1
|
Meneau F, Lapébie P, Daldello EM, Le T, Chevalier S, Assaf S, Houliston E, Jessus C, Miot M. ARPP19 phosphorylation site evolution and the switch in cAMP control of oocyte maturation in vertebrates. Development 2024; 151:dev202655. [PMID: 39576213 DOI: 10.1242/dev.202655] [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: 01/12/2024] [Accepted: 10/28/2024] [Indexed: 12/06/2024]
Abstract
cAMP-PKA signaling initiates the crucial process of oocyte meiotic maturation in many animals, but inhibits it in vertebrates. To address this 'cAMP paradox', we exchanged the key PKA substrate ARPP19 between representative species, the vertebrate Xenopus and the cnidarian Clytia, comparing its phosphorylation and function. We found that, as in Xenopus, Clytia maturing oocytes undergo ARPP19 phosphorylation on a highly conserved Gwl site, which inhibits PP2A and promotes M-phase entry. In contrast, despite a PKA phosphorylation signature motif recognizable across most animals, Clytia ARPP19 was only poorly phosphorylated by PKA in vitro and in vivo. Furthermore, unlike Xenopus ARPP19, exogenous Clytia ARPP19 did not delay Xenopus oocyte maturation. We conclude that, in Clytia, ARPP19 does not intervene in oocyte maturation initiation because of both poor recognition by PKA and the absence of effectors that mediate vertebrate oocyte prophase arrest. We propose that ancestral ARPP19 phosphorylated by Gwl has retained a key role in M-phase across eukaryotes and has acquired new functions during animal evolution mediated by enhanced PKA phosphorylation, allowing co-option into oocyte maturation regulation in the vertebrate lineage.
Collapse
Affiliation(s)
- Ferdinand Meneau
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine, LBD - IBPS, F-75005 Paris, France
| | - Pascal Lapébie
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-mer (LBDV), F-06230 Villefranche-sur-mer, France
| | - Enrico Maria Daldello
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine, LBD - IBPS, F-75005 Paris, France
| | - Tran Le
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine, LBD - IBPS, F-75005 Paris, France
| | - Sandra Chevalier
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-mer (LBDV), F-06230 Villefranche-sur-mer, France
| | - Sarah Assaf
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-mer (LBDV), F-06230 Villefranche-sur-mer, France
| | - Evelyn Houliston
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-mer (LBDV), F-06230 Villefranche-sur-mer, France
| | - Catherine Jessus
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine, LBD - IBPS, F-75005 Paris, France
| | - Marika Miot
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine, LBD - IBPS, F-75005 Paris, France
| |
Collapse
|
2
|
Santoni M, Meneau F, Sekhsoukh N, Castella S, Le T, Miot M, Daldello EM. Unraveling the interplay between PKA inhibition and Cdk1 activation during oocyte meiotic maturation. Cell Rep 2024; 43:113782. [PMID: 38358892 DOI: 10.1016/j.celrep.2024.113782] [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: 09/03/2023] [Revised: 12/19/2023] [Accepted: 01/25/2024] [Indexed: 02/17/2024] Open
Abstract
Oocytes are arrested in prophase I. In vertebrates, meiotic resumption is triggered by hormonal stimulation that results in cAMP-dependent protein kinase (PKA) downregulation leading to Cdk1 activation. Yet the pathways connecting PKA to Cdk1 remain unclear. Here, we identify molecular events triggered by PKA downregulation occurring upstream of Cdk1 activation. We describe a two-step regulation controlling cyclin B1 and Mos accumulation, which depends on both translation and stabilization. Cyclin B1 accumulation is triggered by PKA inhibition upstream of Cdk1 activation, while its translation requires Cdk1 activity. Conversely, Mos translation initiates in response to the hormone, but the protein accumulates only downstream of Cdk1. Furthermore, two successive translation waves take place, the first controlled by PKA inhibition and the second by Cdk1 activation. Notably, Arpp19, an essential PKA effector, does not regulate the early PKA-dependent events. This study elucidates how PKA downregulation orchestrates multiple pathways that converge toward Cdk1 activation and induce the oocyte G2/M transition.
Collapse
Affiliation(s)
- Martina Santoni
- Sorbonne Université-CNRS, Laboratoire de Biologie du Développement Institut de Biologie Paris Seine, LBD-IBPS, 75005 Paris, France
| | - Ferdinand Meneau
- Sorbonne Université-CNRS, Laboratoire de Biologie du Développement Institut de Biologie Paris Seine, LBD-IBPS, 75005 Paris, France
| | - Nabil Sekhsoukh
- Sorbonne Université-CNRS, Laboratoire de Biologie du Développement Institut de Biologie Paris Seine, LBD-IBPS, 75005 Paris, France
| | - Sandrine Castella
- Sorbonne Université-CNRS, Laboratoire de Biologie du Développement Institut de Biologie Paris Seine, LBD-IBPS, 75005 Paris, France
| | - Tran Le
- Sorbonne Université-CNRS, Laboratoire de Biologie du Développement Institut de Biologie Paris Seine, LBD-IBPS, 75005 Paris, France
| | - Marika Miot
- Sorbonne Université-CNRS, Laboratoire de Biologie du Développement Institut de Biologie Paris Seine, LBD-IBPS, 75005 Paris, France
| | - Enrico Maria Daldello
- Sorbonne Université-CNRS, Laboratoire de Biologie du Développement Institut de Biologie Paris Seine, LBD-IBPS, 75005 Paris, France.
