Indexed: 07/11/2024] Open
Abstract
Steroidogenesis is associated with circadian clock genes. However, the regulation of steroid hormone production in sow granulosal cells by Per2, a crucial circadian regulator, remains unexplored. In this study, we have identified the presence of Per2 in ovarian granulosa cells and have observed its circadian expression pattern. Employing siRNA to interfere with Per2 expression, our investigation revealed that Per2 knockdown notably elevated progesterone (P4) levels along with increasing the expression of StAR but interference of Per2 did not alter the rhythm of clock-related gene (Bmal1, Clock, Per1, and Cry1) in granulosa cells. Subsequent mechanistic analysis showed that Per2 formed complexes with PPARγ and interference with Per2 promoted the formation of the PPARγ:RXRα heterodimer. Importantly, we uncovered that PPARγ:RXRα heterodimer could control the expression of StAR via direct peroxisome proliferator response element binding to its promoter to regulate its activity, and knockdown of Per2 promoted the transcription of StAR via increasing the binding of PPARγ:RXRα ligands. Altogether, these findings indicated a noncanonical role of Per2 in controlling PPARγ:RXRα binding to regulate transcription of StAR and progesterone synthesis, thus revealing potential avenues of pharmacological and therapeutic intervention.
Collapse
Affiliation(s)
- Zelin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Jianyong Cheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Li Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Xiaoya Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Qingwang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| |
Collapse
|
5
|
Motta G, Thangaraj SV, Padmanabhan V. Developmental Programming: Impact of Prenatal Exposure to Bisphenol A on Senescence and Circadian Mediators in the Liver of Sheep. TOXICS 2023; 12:15. [PMID: 38250971 PMCID: PMC10818936 DOI: 10.3390/toxics12010015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024]
Abstract
Prenatal exposure to endocrine disruptors such as bisphenol A (BPA) plays a critical role in the developmental programming of liver dysfunction that is characteristic of nonalcoholic fatty liver disease (NAFLD). Circadian and aging processes have been implicated in the pathogenesis of NAFLD. We hypothesized that the prenatal BPA-induced fatty-liver phenotype of female sheep is associated with premature hepatic senescence and disruption in circadian clock genes. The expression of circadian rhythm and aging-associated genes, along with other markers of senescence such as telomere length, mitochondrial DNA copy number, and lipofuscin accumulation, were evaluated in the liver tissue of control and prenatal BPA groups. Prenatal BPA exposure significantly elevated the expression of aging-associated genes GLB1 and CISD2 and induced large magnitude differences in the expression of other aging genes-APOE, HGF, KLOTHO, and the clock genes PER2 and CLOCK-in the liver; the other senescence markers remained unaffected. Prenatal BPA-programmed aging-related transcriptional changes in the liver may contribute to pathological changes in liver function, elucidating the involvement of aging genes in the pathogenesis of liver steatosis.
Collapse
Affiliation(s)
| | | | - Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48105, USA; (G.M.); (S.V.T.)
| |
Collapse
|
6
|
Nomura S, Hosono T, Ono M, Daikoku T, Michihiro M, Kagami K, Iizuka T, Chen Y, Shi Y, Morishige JI, Fujiwara T, Fujiwara H, Ando H. Desynchronization between Food Intake and Light Stimulations Induces Uterine Clock Quiescence in Female Mice. J Nutr 2023; 153:2283-2290. [PMID: 37336322 DOI: 10.1016/j.tjnut.2023.06.018] [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: 05/18/2023] [Accepted: 06/15/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Dysmenorrhea is associated with breakfast skipping in young women, suggesting that fasting in the early active phase disrupts uterine functions. OBJECTIVES To investigate the possible involvement of the uterine clock system in fasting-induced uterine dysfunction, we examined core clock gene expressions in the uterus using a 28-h interval-fed mouse model. METHODS Young female mice (8 wk of age) were divided into 3 groups: group I (ad libitum feeding), group II (time-restricted feeding, initial 4 h of the active period every day), and group III (time-restricted feeding for 8 h with a 28-h cycle). Groups II and III have the same fasting interval of 20 h. After analyzing feeding and wheel running behaviors during 2 wk of dietary restriction, mice were sacrificed at 4-h intervals, and the expression profiles of clock genes in the uterus and liver were examined by qPCR. RESULTS The mice in group I took food mainly during the dark phase and those in group II during the initial 4 h of the dark phase, whereas those in group III delayed feeding time by 4 h per cycle. In all groups, spontaneous wheel running was observed during the dark phase. There was no difference in the quantity of feeding and the amount of running exercise among the 3 groups during the second week. The mRNA expressions of peripheral clock genes, Bmal1, Clock, Per1, Per2, Cry1, Nr1d1, and Dbp and a clock-controlled gene, Fabp1, in the uterus showed rhythmic oscillations with normal sequential expression cascade in groups I and II, whereas their expressions decreased and circadian cycles disappeared in group III. In contrast, liver core clock genes in group III showed clear circadian cycles. CONCLUSIONS Fluctuations in the timing of the first food intake impair the uterine clock oscillator system to reduce clock gene expressions and abolish their circadian rhythms.
Collapse
Affiliation(s)
- Satoshi Nomura
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Takashi Hosono
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Masanori Ono
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo, Japan.
| | - Takiko Daikoku
- Division of Animal Disease Model, Research Center for Experimental Modeling of Human Disease, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Mieda Michihiro
- Department of Integrative Neurophysiology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Kyosuke Kagami
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Takashi Iizuka
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Yuchen Chen
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Yifan Shi
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Jun-Ichi Morishige
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Tomoko Fujiwara
- Department of Social Work and Life Design, Kyoto Notre Dame University, Kyoto, Japan
| | - Hiroshi Fujiwara
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan.
| | - Hitoshi Ando
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
7
|
Furtado A, Costa D, Lemos MC, Cavaco JE, Santos CRA, Quintela T. The impact of biological clock and sex hormones on the risk of disease. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 137:39-81. [PMID: 37709381 DOI: 10.1016/bs.apcsb.2023.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Molecular clocks are responsible for defining 24-h cycles of behaviour and physiology that are called circadian rhythms. Several structures and tissues are responsible for generating these circadian rhythms and are named circadian clocks. The suprachiasmatic nucleus of the hypothalamus is believed to be the master circadian clock receiving light input via the optic nerve and aligning internal rhythms with environmental cues. Studies using both in vivo and in vitro methodologies have reported the relationship between the molecular clock and sex hormones. The circadian system is directly responsible for controlling the synthesis of sex hormones and this synthesis varies according to the time of day and phase of the estrous cycle. Sex hormones also directly interact with the circadian system to regulate circadian gene expression, adjust biological processes, and even adjust their own synthesis. Several diseases have been linked with alterations in either the sex hormone background or the molecular clock. So, in this chapter we aim to summarize the current understanding of the relationship between the circadian system and sex hormones and their combined role in the onset of several related diseases.
Collapse
Affiliation(s)
- André Furtado
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Portugal
| | - Diana Costa
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Portugal
| | - Manuel C Lemos
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Portugal
| | - J Eduardo Cavaco
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Portugal
| | - Cecília R A Santos
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Portugal
| | - Telma Quintela
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Portugal; UDI-IPG, Unidade de Investigação para o Desenvolvimento do Interior, Instituto Politécnico da Guarda, Guarda, Portugal.
| |
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
|
Ono M, Ando H, Daikoku T, Fujiwara T, Mieda M, Mizumoto Y, Iizuka T, Kagami K, Hosono T, Nomura S, Toyoda N, Sekizuka-Kagami N, Maida Y, Kuji N, Nishi H, Fujiwara H. The Circadian Clock, Nutritional Signals and Reproduction: A Close Relationship. Int J Mol Sci 2023; 24:ijms24021545. [PMID: 36675058 PMCID: PMC9865912 DOI: 10.3390/ijms24021545] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/08/2023] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
The circadian rhythm, which is necessary for reproduction, is controlled by clock genes. In the mouse uterus, the oscillation of the circadian clock gene has been observed. The transcription of the core clock gene period (Per) and cryptochrome (Cry) is activated by the heterodimer of the transcription factor circadian locomotor output cycles kaput (Clock) and brain and muscle Arnt-like protein-1 (Bmal1). By binding to E-box sequences in the promoters of Per1/2 and Cry1/2 genes, the CLOCK-BMAL1 heterodimer promotes the transcription of these genes. Per1/2 and Cry1/2 form a complex with the Clock/Bmal1 heterodimer and inactivate its transcriptional activities. Endometrial BMAL1 expression levels are lower in human recurrent-miscarriage sufferers. Additionally, it was shown that the presence of BMAL1-depleted decidual cells prevents trophoblast invasion, highlighting the importance of the endometrial clock throughout pregnancy. It is widely known that hormone synthesis is disturbed and sterility develops in Bmal1-deficient mice. Recently, we discovered that animals with uterus-specific Bmal1 loss also had poor placental development, and these mice also had intrauterine fetal death. Furthermore, it was shown that time-restricted feeding controlled the uterine clock's circadian rhythm. The uterine clock system may be a possibility for pregnancy complications, according to these results. We summarize the most recent research on the close connection between the circadian clock and reproduction in this review.
Collapse
Affiliation(s)
- Masanori Ono
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo 160-0023, Japan
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
- Correspondence: ; Tel.: +81-3-3342-6111
| | - Hitoshi Ando
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
| | - Takiko Daikoku
- Institute for Experimental Animals, Advanced Science Research Center, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
| | - Tomoko Fujiwara
- Department of Social Work and Life Design, Kyoto Notre Dame University, Kyoto 606-0848, Japan
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
| | - Yasunari Mizumoto
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
| | - Takashi Iizuka
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
| | - Kyosuke Kagami
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
| | - Takashi Hosono
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
| | - Satoshi Nomura
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
| | - Natsumi Toyoda
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
- Institute for Experimental Animals, Advanced Science Research Center, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
| | - Naomi Sekizuka-Kagami
- Department of Nursing, College of Medical, Pharmaceutical, and Health Sciences, Kanazawa University, Kanazawa 920-8641, Japan
| | - Yoshiko Maida
- Department of Nursing, College of Medical, Pharmaceutical, and Health Sciences, Kanazawa University, Kanazawa 920-8641, Japan
| | - Naoaki Kuji
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Hirotaka Nishi
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Hiroshi Fujiwara
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
| |
Collapse
|
10
|
Lecour S, Du Pré BC, Bøtker HE, Brundel BJJM, Daiber A, Davidson SM, Ferdinandy P, Girao H, Gollmann-Tepeköylü C, Gyöngyösi M, Hausenloy DJ, Madonna R, Marber M, Perrino C, Pesce M, Schulz R, Sluijter JPG, Steffens S, Van Linthout S, Young ME, Van Laake LW. Circadian rhythms in ischaemic heart disease: key aspects for preclinical and translational research: position paper of the ESC working group on cellular biology of the heart. Cardiovasc Res 2022; 118:2566-2581. [PMID: 34505881 DOI: 10.1093/cvr/cvab293] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 06/04/2021] [Accepted: 09/07/2021] [Indexed: 12/11/2022] Open
Abstract
Circadian rhythms are internal regulatory processes controlled by molecular clocks present in essentially every mammalian organ that temporally regulate major physiological functions. In the cardiovascular system, the circadian clock governs heart rate, blood pressure, cardiac metabolism, contractility, and coagulation. Recent experimental and clinical studies highlight the possible importance of circadian rhythms in the pathophysiology, outcome, or treatment success of cardiovascular disease, including ischaemic heart disease. Disturbances in circadian rhythms are associated with increased cardiovascular risk and worsen outcome. Therefore, it is important to consider circadian rhythms as a key research parameter to better understand cardiac physiology/pathology, and to improve the chances of translation and efficacy of cardiac therapies, including those for ischaemic heart disease. The aim of this Position Paper by the European Society of Cardiology Working Group Cellular Biology of the Heart is to highlight key aspects of circadian rhythms to consider for improvement of preclinical and translational studies related to ischaemic heart disease and cardioprotection. Applying these considerations to future studies may increase the potential for better translation of new treatments into successful clinical outcomes.
