1
|
Bralewska M, Pietrucha T, Sakowicz A. The Role of Catestatin in Preeclampsia. Int J Mol Sci 2024; 25:2461. [PMID: 38473713 DOI: 10.3390/ijms25052461] [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: 01/18/2024] [Revised: 02/14/2024] [Accepted: 02/18/2024] [Indexed: 03/14/2024] Open
Abstract
Preeclampsia (PE) is a unique pregnancy disorder affecting women across the world. It is characterized by the new onset of hypertension with coexisting end-organ damage. Although the disease has been known for centuries, its exact pathophysiology and, most importantly, its prevention remain elusive. The basis of its associated molecular changes has been attributed to the placenta and the hormones regulating its function. One such hormone is chromogranin A (CgA). In the placenta, CgA is cleaved to form a variety of biologically active peptides, including catestatin (CST), known inter alia for its vasodilatory effects. Recent studies indicate that the CST protein level is diminished both in patients with hypertension and those with PE. Therefore, the aim of the present paper is to review the most recent and most relevant in vitro, in vivo, and clinical studies to provide an overview of the proposed impact of CST on the molecular processes of PE and to consider the possibilities for future experiments in this area.
Collapse
Affiliation(s)
- Michalina Bralewska
- Department of Medical Biotechnology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
| | - Tadeusz Pietrucha
- Department of Medical Biotechnology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
| | - Agata Sakowicz
- Department of Medical Biotechnology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
| |
Collapse
|
2
|
Fleming T, Tachizawa M, Nishiike Y, Koiwa A, Homan Y, Okubo K. Estrogen-dependent expression and function of secretogranin 2a in female-specific peptidergic neurons. PNAS NEXUS 2023; 2:pgad413. [PMID: 38111823 PMCID: PMC10726998 DOI: 10.1093/pnasnexus/pgad413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 11/13/2023] [Indexed: 12/20/2023]
Abstract
Secretogranin 2 (Scg2) is a member of the secretogranin/chromogranin family of proteins that is involved in neuropeptide and hormone packaging to secretory granules and serves as a precursor for several secreted pleiotropic peptides. A recent study in zebrafish showed that the teleost Scg2 orthologs, scg2a and scg2b, play an important role in mating behavior, but its modes of action and regulatory mechanisms remain unclear. In this study, we identify scg2a in another teleost species, medaka, by transcriptomic analysis as a gene that is expressed in an ovarian secretion-dependent manner in a group of neurons relevant to female sexual receptivity, termed FeSP neurons. Investigation of scg2a expression in the FeSP neurons of estrogen receptor (Esr)-deficient medaka revealed that it is dependent on estrogen signaling through Esr2b, the major determinant of female-typical mating behavior. Generation and characterization of scg2a-deficient medaka showed no overt changes in secretory granule packaging in FeSP neurons. This, along with the observation that Scg2a and neuropeptide B, a major neuropeptide produced by FeSP neurons, colocalize in a majority of secretory granules, suggests that Scg2a mainly serves as a precursor for secreted peptides that act in conjunction with neuropeptide B. Further, scg2a showed sexually biased expression in several brain nuclei implicated in mating behavior. However, we found no significant impact of scg2a deficiency on the performance of mating behavior in either sex. Collectively, our results indicate that, although perhaps not essential for mating behavior, scg2a acts in an estrogen/Esr2b signaling-dependent manner in neurons that are relevant to female sexual receptivity.