| |
Collapse
|
3
|
Das D, Arur S. Regulation of oocyte maturation: Role of conserved ERK signaling. Mol Reprod Dev 2022; 89:353-374. [PMID: 35908193 PMCID: PMC9492652 DOI: 10.1002/mrd.23637] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/12/2022] [Accepted: 07/18/2022] [Indexed: 11/11/2022]
Abstract
During oogenesis, oocytes arrest at meiotic prophase I to acquire competencies for resuming meiosis, fertilization, and early embryonic development. Following this arrested period, oocytes resume meiosis in response to species-specific hormones, a process known as oocyte maturation, that precedes ovulation and fertilization. Involvement of endocrine and autocrine/paracrine factors and signaling events during maintenance of prophase I arrest, and resumption of meiosis is an area of active research. Studies in vertebrate and invertebrate model organisms have delineated the molecular determinants and signaling pathways that regulate oocyte maturation. Cell cycle regulators, such as cyclin-dependent kinase (CDK1), polo-like kinase (PLK1), Wee1/Myt1 kinase, and the phosphatase CDC25 play conserved roles during meiotic resumption. Extracellular signal-regulated kinase (ERK), on the other hand, while activated during oocyte maturation in all species, regulates both species-specific, as well as conserved events among different organisms. In this review, we synthesize the general signaling mechanisms and focus on conserved and distinct functions of ERK signaling pathway during oocyte maturation in mammals, non-mammalian vertebrates, and invertebrates such as Drosophila and Caenorhabditis elegans.
Collapse
Affiliation(s)
- Debabrata Das
- Department of Genetics, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Swathi Arur
- Department of Genetics, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
4
|
Muthaiyan Shanmugam M, Manoj H. Microinjection for Single-Cell Analysis and Therapy. HANDBOOK OF SINGLE-CELL TECHNOLOGIES 2022:81-107. [DOI: 10.1007/978-981-10-8953-4_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
5
|
Lemonnier T, Daldello EM, Poulhe R, Le T, Miot M, Lignières L, Jessus C, Dupré A. The M-phase regulatory phosphatase PP2A-B55δ opposes protein kinase A on Arpp19 to initiate meiotic division. Nat Commun 2021; 12:1837. [PMID: 33758202 PMCID: PMC7988065 DOI: 10.1038/s41467-021-22124-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 02/25/2021] [Indexed: 02/06/2023] Open
Abstract
Oocytes are held in meiotic prophase for prolonged periods until hormonal signals trigger meiotic divisions. Key players of M-phase entry are the opposing Cdk1 kinase and PP2A-B55δ phosphatase. In Xenopus, the protein Arpp19, phosphorylated at serine 67 by Greatwall, plays an essential role in inhibiting PP2A-B55δ, promoting Cdk1 activation. Furthermore, Arpp19 has an earlier role in maintaining the prophase arrest through a second serine (S109) phosphorylated by PKA. Prophase release, induced by progesterone, relies on Arpp19 dephosphorylation at S109, owing to an unknown phosphatase. Here, we identified this phosphatase as PP2A-B55δ. In prophase, PKA and PP2A-B55δ are simultaneously active, suggesting the presence of other important targets for both enzymes. The drop in PKA activity induced by progesterone enables PP2A-B55δ to dephosphorylate S109, unlocking the prophase block. Hence, PP2A-B55δ acts critically on Arpp19 on two distinct sites, opposing PKA and Greatwall to orchestrate the prophase release and M-phase entry.
Collapse
Affiliation(s)
- Tom Lemonnier
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Paris, France
| | - Enrico Maria Daldello
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Paris, France
| | - Robert Poulhe
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Paris, France
| | - Tran Le
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Paris, France
| | - Marika Miot
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Paris, France
| | | | - Catherine Jessus
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Paris, France
| | - Aude Dupré
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Paris, France.