Collapse
Affiliation(s)
- Sandrine Lecour
- Department of Medicine, Hatter Institute for Cardiovascular Research in Africa, University of Cape Town, Cape Town, South Africa
| | - Bastiaan C Du Pré
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Andreas Daiber
- Department of Cardiology, Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Henrique Girao
- Faculty of Medicine, Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), Clinical Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | | | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University Singapore, Singapore
- The Hatter Cardiovascular Institute, University College London, London, UK
- Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taichung City, Taiwan
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Pisa, Italy
- Department of Internal Medicine, University of Texas Medical School in Houston, Houston, TX, USA
| | - Michael Marber
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, London, UK
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Joost P G Sluijter
- Department of Cardiology, Experimental Cardiology Laboratory, Regenerative Medicine Center, Circulatory Health Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies & Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité, University Medicine Berlin, Berlin 10178, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Martin E Young
- Division of Cardiovascular Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Linda W Van Laake
- Cardiology and UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
11
|
Guedes Linhares SS, da Silva Rodrigues Meurer Y, Aquino A, Aquino Câmara D, Mateus Brandão LE, Dierschnabel AL, Porto Fiuza F, Hypólito Lima R, Engelberth RC, Cavalcante JS. Effects of prenatal exposure to fluoxetine on circadian rhythmicity in the locomotor activity and neuropeptide Y and 5-HT expression in male and female adult Wistar rats. Int J Dev Neurosci 2022; 82:407-422. [PMID: 35481929 DOI: 10.1002/jdn.10189] [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: 10/21/2021] [Revised: 03/04/2022] [Accepted: 04/03/2022] [Indexed: 11/07/2022] Open
Abstract
Serotonin (5-HT) reuptake inhibitors, such as fluoxetine, are the most prescribed antidepressant for maternal depression. In this sense, it exposes mothers and the brains of infants to increased modulatory and trophic effects of serotonergic neurotransmission. 5-HT promotes essential brain changes throughout its development, which include neuron migration, differentiation, and organization of neural circuitries related to emotional, cognitive, and circadian behavior. Early exposure to the SSRIs induces long-term effects on behavioral and neural serotonergic signalization. We have aimed to evaluate the circadian rhythm of locomotor activity and the neurochemical content, neuropeptide Y (NPY) and 5-HT in three brain areas: intergeniculate leaflet (IGL), suprachiasmatic nuclei (SCN) and raphe nuclei (RN), at two zeitgebers (ZT6 and ZT18), in male and female rat's offspring early exposed (developmental period GD13-GD21) to fluoxetine (20mg/kg). First, we have conducted daily records of the locomotor activity rhythm using activity sensors coupled to individual cages over four weeks. We have lastly evaluated the immunoreactivity of NPY in both SCN and IGL, and as well the 5-HT expression in the dorsal and medial RN. In summary, our results showed that (1) prenatal fluoxetine affects phase entrainment of the rest/activity rhythm at ZT6 and ZT18, more in male than female specimens, and (2) modulates the NPY and 5-HT expression. Here, we show male rats are more susceptible to phase entrainment and the NPY and 5-HT misexpression compared to female ones. The sex differences induced by early exposure to fluoxetine in both the circadian rhythm of locomotor activity and the neurochemical expression into SCN, IGL, and midbrain raphe are an important highlight in the present work. Thus, our results may help to improve the knowledge on neurobiological mechanisms of circadian rhythms and are relevant to understanding the "broken brains" and behavioral abnormalities of offspring early exposed to antidepressants.
Collapse
Affiliation(s)
- Sara Sophia Guedes Linhares
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Ywlliane da Silva Rodrigues Meurer
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Antonio Aquino
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Diego Aquino Câmara
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | | | - Aline Lima Dierschnabel
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Felipe Porto Fiuza
- Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Graduate Program in Neuroengineering, Macaíba, Brazil
| | - Ramon Hypólito Lima
- Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Graduate Program in Neuroengineering, Macaíba, Brazil
| | - Rovena Clara Engelberth
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Jeferson Souza Cavalcante
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| |
Collapse
|
12
|
Continuous artificial light potentially disrupts central and peripheral reproductive clocks leading to altered uterine physiology and reduced pregnancy success in albino mice. Photochem Photobiol Sci 2022; 21:1217-1232. [PMID: 35399124 DOI: 10.1007/s43630-022-00210-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 03/11/2022] [Indexed: 10/18/2022]
Abstract
AIMS The mechanism behind clock coordination in female reproductive disorders is poorly understood despite the known importance of coordinated and synchronized timing of central and clocks in reproductive organs. We investigated the effect of continuous artificial light (LL) on the central and peripheral reproductive clock gene (Bmal1, Clock, Per1, Per2 and Cry1) and its downstream regulators (Hgf, PR-A and HOXA10) during non-pregnancy and pregnancy phases of female mice. MAIN METHODS Mice (n = 60) in two sets, were maintained under continuous light (LL) and natural day cycle (LD;12L: 12D) for both non-pregnant and pregnant study. Tissues from hypothalamus-containing SCN, ovary, uterus and serum were collected at different zeitgeber time points (ZT; at 4-h intervals across 24-h periods). KEY FINDINGS LL exposure desynchronized the expressions of the clock mRNAs (Bmal1, Clock, Per1, Per2 and Cry1) in SCN, ovary, and uterus along with Hgf mRNA rhythm. LL significantly increased the thickness of endometrial tissues. Furthermore, the pregnant study revealed lower serum progesterone level during peri- and post-implantation under LL along with downregulated expression of progesterone receptor (PR) as well as progesterone dependent uterine Homeobox A-10 (Hoxa10) proteins with lowered pregnancy outcomes. SIGNIFICANCE Our result suggests that LL disrupted the circadian coordination between central and clock genes in reproductive tissue leading to interrupted uterine physiology and altered pregnancy in mice. This led us to propose that duration of light exposure at work-places or home for females is very important in prevention of pregnancy anomalies.
Collapse
|
13
|
Modelling Female Physiology from Head to Toe: Impact of Sex Hormones, Menstrual Cycle, and Pregnancy. J Theor Biol 2022; 540:111074. [PMID: 35227731 DOI: 10.1016/j.jtbi.2022.111074] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 12/14/2022]
|
14
|
Scariot PP, Gobatto CA, Polisel EE, Gomes AE, Beck WR, Manchado-Gobatto FB. Early-life mice housed in standard stocking density reduce the spontaneous physical activity and increase visceral fat deposition before reaching adulthood. Lab Anim 2022; 56:344-355. [PMID: 35062839 DOI: 10.1177/00236772211065915] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Laboratory rodents spend the entire day housed in standard cages that provide a restricted area for movements and might, therefore, limit physical activity. However, it has not been tested in immature rodents of ages ranging from weaning to adulthood (adolescence period) whether the restricted area per animal does actually reduce physical activity and impact the body composition. We analyzed the spontaneous physical activity and feeding behavior during the adolescence of mice kept in two different housing conditions (standard stocking density (SSD) versus low stocking density (LSD)). We aimed to compare the body composition between SSD and LSD groups before they reached adulthood. Differential housing began at four weeks of age and was maintained for four weeks until euthanasia at eight weeks of age. The SSD group had a floor space of 88 cm2 available per animal, while LSD mice were housed with a floor space of 320 cm2 per animal, increasing the individual radius for movement more than three-fold compared with standard requirements. Mice kept in SSD exhibit lower spontaneous physical activity than mice kept in LSD. Early-life exposure to reduced physical activity in mice housed in SSD resulted in greater visceral fat accumulation before adulthood. An environment enabling/stimulating physical activity should be established for rodents as early as possible. This study will be helpful in showing that mice kept in SSD are early exposed to a reduced physical activity already in the adolescence period. Our findings could raise reflections about the translatability of rodents kept in SSD to healthy active humans.
Collapse
Affiliation(s)
- Pedro Pm Scariot
- Laboratory of Applied Sport Physiology, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Claudio A Gobatto
- Laboratory of Applied Sport Physiology, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Emanuel Ec Polisel
- Laboratory of Applied Sport Physiology, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Ana Ec Gomes
- Laboratory of Applied Sport Physiology, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Wladimir R Beck
- Laboratory of Endocrine Physiology and Physical Exercise, Department of Physiological Sciences, Federal University of São Carlos, Brazil
| | - Fúlvia B Manchado-Gobatto
- Laboratory of Applied Sport Physiology, School of Applied Sciences, University of Campinas, Limeira, Brazil
| |
Collapse
|
15
|
Zhang J, Zhao L, Li Y, Dong H, Zhang H, Zhang Y, Ma T, Yang L, Gao D, Wang X, Jiang H, Li C, Wang A, Jin Y, Chen H. Circadian clock regulates granulosa cell autophagy through NR1D1-mediated inhibition of ATG5. Am J Physiol Cell Physiol 2021; 322:C231-C245. [PMID: 34936504 DOI: 10.1152/ajpcell.00267.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Autophagy of granulosa cells (GCs) is involved in follicular atresia, which occurs repeatedly during the ovarian development cycle. Several circadian clock genes are rhythmically expressed in both rodent ovarian tissues and GCs. Nuclear receptor subfamily 1 group D member 1 (NR1D1), an important component of the circadian clock system, is involved in the autophagy process through the regulation of autophagy-related genes. However, there are no reports illustrating the role of the circadian clock system in mouse GC autophagy. In the present study, we found that core circadian clock genes (Bmal1, Per2, Nr1d1, and Dbp) and an autophagy-related gene (Atg5) exhibited rhythmic expression patterns across 24 h in mouse ovaries and primary GCs. Treatment with SR9009, an agonist of NR1D1, significantly reduced the expression of Bmal1, Per2, and Dbp in mouse GCs. ATG5 expression was significantly attenuated by SR9009 treatment in mouse GCs. Conversely, Nr1d1 knockdown increased ATG5 expression in mouse GCs. Decreased NR1D1 expression at both the mRNA and protein levels was detected in the ovaries of Bmal1-/- mice, along with elevated expression of ATG5. Dual-luciferase reporter assay and electrophoretic mobility shift assay showed that NR1D1 inhibited Atg5 transcription by binding to two putative retinoic acid-related orphan receptor response elements within the promoter. In addition, rapamycin-induced autophagy and ATG5 expression were partially reversed by SR9009 treatment in mouse GCs. Taken together, our current data demonstrated that the circadian clock regulates GC autophagy through NR1D1-mediated inhibition of ATG5 expression, and thus, plays a role in maintaining autophagy homeostasis in GCs.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Lijia Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Yating Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Hao Dong
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Haisen Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Yu Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Tiantian Ma
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Luda Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Dengke Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaoyu Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Haizhen Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Chao Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Aihua Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural affairs, Northwest A&F University, Yangling, Shaanxi, China.,Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yaping Jin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Huatao Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural affairs, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
16
|
Han Q, He X, Di R, Chu M. Comparison of expression patterns of six canonical clock genes of follicular phase and luteal phase in Small-tailed Han sheep. Arch Anim Breed 2021; 64:457-466. [PMID: 34746369 PMCID: PMC8567854 DOI: 10.5194/aab-64-457-2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 08/30/2021] [Indexed: 12/24/2022] Open
Abstract
The circadian rhythm is a biological rhythm that is closely related to
the rhythmic expression of a series of clock genes. Results from several
studies have indicated that clock genes are associated with the estrous cycle in
female animals. Until now, the relationship between estrus cycle transition
and clock gene expression in reproductive-axis-related tissues has remained
unknown in Small-tailed Han (STH) sheep. This study was conducted to analyze
the expression patterns of six canonical clock genes (Clock, BMAL1, Per1, Per2, Cry1, and Cry2) in the follicle
phase and luteal phase of STH sheep. We found that all six genes were
expressed in the brain, cerebellum, hypothalamus, pituitary, ovary, uterus,
and oviduct in follicle and luteal phases. The results indicated that Clock expression
was significantly higher in the cerebellum, hypothalamus, and uterus of
the luteal phase than that of the follicle phase, whereas BMAL1 expression was
significantly higher in the hypothalamus of the luteal phase than that of
the follicle phase. Per1 expression was significantly higher in the brain,
cerebellum, hypothalamus, and pituitary of the luteal phase than that of the follicle
phase, and Per2 expression was significantly higher in the hypothalamus,
pituitary, and uterus of the luteal phase than that of the follicle phase. Cry1
expression was significantly higher in the brain, cerebellum, and
hypothalamus of the luteal phase than that of the follicle phase, whereas Cry2 expression
was significantly higher in the pituitary of the luteal phase than that of the
follicle phase. The clock gene expression in all tissues was different
between follicle and luteal phases, but all clock gene mRNA levels were
found to exhibit higher expression among seven tissues in the luteal
phase. Our results suggest that estrous cycles may be associated
with clock gene expression in the STH sheep. This is the first study to
systematically analyze the expression patterns of clock genes of different
estrous cycle in ewes, which could form a basis for further studies to
develop the relationship between clock genes and the estrous cycle.