Collapse
Affiliation(s)
- Thomas Fleming
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Masaya Tachizawa
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Yuji Nishiike
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Ai Koiwa
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Yuki Homan
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Kataaki Okubo
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| |
Collapse
|
3
|
Guillemot J, Guérin M, Cailleux AF, Lopez AG, Kuhn JM, Anouar Y, Yon L. Characterization of the EM66 Biomarker in the Pituitary and Plasma of Healthy Subjects With Different Gonadotroph Status and Patients With Gonadotroph Tumor. Front Endocrinol (Lausanne) 2019; 10:102. [PMID: 30853937 PMCID: PMC6395403 DOI: 10.3389/fendo.2019.00102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 02/04/2019] [Indexed: 12/05/2022] Open
Abstract
Granins and their derived-peptides are useful markers of secretion from normal and tumoral neuroendocrine cells. The need to identify new diagnostic markers for neuroendocrine tumors, including pituitary tumors prompted us to determine plasma levels of the secretogranin II-derived peptide EM66 in healthy volunteers with different gonadotroph status and to evaluate its usefulness as a circulating marker for the diagnosis of gonadotroph tumor. Using a radioimmunoassay, we determined plasma EM66 concentrations in healthy men and women volunteers in different physiological conditions in relation with the gonadotroph function. Our results revealed that in men, in women with or without contraception, in pregnant or post-menopausal women, plasma EM66 concentrations are not significantly different, and did not show any correlation with gonadotropin levels. In addition, stimulation or inhibition tests of the gonadotroph axis had no effect on EM66 levels, whatever the group of healthy volunteers investigated while gonadotropin levels showed the expected variations. Immunohistochemical experiments and HPLC analysis showed the occurrence of EM66 in pituitary gonadotroph, lactotroph and corticotroph tumors but not in somatotroph tumor. In patients with gonadotroph or lactotroph tumor, plasma EM66 levels were 1.48 (0.82-4.38) ng/ml and 2.49 (1.19-3.54) ng/ml, respectively. While median value of EM66 was significantly lower in patients with gonadotroph tumor compared to healthy volunteers [2.59 (0.62-4.95) ng/ml], plasma EM66 concentrations were in the same range as normal values and did not show any correlation with gonadotropin levels. These results show that plasma EM66 levels are independent of the activity of the gonadotroph axis in healthy volunteers and, while EM66 levels are reduced in gonadotroph tumors, plasma EM66 does not provide a helpful marker for the diagnosis of these tumors.
Collapse
Affiliation(s)
- Johann Guillemot
- Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Normandie Univ, UNIROUEN, INSERM, Rouen, France
| | - Marlène Guérin
- Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Normandie Univ, UNIROUEN, INSERM, Rouen, France
| | - Anne-Françoise Cailleux
- Endocrinology, Diabetes and Metabolism Department, Normandie Univ, UNIROUEN, Rouen University Hospital, INSERM CIC-CRB, Rouen, France
| | - Antoine-Guy Lopez
- Department of Endocrinology, Diabetes and Metabolic Diseases, Normandie Univ, UNIROUEN, Rouen University Hospital, Rouen, France
| | - Jean-Marc Kuhn
- Endocrinology, Diabetes and Metabolism Department, Normandie Univ, UNIROUEN, Rouen University Hospital, INSERM CIC-CRB, Rouen, France
| | - Youssef Anouar
- Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Normandie Univ, UNIROUEN, INSERM, Rouen, France
| | - Laurent Yon
- Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Normandie Univ, UNIROUEN, INSERM, Rouen, France
- *Correspondence: Laurent Yon
| |
Collapse
|
4
|
Da Fonte DF, Xing L, Mikwar M, Trudeau VL. Secretoneurin-A inhibits aromatase B (cyp19a1b) expression in female goldfish (Carassius auratus) radial glial cells. Gen Comp Endocrinol 2018; 257:106-112. [PMID: 28487180 DOI: 10.1016/j.ygcen.2017.04.014] [Citation(s) in RCA: 9] [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: 01/27/2017] [Revised: 04/20/2017] [Accepted: 04/26/2017] [Indexed: 12/29/2022]
Abstract
In the teleost brain, radial glial cells (RGCs) are the main macroglia and are stem-like progenitors that express key steroidogenic enzymes, including the estrogen-synthesizing enzyme, aromatase B (cyp19a1b). As a result, RGCs are integral to neurogenesis and neurosteroidogenesis, however little is known about the regulatory factors and signaling mechanisms that control these functions. A potential new role of the secretogranin II-derived neuropeptide secretoneurin A (SNa) in the control of goldfish (Carassius auratus) RGC function is the subject of this study. Immunohistochemistry revealed a close neuroanatomical relationship between RGCs and soma of SNa-immunoreactive magnocellular and parvocellular neurons in the preoptic nucleus of female goldfish. Five hours following intracerebroventricular injection of 0.2ng/g SNa cyp19a1b mRNA levels were decreased by 86% (P<0.05) in the hypothalamus and by 88% (P<0.05) in the telencephalon. In vitro, 24 h incubation with 500nM SNa decreased cyp19a1b mRNA by 51% (P<0.05) in cultured RGCs. These data provide evidence that SNa can regulate aromatase expression in goldfish RGCs. By regulating neuroestrogen production in RGCs SNa may therefore be implicated in the control of major estrogen-dependent functions of the preoptic region such as reproductive behavior and osmoregulation.