| |
Collapse
|
6
|
O'Shea LC, Fair T, Hensey C. X-linked α-thalassemia with mental retardation is downstream of protein kinase A in the meiotic cell cycle signaling cascade in Xenopus oocytes and is dynamically regulated in response to DNA damage†. Biol Reprod 2020; 100:1238-1249. [PMID: 30649195 DOI: 10.1093/biolre/ioz001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 10/19/2018] [Accepted: 01/08/2019] [Indexed: 12/19/2022] Open
Abstract
X-linked α-thalassemia with mental retardation (ATRX) is a chromatin remodeling protein that belongs to the SWItch/sucrose non-fermentable (SWI2/SNF2) family of helicase/ATPases. During meiosis, ATRX is necessary for heterochromatin formation and maintenance of chromosome stability in order to ensure proper assembly of the metaphase II spindle. Previously, we established ATRX as a novel progesterone regulated protein during bovine meiotic maturation, in addition to being dynamically regulated in response to DNA damage in oocytes. In the present study, we utilize the Xenopus laevis model system to further elucidate the signaling pathways regulating ATRX expression within the oocyte. Here, we present an analysis of endogenous ATRX protein expression during oogenesis, oocyte meiotic maturation, and early embryonic development. ATRX expression is dynamically regulated as evidenced by loss of the protein in metaphase II of meiosis. The downstream activation of meiosis via protein kinase A inhibition resulted in a similar decrease in ATRX protein expression. We demonstrate that the ATRX protein is detected in ubiquitin immuno-precipitates from germinal vesicle oocyte extracts and experimentally demonstrate that proteosomal degradation is responsible for the decreased expression of ATRX during meiosis. ATRX expression is significantly increased in response to gamma-irradiation induced DNA damage in oocytes and embryos. This increased expression is independent of p53 protein expression in apoptotic embryos, as determined by the expression of active caspase-3. Thus, regulation of ATRX protein expression impacts on G2-M progression and ultimately has consequences for cell survival.
Collapse
Affiliation(s)
| | - Trudee Fair
- UCD School of Agriculture and Food Science, Dublin, Ireland
| | - Carmel Hensey
- UCD School of Bimolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
7
|
Jessus C, Munro C, Houliston E. Managing the Oocyte Meiotic Arrest-Lessons from Frogs and Jellyfish. Cells 2020; 9:E1150. [PMID: 32392797 PMCID: PMC7290932 DOI: 10.3390/cells9051150] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 12/11/2022] Open
Abstract
During oocyte development, meiosis arrests in prophase of the first division for a remarkably prolonged period firstly during oocyte growth, and then when awaiting the appropriate hormonal signals for egg release. This prophase arrest is finally unlocked when locally produced maturation initiation hormones (MIHs) trigger entry into M-phase. Here, we assess the current knowledge of the successive cellular and molecular mechanisms responsible for keeping meiotic progression on hold. We focus on two model organisms, the amphibian Xenopus laevis, and the hydrozoan jellyfish Clytia hemisphaerica. Conserved mechanisms govern the initial meiotic programme of the oocyte prior to oocyte growth and also, much later, the onset of mitotic divisions, via activation of two key kinase systems: Cdk1-Cyclin B/Gwl (MPF) for M-phase activation and Mos-MAPkinase to orchestrate polar body formation and cytostatic (CSF) arrest. In contrast, maintenance of the prophase state of the fully-grown oocyte is assured by highly specific mechanisms, reflecting enormous variation between species in MIHs, MIH receptors and their immediate downstream signalling response. Convergence of multiple signalling pathway components to promote MPF activation in some oocytes, including Xenopus, is likely a heritage of the complex evolutionary history of spawning regulation, but also helps ensure a robust and reliable mechanism for gamete production.
Collapse
Affiliation(s)
- Catherine Jessus
- Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine, LBD - IBPS, Sorbonne Université, CNRS, F-75005 Paris, France
| | - Catriona Munro
- Laboratoire de Biologie du Développement de Villefranche-sur-mer (LBDV), Sorbonne Université, CNRS, 06230 Villefranche-sur-mer, France;
- Inserm, Center for Interdisciplinary Research in Biology, Collège de France, PSL Research University, CNRS, 75005 Paris, France
| | - Evelyn Houliston
- Laboratoire de Biologie du Développement de Villefranche-sur-mer (LBDV), Sorbonne Université, CNRS, 06230 Villefranche-sur-mer, France;
| |
Collapse
|
8
|
Dupré AI, Haccard O, Jessus C. The greatwall kinase is dominant over PKA in controlling the antagonistic function of ARPP19 in Xenopus oocytes. Cell Cycle 2017; 16:1440-1452. [PMID: 28722544 DOI: 10.1080/15384101.2017.1338985] [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] [Indexed: 01/25/2023] Open
Abstract
The small protein ARPP19 plays a dual role during oocyte meiosis resumption. In Xenopus, ARPP19 phosphorylation at S109 by PKA is necessary for maintaining oocytes arrested in prophase of the first meiotic division. Progesterone downregulates PKA, leading to the dephosphorylation of ARPP19 at S109. This initiates a transduction pathway ending with the activation of the universal inducer of M-phase, the kinase Cdk1. This last step depends on ARPP19 phosphorylation at S67 by the kinase Greatwall. Hence, phosphorylated by PKA at S109, ARPP19 restrains Cdk1 activation while when phosphorylated by Greatwall at S67, ARPP19 becomes an inducer of Cdk1 activation. Here, we investigate the functional interplay between S109 and S67-phosphorylations of ARPP19. We show that both PKA and Gwl phosphorylate ARPP19 independently of each other and that Cdk1 is not directly involved in regulating the biological activity of ARPP19. We also show that the phosphorylation of ARPP19 at S67 that activates Cdk1, is dominant over the inhibitory S109 phosphorylation. Therefore our results highlight the importance of timely synchronizing ARPP19 phosphorylations at S109 and S67 to fully activate Cdk1.