Collapse
Affiliation(s)
- Qi Han
- Key Laboratory of Animal Genetics and Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Xiaoyun He
- Key Laboratory of Animal Genetics and Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Ran Di
- Key Laboratory of Animal Genetics and Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Mingxing Chu
- Key Laboratory of Animal Genetics and Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| |
Collapse
|
17
|
Hoffmann HM, Meadows JD, Breuer JA, Yaw AM, Nguyen D, Tonsfeldt KJ, Chin AY, Devries BM, Trang C, Oosterhouse HJ, Lee JS, Doser JW, Gorman MR, Welsh DK, Mellon PL. The transcription factors SIX3 and VAX1 are required for suprachiasmatic nucleus circadian output and fertility in female mice. J Neurosci Res 2021; 99:2625-2645. [PMID: 34212416 PMCID: PMC8577618 DOI: 10.1002/jnr.24864] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 05/10/2021] [Indexed: 12/12/2022]
Abstract
The homeodomain transcription factors sine oculis homeobox 3 (Six3) and ventral anterior homeobox 1 (Vax1) are required for brain development. Their expression in specific brain areas is maintained in adulthood, where their functions are poorly understood. To identify the roles of Six3 and Vax1 in neurons, we conditionally deleted each gene using Synapsincre , a promoter targeting maturing neurons, and generated Six3syn and Vax1syn mice. Six3syn and Vax1syn females, but not males, had reduced fertility, due to impairment of the luteinizing hormone (LH) surge driving ovulation. In nocturnal rodents, the LH surge requires a precise timing signal from the brain's circadian pacemaker, the suprachiasmatic nucleus (SCN), near the time of activity onset. Indeed, both Six3syn and Vax1syn females had impaired rhythmic SCN output, which was associated with weakened Period 2 molecular clock function in both Six3syn and Vax1syn mice. These impairments were associated with a reduction of the SCN neuropeptide vasoactive intestinal peptide in Vax1syn mice and a modest weakening of SCN timekeeping function in both Six3syn and Vax1syn mice. Changes in SCN function were associated with mistimed peak PER2::LUC expression in the SCN and pituitary in both Six3syn and Vax1syn females. Interestingly, Six3syn ovaries presented reduced sensitivity to LH, causing reduced ovulation during superovulation. In conclusion, we have identified novel roles of the homeodomain transcription factors SIX3 and VAX1 in neurons, where they are required for proper molecular circadian clock function, SCN rhythmic output, and female fertility.
Collapse
Affiliation(s)
- Hanne M. Hoffmann
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA
| | - Jason D. Meadows
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Joseph A. Breuer
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Alexandra M. Yaw
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA
| | - Duong Nguyen
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA
| | - Karen J. Tonsfeldt
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Austin Y. Chin
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Brooke M. Devries
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA
| | - Crystal Trang
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Haley J. Oosterhouse
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Jessica Sora Lee
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Jeffrey W. Doser
- CANR Statistical Consulting Center, Michigan State University, East Lansing, MI, USA
| | - Michael R. Gorman
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
- Department of Psychology, University of California, San Diego, La Jolla, CA, USA
| | - David K. Welsh
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Pamela L. Mellon
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
18
|
Lopes-Júnior LC, Veronez LC. Circadian rhythms disruption in cancer. BIOL RHYTHM RES 2021. [DOI: 10.1080/09291016.2021.1951470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Luís Carlos Lopes-Júnior
- Postgraduate Program in Nutrition and Health in Sciences. Health Sciences Center at the Universidade Federal Do Espírito Santo (UFES), Vitória, ES, Brazil
| | - Luciana Chain Veronez
- BSc in Biology., Ph.D. In Immunology. Post-doctoral Fellow at the Department of Childcare and Pediatrics at the Ribeirão PretoMedical School at the University of São Paulo (USP). (FMRP-USP)., Ribeirão Preto, SP, Brazil
| |
Collapse
|
19
|
Alvord VM, Kantra EJ, Pendergast JS. Estrogens and the circadian system. Semin Cell Dev Biol 2021; 126:56-65. [PMID: 33975754 DOI: 10.1016/j.semcdb.2021.04.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 11/26/2022]
Abstract
Circadian rhythms are ~24 h cycles of behavior and physiology that are generated by a network of molecular clocks located in nearly every tissue in the body. In mammals, the circadian system is organized hierarchically such that the suprachiasmatic nucleus (SCN) is the main circadian clock that receives light information from the eye and entrains to the light-dark cycle. The SCN then coordinates the timing of tissue clocks so internal rhythms are aligned with environmental cycles. Estrogens interact with the circadian system to regulate biological processes. At the molecular level, estrogens and circadian genes interact to regulate gene expression and cell biology. Estrogens also regulate circadian behavior across the estrous cycle. The timing of ovulation during the estrous cycle requires coincident estrogen and SCN signals. Studies using circadian gene reporter mice have also elucidated estrogen regulation of peripheral tissue clocks and metabolic rhythms. This review synthesizes current understanding of the interplay between estrogens and the circadian system, with a focus on female rodents, in regulating molecular, physiological, and behavioral processes.
Collapse
|
20
|
Hosono T, Ono M, Daikoku T, Mieda M, Nomura S, Kagami K, Iizuka T, Nakata R, Fujiwara T, Fujiwara H, Ando H. Time-Restricted Feeding Regulates Circadian Rhythm of Murine Uterine Clock. Curr Dev Nutr 2021; 5:nzab064. [PMID: 33981944 PMCID: PMC8099714 DOI: 10.1093/cdn/nzab064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/23/2021] [Accepted: 04/07/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Skipping breakfast is associated with dysmenorrhea in young women. This suggests that the delay of food intake in the active phase impairs uterine functions by interfering with circadian rhythms. OBJECTIVES To examine the relation between the delay of feeding and uterine circadian rhythms, we investigated the effects of the first meal occasion in the active phase on the uterine clock. METHODS Zeitgeber time (ZT) was defined as ZT0 (08:45) with lights on and ZT12 (20:45) with lights off. Young female mice (8 wk of age) were divided into 3 groups: group I (ad libitum consumption), group II (time-restricted feeding during ZT12-16, initial 4 h of the active period), and group III (time-restricted feeding during ZT20-24, last 4 h of the active period, a breakfast-skipping model). After 2 wk of dietary restriction, mice in each group were killed at 4-h intervals and the expression profiles of uterine clock genes, Bmal1 (brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein 1), Per1 (period circadian clock 1), Per2, and Cry1 (cryptochrome 1), were examined. RESULTS qPCR and western blot analyses demonstrated synchronized circadian clock gene expression within the uterus. Immunohistochemical analysis confirmed that BMAL1 protein expression was synchronized among the endometrium and myometrium. In groups I and II, mRNA expression of Bmal1 was elevated after ZT12 at the start of the active phase. In contrast, Bmal1 expression was elevated just after ZT20 in group III, showing that the uterine clock rhythm had shifted 8 h backward. The changes in BMAL1 protein expression were confirmed by western blot analysis. CONCLUSIONS This study is the first to indicate that time-restricted feeding regulates a circadian rhythm of the uterine clock that is synchronized throughout the uterine body. These findings suggest that the uterine clock system is a new candidate to explain the etiology of breakfast skipping-induced uterine dysfunction.
Collapse
Affiliation(s)
- Takashi Hosono
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Masanori Ono
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Takiko Daikoku
- Institute for Experimental Animals, Advanced Science Research Center, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Satoshi Nomura
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Kyosuke Kagami
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Takashi Iizuka
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Rieko Nakata
- Department of Food Science and Nutrition, Nara Women's University, Nara, Japan
| | - Tomoko Fujiwara
- Department of Social Work and Life Design, Kyoto Notre Dame University, Kyoto, Japan
| | - Hiroshi Fujiwara
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Hitoshi Ando
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
21
|
Zhao L, Yang L, Zhang J, Xiao Y, Wu M, Ma T, Wang X, Zhang L, Jiang H, Chao HW, Wang A, Jin Y, Chen H. Bmal1 promotes prostaglandin E 2 synthesis by upregulating Ptgs2 transcription in response to increasing estradiol levels in day 4 pregnant mice. Am J Physiol Endocrinol Metab 2021; 320:E747-E759. [PMID: 33554778 DOI: 10.1152/ajpendo.00466.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/31/2021] [Indexed: 12/28/2022]
Abstract
Prostaglandin G/H synthase 2 (PTGS2) is a rate-limiting enzyme in prostaglandin synthesis. The present study assessed the role of the uterine circadian clock on Ptgs2 transcription in response to steroid hormones during early pregnancy. We demonstrated that the core clock genes (Bmal1, Per2, Nr1d1, and Dbp), Vegf, and Ptgs2, and their encoded proteins, have rhythmic expression in the mouse uterus from days 3.5 to 4.5 (D3.5-4.5) of pregnancy. Progesterone (P4) treatment of cultured uterus endometrial stromal cells (UESCs) isolated from mPer2Luciferase reporter gene knock-in mice on D4 induced a phase shift in PER2::LUCIFERASE oscillations. This P4-induced phase shift of PER2::LUCIFERASE oscillations was significantly attenuated by the P4 antagonist RU486. Additionally, the amplitude of PER2::LUCIFERASE oscillations was increased by estradiol (E2) treatment in the presence of P4. Consistently, the mRNA levels of clock genes (Bmal1 and Per2), Vegf, and Ptgs2 were markedly increased by E2 treatment of UESCs in the presence of P4. Treatment with E2 also promoted prostaglandin E2 (PGE2) synthesis by UESCs. Depletion of Bmal1 in UESCs by small-interfering RNA (siRNA) decreased the transcript levels of clock genes (Nr1d1 and Dbp), Vegf, and Ptgs2 compared with nonsilencing siRNA treatment. Bmal1 knockdown also inhibited PGE2 synthesis. Moreover, the mRNA expression levels of clock genes (Nr1d1 and Dbp), Vegf, and Ptgs2, and their respective proteins were significantly decreased in the uterus of Bmal1-/- mice. Thus, these data suggest that Bmal1 in mice promotes PGE2 synthesis by upregulating Ptgs2 in response to increases in E2 on D4 of pregnancy.NEW & NOTEWORTHY Rhythmic expression of Bmal1 and Ptgs2 was observed in the uterus isolated from D3.5-4.5 of pregnant mice. E2 increased the expression of Bmal1 and Ptg2 in UESCs isolated from mice on D4. The expression of Ptgs2 was significantly decreased in Bmal1-siRNA treated UESCs. Bmal1 knockdown also inhibited PGE2 synthesis. Thus, these data suggest that Bmal1 in mice promotes PGE2 synthesis by upregulating Ptgs2 in response to increases in E2 on D4 of pregnancy.