Collapse
Affiliation(s)
- Dillon F Da Fonte
- Department of Biology, University of Ottawa, Ontario K1N 6N5, Canada
| | - Lei Xing
- Department of Biology, University of Ottawa, Ontario K1N 6N5, Canada
| | - Myy Mikwar
- Department of Biology, University of Ottawa, Ontario K1N 6N5, Canada
| | - Vance L Trudeau
- Department of Biology, University of Ottawa, Ontario K1N 6N5, Canada.
| |
Collapse
|
5
|
Pandey K, Mizukami Y, Watanabe K, Sakaguti S, Kadokawa H. Deep sequencing of the transcriptome in the anterior pituitary of heifers before and after ovulation. J Vet Med Sci 2017; 79:1003-1012. [PMID: 28442638 PMCID: PMC5487774 DOI: 10.1292/jvms.16-0531] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We aimed to determine gene expression patterns in the anterior pituitary (AP) of heifers
before and after ovulation via deep sequencing of the transcriptome (RNA-seq) to identify
new genes and clarify important pathways. Heifers were slaughtered on the estrus day
(pre-ovulation; n=5) or 3 days after ovulation (post-ovulation; n=5) for AP collection. We
randomly selected 4 pre-ovulation and 4 post-ovulation APs, and the ribosomal RNA-depleted
poly (A)+RNA were prepared to assemble next-generation sequencing libraries. The bovine
APs expressed 12,769 annotated genes at pre- or post-ovulation. The sum of the reads per
kilobase of exon model per million mapped reads (RPKM) values of all transcriptomes were
599,676 ± 38,913 and 668,209 ± 23,690, and 32.2 ± 2.6% and 44.0 ± 4.4% of these
corresponded to the AP hormones in the APs of pre- and post-ovulation heifers,
respectively. The bovine AP showed differential expression of 396 genes
(P<0.05) in the pre- and post-ovulation APs. The 396 genes included
two G-protein-coupled receptor (GPCR) genes (GPR61 and
GPR153) and those encoding 13 binding proteins. The AP also expressed
259 receptor and other 364 binding proteins. Moreover, ingenuity pathway analysis for the
396 genes revealed (P=2.4 × 10−3) a canonical pathway linking
GPCR to cytoskeleton reorganization, actin polymerization, microtubule growth, and gene
expression. Thus, the present study clarified the novel genes found to be differentially
expressed before and after ovulation and clarified an important pathway in the AP.
Collapse
Affiliation(s)
- Kiran Pandey
- Joint Faculty of Veterinary Medicine, Yamaguchi University, Yoshida 1677-1, Yamaguchi-shi, Yamaguchi 753-8515, Japan
| | - Yoichi Mizukami
- Center for Gene Research, Yamaguchi University, Minami Kogushi 1-1-1, Ube-shi, Yamaguchi 755-8505, Japan
| | - Kenji Watanabe
- Center for Gene Research, Yamaguchi University, Minami Kogushi 1-1-1, Ube-shi, Yamaguchi 755-8505, Japan
| | - Syuiti Sakaguti
- Institute of Radioisotope Research and Education, Yamaguchi University, Minami Kogushi 1-1-1, Ube-shi, Yamaguchi 755-8505, Japan
| | - Hiroya Kadokawa
- Joint Faculty of Veterinary Medicine, Yamaguchi University, Yoshida 1677-1, Yamaguchi-shi, Yamaguchi 753-8515, Japan
| |
Collapse
|
6
|
Gomi H, Morikawa S, Shinmura N, Moki H, Yasui T, Tsukise A, Torii S, Watanabe T, Maeda Y, Hosaka M. Expression of Secretogranin III in Chicken Endocrine Cells: Its Relevance to the Secretory Granule Properties of Peptide Prohormone Processing and Bioactive Amine Content. J Histochem Cytochem 2015; 63:350-66. [PMID: 25673289 PMCID: PMC4409946 DOI: 10.1369/0022155415575032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 02/05/2015] [Indexed: 01/27/2023] Open
Abstract
The expression of secretogranin III (SgIII) in chicken endocrine cells has not been investigated. There is limited data available for the immunohistochemical localization of SgIII in the brain, pituitary, and pancreatic islets of humans and rodents. In the present study, we used immunoblotting to reveal the similarities between the expression patterns of SgIII in the common endocrine glands of chickens and rats. The protein-protein interactions between SgIII and chromogranin A (CgA) mediate the sorting of CgA/prohormone core aggregates to the secretory granule membrane. We examined these interactions using co-immunoprecipitation in chicken endocrine tissues. Using immunohistochemistry, we also examined the expression of SgIII in a wide range of chicken endocrine glands and gastrointestinal endocrine cells (GECs). SgIII was expressed in the pituitary, pineal, adrenal (medullary parts), parathyroid, and ultimobranchial glands, but not in the thyroid gland. It was also expressed in GECs of the stomach (proventriculus and gizzard), small and large intestines, and pancreatic islet cells. These SgIII-expressing cells co-expressed serotonin, somatostatin, gastric inhibitory polypeptide, glucagon-like peptide-1, glucagon, or insulin. These results suggest that SgIII is expressed in the endocrine cells that secrete peptide hormones, which mature via the intragranular enzymatic processing of prohormones and physiologically active amines in chickens.