Collapse
Affiliation(s)
- Aude-Isabelle Dupré
- a Sorbonne Universités, UPMC Univ Paris 06, CNRS, Biologie du développement - Institut de Biologie Paris Seine (LBD - IBPS) , Paris , France
| | - Olivier Haccard
- a Sorbonne Universités, UPMC Univ Paris 06, CNRS, Biologie du développement - Institut de Biologie Paris Seine (LBD - IBPS) , Paris , France
| | - Catherine Jessus
- a Sorbonne Universités, UPMC Univ Paris 06, CNRS, Biologie du développement - Institut de Biologie Paris Seine (LBD - IBPS) , Paris , France
| |
Collapse
|
9
|
Dupré A, Daldello EM, Nairn AC, Jessus C, Haccard O. Phosphorylation of ARPP19 by protein kinase A prevents meiosis resumption in Xenopus oocytes. Nat Commun 2015; 5:3318. [PMID: 24525567 PMCID: PMC4014304 DOI: 10.1038/ncomms4318] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 01/24/2014] [Indexed: 11/13/2022] Open
Abstract
During oogenesis, oocytes are arrested in prophase and resume meiosis by activating the kinase Cdk1 upon hormonal stimulation. In all vertebrates, release from prophase arrest relies on protein kinase A (PKA) downregulation and on the dephosphorylation of a long-sought but still unidentified substrate. Here we show that ARPP19 is the PKA substrate whose phosphorylation at serine 109 is necessary and sufficient for maintaining Xenopus oocytes arrested in prophase. By downregulating PKA, progesterone, the meiotic inducer in Xenopus, promotes partial dephosphorylation of ARPP19 that is required for the formation of a threshold level of active Cdk1. Active Cdk1 then initiates MPF autoamplification loop that occurs independently of both PKA and ARPP19 phosphorylation at serine 109 but requires the Greatwall-dependent phosphorylation of ARPP19 at serine 67. Therefore, ARPP19 stands at a crossroads in the meiotic M-phase control network by integrating differential effects of PKA and Greatwall, two essential kinases for meiosis resumption.
Collapse
Affiliation(s)
- Aude Dupré
- 1] Sorbonne Universités, UPMC Univ Paris 06, UMR7622-Biologie du Développement, Paris F-75005, France [2] CNRS, UMR7622-Biologie du Développement, Paris F-75005, France [3]
| | - Enrico M Daldello
- 1] Sorbonne Universités, UPMC Univ Paris 06, UMR7622-Biologie du Développement, Paris F-75005, France [2] CNRS, UMR7622-Biologie du Développement, Paris F-75005, France [3] Sorbonne Universités, UPMC Univ Paris 06, IFD, 4 Place Jussieu, cedex 05, Paris 75252, France [4]
| | - Angus C Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut 06508, USA
| | - Catherine Jessus
- 1] Sorbonne Universités, UPMC Univ Paris 06, UMR7622-Biologie du Développement, Paris F-75005, France [2] CNRS, UMR7622-Biologie du Développement, Paris F-75005, France
| | - Olivier Haccard
- 1] Sorbonne Universités, UPMC Univ Paris 06, UMR7622-Biologie du Développement, Paris F-75005, France [2] CNRS, UMR7622-Biologie du Développement, Paris F-75005, France
| |
Collapse
|
10
|
Ser 15 of WEE1B is a potential PKA phosphorylation target in G2/M transition in one-cell stage mouse embryos. Mol Med Rep 2013; 7:1929-37. [DOI: 10.3892/mmr.2013.1437] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 03/28/2013] [Indexed: 11/05/2022] Open
|
11
|
Hennenberg M, Strittmatter F, Walther S, Hedlund P, Andersson KE, Stief CG, Schlenker B, Gratzke C. α1-adrenoceptor activation induces phosphorylation of β2-adrenoceptors in human prostate tissue. BJU Int 2011; 108:922-8. [PMID: 21371241 DOI: 10.1111/j.1464-410x.2010.10021.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE • To test whether β1-adrenoceptor activation leads to phosphorylation of the β2-adrenoceptor in human prostate tissue. PATIENTS AND METHODS • Prostate tissue from patients undergoing radical prostatectomy was stimulated in vitro with the α1-adrenergic agonist phenylephrine (10 µM). • α2-adrenoceptor phosphorylation at serines 345/346 was studied using Western blot analysis with a phospho-specific antibody. • The role of second messenger kinases was assessed by studying the effects of the protein kinase C (PKC) inhibitor Ro 31-8425 and the protein kinase A (PKA) inhibitor H89 on phenylephrine-induced phosphorylation. • The expression of G protein-coupled receptor kinases (GRKs) 2/3 was analysed using quantitative reverse-transcriptase-polymerase chain reaction (RT-PCR), Western blot analysis and immunohistochemistry. RESULTS • Stimulation of prostate tissue with phenylephrine resulted in phosphorylation of the β2-adrenoceptor (5, 10 and 20 min after stimulation). • This α1-adrenoceptor-induced phosphorylation of β2-adrenoceptors was resistant to inhibition of PKC and PKA. • Changes in phosphorylation levels were not attributable to changes in receptor levels, as these remained constant during stimulation. • RT-PCR and Western blot analysis showed expression of GRK2/3 in human prostate tissues. • Immunohistochemical staining showed that GRK2/3 expression in human prostate tissue is located to stromal and smooth muscle cells. CONCLUSIONS • Activation of α1-adrenoceptors causes phosphorylation of β2-adrenoceptors in the human prostate. This may enhance α1-adrenergic contraction and is possibly mediated by GRK2, which is expressed in prostate smooth muscle. • Mutual regulation between different adrenergic receptors might be involved in the therapeutic effects of α1-blockers in patients with benign prostate hyperplasia.