Collapse
Affiliation(s)
- Lijia Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Luda Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Jing Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Yaoyao Xiao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Meina Wu
- Key Laboratory of Cellular Physiology, Department of Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China
| | - Tiantian Ma
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Xiaoyu Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Linlin Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Haizhen Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Hsu-Wen Chao
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Aihua Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, China
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Shaanxi, China
| | - Yaping Jin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Huatao Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, China
| |
Collapse
|
22
|
Bottalico LN, Weljie AM. Cross-species physiological interactions of endocrine disrupting chemicals with the circadian clock. Gen Comp Endocrinol 2021; 301:113650. [PMID: 33166531 PMCID: PMC7993548 DOI: 10.1016/j.ygcen.2020.113650] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 10/09/2020] [Accepted: 10/17/2020] [Indexed: 02/06/2023]
Abstract
Endocrine disrupting chemicals (EDCs) are endocrine-active chemical pollutants that disrupt reproductive, neuroendocrine, cardiovascular and metabolic health across species. The circadian clock is a transcriptional oscillator responsible for entraining 24-hour rhythms of physiology, behavior and metabolism. Extensive bidirectional cross talk exists between circadian and endocrine systems and circadian rhythmicity is present at all levels of endocrine control, from synthesis and release of hormones, to sensitivity of target tissues to hormone action. In mammals, a range of hormones directly alter clock gene expression and circadian physiology via nuclear receptor (NR) binding and subsequent genomic action, modulating physiological processes such as nutrient and energy metabolism, stress response, reproductive physiology and circadian behavioral rhythms. The potential for EDCs to perturb circadian clocks or circadian-driven physiology is not well characterized. For this reason, we explore evidence for parallel endocrine and circadian disruption following EDC exposure across species. In the reviewed studies, EDCs dysregulated core clock and circadian rhythm network gene expression in brain and peripheral organs, and altered circadian reproductive, behavioral and metabolic rhythms. Circadian impacts occurred in parallel to endocrine and metabolic alterations such as impaired fertility and dysregulated metabolic and energetic homeostasis. Further research is warranted to understand the nature of interaction between circadian and endocrine systems in mediating physiological effects of EDC exposure at environmental levels.
Collapse
Affiliation(s)
- Lisa N Bottalico
- Department of Systems Pharmacology and Translational Therapeutics, Institute for Translational Medicine and Therapeutics, Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Aalim M Weljie
- Department of Systems Pharmacology and Translational Therapeutics, Institute for Translational Medicine and Therapeutics, Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
23
|
Yaw AM, McLane-Svoboda AK, Hoffmann HM. Shiftwork and Light at Night Negatively Impact Molecular and Endocrine Timekeeping in the Female Reproductive Axis in Humans and Rodents. Int J Mol Sci 2020; 22:E324. [PMID: 33396885 PMCID: PMC7795361 DOI: 10.3390/ijms22010324] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/24/2020] [Accepted: 12/25/2020] [Indexed: 01/17/2023] Open
Abstract
Shiftwork, including work that takes place at night (nightshift) and/or rotates between day and nightshifts, plays an important role in our society, but is associated with decreased health, including reproductive dysfunction. One key factor in shiftwork, exposure to light at night, has been identified as a likely contributor to the underlying health risks associated with shiftwork. Light at night disrupts the behavioral and molecular circadian timekeeping system, which is important for coordinated timing of physiological processes, causing mistimed hormone release and impaired physiological functions. This review focuses on the impact of shiftwork on reproductive function and pregnancy in women and laboratory rodents and potential underlying molecular mechanisms. We summarize the negative impact of shiftwork on female fertility and compare these findings to studies in rodent models of light shifts. Light-shift rodent models recapitulate several aspects of reproductive dysfunction found in shift workers, and their comparison with human studies can enable a deeper understanding of physiological and hormonal responses to light shifts and the underlying molecular mechanisms that may lead to reproductive disruption in human shift workers. The contributions of human and rodent studies are essential to identify the origins of impaired fertility in women employed in shiftwork.
Collapse
Affiliation(s)
| | | | - Hanne M. Hoffmann
- Department of Animal Science and the Reproductive and Developmental Science Program, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
24
|
Pilorz V, Kolms B, Oster H. Rapid Jetlag Resetting of Behavioral, Physiological, and Molecular Rhythms in Proestrous Female Mice. J Biol Rhythms 2020; 35:612-627. [PMID: 33140660 DOI: 10.1177/0748730420965291] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A gradual adaptation to a shifted light-dark (LD) cycle is a key element of the circadian clock system and believed to be controlled by the central circadian pacemaker, the suprachiasmatic nucleus (SCN). Endocrine factors have a strong influence on the regulation of the circadian clock network and alter acute photic responses of the SCN clock. In females, endocrine function depends on the stage of the ovarian cycle. So far, however, little is known about the effect of the estrous cycle on behavioral and molecular responses to shifts in the LD rhythm. Based on this, we investigated whether estrous state affects the kinetics of phase shift during jetlag in behavior, physiology, and molecular clock rhythms in the SCN and in peripheral tissues. Female mice exposed to an advanced LD phase at proestrous or metestrous showed different phase-shift kinetics, with proestrous females displaying accelerated adaptation in behavior and physiology. Constant darkness release experiments suggest that these fast phase shifts do not reflect resetting of the SCN pacemaker. Explant experiments on SCN, adrenal gland, and uterus confirmed this finding with proestrous females showing significantly faster clock phase shifts in peripheral tissues compared with the SCN. Together, these findings provide strong evidence for an accelerated adaptation of proestrous compared with metestrous females to new LD conditions that is accompanied by rapid behavioral, physiological, and molecular rhythm resetting. Not only do these findings open up a new avenue to understand the effect of estrous cycle on the clock network under changing environmental conditions but also imply a greater susceptibility in proestrous females.
Collapse
Affiliation(s)
- Violetta Pilorz
- Institute of Neurobiology, Center of Brain, Behavior and Metabolism, University of Lübeck, Luebeck, Germany
| | - Beke Kolms
- Institute of Neurobiology, Center of Brain, Behavior and Metabolism, University of Lübeck, Luebeck, Germany
| | - Henrik Oster
- Institute of Neurobiology, Center of Brain, Behavior and Metabolism, University of Lübeck, Luebeck, Germany
| |
Collapse
|
25
|
Zhao Y, Wu Y, Wang J, Liao C, Mi X, Chen F. Circadian transcription factor Dbp promotes rat calvarial osteoprogenitors osteogenic differentiation through Kiss1/GnRH/E2 signaling pathway loop. J Cell Biochem 2020; 122:166-179. [PMID: 32830342 DOI: 10.1002/jcb.29836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/19/2020] [Accepted: 07/24/2020] [Indexed: 11/06/2022]
Abstract
To determine the mechanism by which D-site-binding protein (Dbp) regulates rat calvarial osteoprogenitors (OPCs) osteogenic differentiation. α-Smooth muscle actin (α-SMA) + rat calvarial OPCs were extracted and purified using immunomagnetic beads. Cells were transduced with Dbp-lentivirus and divided into Dbp knockdown, Dbp overexpression and vehicle groups. After osteogenic induction for 21 days, Alizarin red staining and alkaline phosphatase (ALP) activity were examined. Expression levels of Runx2, Ocn, Osterix, Bmp4, Kiss1, and GnRH were determined using a quantitative real-time polymerase chain reaction. The observed changes in Kisspeptin, GnRH, ERα, and Runx2 were further validated via Western blot analysis. Furthermore, E2 and GnRH secretion levels were detected via an enzyme-linked immunosorbent assay (ELISA). Chromatin immunoprecipitation (ChIP) and luciferase assay were used to assess the effects of Dbp on the Kiss1 gene promoter. The coexpression of Dbp and Kisspeptin or GnRH was also evaluated via immunofluorescence. Following osteogenic induction, Dbp overexpression significantly increased calcium nodule formation and ALP activity, as well as Runx2, Ocn, Osterix, Bmp4, Kiss1, and GnRH messenger RNA expression, while Dbp knockdown presented the opposite results. Western blot analysis and ELISA results showed that Dbp significantly promotes Runx2, E2/ERα, Kisspeptin, and GnRH expression. These findings were confirmed by the ChIP assay, which indicated that the estrogen receptor promotes Kisspeptin expression after binding to the Kiss1 gene promoter, which is regulated by Dbp. Immunofluorescence assay showed that Dbp coexpression with Kisspeptin or GnRH varied depending on Dbp expression levels. Collectively, the circadian transcription factor Dbp promotes α-SMA + rat calvarial OPCs osteoblastic differentiation through Kiss1/GnRH/E2 signaling pathway loop.
Collapse
Affiliation(s)
- Yanhui Zhao
- Department of Orthodontics, School & Hospital of Stomatology, Engineering Researching Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China
| | - Yanan Wu
- Department of Orthodontics, School & Hospital of Stomatology, Engineering Researching Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China
| | - Jie Wang
- Department of Orthodontics, School & Hospital of Stomatology, Engineering Researching Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China
| | - Chongshan Liao
- Department of Orthodontics, School & Hospital of Stomatology, Engineering Researching Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China
| | - Xiaohui Mi
- Department of Orthodontics, School & Hospital of Stomatology, Engineering Researching Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China
| | - Fengshan Chen
- Department of Orthodontics, School & Hospital of Stomatology, Engineering Researching Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China
| |
Collapse
|
26
|
A high-salt/high fat diet alters circadian locomotor activity and glucocorticoid synthesis in mice. PLoS One 2020; 15:e0233386. [PMID: 32437460 PMCID: PMC7241774 DOI: 10.1371/journal.pone.0233386] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/03/2020] [Indexed: 12/30/2022] Open
Abstract
Salt is an essential nutrient; however, excessive salt intake is a prominent public health concern worldwide. Various physiological functions are associated with circadian rhythms, and disruption of circadian rhythms is a prominent risk factor for cardiovascular diseases, cancer, and immune disease. Certain nutrients are vital regulators of peripheral circadian clocks. However, the role of a high-fat and high-salt (HFS) diet in the regulation of circadian gene expression is unclear. This study aimed to investigate the effect of an HFS diet on rhythms of locomotor activity, caecum glucocorticoid secretion, and clock gene expression in mice. Mice administered an HFS diet displayed reduced locomotor activity under normal light/dark and constant dark conditions in comparison with those administered a normal diet. The diurnal rhythm of caecum glucocorticoid secretion and the expression levels of glucocorticoid-related genes and clock genes in the adrenal gland were disrupted with an HFS diet. These results suggest that an HFS diet alters locomotor activity, disrupts circadian rhythms of glucocorticoid secretion, and downregulates peripheral adrenal gland circadian clock genes.
Collapse
|
27
|
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
|
28
|
Silva CC, Domínguez R. Clock control of mammalian reproductive cycles: Looking beyond the pre-ovulatory surge of gonadotropins. Rev Endocr Metab Disord 2020; 21:149-163. [PMID: 31828563 DOI: 10.1007/s11154-019-09525-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Several aspects of the physiology and behavior of organisms are expressed rhythmically with a 24-h periodicity and hence called circadian rhythms. Such rhythms are thought to be an adaptive response that allows to anticipate cyclic events in the environment. In mammals, the circadian system is a hierarchically organized net of endogenous oscillators driven by the hypothalamic suprachiasmatic nucleus (SCN). This system is synchronized by the environment throughout afferent pathways and in turn it organizes the activity of tissues by means of humoral secretions and neuronal projections. It has been shown that reproductive cycles are regulated by the circadian system. In rodents, the lesion of the SCN results on alterations of the estrous cycle, sexual behavior, tonic and phasic secretion of gonadotropin releasing hormone (GnRH)/gonadotropins and in the failure of ovulation. Most of the studies regarding the circadian control of reproduction, in particular of ovulation, have only focused on the participation of the SCN in the triggering of the proestrus surge of gonadotropins. Here we review aspects of the evolution and organization of the circadian system with particular focus on its relationship with the reproductive cycle of laboratory rodents. Experimental evidence of circadian control of neuroendocrine events indispensable for ovulation that occur prior to proestrus are discussed. In order to offer a working model of the circadian regulation of reproduction, its participation on aspects ranging from gamete production, neuroendocrine regulation, sexual behavior, mating coordination, pregnancy and deliver of the product should be assessed experimentally.