Collapse
Affiliation(s)
- Hiroshi Gomi
- Department of Veterinary Anatomy, College of Bioresource Sciences, Nihon University, Fujisawa, Japan (HG, SM, NS, HM, TY, AT)
| | - Satomi Morikawa
- Department of Veterinary Anatomy, College of Bioresource Sciences, Nihon University, Fujisawa, Japan (HG, SM, NS, HM, TY, AT)
| | - Naoki Shinmura
- Department of Veterinary Anatomy, College of Bioresource Sciences, Nihon University, Fujisawa, Japan (HG, SM, NS, HM, TY, AT)
| | - Hiroaki Moki
- Department of Veterinary Anatomy, College of Bioresource Sciences, Nihon University, Fujisawa, Japan (HG, SM, NS, HM, TY, AT)
| | - Tadashi Yasui
- Department of Veterinary Anatomy, College of Bioresource Sciences, Nihon University, Fujisawa, Japan (HG, SM, NS, HM, TY, AT)
| | - Azuma Tsukise
- Department of Veterinary Anatomy, College of Bioresource Sciences, Nihon University, Fujisawa, Japan (HG, SM, NS, HM, TY, AT)
| | - Seiji Torii
- Laboratory of Secretion Biology, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan (ST)
| | - Tsuyoshi Watanabe
- Department of Microscopic Anatomy and Cell Biology, Asahikawa Medical College, Asahikawa, Japan (TW)
| | - Yoshinori Maeda
- Laboratory of Molecular Life Sciences, Department of Biotechnology, Akita Prefectural University, Akita, Japan (YM, MH)
| | - Masahiro Hosaka
- Laboratory of Molecular Life Sciences, Department of Biotechnology, Akita Prefectural University, Akita, Japan (YM, MH)
| |
Collapse
|
7
|
Trudeau VL, Martyniuk CJ, Zhao E, Hu H, Volkoff H, Decatur WA, Basak A. Is secretoneurin a new hormone? Gen Comp Endocrinol 2012; 175:10-8. [PMID: 22036841 DOI: 10.1016/j.ygcen.2011.10.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2011] [Revised: 10/05/2011] [Accepted: 10/12/2011] [Indexed: 11/25/2022]
Abstract
Numerous small potentially bioactive peptides are derived from the selective processing of the ~600 amino acid secretogranin II (SgII) precursor, but only the 31-42 amino acid segment termed secretoneurin (SN) is well-conserved from sharks to mammals. Both SNa and SNb paralogs have been identified in some teleosts, likely arising as a result of the specific genome duplication event in this lineage. Only one copy of the putative lamprey SgII (188 amino acids) could be identified which gives rise to a divergent agnathan SN that contains the signature YTPQ-X-LA-X(7)-EL sequence typical of the central core of all known SN peptides. In rodent models, SN has regulatory effects on neuroinflammation and neurotransmitter release, and possesses therapeutic potential for the induction of angiogenesis. The wide distribution of SN in neuroendocrine neurons and pituitary cells suggests important endocrine roles. The clearest example of the endocrine action of SN is the stimulatory effects on pituitary luteinizing hormone release from goldfish pituitary and mouse LβT2 gonadotroph cells, indicative of an important role in reproduction. Several lines of evidence suggest that the SN receptor is most likely a G-protein coupled protein. Microarray analysis of SN effects on dispersed goldfish pituitary cells in vitro reveals novel SN actions that include effects on genes involved in notch signaling and the guanylate cyclase pathway. Intracerebroventricular injection of SN increases feeding and locomotory behaviors in goldfish. Given that SgII appeared early in vertebrate evolution, SN is an old peptide with emerging implications as a new multifunctional hormone.