Collapse
|
12
|
Marteil G, Richard-Parpaillon L, Kubiak JZ. Role of oocyte quality in meiotic maturation and embryonic development. Reprod Biol 2010; 9:203-24. [PMID: 19997475 DOI: 10.1016/s1642-431x(12)60027-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The quality of oocytes plays a key role in a proper embryo development. In humans, oocytes of poor quality may be the cause of women infertility and an important obstacle in successful in vitro fertilization (IVF). The competence of oocytes depends on numerous processes taking place during the whole oogenesis, but its final steps such as oocyte maturation, seem to be of key importance. In this paper, we overview factors involved in the development of a fully functional female gamete with Xenopus laevis as a major experimental model. Modern approaches, e.g. proteomic analysis, enable the identification of novel proteins involved in oocyte development. EP45, called also Seryp or pNiXa, which belongs to the serpin (serine protease inhibitors) super-family is one of such recently analyzed proteins. This protein seems to be involved in the stimulation of meiotic maturation and embryo development. EP45 is potentially a key factor in correct oocyte development and determining the quality of oocytes.
Collapse
Affiliation(s)
- Gaëlle Marteil
- CNRS-UMR 6061, University of Rennes 1, IFR 140 GFAS, Rennes, France
| | | | | |
Collapse
|
13
|
Dehennaut V, Lefebvre T, Leroy Y, Vilain JP, Michalski JC, Bodart JF. Survey of O-GlcNAc level variations in Xenopus laevis from oogenesis to early development. Glycoconj J 2008; 26:301-11. [PMID: 18633701 DOI: 10.1007/s10719-008-9166-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Revised: 06/13/2008] [Accepted: 06/20/2008] [Indexed: 02/03/2023]
Abstract
Little is known about the impact of O-linked-N-acetylglucosaminylation (O-GlcNAc) in gametes production and developmental processes. Here we investigated changes in O-GlcNAc, UDP-GlcNAc and O-GlcNAc transferase (OGT) levels in Xenopus laevis from oogenesis to embryo hatching. We showed that in comparison to stage VI, stages I-V oocytes expressed higher levels of O-GlcNAc correlating changes in OGT expression, but not in UDP-GlcNAc pools. Upon progesterone stimulation, an O-GlcNAc level burst occurred during meiotic resumption long before MPF and Mos-Erk2 pathways activations. Finally, we observed high levels of O-GlcNAc, UDP-GlcNAc and OGT during segmentation that decreased concomitantly at the onset of gastrulation. Nevertheless, no correlation between the glycosylation, the nucleotide-sugar and the glycosyltransferase was observed after neurulation. Our results show that O-GlcNAc is regulated throughout oogenesis and development within a complex pattern and suggest that dysfunctions in the dynamics of this glycosylation could lead to developmental abnormalities.
Collapse
Affiliation(s)
- Vanessa Dehennaut
- UMR-CNRS 8576, Unité de Glycobiologie Structurale et Fonctionnelle, IFR 147, USTL, 59655, Villeneuve d'Ascq, France
| | | | | | | | | | | |
Collapse
|
14
|
Tosco M, Faelli A, Gastaldi G, Paulmichl M, Orsenigo MN. Endogenous lactate transport in Xenopus laevis oocyte: dependence on cytoskeleton and regulation by protein kinases. J Comp Physiol B 2008; 178:457-63. [PMID: 18180930 DOI: 10.1007/s00360-007-0238-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2007] [Revised: 12/04/2007] [Accepted: 12/06/2007] [Indexed: 11/28/2022]
Abstract
Carbon flux in Xenopus laevis oocyte is glycogenic and an endogenous monocarboxylate transporter is responsible for intracellular lactate uptake. The aim of the present study was to determine if direct activation of protein kinases C and A modulates the activity of lactate transporter, as well as to investigate the possible role of cytoskeleton in these regulatory phenomena. The modulation was studied in isolated Xenopus oocytes of stage V-VI by measuring (14)C-lactate uptake, both in the absence and in the presence of cytoskeletal-perturbing toxins. We found that the basal lactate transporter activity depends on the integrity of the cytoskeleton since it is partially inhibited by cytoskeleton disorganisation. Both PKA and PKC activation caused a significant decrease in transport activity and this decrease could be blocked by specific protein kinase inhibitors. The evidenced effects were not additive. Transport inhibition was annulled by agents that destabilize actin filaments or microtubules. We conclude that both protein kinases A and C, whose effects are mediated by cytoskeleton, negatively regulate the endogenous lactate transporter of Xenopus oocyte, suggesting that these kinases may have a role in the control of cytosolic pyruvate/lactate pool in the oocyte.