Collapse
Affiliation(s)
- Carlos-Camilo Silva
- Chronobiology of Reproduction Research Lab-UIBR, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, México City, Mexico
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, México City, Mexico
| | - Roberto Domínguez
- Chronobiology of Reproduction Research Lab-UIBR, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, México City, Mexico.
| |
Collapse
|
29
|
Sen A, Hoffmann HM. Role of core circadian clock genes in hormone release and target tissue sensitivity in the reproductive axis. Mol Cell Endocrinol 2020; 501:110655. [PMID: 31756424 PMCID: PMC6962569 DOI: 10.1016/j.mce.2019.110655] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/15/2019] [Accepted: 11/16/2019] [Indexed: 12/17/2022]
Abstract
Precise timing in hormone release from the hypothalamus, the pituitary and ovary is critical for fertility. Hormonal release patterns of the reproductive axis are regulated by a feedback loop within the hypothalamic-pituitary-gonadal (HPG) axis. The timing and rhythmicity of hormone release and tissue sensitivity in the HPG axis is regulated by circadian clocks located in the hypothalamus (suprachiasmatic nucleus, kisspeptin and GnRH neurons), the pituitary (gonadotrophs), the ovary (theca and granulosa cells), the testis (Leydig cells), as well as the uterus (endometrium and myometrium). The circadian clocks integrate environmental and physiological signals to produce cell endogenous rhythms generated by a transcriptional-translational feedback loop of transcription factors that are collectively called the "molecular clock". This review specifically focuses on the contribution of molecular clock transcription factors in regulating hormone release patterns in the reproductive axis, with an emphasis on the female reproductive system. Specifically, we discuss the contributions of circadian rhythms in distinct neuronal populations of the female hypothalamus, the molecular clock in the pituitary and its overall impact on female and male fertility.
Collapse
Affiliation(s)
- Aritro Sen
- Department of Animal Science and the Reproductive and Developmental Science Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Hanne M Hoffmann
- Department of Animal Science and the Reproductive and Developmental Science Program, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
30
|
Pepe G, Locati M, Della Torre S, Mornata F, Cignarella A, Maggi A, Vegeto E. The estrogen-macrophage interplay in the homeostasis of the female reproductive tract. Hum Reprod Update 2019; 24:652-672. [PMID: 30256960 DOI: 10.1093/humupd/dmy026] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 08/10/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Estrogens are known to orchestrate reproductive events and to regulate the immune system during infections and following tissue damage. Recent findings suggest that, in the absence of any danger signal, estrogens trigger the physiological expansion and functional specialization of macrophages, which are immune cells that populate the female reproductive tract (FRT) and are increasingly being recognized to participate in tissue homeostasis beyond their immune activity against infections. Although estrogens are the only female gonadal hormones that directly target macrophages, a comprehensive view of this endocrine-immune communication and its involvement in the FRT is still missing. OBJECTIVE AND RATIONALE Recent accomplishments encourage a revision of the literature on the ability of macrophages to respond to estrogens and induce tissue-specific functions required for reproductive events, with the aim to envision macrophages as key players in FRT homeostasis and mediators of the regenerative and trophic actions of estrogens. SEARCH METHODS We conducted a systematic search using PubMed and Ovid for human, animal (rodents) and cellular studies published until 2018 on estrogen action in macrophages and the activity of these cells in the FRT. OUTCOMES Our search identified the remarkable ability of macrophages to activate biochemical processes in response to estrogens in cell culture experiments. The distribution at specific locations, interaction with selected cells and acquisition of distinct phenotypes of macrophages in the FRT, as well as the cyclic renewal of these properties at each ovarian cycle, demonstrate the involvement of these cells in the homeostasis of reproductive events. Moreover, current evidence suggests an association between estrogen-macrophage signaling and the generation of a tolerant and regenerative environment in the FRT, although a causative link is still missing. WIDER IMPLICATIONS Dysregulation of the functions and estrogen responsiveness of FRT macrophages may be involved in infertility and estrogen- and macrophage-dependent gynecological diseases, such as ovarian cancer and endometriosis. Thus, more research is needed on the physiology and pharmacological control of this endocrine-immune interplay.
Collapse
Affiliation(s)
- Giovanna Pepe
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, University of Milan, via Balzaretti, 9 Milan, Italy
| | - Massimo Locati
- Humanitas Clinical and Research Center, Segrate, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, via fratelli Cervi, Segrate, Italy
| | - Sara Della Torre
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, University of Milan, via Balzaretti, 9 Milan, Italy
| | - Federica Mornata
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, University of Milan, via Balzaretti, 9 Milan, Italy
| | - Andrea Cignarella
- Department of Medicine, University of Padua, Largo Meneghetti 2, Padua, Italy
| | - Adriana Maggi
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, University of Milan, via Balzaretti, 9 Milan, Italy
| | - Elisabetta Vegeto
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, University of Milan, via Balzaretti, 9 Milan, Italy
| |
Collapse
|
31
|
Xu J, Gao B, Shi MR, Yu H, Huang LY, Chen P, Li YH. Copulation Exerts Significant Effects on mRNA Expression of Cryptochrome Genes in a Moth. JOURNAL OF INSECT SCIENCE (ONLINE) 2019; 19:3. [PMID: 30817821 PMCID: PMC6394973 DOI: 10.1093/jisesa/iez016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 01/23/2019] [Accepted: 01/31/2019] [Indexed: 05/12/2023]
Abstract
It is recognized that the behavioral rhythms of organisms are controlled by the circadian clock, while the reverse direction, i.e., whether changes in physiology and behavior react to the internal rhythms, is unclear. Cryptochromes (CRYs) are photolyase-like flavoproteins with blue-light receptor function and other functions on circadian clock and migration in animals. Here, we cloned the full-length cDNA of CRY1 and CRY2 in Spodoptera litura (Fabricius, 1775) (Lepidoptera: Noctuidae). Sl-CRYs show high similarity to orthologs from other insects, and their conserved regions contain a DNA photolyase domain and a FAD-binding seven domain. The expression levels of both genes were relatively low during the larval stage, which increased during the pupal stage and then peaked at the adult stage. The expression of Sl-CRY1 and Sl-CRY2 showed differences between males and females and between scotophase and photophase. Further, our study demonstrated that copulation has a significant effect on the expression of Sl-CRYs. More interestingly, the changes in the expression of Sl-CRY1 and Sl-CRY2 due to copulation showed the same trend in both sexes, in which the expression levels of both genes in copulated males and females decreased in the subsequent scotophase after copulation and then increased significantly in the following photophase. Considering the nature of the dramatic changes in reproductive behavior and physiology after copulation in S. litura, we propose that the changes in the expression of Sl-CRYs after copulation could have some function in the reproductive process.
Collapse
Affiliation(s)
- Jin Xu
- Yunnan Academy of Biodiversity, Southwest Forestry University, Kunming, China
| | - Bo Gao
- School of Life Sciences, Yunnan University, Kunming, China
| | - Min-Rui Shi
- Yunnan Academy of Biodiversity, Southwest Forestry University, Kunming, China
| | - Hong Yu
- Yunnan Academy of Biodiversity, Southwest Forestry University, Kunming, China
| | - Ling-Yan Huang
- School of Life Sciences, Yunnan University, Kunming, China
| | - Peng Chen
- Yunnan Academy of Forestry, Kunming, China
| | - Yong-He Li
- Yunnan Academy of Biodiversity, Southwest Forestry University, Kunming, China
| |
Collapse
|
32
|
de la Vega MDC, Delsouc MB, Ponce I, Ragusa V, Vallcaneras S, Anzulovich AC, Casais M. Circadian rhythms of factors involved in luteal regression are modified in p55 tumour necrosis factor receptor (TNFRp55)-deficient mice. Reprod Fertil Dev 2018; 30:1651-1665. [PMID: 29903342 DOI: 10.1071/rd18058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/05/2018] [Indexed: 12/17/2023] Open
Abstract
The rhythm of factors involved in luteal regression is crucial in determining the physiological duration of the oestrous cycle. Given the role of tumour necrosis factor (TNF)-α in luteal function and circadian regulation and that most of the effects of TNF-α are mediated by p55 TNF receptor (TNFRp55), the aims of the present study were to analyse the following during the luteal regression phase in the ovary of mice: (1) whether the pattern of expression of progesterone (P4) and the enzymes involved in the synthesis and degradation of P4 is circadian and endogenous (the rhythm persists in constant conditions, (i.e., constant darkness) with a period of about 24 hours); (2) circadian oscillations in clock gene expression; (3) whether there are daily variations in the expression of key genes involved in apoptosis and antioxidant mechanisms; and (4) the consequences of TNFRp55 deficiency. P4 was found to oscillate circadianally following endogenous rhythms of clock factors. Of note, TNFRp55 deficiency modified the circadian oscillation in P4 concentrations and its enzymes involved in the synthesis and degradation of P4, probably as a consequence of changes in the circadian oscillations of brain and muscle ARNT-Like protein 1 (Bmal1) and Cryptochrome 1 (Cry1). Furthermore, TNFRp55 deficiency modified the circadian rhythms of apoptosis genes, as well as antioxidant enzymes and peroxidation levels in the ovary in dioestrus. The findings of the present study strengthen the hypothesis that dysregulation of TNF-α signalling may be a potential cause for altered circadian and menstrual cycling in some gynaecological diseases.
Collapse
Affiliation(s)
- Magali Del C de la Vega
- Laboratorio de Biología de la Reproducción, Facultad de Química, Bioquímica y Farmacia, Instituto Multidisciplinario de Investigaciones Biológicas, San Luis, Universidad Nacional de San Luis, Ejército de los Andes 950, CP D5700HHW, San Luis, Argentina
| | - María B Delsouc
- Laboratorio de Biología de la Reproducción, Facultad de Química, Bioquímica y Farmacia, Instituto Multidisciplinario de Investigaciones Biológicas, San Luis, Universidad Nacional de San Luis, Ejército de los Andes 950, CP D5700HHW, San Luis, Argentina
| | - Ivana Ponce
- Laboratorio de Cronobiología, Facultad de Química, Bioquímica y Farmacia, Instituto Multidisciplinario de Investigaciones Biológicas, San Luis, Universidad Nacional de San Luis, Ejército de los Andes 950, CP D5700HHW, San Luis, Argentina
| | - Vicente Ragusa
- Laboratorio de Biología de la Reproducción, Facultad de Química, Bioquímica y Farmacia, Instituto Multidisciplinario de Investigaciones Biológicas, San Luis, Universidad Nacional de San Luis, Ejército de los Andes 950, CP D5700HHW, San Luis, Argentina
| | - Sandra Vallcaneras
- Laboratorio de Biología de la Reproducción, Facultad de Química, Bioquímica y Farmacia, Instituto Multidisciplinario de Investigaciones Biológicas, San Luis, Universidad Nacional de San Luis, Ejército de los Andes 950, CP D5700HHW, San Luis, Argentina
| | - Ana C Anzulovich
- Laboratorio de Cronobiología, Facultad de Química, Bioquímica y Farmacia, Instituto Multidisciplinario de Investigaciones Biológicas, San Luis, Universidad Nacional de San Luis, Ejército de los Andes 950, CP D5700HHW, San Luis, Argentina
| | - Marilina Casais
- Laboratorio de Biología de la Reproducción, Facultad de Química, Bioquímica y Farmacia, Instituto Multidisciplinario de Investigaciones Biológicas, San Luis, Universidad Nacional de San Luis, Ejército de los Andes 950, CP D5700HHW, San Luis, Argentina
| |
Collapse
|
33
|
Mizuta S, Sugiyama M, Tokuda IT, Nakamura W, Nakamura TJ. Photic phase-response curves for cycling female mice. Horm Behav 2018; 105:41-46. [PMID: 30031017 DOI: 10.1016/j.yhbeh.2018.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 07/16/2018] [Accepted: 07/16/2018] [Indexed: 10/28/2022]
Abstract
The photic entrainment system is critical for the internal circadian clock to be synchronized by external time cues. In nocturnal rodents, exposure to light during the early subjective night causes a phase delay, whereas it causes a phase advance during the late subjective night. This is represented by a phase-response curve (PRC). The PRC of females has not been well studied due to their estrous cycles. Our aim in this study was to understand the characteristics of photic entrainment in female cycling rodents and identify differences in photic entrainment among the stages of the estrous cycle. To establish two types of PRC, immediate PRC (iPRC) and steady state PRC (ssPRC), in each stage of the estrous cycle, we recorded circadian rhythms of wheel running activity, applying a 15-min light pulse to cycling female mice in constant darkness. In the iPRC, which was evaluated on the next day of the light pulse, the amount of phase shift in the diestrus was larger than that in the metestrus stage at circadian time (CT) 2. Similarly, the amount of phase shift in metestrus was larger than that in proestrus at CT 10. In the ssPRC, which was evaluated after completion of a new steady state, no significant estrous variations in the amount of photic phase shifts were detected for any CTs. Although these results indicate that the intrinsic photic entrainment system is not influenced by the estrous cycle, it may affect photoreception and cause sudden behavioral changes.