Collapse
Affiliation(s)
- Vance L Trudeau
- Department of Biology, Centre for Advanced Research in Environmental Genomics, University of Ottawa, 30 Marie Curie, Ottawa, ON, Canada.
| | | | | | | | | | | | | |
Collapse
|
8
|
Hosaka M, Watanabe T. Secretogranin III: a bridge between core hormone aggregates and the secretory granule membrane. Endocr J 2010; 57:275-86. [PMID: 20203425 DOI: 10.1507/endocrj.k10e-038] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Secretory granules in endocrine cells selectively store bioactive peptide hormones and amines, which are secreted in a regulated manner upon appropriate stimulation. In addition to bioactive substances, various proteins and lipids characteristic of secretory granules are likely recruited to a restricted space at the trans-Golgi Network (TGN), and the space then matures to the secretory granule. Although experimental findings so far have strongly suggested that aggregation- and receptor-mediated processes are essential for the formation of secretory granules, the putative link between these two processes remains to be clarified. Recently, secretogranin III (SgIII) has been identified as a specific binding protein for chromogranin A (CgA), a representative constituent of the core aggregate within secretory granules, and it was later revealed that SgIII can also bind to the cholesterol-rich membrane domain at the TGN. Based on its multifaceted binding properties, SgIII may act as a central player in the formation of cholesterol-rich membrane platforms. Upon these platforms, essential processes for secretory granule biogenesis coordinately occur; that is, selective recruitment of prohormones, processing and modifying of prohormones, and condensation of mature hormones as an aggregate. This review summarizes the findings and theoretical concepts on the issue to date and then focuses on the putative role of SgIII in secretory granule biogenesis in endocrine cells.
Collapse
Affiliation(s)
- Masahiro Hosaka
- Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan.
| | | |
Collapse
|
9
|
Zhao E, Zhang D, Basak A, Trudeau VL. New insights into granin-derived peptides: evolution and endocrine roles. Gen Comp Endocrinol 2009; 164:161-74. [PMID: 19523383 DOI: 10.1016/j.ygcen.2009.01.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2008] [Revised: 12/31/2008] [Accepted: 01/20/2009] [Indexed: 11/22/2022]
Abstract
The granin protein family is composed of two chromogranin and five secretogranin members that are acidic, heat-stable proteins in secretory granules in cells of the nervous and endocrine systems. We report that there is little evidence for evolutionary relationships among the granins except for the chromogranin group. The main granin members, including chromogranin A and B, and secretogranin II are moderately conserved in the vertebrates. Several small bioactive peptides can be generated by proteolysis from those homologous domains existing within the granin precursors, reflecting the conservation of biological activities in different vertebrates. In this context, we focus on reviewing the distribution and function of the major granin-derived peptides, including vasostatin, bovine CgB(1-41) and secretoneurin in vertebrate endocrine systems, especially those associated with growth, glucose metabolism and reproduction.
Collapse
Affiliation(s)
- E Zhao
- Centre for Advanced Research in Environmental Genomics, Department of Biology, University of Ottawa, 30 Marie Curie, Ottawa, ON, Canada
| | | | | | | |
Collapse
|
10
|
Xie J, Roberson MS. 3', 5'-cyclic adenosine 5'-monophosphate response element-dependent transcriptional regulation of the secretogranin II gene promoter depends on gonadotropin-releasing hormone-induced mitogen-activated protein kinase activation and the transactivator activating transcription factor 3. Endocrinology 2008; 149:783-92. [PMID: 17962349 PMCID: PMC2219298 DOI: 10.1210/en.2007-0694] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Previous studies demonstrated that GnRH-induced secretogranin II (SgII) promoter regulation required a consensus cAMP response element (CRE) and protein kinase A/CRE binding protein. The present studies examined the role of additional components of the GnRH signaling network on SgII promoter activity with particular attention devoted to CRE-dependent gene regulation. Disruption of the SgII CRE by mutagenesis resulted in inhibition of GnRH agonist (GnRHa) induction of this promoter in alphaT3-1 cells. Pharmacological and dominant-negative inhibition of the ERK and c-Jun N-terminal kinase (JNK) signaling pathways revealed that GnRHa-induced SgII promoter activity required functional JNK and ERK modules. Combined inhibition of both pathways nearly abolished GnRHa-induced SgII promoter activity. Specific induction of the ERK cascade alone using overexpression of Raf-CAAX was not sufficient to activate the SgII gene promoter. In contrast, overexpression of the catalytic domain of the more pleiotropic MAPK activator, MAPK/ERK kinase-1, was sufficient to induce SgII promoter activity. The effect(s) of mitogen-activated protein/ERK kinase-1 on SgII promoter activity was CRE dependent and was reversed by the combined pharmacological inhibition of both JNK and ERK modules. CRE DNA binding studies demonstrated the recruitment of activating transcription factor (ATF)-3 and c-Jun to the CRE after administration of GnRHa to alphaT3-1 cells. Specific small interfering RNA knockdown of ATF3 reduced ATF3 DNA binding and the effect of GnRHa on the SgII promoter. These studies support the conclusion that MAPK signaling and ATF3 action are essential for full SgII promoter activation by GnRHa through a canonical CRE. Moreover, we suggest that within the GnRH signaling network, CRE-dependent gene regulation in general may be mediated primarily through the immediate early response gene ATF3.