Collapse
Affiliation(s)
- Marisa Tosco
- Dipartimento di Scienze Biomolecolari e Biotecnologie, Università di Milano, Milano, Italy.
| | | | | | | | | |
Collapse
|
15
|
Lu CC, Tsai SC. The cyclic AMP-dependent protein kinase A pathway is involved in progesterone effects on calcitonin secretion from TT cells. Life Sci 2007; 81:1411-20. [PMID: 17963787 DOI: 10.1016/j.lfs.2007.08.046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2007] [Revised: 08/28/2007] [Accepted: 08/28/2007] [Indexed: 10/22/2022]
Abstract
It is well known that gonadal steroid hormones influence the level of plasma calcitonin (CT), but the mechanism by which progesterone affects CT secretion is not clear. Immortalized TT cells are a reliable model system for studying the endocrine function of human parafollicular cells. In the present study, the effects of progesterone on CT secretion were examined in TT cells. TT cells were incubated in medium containing vehicle (DMSO), progesterone or BSA-progesterone for 60 or 150 min, and then the levels of CT in the medium, progesterone receptors, cAMP accumulation and CT mRNA expression were measured. To study the correlation between progesterone effects and the cAMP-dependent protein kinase A (PKA) pathway, cell lysates or cells in 24-well plates were treated with either vehicle or progesterone plus RU486, SQ22536, KT5720, or 3-isobutyl-1-methylxanthine. Then, adenylyl cyclase and protein kinase A (PKA) activities were measured in the cell lysates, and the CT levels were measured in the medium from the 24-well plate. The activated cAMP response element binding protein (P-CREB) was also measured by immunofluorescence. Administration of 1 microM progesterone or 500 nM BSA-progesterone increased the secretion of CT by 381% and 100%, respectively. Progesterone receptors A and B were downregulated by progesterone treatment. The cAMP concentration, adenylyl cyclase and PKA activity, CT mRNA expression, and nuclear P-CREB concentrations all showed an increase after progesterone treatment. RU486, SQ22536 and KT5720 inhibited the progesterone-stimulated effects. These results suggest that a cAMP-dependent PKA pathway is involved in progesterone-stimulated effects on CT secretion from TT cells.
Collapse
Affiliation(s)
- Chien-Chen Lu
- Division of Research and Development, Mackay Medicine, Nursing and Management College, Taipei, Taiwan, ROC.
| | | |
Collapse
|
16
|
Morikawa M, Seki M, Kume S, Endo T, Nishimura Y, Kano K, Naito K. Meiotic resumption of porcine immature oocytes is prevented by ooplasmic Gsalpha functions. J Reprod Dev 2007; 53:1151-7. [PMID: 17693700 DOI: 10.1262/jrd.19055] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A high cyclic adenosine monophosphate (cAMP) level in fully-grown immature oocytes prevents meiotic resumption. In Xenopus, inhibitory cAMP is synthesized within oocytes depending on a stimulatory alpha-subunit of G-protein (Gsalpha). In the present study, we examined whether ooplasmic Gsalpha is involved in meiotic arrest of porcine oocytes. First, we studied the presence of Gsalpha molecules in porcine oocytes by immunoblotting, and this suggested the presence of reported isoforms (45 and 48 kDa) not only in cumulus cells but also in porcine oocytes. Then we injected an anti-Gsalpha antibody into porcine immature oocytes and found that inhibition of ooplasmic Gsalpha functions significantly promoted germinal vesicle breakdown of the oocytes, whose spontaneous meiotic resumption was prevented by 3-isobutyl-l-methylxanthine (IBMX) treatment. Although cyclin B synthesis and M-phase promoting factor (MPF) activation were largely prevented until 30 h of culture in IBMX-treated oocytes, injection of anti-Gsalpha antibody into these oocytes partially recovered cyclin B synthesis and activated MPF activity at 30 h. These results suggest that meiotic resumption of porcine oocytes is prevented by ooplasmic Gsalpha, which may stimulate cAMP synthesis within porcine oocytes, and that synthesized cAMP prevents meiotic resumption of oocytes through the signaling pathways involved in MPF activation.