Collapse
Affiliation(s)
- Shuto Mizuta
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, Kawasaki, Kanagawa 214-8571, Japan
| | - Mizuki Sugiyama
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, Kawasaki, Kanagawa 214-8571, Japan
| | - Isao T Tokuda
- Department of Mechanical Engineering, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan
| | - Wataru Nakamura
- Department of Oral-Chrono Physiology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki 852-8588, Japan
| | - Takahiro J Nakamura
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, Kawasaki, Kanagawa 214-8571, Japan.
| |
Collapse
|
34
|
Mark PJ, Crew RC, Wharfe MD, Waddell BJ. Rhythmic Three-Part Harmony: The Complex Interaction of Maternal, Placental and Fetal Circadian Systems. J Biol Rhythms 2017; 32:534-549. [PMID: 28920512 DOI: 10.1177/0748730417728671] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
From the perspective of circadian biology, mammalian pregnancy presents an unusual biological scenario in which an entire circadian system (i.e., that of the fetus) is embodied within another (i.e., that of the mother). Moreover, both systems are likely to be influenced at their interface by a third player, the placenta. Successful pregnancy requires major adaptations in maternal physiology, many of which involve circadian changes that support the high metabolic demands of the growing fetus. A functional role for maternal circadian adaptations is implied by the effects of circadian disruption, which result in pregnancy complications including higher risks for miscarriage, preterm labor, and low birth weight. Various aspects of fetal physiology lead to circadian variation, at least in late gestation, but it remains unclear what drives this rhythmicity. It likely involves contributions from the maternal environment and possibly from the placenta and the developing intrinsic molecular clocks within fetal tissues. The role of the placenta is of particular significance because it serves not only to relay signals about the external environment (via the mother) but may also exhibit its own circadian rhythmicity. This review considers how the fetus may be influenced by dynamic circadian signals from the mother and the placenta during gestation, and how, in the face of these changing influences, a new fetal circadian system emerges. Particular emphasis is placed on the role of endocrine signals, most notably melatonin and glucocorticoids, as mediators of maternal-fetal circadian interactions, and on the expression of the clock gene in the 3 compartments. Further study is required to understand how the mother, placenta, and fetus interact across pregnancy to optimize circadian adaptations that support adequate growth and development of the fetus and its transition to postnatal life in a circadian environment.
Collapse
Affiliation(s)
- Peter J Mark
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Rachael C Crew
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Michaela D Wharfe
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Brendan J Waddell
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
35
|
The sweet tooth of the circadian clock. Biochem Soc Trans 2017; 45:871-884. [PMID: 28673939 DOI: 10.1042/bst20160183] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/04/2017] [Accepted: 05/16/2017] [Indexed: 12/19/2022]
Abstract
The endogenous circadian clock is a key regulator of daily metabolic processes. On the other hand, circadian clocks in a broad range of tissues can be tuned by extrinsic and intrinsic metabolic cues. The bidirectional interaction between circadian clocks and metabolism involves both transcriptional and post-translational mechanisms. Nuclear receptors exemplify the transcriptional programs that couple molecular clocks to metabolism. The post-translational modifications of the core clock machinery are known to play a key role in metabolic entrainment of circadian clocks. O-linked N-acetylglucosamine modification (O-GlcNAcylation) of intracellular proteins is a key mediator of metabolic response to nutrient availability. This review highlights our current understanding of the role of protein O-GlcNAcylation in mediating metabolic input and output of the circadian clock.
Collapse
|
36
|
Zhang Z, Lai S, Wang Y, Li L, Yin H, Wang Y, Zhao X, Li D, Yang M, Zhu Q. Rhythmic expression of circadian clock genes in the preovulatory ovarian follicles of the laying hen. PLoS One 2017; 12:e0179019. [PMID: 28604799 PMCID: PMC5467841 DOI: 10.1371/journal.pone.0179019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 05/23/2017] [Indexed: 11/18/2022] Open
Abstract
The circadian clock is reported to play a role in the ovaries in a variety of vertebrate species, including the domestic hen. However, the ovary is an organ that changes daily, and the laying hen maintains a strict follicular hierarchy. The aim of this study was to examine the spatial-temporal expression of several known canonical clock genes in the granulosa and theca layers of six hierarchy follicles. We demonstrated that the granulosa cells (GCs) of the F1-F3 follicles harbored intrinsic oscillatory mechanisms in vivo. In addition, cultured granulosa cells (GCs) from F1 follicles exposed to luteinizing hormone (LH) synchronization displayed Per2 mRNA oscillations, whereas, the less mature GCs (F5 plus F6) displayed no circadian change in Per2 mRNA levels. Cultures containing follicle-stimulating hormone (FSH) combined with LH expressed levels of Per2 mRNA that were 2.5-fold higher than those in cultures with LH or FSH alone. These results show that there is spatial specificity in the localization of clock cells in hen preovulatory follicles. In addition, our results support the hypothesis that gonadotropins provide a cue for the development of the functional cellular clock in immature GCs.
Collapse
Affiliation(s)
- Zhichao Zhang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Shuang Lai
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Yagang Wang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Liang Li
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Huadong Yin
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Yan Wang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Xiaoling Zhao
- College of Animal Science and Technology, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Diyan Li
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Mingyao Yang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Qing Zhu
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
- * E-mail:
| |
Collapse
|
37
|
Sun X, Liu X. Cancer metastasis: enactment of the script for human reproductive drama. Cancer Cell Int 2017; 17:51. [PMID: 28469531 PMCID: PMC5414196 DOI: 10.1186/s12935-017-0421-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 04/24/2017] [Indexed: 12/14/2022] Open
Abstract
Based on compelling evidence from many biological disciplines, we put forth a hypothesis for cancer metastasis. In the hypothesis, the metastatic cascade is depicted as human reproduction in miniature. Illustrated in a reproductive light, the staggering resemblance of cancer metastasis to human reproduction becomes evident despite some ostensible dis-similarities. In parallel to the appearance of primordial germ cells during early embryogenesis, the cancer reproductive saga starts with the separation of metastasis initiating cells (MICs) from cancer initiating cells when the primary cancer is still in its infancy. Prime MICs embark on a journey to the host bone marrow where they undergo further development and regulation. Migrating MICs are guided by the same CXCR4/CYCL12 axis as used in the migration of primordial germ cells to the genital ridge. Like the ovary, the host bone marrow features immune privileges, coolness, hypoxia and acidity which are essential for stemness maintenance and regulation. Opportune activation of the MICs via fusion with bone marrow stem cells triggers a frenzy of cellular proliferation and sets them on the move again. This scenario is akin to oocyte fertilization in the Fallopian tube and its subsequent journey towards the decidum. Just as the human reproductive process is plagued with undesirable outcomes so is the cancer metastasis highly inefficient. The climax of the cancer metastatic drama (colonization) is reached when proliferating MIC clusters attempt to settle down on decidum-like premetastatic sites. Successfully colonized clusters blossom into overt macrometastases only after the execution of sophisticated immunomodulation, angiogenesis and vascular remodeling. Similarly, the implanted blastomere needs to orchestrate these feats before flourishing into a new life. What is more, the cancer reproductive drama seems to be directed by a primordial hypothalamus–pituitary–gonad axis. Pursuing this reproductive trail could lead to new frontiers and breakthroughs in cancer research and therapeutics.
Collapse
Affiliation(s)
- Xichun Sun
- Department of Pathology and Laboratory Medicine, McGuire Holmes Veteran Affairs Medical Center, School of Medicine, Virginia Commonwealth University, 1201 Broad Rock Boulevard, Richmond, VA 23249 USA.,Department of Hepatobiliary Surgery, People's Hospital of Hunan Province, Hunan, China
| | - Xiwu Liu
- Department of Pathology and Laboratory Medicine, McGuire Holmes Veteran Affairs Medical Center, School of Medicine, Virginia Commonwealth University, 1201 Broad Rock Boulevard, Richmond, VA 23249 USA.,Department of Hepatobiliary Surgery, People's Hospital of Hunan Province, Hunan, China
| |
Collapse
|
38
|
Papacleovoulou G, Nikolova V, Oduwole O, Chambers J, Vazquez-Lopez M, Jansen E, Nicolaides K, Parker M, Williamson C. Gestational disruptions in metabolic rhythmicity of the liver, muscle, and placenta affect fetal size. FASEB J 2017; 31:1698-1708. [PMID: 28082353 PMCID: PMC5566176 DOI: 10.1096/fj.201601032r] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 01/03/2017] [Indexed: 11/12/2022]
Abstract
Maternal metabolic adaptations are essential for successful pregnancy outcomes. We investigated how metabolic gestational processes are coordinated, whether there is a functional link with internal clocks, and whether disruptions are related to metabolic abnormalities in pregnancy, by studying day/night metabolic pathways in murine models and samples from pregnant women with normally grown and large-for-gestational age infants. In early mouse pregnancy, expression of hepatic lipogenic genes was up-regulated and uncoupled from the hepatic clock. In late mouse pregnancy, rhythmicity of energy metabolism-related genes in the muscle followed the patterns of internal clock genes in this tissue, and coincided with enhanced lipid transporter expression in the fetoplacental unit. Diurnal triglyceride patterns were disrupted in human placentas from pregnancies with large-for-gestational age infants and this overlapped with an increase in BMAL1 expression. Metabolic adaptations in early pregnancy are uncoupled from the circadian clock, whereas in late pregnancy, energy availability is mediated by coordinated muscle-placenta metabolic adjustments linked to internal clocks. Placental triglyceride oscillations in the third trimester of human pregnancy are lost in large-for-gestational age infants and may be regulated by BMAL1. In summary, disruptions in metabolic and circadian rhythmicity are associated with increased fetal size, with implications for the pathogenesis of macrosomia.-Papacleovoulou, G., Nikolova, V., Oduwole, O., Chambers, J., Vazquez-Lopez, M., Jansen, E., Nicolaides, K., Parker, M., Williamson, C. Gestational disruptions in metabolic rhythmicity of the liver, muscle, and placenta affect fetal size.
Collapse
Affiliation(s)
| | - Vanya Nikolova
- Division of Women's Health, Guy's Campus, King's College London, London, United Kingdom
| | - Olayiwola Oduwole
- Institute of Reproductive and Developmental Biology, Surgery and Cancer, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Jenny Chambers
- Women's Health Research Centre, Surgery and Cancer, Faculty of Medicine, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Marta Vazquez-Lopez
- Women's Health Research Centre, Surgery and Cancer, Faculty of Medicine, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Eugene Jansen
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, The Netherlands; and
| | - Kypros Nicolaides
- Harris Birthright Centre for Fetal Medicine, King's College London, London, United Kingdom
| | - Malcolm Parker
- Institute of Reproductive and Developmental Biology, Surgery and Cancer, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Catherine Williamson
- Division of Women's Health, Guy's Campus, King's College London, London, United Kingdom;
- Institute of Reproductive and Developmental Biology, Surgery and Cancer, Hammersmith Hospital, Imperial College London, London, United Kingdom
| |
Collapse
|
39
|
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
|
40
|
The choroid plexus as a sex hormone target: Functional implications. Front Neuroendocrinol 2017; 44:103-121. [PMID: 27998697 DOI: 10.1016/j.yfrne.2016.12.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/25/2016] [Accepted: 12/12/2016] [Indexed: 12/21/2022]
Abstract
The choroid plexuses (CPs) are highly vascularized branched structures that protrude into the ventricles of the brain, and form a unique interface between the blood and the cerebrospinal fluid (CSF). In recent years, novel functions have been attributed to this tissue such as in immune and chemical surveillance of the central nervous system, brain development, adult neurogenesis and circadian rhythm regulation. Sex hormones (SH) are widely recognized as modulators in several neurodegenerative diseases, and there is evidence that estrogens and androgens regulate several fundamental biological functions in the CPs. Therefore, SH are likely to affect the composition of the CSF impacting on brain homeostasis. This review will look at implications of the CPs' sex-related specificities.