Collapse
Affiliation(s)
- Jianjun Xie
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | | |
Collapse
|
11
|
Zhao E, Basak A, Trudeau VL. Secretoneurin stimulates goldfish pituitary luteinizing hormone production. Neuropeptides 2006; 40:275-82. [PMID: 16806466 DOI: 10.1016/j.npep.2006.05.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2006] [Revised: 04/13/2006] [Accepted: 05/02/2006] [Indexed: 10/24/2022]
Abstract
Secretoneurin (SN), a 33-34 amino acid neuropeptide is derived from endoproteolysis of secretogranin II (SgII), a member protein of the chromogranin family. SN is widely distributed in various tissues of vertebrates especially in pituitary and hypothalamus, and is a potential new hormone. In vivo, i.p. injection of SN increased luteinizing hormone (LH) release in goldfish pretreated with the dopamine antagonist domperidone. In 6-h static incubation of goldfish pituitary fragments, 10 and 100 nM but not 1 nM concentrations of goldfish SN had a direct stimulatory effect to increase LH release by 2.3- and 1.5-fold (p<0.05), respectively. In addition, 500 nM SN induced a 2.6-fold increase in LHbeta subunit messenger RNA (mRNA) levels in pituitary fragments, regardless of whether LHbeta mRNA levels were expressed relative to 18S ribosomal RNA or beta-actin mRNA. We suggest that the stimulatory actions of SN on LH release may be a part of a paracrine or autocrine feedback loop in the pituitary.
Collapse
Affiliation(s)
- E Zhao
- Centre for Advanced Research in Environmental Genomics, Department of Biology, University of Ottawa, Ont., Canada K1N 6N5
| | | | | |
Collapse
|
12
|
Montero-Hadjadje M, Pelletier G, Yon L, Li S, Guillemot J, Magoul R, Tillet Y, Vaudry H, Anouar Y. Biochemical characterization and immunocytochemical localization of EM66, a novel peptide derived from secretogranin II, in the rat pituitary and adrenal glands. J Histochem Cytochem 2003; 51:1083-95. [PMID: 12871990 DOI: 10.1177/002215540305100812] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Characterization of secretogranin II (SgII) mRNA in various vertebrates has revealed selective conservation of the amino acid sequences of two regions of the protein, i.e., the bioactive peptide secretoneurin and a flanking novel peptide that we named EM66. To help elucidate the possible role of EM66, we examined the occurrence as well as the cellular and subcellular distribution of EM66 in rat pituitary and adrenal glands by using a polyclonal antibody raised against the recombinant human EM66 peptide. High-performance liquid chromatography (HPLC) analysis of rat pituitary and adrenal extracts combined with a radioimmunoassay resolved EM66-immunoreactive material exhibiting the same retention time as recombinant EM66. In the rat pituitary, double-labeling immunohistochemical (IHC) studies showed that EM66 immunoreactivity (IR) was present in gonadotrophs, lactotrophs, thyrotrophs, and melanotrophs, whereas corticotrophs were devoid of labeling. EM66-IR was also observed in nerve endings in the neural lobe. Immunocytochemical staining at the electron microscopic level revealed that EM66-IR is sequestered in the secretory granules within gonadotrophs and lactotrophs. In the adrenal medulla, double IHC labeling showed that EM66-IR occurs exclusively in epinephrine-synthesizing cells. At the ultrastructural level, EM66-IR was seen in chromaffin vesicles of adrenomedullary cells. These results demonstrate that post-translational processing of SgII generates a novel peptide that exhibits a cell-specific distribution in the rat pituitary and adrenal glands where it is stored in secretory granules, supporting the notion that EM66 may play a role in the endocrine system.