Collapse
Affiliation(s)
- Marie Morikawa
- Laboratory of Applied Genetics, Graduate School of Agriculture and Life Science, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
17
|
Gill A, Hammes SR. G beta gamma signaling reduces intracellular cAMP to promote meiotic progression in mouse oocytes. Steroids 2007; 72:117-23. [PMID: 17178138 PMCID: PMC1853321 DOI: 10.1016/j.steroids.2006.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2006] [Accepted: 11/12/2006] [Indexed: 10/23/2022]
Abstract
In nearly every vertebrate species, elevated intracellular cAMP maintains oocytes in prophase I of meiosis. Prior to ovulation, gonadotropins trigger various intra-ovarian processes, including the breakdown of gap junctions, the activation of EGF receptors, and the secretion of steroids. These events in turn decrease intracellular cAMP levels in select oocytes to allow meiotic progression, or maturation, to resume. Studies suggest that cAMP levels are kept elevated in resting oocytes by constitutive G protein signaling, and that the drop in intracellular cAMP that accompanies maturation may be due in part to attenuation of this inhibitory G protein-mediated signaling. Interestingly, one of these G protein regulators of meiotic arrest is the Galpha(s) protein, which stimulates adenylyl cyclase to raise intracellular cAMP in two important animal models of oocyte development: Xenopus leavis frogs and mice. In addition to G(alpha)(s), constitutive Gbetagamma activity similarly stimulates adenylyl cyclase to raise cAMP and prevent maturation in Xenopus oocytes; however, the role of Gbetagamma in regulating meiosis in mouse oocytes has not been examined. Here we show that Gbetagamma does not contribute to the maintenance of murine oocyte meiotic arrest. In fact, contrary to observations in frog oocytes, Gbetagamma signaling in mouse oocytes reduces cAMP and promotes oocyte maturation, suggesting that Gbetagamma might in fact play a positive role in promoting oocyte maturation. These observations emphasize that, while many general concepts and components of meiotic regulation are conserved from frogs to mice, specific differences exist that may lead to important insights regarding ovarian development in vertebrates.
Collapse
Affiliation(s)
- Arvind Gill
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd., Dallas, Texas 75390-8857, USA
| | - Stephen R Hammes
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd., Dallas, Texas 75390-8857, USA
- Corresponding author: Phone: 214-648-3749, FAX: 214-648-7934,
| |
Collapse
|
18
|
Charlesworth A, Wilczynska A, Thampi P, Cox LL, MacNicol AM. Musashi regulates the temporal order of mRNA translation during Xenopus oocyte maturation. EMBO J 2006; 25:2792-801. [PMID: 16763568 PMCID: PMC1500856 DOI: 10.1038/sj.emboj.7601159] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2005] [Accepted: 04/28/2006] [Indexed: 11/08/2022] Open
Abstract
A strict temporal order of maternal mRNA translation is essential for meiotic cell cycle progression in oocytes of the frog Xenopus laevis. The molecular mechanisms controlling the ordered pattern of mRNA translational activation have not been elucidated. We report a novel role for the neural stem cell regulatory protein, Musashi, in controlling the translational activation of the mRNA encoding the Mos proto-oncogene during meiotic cell cycle progression. We demonstrate that Musashi interacts specifically with the polyadenylation response element in the 3' untranslated region of the Mos mRNA and that this interaction is necessary for early Mos mRNA translational activation. A dominant inhibitory form of Musashi blocks maternal mRNA cytoplasmic polyadenylation and meiotic cell cycle progression. Our data suggest that Musashi is a target of the initiating progesterone signaling pathway and reveal that late cytoplasmic polyadenylation element-directed mRNA translation requires early, Musashi-dependent mRNA translation. These findings indicate that Musashi function is necessary to establish the temporal order of maternal mRNA translation during Xenopus meiotic cell cycle progression.
Collapse
Affiliation(s)
- Amanda Charlesworth
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Anna Wilczynska
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Prajitha Thampi
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Linda L Cox
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- The Arkansas Cancer Research Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Neurobiology and Developmental Sciences, The Arkansas Cancer Research Center, Slot 814, University of Arkansas for Medical Sciences, 4301 West Markham, Little Rock, AR 72205, USA. Tel.: +1 501 686 8164; Fax: +1 501 686 6517; E-mail:
| |
Collapse
|
19
|
Gaffré M, Dupré A, Valuckaite R, Suziedelis K, Jessus C, Haccard O. Deciphering the H-Ras pathway in Xenopus oocyte. Oncogene 2006; 25:5155-62. [PMID: 16607282 DOI: 10.1038/sj.onc.1209523] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Xenopus oocytes are arrested in prophase of the first meiotic division. In response to progesterone, they re-enter meiosis and arrest again in metaphase of the second meiotic division. This process, called meiotic maturation, is under the control of the Cyclin B-Cdc2 complex, M phase promoting factor (MPF). Injection of a constitutively active Xenopus H-Ras protein activates MPF, suggesting that Ras proteins could be implicated in the progesterone transduction pathway. The aim of this study was (1) to elucidate the pathway triggered by H-Ras leading to MPF activation in Xenopus oocytes and (2) to investigate whether endogenous H-Ras is involved in the physiological process of meiotic maturation. We generated three constitutively active double mutants, each of them recruiting a single effector in mammalian cells, mitogen-activated protein kinase (MAPK), phosphatidylinositol 3-kinase (PI3K) or RalGDS. Our results show that the activation of a PI3K-related enzyme is crucial for H-Ras-induced MPF activation, whereas the recruitment of either MAPK or RalGDS is not. However, although the H-Ras/PI3K pathway is functional in Xenopus oocytes, it is not the physiological transducer of progesterone responsible for meiotic resumption.