Collapse
|
41
|
Pegolo S, Cecchinato A, Mach N, Babbucci M, Pauletto M, Bargelloni L, Schiavon S, Bittante G. Transcriptomic Changes in Liver of Young Bulls Caused by Diets Low in Mineral and Protein Contents and Supplemented with n-3 Fatty Acids and Conjugated Linoleic Acid. PLoS One 2016; 11:e0167747. [PMID: 27930681 PMCID: PMC5145186 DOI: 10.1371/journal.pone.0167747] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 11/19/2016] [Indexed: 02/04/2023] Open
Abstract
The aim of the present study was to identify transcriptional modifications and regulatory networks accounting for physiological and metabolic responses to specific nutrients in the liver of young Belgian Blue × Holstein bulls using RNA-sequencing. A larger trial has been carried out in which animals were fed with different diets: 1] a conventional diet; 2] a low-protein/low-mineral diet (low-impact diet) and 3] a diet enriched in n-3 fatty acids (FAs), conjugated linoleic acid (CLA) and vitamin E (nutraceutical diet). The initial hypothesis was that the administration of low-impact and nutraceutical diets might influence the transcriptional profiles in bovine liver and the resultant nutrient fluxes, which are essential for optimal liver function and nutrient interconversion. Results showed that the nutraceutical diet significantly reduced subcutaneous fat covering in vivo and liver pH. Dietary treatments did not affect overall liver fat content, but significantly modified the liver profile of 33 FA traits (out of the total 89 identified by gas-chromatography). In bulls fed nutraceutical diet, the percentage of n-3 and CLA FAs increased around 2.5-fold compared with the other diets, whereas the ratio of n6/n3 decreased 2.5-fold. Liver transcriptomic analyses revealed a total of 198 differentially expressed genes (DEGs) when comparing low-impact, nutraceutical and conventional diets, with the nutraceutical diet showing the greatest effects on liver transcriptome. Functional analyses using ClueGo and Ingenuity Pathway Analysis evidenced that DEGs in bovine liver were variously involved in energy reserve metabolic process, glutathione metabolism, and carbohydrate and lipid metabolism. Modifications in feeding strategies affected key transcription factors regulating the expression of several genes involved in fatty acid metabolism, e.g. insulin-induced gene 1, insulin receptor substrate 2, and RAR-related orphan receptor C. This study provides noteworthy insights into the molecular changes occurring as a result of nutrient variation in diets (aimed at reducing the environmental impact and improving human health) and broadens our understanding of the relationship between nutrients variation and phenotypic effects.
Collapse
Affiliation(s)
- Sara Pegolo
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, Legnaro, Padova, Italy
- * E-mail:
| | - Alessio Cecchinato
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, Legnaro, Padova, Italy
| | - Núria Mach
- Animal Genetics and Integrative Biology unit (GABI), INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Massimiliano Babbucci
- Department of Comparative Biomedicine and Food Science, University of Padova, Legnaro, Padova, Italy
| | - Marianna Pauletto
- Department of Comparative Biomedicine and Food Science, University of Padova, Legnaro, Padova, Italy
| | - Luca Bargelloni
- Department of Comparative Biomedicine and Food Science, University of Padova, Legnaro, Padova, Italy
| | - Stefano Schiavon
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, Legnaro, Padova, Italy
| | - Giovanni Bittante
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, Legnaro, Padova, Italy
| |
Collapse
|
42
|
Nagy Z, Marta A, Butz H, Liko I, Racz K, Patocs A. Modulation of the circadian clock by glucocorticoid receptor isoforms in the H295R cell line. Steroids 2016; 116:20-27. [PMID: 27725099 DOI: 10.1016/j.steroids.2016.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 10/03/2016] [Accepted: 10/04/2016] [Indexed: 11/25/2022]
Abstract
Peripheral clocks are set by different nervous, hormonal and metabolic stimuli, and regulate the circadian expression of several genes. We investigated whether a peripheral clock could be induced in the human adrenocortical cell line H295R and whether glucocorticoid receptor isoforms (GRα and GRß) are involved in this clock system. After synchronization of cells with serum shock, the rhythmic oscillation of clock genes PER1, PER2, REV-ERBα, and ARNTL was confirmed. In addition, H295R cells even without serum shock showed rhythmic expression of PER1, PER2, CRY1 and ARNTL. Glucocorticoid treatment induced a rapid response of PER1, PER2 and CRY1 in a GRα-dependent manner. Continuous glucocorticoid stimulation after 6h caused suppression of REV-ERBα. Administration of a GR antagonist, RU486, disrupted the circadian oscillation of clock genes and prevented the acute changes in PER1, PER2 and CRY1 levels. Overexpression of the GRß isoform alone did not alter the expression of the examined clock genes, but did prevent the GRα-related suppression of REV-ERBα. These alterations occurred independently from ACTH and CRH. Our data demonstrate that a peripheral clock system is present in a human adrenocortical cell line and that periodic oscillations of clock genes are influenced by glucocorticoids, mainly through GRα.
Collapse
Affiliation(s)
- Zsolt Nagy
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University "Lendulet" Hereditary Endocrine Tumors Research Group, Budapest, Hungary
| | - Alexa Marta
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Henriett Butz
- Hungarian Academy of Sciences-Semmelweis University Molecular Medicine Research Group, Budapest, Hungary
| | - Istvan Liko
- Hungarian Academy of Sciences-Semmelweis University "Lendulet" Hereditary Endocrine Tumors Research Group, Budapest, Hungary
| | - Karoly Racz
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University Molecular Medicine Research Group, Budapest, Hungary
| | - Attila Patocs
- Hungarian Academy of Sciences-Semmelweis University "Lendulet" Hereditary Endocrine Tumors Research Group, Budapest, Hungary; Department of Laboratory Medicine Institute, Faculty of Medicine, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
43
|
Wharfe MD, Wyrwoll CS, Waddell BJ, Mark PJ. Pregnancy-induced changes in the circadian expression of hepatic clock genes: implications for maternal glucose homeostasis. Am J Physiol Endocrinol Metab 2016; 311:E575-86. [PMID: 27406739 DOI: 10.1152/ajpendo.00060.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/06/2016] [Indexed: 01/26/2023]
Abstract
Adaptations in maternal carbohydrate metabolism are particularly important in pregnancy because glucose is the principal energy substrate used by the fetus. As metabolic homeostasis is intricately linked to the circadian system via the rhythmic expression of clock genes, it is likely that metabolic adaptations during pregnancy also involve shifts in maternal circadian function. We hypothesized that maternal adaptation in pregnancy involves changes in the hepatic expression of clock genes, which drive downstream shifts in circadian expression of glucoregulatory genes. Maternal liver and plasma (n = 6-8/group) were collected across 24-h periods (0800, 1200, 1600, 2000, 0000, 0400) from C57Bl/6J mice under isoflurane-nitrous oxide anesthesia prior to and on days 6, 10, 14 and 18 of pregnancy (term = day 19). Hepatic expression of clock genes and glucoregulatory genes was determined by RT-qPCR. Hepatic clock gene expression was substantially altered across pregnancy, most notably in late gestation when the circadian rhythmicity of several clock genes was attenuated (≤64% reduced amplitude on day 18). These changes were associated with a similar decline in rhythmicity of the key glucoregulatory genes Pck1, G6Pase, and Gk, and by day 18, Pck1 was no longer rhythmic. Overall, our data show marked adaptations in the liver clock during mouse pregnancy, changes that may contribute to the altered circadian variation in glucoregulatory genes near term. We propose that the observed reduction of daily oscillations in glucose metabolism ensure a sustained supply of glucose to meet the high demands of fetal growth.
Collapse
Affiliation(s)
- Michaela D Wharfe
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, Australia
| | - Caitlin S Wyrwoll
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, Australia
| | - Brendan J Waddell
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, Australia
| | - Peter J Mark
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, Australia
| |
Collapse
|
44
|
Sen A, Sellix MT. The Circadian Timing System and Environmental Circadian Disruption: From Follicles to Fertility. Endocrinology 2016; 157:3366-73. [PMID: 27501186 DOI: 10.1210/en.2016-1450] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The internal or circadian timing system is deeply integrated in female reproductive physiology. Considerable details of rheostatic timing function in the neuroendocrine control of pituitary hormone secretion, adenohypophyseal hormone gene expression and secretion, gonadal steroid hormone biosynthesis and secretion, ovulation, implantation, and parturition have been reported. The molecular clock, an autonomous feedback loop oscillator of interacting transcriptional regulators, dictates the timing and amplitude of gene expression in each tissue of the female hypothalamic-pituitary-gonadal (HPG) axis. Although multiple targets of the molecular clock have been identified, many associated with critical physiological functions in the HPG axis, the full extent of clock-driven gene expression and physiology in this critical system remains unknown. Environmental circadian disruption (ECD), the disturbance of temporal relationships within and between internal clocks (brain and periphery), and external timing cues (eg, light, nutrients, social cues) due to rotating/night shift work or transmeridian travel have been linked to reproductive dysfunction and subfertility. Moreover, ECD resulting from exposure to endocrine disrupting chemicals, environmental toxins, and/or irregular hormone levels during sexual development can also reduce fertility. Thus, perturbations that disturb clock function at the molecular, cellular or systemic level correlate with significant declines in female reproductive function. Here we briefly review the evidence for molecular clock function in each tissue of the female HPG axis (GnRH neuron, pituitary, uterus, oviduct, and ovary), describe the human epidemiological and animal data supporting the negative effects of ECD on fertility, and explore the potential for novel chronotherapeutics in women's health and fertility.
Collapse
Affiliation(s)
- Aritro Sen
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester New York 14642
| | - Michael T Sellix
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester New York 14642
| |
Collapse
|
45
|
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
|
46
|
Wharfe MD, Wyrwoll CS, Waddell BJ, Mark PJ. Pregnancy Suppresses the Daily Rhythmicity of Core Body Temperature and Adipose Metabolic Gene Expression in the Mouse. Endocrinology 2016; 157:3320-31. [PMID: 27409644 DOI: 10.1210/en.2016-1177] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Maternal adaptations in lipid metabolism are crucial for pregnancy success due to the role of white adipose tissue as an energy store and the dynamic nature of energy needs across gestation. Because lipid metabolism is regulated by the rhythmic expression of clock genes, it was hypothesized that maternal metabolic adaptations involve changes in both adipose clock gene expression and the rhythmic expression of downstream metabolic genes. Maternal core body temperature (Tc) was investigated as a possible mechanism driving pregnancy-induced changes in clock gene expression. Gonadal adipose tissue and plasma were collected from C57BL/6J mice before and on days 6, 10, 14, and 18 of pregnancy (term 19 d) at 4-hour intervals across a 24-hour period. Adipose expression of clock genes and downstream metabolic genes were determined by quantitative RT-PCR, and Tc was measured by intraperitoneal temperature loggers. Adipose clock gene expression showed robust rhythmicity throughout pregnancy, but absolute levels varied substantially across gestation. Rhythmic expression of the metabolic genes Lipe, Pnpla2, and Lpl was clearly evident before pregnancy; however, this rhythmicity was lost with the onset of pregnancy. Tc rhythm was significantly altered by pregnancy, with a 65% decrease in amplitude by term and a 0.61°C decrease in mesor between days 6 and 18. These changes in Tc, however, did not appear to be linked to adipose clock gene expression across pregnancy. Overall, our data show marked adaptations in the adipose clock in pregnancy, with an apparent decoupling of adipose clock and lipolytic/lipogenic gene rhythms from early in gestation.