Collapse
Affiliation(s)
- Maité Montero-Hadjadje
- European Institute for Peptide Research (IFRMP 23), Laboratory of Cellular and Molecular Neuroendocrinology, INSERM U413, UA CNRS, University of Rouen, Mont-Saint-Aignan, France
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Buhler DR, Miranda CL, Henderson MC, Yang YH, Lee SJ, Wang-Buhler JL. Effects of 17beta-estradiol and testosterone on hepatic mRNA/protein levels and catalytic activities of CYP2M1, CYP2K1, and CYP3A27 in rainbow trout (Oncorhynchus mykiss). Toxicol Appl Pharmacol 2000; 168:91-101. [PMID: 11032764 DOI: 10.1006/taap.1999.9016] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
There is growing concern that exposure to chemicals in the environment can disrupt the endocrine systems of wildlife and humans, causing reproductive problems or other adverse effects. The expression of many cytochrome P450s (CYPs) is under hormonal control, hence, levels of these enzymes can be affected by exposure to endocrine-disrupting chemicals. Previous research has reported that treatment of fish and other animals with the estrogenic and androgenic hormones 17beta-estradiol (E2) and testosterone (T) alters the P450 content or enzyme activities in the treated animals. However, the results of many of these studies are either incomplete or in disagreement and in most cases the effect on specific P450 forms has not been determined. Therefore, to better understand the effects of gonadal hormones on the expression of P450s and their associated enzyme activities, it was of interest to undertake a comprehensive investigation of the transcriptional and translational expression of three constitutive hepatic P450s in the rainbow trout (Oncorhynchus mykiss) following hormone exposure. Accordingly, juvenile trout were injected intraperitoneally with propylene glycol vehicle and the most active estrogenic and androgenic hormones E2 (3 mg/kg) or T (3 mg/kg) on days 1, 4, 7, 13, and 15 and euthanized on day 19. After treatment with E2, hepatic microsomes showed significantly lower levels (percentage of control) in total P450 contents (52%), lauric acid hydroxylase (32%), and 6beta-progesterone hydroxylase activities (27%), [(3)H]aflatoxin-DNA binding (31%), and the protein levels of individual cytochrome P450s (CYPs) LMC1 (CYP2M1), LMC2, (CYP2K1), and LMC5 (CYP3A27) (average for three isoforms a reduction to 29% of control values) with only minor differences between sexes. Treatment with T had either no effect or resulted in small increases in total P450 in males (42%), in lauric acid hydroxylase in females (24%), and in 6beta-progesterone hydroxylase activity in males (21%). Biological variabilities among fish were high and a polymorphic or new LMC2-like form was detected at about 52 kDa in some liver microsomal samples after exposure of fish to either hormone. Female liver RNAs were analyzed through Northern blots and an average decrease of 94% in CYP2 M1, CYP2K1, and CYP3A27 mRNA levels occurred in the E2-treated trout. In livers from T-treated trout, the changes of mRNA levels of CYP2M1 and CYP3A27 were negligible, but CYP2K1 mRNA level decreased by about 60%. Additional CYP2K1 cDNA hybridizable mRNAs were seen in some fish as faint bands at about 2.8 kb for both hormone treatments. Results of this study, therefore, indicated that E2 down-regulated while T produced small but variable effects on the hepatic mRNA/protein levels of CYP2K1, CYP2M1, and CYP3A27 in juvenile rainbow trout. This study, therefore, suggests that exposure of fish and other wildlife to environmental endocrine disruptors, especially estrogen mimics, can adversely affect a number of physiological processes through mechanisms involving altered levels of expression of specific P450 isozymes.