Collapse
Affiliation(s)
- M Gaffré
- Laboratoire de Biologie du Développement, UMR-CNRS 7622, Université Pierre et Marie Curie, Paris, France
| | | | | | | | | | | |
Collapse
|
20
|
Wang J, Liu XJ. Monitoring protein kinase A activities using expressed substrate in live cells. Methods Mol Biol 2006; 322:425-33. [PMID: 16739741 DOI: 10.1007/978-1-59745-000-3_30] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Protein kinase A (PKA) activity is regulated by intracellular cyclic adenosine monophosphate. Conventional protein kinase assays after cell lysis are hence not suitable for analyzing PKA activities. In this chapter, we describe a new method for monitoring PKA activity in live cells. A triparti substrate for PKA (Myr-HA-beta2AR-C) is constructed that contains an N-terminal myristylation sequence followed by an antigenic hemagglutinin epitope tag and a substrate motif (the C-terminal tail of human beta2 adrenergic receptor). The PKA phosphorylation status of the substrate in frog oocytes is determined either by two-dimensional electrophoresis followed by HA epitope immunoblotting or by direct SDS-PAGE followed by immunoblotting using anti-P-beta2 adrenergic receptor antibodies specifically recognizing the PKA-phosphorylated C-terminus. We also describe the application of this strategy in mammalian somatic cells through DNA transfection. Myr-HA-beta2AR-C should be widely adaptable as an in vivo PKA activity indicator.
Collapse
Affiliation(s)
- Jing Wang
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ontario, Canada
| | | |
Collapse
|
21
|
Barnard DC, Cao Q, Richter JD. Differential phosphorylation controls Maskin association with eukaryotic translation initiation factor 4E and localization on the mitotic apparatus. Mol Cell Biol 2005; 25:7605-15. [PMID: 16107707 PMCID: PMC1190291 DOI: 10.1128/mcb.25.17.7605-7615.2005] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Several cytoplasmic polyadenylation element (CPE)-containing mRNAs that are repressed in Xenopus oocytes become active during meiotic maturation. A group of factors that are anchored to the CPE are responsible for this repression and activation. Two of the most important are CPEB, which binds directly to the CPE, and Maskin, which associates with CPEB. In oocytes, Maskin also binds eukaryotic translation initiation factor 4E (eIF4E), an interaction that excludes eIF4G and prevents formation of the eIF4F initiation complex. When the oocytes are stimulated to reenter the meiotic divisions (maturation), CPEB promotes cytoplasmic polyadenylation. The newly elongated poly(A) tail becomes bound by poly(A) binding protein (PABP), which in turn binds eIF4G and helps it displace Maskin from eIF4E, thereby inducing translation. Here we show that Maskin undergoes several phosphorylation events during oocyte maturation, some of which are important for its dissociation from eIF4E and translational activation of CPE-containing mRNA. These sites are T58, S152, S311, S343, S453, and S638 and are phosphorylated by cdk1. Mutation of these sites to alanine alleviates the cdk1-induced dissociation of Maskin from eIF4E. Prior to maturation, Maskin is phosphorylated on S626 by protein kinase A. While this modification has no detectable effect on translation during oocyte maturation, it is critical for this protein to localize on the mitotic apparatus in somatic cells. These results show that Maskin activity and localization is controlled by differential phosphorylation.
Collapse
Affiliation(s)
- Daron C Barnard
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, 01605, USA
| | | | | |
Collapse
|
22
|
Eyers PA, Liu J, Hayashi NR, Lewellyn AL, Gautier J, Maller JL. Regulation of the G(2)/M transition in Xenopus oocytes by the cAMP-dependent protein kinase. J Biol Chem 2005; 280:24339-46. [PMID: 15860459 DOI: 10.1074/jbc.m412442200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Vertebrate oocytes are arrested in G(2) phase of the cell cycle at the prophase border of meiosis I. Progesterone treatment of Xenopus oocytes releases the G(2) block and promotes entry into the M phases of meiosis I and II. Substantial evidence indicates that the release of the G(2) arrest requires a decrease in cAMP and reduced activity of the cAMP-dependent protein kinase (PKAc). It has been reported and we confirm here that microinjection of either wild type or kinase-dead K72R PKAc inhibits progesterone-dependent release of the G(2) arrest with equal potency and that inhibition can be reversed by a second injection of the heat-stable inhibitor of PKAc, PKI. However, a mutant enzyme predicted to be completely kinase-dead from the crystal structure of PKAc, K72H PKAc, was much less inhibitory when carrying additional mutations that block interaction with either type I or type II regulatory subunit. Moreover, inhibition by K72H PKAc was reversed by PKI at a 30-fold lower concentration and with more rapid kinetics compared with wild type PKAc. K72R PKAc was found to have low but detectable activity after incubation in an oocyte extract. These results indicate that inhibition of the progesterone-dependent G(2)/M transition in oocytes after microinjection of dead PKAc reflects either low residual activity or binding to regulatory subunits with a resulting net increase in the level of endogenous wild type PKAc. Consistent with this hypothesis, the induction of mitosis in Xenopus egg extracts by the addition of cyclin B was blocked by wild type PKAc but not by K72H PKAc. The identification of substrates for PKAc that maintain cell cycle arrest in G(2) remains an important goal for future work.
Collapse
Affiliation(s)
- Patrick A Eyers
- Howard Hughes Medical Institute and Department of Pharmacology, University of Colorado School of Medicine, Denver, Colorado 80262, USA
| | | | | | | | | | | |
Collapse
|