Collapse
Affiliation(s)
- Michaela D Wharfe
- School of Anatomy, Physiology, and Human Biology, The University of Western Australia, Perth 6009, Australia
| | - Caitlin S Wyrwoll
- School of Anatomy, Physiology, and Human Biology, The University of Western Australia, Perth 6009, Australia
| | - Brendan J Waddell
- School of Anatomy, Physiology, and Human Biology, The University of Western Australia, Perth 6009, Australia
| | - Peter J Mark
- School of Anatomy, Physiology, and Human Biology, The University of Western Australia, Perth 6009, Australia
| |
Collapse
|
47
|
Bayerl DS, Kaczmarek V, Jurek B, van den Burg EH, Neumann ID, Gaßner BM, Klampfl SM, Bosch OJ. Antagonism of V1b receptors promotes maternal motivation to retrieve pups in the MPOA and impairs pup-directed behavior during maternal defense in the mpBNST of lactating rats. Horm Behav 2016; 79:18-27. [PMID: 26747375 DOI: 10.1016/j.yhbeh.2015.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 12/26/2015] [Accepted: 12/29/2015] [Indexed: 01/30/2023]
Abstract
Recent studies using V1b receptor (V1bR) knockout mice or central pharmacological manipulations in lactating rats highlighted the influence of this receptor for maternal behavior. However, its role in specific brain sites known to be important for maternal behavior has not been investigated to date. In the present study, we reveal that V1bR mRNA (qPCR) and protein levels (Western blot) within either the medial preoptic area (MPOA) or the medial-posterior part of the bed nucleus of the stria terminalis (mpBNST) did not differ between virgin and lactating rats. Furthermore, we characterized the effects of V1bR blockade via bilateral injections of the receptor subtype-specific antagonist SSR149415 within the MPOA or the mpBNST on maternal behavior (maternal care under non-stress and stress conditions, maternal motivation to retrieve pups in a novel environment, maternal aggression) and anxiety-related behavior in lactating rats. Blocking V1bR within the MPOA increased pup retrieval, whereas within the mpBNST it decreased pup-directed behavior, specifically licking/grooming the pups, during the maternal defense test. In addition, immediately after termination of the maternal defense test, V1bR antagonism in both brain regions reduced nursing, particularly arched back nursing. Anxiety-related behavior was not affected by V1bR antagonism in either brain region. In conclusion our data indicate that V1bR antagonism significantly modulates different aspects of maternal behavior in a brain region-dependent manner.
Collapse
Affiliation(s)
- Doris S Bayerl
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, 93053 Regensburg, Germany.
| | - Veronika Kaczmarek
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, 93053 Regensburg, Germany.
| | - Benjamin Jurek
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, 93053 Regensburg, Germany.
| | - Erwin H van den Burg
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, 93053 Regensburg, Germany.
| | - Inga D Neumann
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, 93053 Regensburg, Germany.
| | - Barbara M Gaßner
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, 93053 Regensburg, Germany.
| | - Stefanie M Klampfl
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, 93053 Regensburg, Germany.
| | - Oliver J Bosch
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, 93053 Regensburg, Germany.
| |
Collapse
|
48
|
Wharfe MD, Mark PJ, Wyrwoll CS, Smith JT, Yap C, Clarke MW, Waddell BJ. Pregnancy-induced adaptations of the central circadian clock and maternal glucocorticoids. J Endocrinol 2016; 228:135-47. [PMID: 26883207 DOI: 10.1530/joe-15-0405] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/22/2015] [Indexed: 01/21/2023]
Abstract
Maternal physiological adaptations, such as changes to the hypothalamic-pituitary-adrenal (HPA) axis, are central to pregnancy success. Circadian variation of the HPA axis is dependent on clock gene rhythms in the hypothalamus, but it is not known whether pregnancy-induced changes in maternal glucocorticoid levels are mediated via this central clock. We hypothesized that hypothalamic expression of clock genes changes across mouse pregnancy and this is linked to altered HPA activity. The anterior hypothalamus and maternal plasma were collected from C57Bl/6J mice prior to pregnancy and on days 6, 10, 14 and 18 of gestation (term=d19), across a 24-h period (0800, 1200, 1600, 2000, 0000, 0400 h). Hypothalamic expression of clock genes and Crh was determined by qPCR, plasma ACTH concentration measured by Milliplex assay and plasma corticosterone concentration by LC-MS/MS. Expression of all clock genes varied markedly across gestation, most notably at mid-gestation when levels of each gene were elevated. The pregnancy-induced increase in maternal corticosterone levels (by up to 14-fold on day 14) was not accompanied by a parallel shift in plasma ACTH (28% lower on day 14 compared with non-pregnant levels). Moreover, while circadian rhythmicity in corticosterone was maintained up to day 14 of gestation, this was effectively lost by day 18. Overall, our data show that the central circadian clock undergoes marked adaptations throughout mouse pregnancy, changes that are likely to contribute to maternal physiological adaptations. Importantly, however, neither hypothalamic clock genes nor plasma ACTH levels appear to drive the marked increase in maternal corticosterone after mid-gestation.
Collapse
Affiliation(s)
- Michaela D Wharfe
- School of AnatomyPhysiology and Human Biology, The University of Western Australia, M309, Perth 6009, AustraliaMetabolomics AustraliaThe University of Western Australia, Perth 6009, Australia
| | - Peter J Mark
- School of AnatomyPhysiology and Human Biology, The University of Western Australia, M309, Perth 6009, AustraliaMetabolomics AustraliaThe University of Western Australia, Perth 6009, Australia
| | - Caitlin S Wyrwoll
- School of AnatomyPhysiology and Human Biology, The University of Western Australia, M309, Perth 6009, AustraliaMetabolomics AustraliaThe University of Western Australia, Perth 6009, Australia
| | - Jeremy T Smith
- School of AnatomyPhysiology and Human Biology, The University of Western Australia, M309, Perth 6009, AustraliaMetabolomics AustraliaThe University of Western Australia, Perth 6009, Australia
| | - Cassandra Yap
- School of AnatomyPhysiology and Human Biology, The University of Western Australia, M309, Perth 6009, AustraliaMetabolomics AustraliaThe University of Western Australia, Perth 6009, Australia
| | - Michael W Clarke
- School of AnatomyPhysiology and Human Biology, The University of Western Australia, M309, Perth 6009, AustraliaMetabolomics AustraliaThe University of Western Australia, Perth 6009, Australia
| | - Brendan J Waddell
- School of AnatomyPhysiology and Human Biology, The University of Western Australia, M309, Perth 6009, AustraliaMetabolomics AustraliaThe University of Western Australia, Perth 6009, Australia
| |
Collapse
|
49
|
Qiu C, Gelaye B, Denis M, Tadesse MG, Luque Fernandez MA, Enquobahrie DA, Ananth CV, Sanchez SE, Williams MA. Circadian clock-related genetic risk scores and risk of placental abruption. Placenta 2015; 36:1480-6. [PMID: 26515929 PMCID: PMC5010362 DOI: 10.1016/j.placenta.2015.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 10/06/2015] [Accepted: 10/11/2015] [Indexed: 10/22/2022]
Abstract
INTRODUCTION The circadian clock plays an important role in several aspects of female reproductive biology. Evidence linking circadian clock-related genes to pregnancy outcomes has been inconsistent. We sought to examine whether variations in single nucleotide polymorphisms (SNPs) of circadian clock genes are associated with PA risk. METHODS Maternal blood samples were collected from 470 PA case and 473 controls. Genotyping was performed using the Illumina Cardio-MetaboChip platform. We examined 119 SNPs in 13 candidate genes known to control circadian rhythms (e.g., CRY2, ARNTL, and RORA). Univariate and penalized logistic regression models were fit to estimate odds ratios (ORs); and the combined effect of multiple SNPs on PA risk was estimated using a weighted genetic risk score (wGRS). RESULTS A common SNP in the RORA gene (rs2899663) was associated with a 21% reduced odds of PA (P < 0.05). The odds of PA increased with increasing wGRS (Ptrend < 0.001). The corresponding ORs were 1.00, 1.83, 2.81 and 5.13 across wGRS quartiles. Participants in the highest wGRS quartile had a 5.13-fold (95% confidence interval: 3.21-8.21) higher odds of PA compared to those in the lowest quartile. Although the test for interaction was not significant, the odds of PA was substantially elevated for preeclamptics with the highest wGRS quartile (OR = 14.44, 95%CI: 6.62-31.53) compared to normotensive women in the lowest wGRS quartile. DISCUSSION Genetic variants in circadian rhythm genes may be associated with PA risk. Larger studies are needed to corroborate these findings and to further elucidate the pathogenesis of this important obstetrical complication.
Collapse
Affiliation(s)
- Chunfang Qiu
- Center for Perinatal Studies, Swedish Medical Center, Seattle, WA, USA.
| | - Bizu Gelaye
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Marie Denis
- UMR Amélioration Génétique et Adaptation des Plantes méditerranéennes et tropicales (AGAP), CIRAD, Montpellier, France
| | - Mahlet G Tadesse
- Department of Mathematics and Statistics, Georgetown University, Washington, DC, USA
| | | | - Daniel A Enquobahrie
- Center for Perinatal Studies, Swedish Medical Center, Seattle, WA, USA; Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - Cande V Ananth
- Department of Obstetrics and Gynecology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA; Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Sixto E Sanchez
- Sección de Post Grado, Facultad de Medicina Humana, Universidad San Martín de Porres, Lima, Peru; A.C. PROESA, Lima, Peru; Department of Obstetrics and Gynecology, San Marcos University, Lima, Peru
| | - Michelle A Williams
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
50
|
Chun LE, Woodruff ER, Morton S, Hinds LR, Spencer RL. Variations in Phase and Amplitude of Rhythmic Clock Gene Expression across Prefrontal Cortex, Hippocampus, Amygdala, and Hypothalamic Paraventricular and Suprachiasmatic Nuclei of Male and Female Rats. J Biol Rhythms 2015; 30:417-36. [PMID: 26271538 DOI: 10.1177/0748730415598608] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The molecular circadian clock is a self-regulating transcription/translation cycle of positive (Bmal1, Clock/Npas2) and negative (Per1,2,3, Cry1,2) regulatory components. While the molecular clock has been well characterized in the body's master circadian pacemaker, the hypothalamic suprachiasmatic nucleus (SCN), only a few studies have examined both the positive and negative clock components in extra-SCN brain tissue. Furthermore, there has yet to be a direct comparison of male and female clock gene expression in the brain. This comparison is warranted, as there are sex differences in circadian functioning and disorders associated with disrupted clock gene expression. This study examined basal clock gene expression (Per1, Per2, Bmal1 mRNA) in the SCN, prefrontal cortex (PFC), rostral agranular insula, hypothalamic paraventricular nucleus (PVN), amygdala, and hippocampus of male and female rats at 4-h intervals throughout a 12:12 h light:dark cycle. There was a significant rhythm of Per1, Per2, and Bmal1 in the SCN, PFC, insula, PVN, subregions of the hippocampus, and amygdala with a 24-h period, suggesting the importance of an oscillating molecular clock in extra-SCN brain regions. There were 3 distinct clock gene expression profiles across the brain regions, indicative of diversity among brain clocks. Although, generally, the clock gene expression profiles were similar between male and female rats, there were some sex differences in the robustness of clock gene expression (e.g., females had fewer robust rhythms in the medial PFC, more robust rhythms in the hippocampus, and a greater mesor in the medial amygdala). Furthermore, females with a regular estrous cycle had attenuated aggregate rhythms in clock gene expression in the PFC compared with noncycling females. This suggests that gonadal hormones may modulate the expression of the molecular clock.
Collapse
Affiliation(s)
- Lauren E Chun
- Department of Psychology & Neuroscience, University of Colorado Boulder, USA
| | | | - Sarah Morton
- Department of Psychology & Neuroscience, University of Colorado Boulder, USA
| | - Laura R Hinds
- Department of Psychology & Neuroscience, University of Colorado Boulder, USA
| | - Robert L Spencer
- Department of Psychology & Neuroscience, University of Colorado Boulder, USA
| |
Collapse
|