Collapse
Affiliation(s)
- D R Buhler
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, 97331, USA
| | | | | | | | | | | |
Collapse
|
14
|
Watanabe T, Banno T, Jeziorowski T, Ohsawa Y, Waguri S, Grube D, Uchiyama Y. Effects of sex steroids on secretory granule formation in gonadotropes of castrated male rats with respect to granin expression. Endocrinology 1998; 139:2765-73. [PMID: 9607783 DOI: 10.1210/endo.139.6.6059] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pituitary gonadotropes show sex-related differences in their ultrastructure. Typical gonadotropes of male rats exhibit both large granules, which contain chromogranin A (CgA), and small granules, which contain secretogranin II (SgII). In contrast, typical female rat gonadotropes show only a very few large granules among the numerous small granules. To clarify the nature of the biogenesis of these secretory granules and the effects of sex steroids, the ultrastructural and immunocytochemical changes in gonadotropes were examined in castrated male rats supplied with a testosterone or estradiol implant. In castrated rats, pituitary expression and plasma levels of LH increased drastically, but the pituitary content of CgA decreased. The majority of gonadotropes then showed features of "castration cells" containing many small secretory granules. A testosterone implant to castrated rats remarkably suppressed the expression and circulating levels of LH and increased the CgA content in the pituitary to near-normal levels. In this situation, immunocytochemical studies demonstrated that gonadotropes again exhibited large and small secretory granules with the respective localization of CgA and SgII. On the contrary, in castrated rats supplied with an estradiol implant, the expression and content of CgA in the pituitary were remarkably suppressed, and large secretory granules disappeared from gonadotropes. These results suggest that the expression of CgA in gonadotropes is regulated differently by male and female sex steroids. These different effects of androgen and estrogen on the expression level of CgA are closely associated with the sex-related differences in the ultrastructure of secretory granules within gonadotropes.
Collapse
Affiliation(s)
- T Watanabe
- Department of Cell Biology and Anatomy I, Osaka University Medical School, Japan.
| | | | | | | | | | | | | |
Collapse
|
15
|
Iacangelo AL, Eiden LE. Chromogranin A: current status as a precursor for bioactive peptides and a granulogenic/sorting factor in the regulated secretory pathway. REGULATORY PEPTIDES 1995; 58:65-88. [PMID: 8577930 DOI: 10.1016/0167-0115(95)00069-n] [Citation(s) in RCA: 128] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- A L Iacangelo
- Section on Molecular Neuroscience, NIMH, NIH, Bethesda, MD 20892-4090, USA
| | | |
Collapse
|
16
|
Fischer-Colbrie R, Laslop A, Kirchmair R. Secretogranin II: molecular properties, regulation of biosynthesis and processing to the neuropeptide secretoneurin. Prog Neurobiol 1995; 46:49-70. [PMID: 7568909 DOI: 10.1016/0301-0082(94)00060-u] [Citation(s) in RCA: 183] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Secretogranin II is an acidic secretory protein in large dense core vesicles of endocrine, neuroendocrine and neuronal tissues. It comprises, together with chromogranins A and B, the class of proteins collectively called chromogranins. In this review the physico-chemical properties, genomic organization, tissue distribution, synthesis regulation, ontogeny and physiological function of this protein are discussed. Secretogranin II gained interest recently for mainly three reasons: (1) secretogranin II is an excellent marker for the regulated secretory pathway due to its simple and specific metabolic labeling by inorganic sulfate; (2) secretogranin II occurs in a variety of neoplasms arising from endocrine and neuroendocrine cells and was shown to be a useful histological tumor marker for these cells; (3) secretogranin II is the precursor of the recently discovered neuropeptide secretoneurin which induces dopamine release in the striatum of the rat brain.
Collapse
|
17
|
Abstract
The anterior pituitary (AP) has been shown to contain a wide variety of bioactive peptides: brain-gut peptides, growth factors, hypothalamic releasing factors, posterior lobe peptides, opioids, and various other peptides. The localization of most of these peptides was first established by immunocytochemical methods and some of the peptides were localized in identified cell types. Although intracellular localization of a peptide may be the consequence of internalization from the plasma compartment, there is evidence for local synthesis of most of these peptides in the AP based on the identification of their messenger-RNA (mRNA). In several cases the release of the peptide from the AP cell has been shown and regulation of synthesis, storage and release have also been described. Because the amount of most of the AP peptides is very low (except for POMC peptides and galanin), endocrine functions are not expected. There is more evidence for paracrine, autocrine, or intracrine roles in growth, differentiation, and regeneration, or in the control of hormone release. To demonstrate such functions, in vitro AP experiments have been designed to avoid the interference of hypothalamic or peripheral hormones. The strategy is first to show a direct effect of the peptide after adding it to the in vitro system and, secondly, to explore if the endogenous AP peptide has a similar action by using blockers of peptide receptors or antisera immunoneutralizing the peptide.
Collapse
Affiliation(s)
- H Houben
- University of Leuven, School of Medicine, Department of Pharmacology, Belgium
| | | |
Collapse
|