24
views
0
recommends
+1 Recommend
1 collections
    0
    shares

         An official journal of the Society for Reproduction and Fertility. Learn more

      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Core clock gene Bmal1 deprivation impairs steroidogenesis in mice luteinized follicle cells

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Luteinization is the event of corpus luteum formation, a way of follicle cells transformation and a process of steroidogenesis alteration. As the core clock gene, Bmal1 was involved in the regulation of ovulation process and luteal function afterwards. Till now, the underlying roles of luteinization played by Bmal1 remain unknown. To explore the unique role of Bmal1 in luteal steroidogenesis and its underlying pathway, we investigated the luteal hormone synthesis profile in Bmal1 knockout female mice. We found that luteal hormone synthesis was notably impaired, and phosphorylation of PI3K/NfκB pathway was significantly activated. Then, the results were verified in in vitro cultured cells, including isolated Bmal1 interference granulosa cells (GCs) and theca cells (TCs), respectively. Hormones levels of supernatant culture media and mRNA expressions of steroidogenesis-associated genes ( star, Hsd3β2, cyp19a1 in GCs, Lhcgr, star, Hsd3β2, cyp17a1 in TCs) were mutually decreased, while the phosphorylation of PI3K/NfκB was promoted during in vitro luteinization. After PI3K specific-inhibitor LY294002 intervention, mRNA expressions of Lhcgr and Hsd3β2 were partially rescued in Bmal1 interference TCs, together with significantly increased androstenedione and T synthesis. Further exploration in TCs demonstrated BMAL1 interacted directly but negatively with NfκB p65 (RelA), a subunit which was supposed as a mediator in Bmal1-governed PI3K signaling regulation. Taken together, we verified the novel role of Bmal1 in luteal steroidogenesis, achieving by negative interplay with RelA-mediated PI3K/NfκB pathway.

          Related collections

          Most cited references47

          • Record: found
          • Abstract: found
          • Article: not found

          Coordinated transcription of key pathways in the mouse by the circadian clock.

          In mammals, circadian control of physiology and behavior is driven by a master pacemaker located in the suprachiasmatic nuclei (SCN) of the hypothalamus. We have used gene expression profiling to identify cycling transcripts in the SCN and in the liver. Our analysis revealed approximately 650 cycling transcripts and showed that the majority of these were specific to either the SCN or the liver. Genetic and genomic analysis suggests that a relatively small number of output genes are directly regulated by core oscillator components. Major processes regulated by the SCN and liver were found to be under circadian regulation. Importantly, rate-limiting steps in these various pathways were key sites of circadian control, highlighting the fundamental role that circadian clocks play in cellular and organismal physiology.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Transcriptional architecture of the mammalian circadian clock

            Next-generation sequencing approaches have yielded new insights into circadian function. Here, Takahashi reviews genome-wide analyses of the clock transcriptional feedback loop in mammals, which reveal a global circadian regulation of transcription factor occupancy, RNA polymerase II recruitment and initiation, nascent transcription and chromatin remodelling.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              A circadian gene expression atlas in mammals: implications for biology and medicine.

              To characterize the role of the circadian clock in mouse physiology and behavior, we used RNA-seq and DNA arrays to quantify the transcriptomes of 12 mouse organs over time. We found 43% of all protein coding genes showed circadian rhythms in transcription somewhere in the body, largely in an organ-specific manner. In most organs, we noticed the expression of many oscillating genes peaked during transcriptional "rush hours" preceding dawn and dusk. Looking at the genomic landscape of rhythmic genes, we saw that they clustered together, were longer, and had more spliceforms than nonoscillating genes. Systems-level analysis revealed intricate rhythmic orchestration of gene pathways throughout the body. We also found oscillations in the expression of more than 1,000 known and novel noncoding RNAs (ncRNAs). Supporting their potential role in mediating clock function, ncRNAs conserved between mouse and human showed rhythmic expression in similar proportions as protein coding genes. Importantly, we also found that the majority of best-selling drugs and World Health Organization essential medicines directly target the products of rhythmic genes. Many of these drugs have short half-lives and may benefit from timed dosage. In sum, this study highlights critical, systemic, and surprising roles of the mammalian circadian clock and provides a blueprint for advancement in chronotherapy.
                Bookmark

                Author and article information

                Journal
                Reproduction
                Reproduction
                REP
                Reproduction (Cambridge, England)
                Bioscientifica Ltd (Bristol )
                1470-1626
                1741-7899
                December 2020
                17 September 2020
                : 160
                : 6
                : 955-967
                Affiliations
                [1 ]Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University , Guangdong, Guangzhou, China
                [2 ]Reproductive Medicine Center , Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
                [3 ]Guangdong Provincial Key Laboratory of Reproductive Medicine , First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
                Author notes
                Correspondence should be addressed to Y Xu; Email: xuyanwen663000@ 123456126.com
                Author information
                http://orcid.org/0000-0001-7313-1608
                http://orcid.org/0000-0001-7313-1608
                Article
                REP-20-0340
                10.1530/REP-20-0340
                7707808
                33112769
                b4a9cfb9-bfba-4e1c-989f-771ef7e505a8
                © 2020 The authors

                This work is licensed under a Creative Commons Attribution 4.0 International License.

                History
                : 16 June 2020
                : 17 September 2020
                Categories
                Research

                Obstetrics & Gynecology
                Obstetrics & Gynecology

                Comments

                Comment on this article