1
|
Ivy JR, Carter RN, Zhao JF, Buckley C, Urquijo H, Rog-Zielinska EA, Panting E, Hrabalkova L, Nicholson C, Agnew EJ, Kemp MW, Morton NM, Stock SJ, Wyrwoll C, Ganley IG, Chapman KE. Glucocorticoids regulate mitochondrial fatty acid oxidation in fetal cardiomyocytes. J Physiol 2021; 599:4901-4924. [PMID: 34505639 DOI: 10.1113/jp281860] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/19/2021] [Indexed: 11/08/2022] Open
Abstract
The late gestational rise in glucocorticoids contributes to the structural and functional maturation of the perinatal heart. Here, we hypothesized that glucocorticoid action contributes to the metabolic switch in perinatal cardiomyocytes from carbohydrate to fatty acid oxidation. In primary mouse fetal cardiomyocytes, dexamethasone treatment induced expression of genes involved in fatty acid oxidation and increased mitochondrial oxidation of palmitate, dependent upon a glucocorticoid receptor (GR). Dexamethasone did not, however, induce mitophagy or alter the morphology of the mitochondrial network. In vivo, in neonatal mice, dexamethasone treatment induced cardiac expression of fatty acid oxidation genes. However, dexamethasone treatment of pregnant C57Bl/6 mice at embryonic day (E)13.5 or E16.5 failed to induce fatty acid oxidation genes in fetal hearts assessed 24 h later. Instead, at E17.5, fatty acid oxidation genes were downregulated by dexamethasone, as was GR itself. PGC-1α, required for glucocorticoid-induced maturation of primary mouse fetal cardiomyocytes in vitro, was also downregulated in fetal hearts at E17.5, 24 h after dexamethasone administration. Similarly, following a course of antenatal corticosteroids in a translational sheep model of preterm birth, both GR and PGC-1α were downregulated in heart. These data suggest that endogenous glucocorticoids support the perinatal switch to fatty acid oxidation in cardiomyocytes through changes in gene expression rather than gross changes in mitochondrial volume or mitochondrial turnover. Moreover, our data suggest that treatment with exogenous glucocorticoids may interfere with normal fetal heart maturation, possibly by downregulating GR. This has implications for clinical use of antenatal corticosteroids when preterm birth is considered a possibility. KEY POINTS: Glucocorticoids are steroid hormones that play a vital role in late pregnancy in maturing fetal organs, including the heart. In fetal cardiomyocytes in culture, glucocorticoids promote mitochondrial fatty acid oxidation, suggesting they facilitate the perinatal switch from carbohydrates to fatty acids as the predominant energy substrate. Administration of a synthetic glucocorticoid in late pregnancy in mice downregulates the glucocorticoid receptor and interferes with the normal increase in genes involved in fatty acid metabolism in the heart. In a sheep model of preterm birth, antenatal corticosteroids (synthetic glucocorticoid) downregulates the glucocorticoid receptor and the gene encoding PGC-1α, a master regulator of energy metabolism. These experiments suggest that administration of antenatal corticosteroids in anticipation of preterm delivery may interfere with fetal heart maturation by downregulating the ability to respond to glucocorticoids.
Collapse
Affiliation(s)
- Jessica R Ivy
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Roderic N Carter
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Jin-Feng Zhao
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Charlotte Buckley
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Helena Urquijo
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Eva A Rog-Zielinska
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Emma Panting
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK.,School of Human Sciences, The University of Western Australia, Crawley, Australia
| | - Lenka Hrabalkova
- The Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Cara Nicholson
- The Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Emma J Agnew
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Matthew W Kemp
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore.,Division of Obstetrics and Gynaecology, The University of Western Australia, Crawley, Western Australia, Australia.,Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Nicholas M Morton
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Sarah J Stock
- The Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK.,Division of Obstetrics and Gynaecology, The University of Western Australia, Crawley, Western Australia, Australia.,The Usher Institute, The University of Edinburgh, Edinburgh, UK
| | - Caitlin Wyrwoll
- School of Human Sciences, The University of Western Australia, Crawley, Australia
| | - Ian G Ganley
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Karen E Chapman
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK.,School of Human Sciences, The University of Western Australia, Crawley, Australia
| |
Collapse
|
2
|
Kim JH, Choi MH. Embryonic Development and Adult Regeneration of the Adrenal Gland. Endocrinol Metab (Seoul) 2020; 35:765-773. [PMID: 33397037 PMCID: PMC7803617 DOI: 10.3803/enm.2020.403] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
The adrenal gland plays a pivotal role in an organism's health span by controlling the endocrine system. Decades of research on the adrenal gland have provided multiscale insights into the development and maintenance of this essential organ. A particularly interesting finding is that founder stem/progenitor cells participate in adrenocortical development and enable the adult adrenal cortex to regenerate itself in response to hormonal stress and injury. Since major advances have been made in understanding the dynamics of the developmental process and the remarkable regenerative capacity of the adrenal gland, understanding the mechanisms underlying adrenal development, maintenance, and regeneration will be of interest to basic and clinical researchers. Here, we introduce the developmental processes of the adrenal gland and discuss current knowledge regarding stem/progenitor cells that regulate adrenal cortex remodeling and regeneration. This review will provide insights into the fascinating ongoing research on the development and regeneration of the adrenal cortex.
Collapse
Affiliation(s)
- Ji-Hoon Kim
- School of Biological Sciences, Seoul National University, Seoul,
Korea
| | - Man Ho Choi
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul,
Korea
| |
Collapse
|
3
|
Maroli G, Braun T. The long and winding road of cardiomyocyte maturation. Cardiovasc Res 2020; 117:712-726. [PMID: 32514522 DOI: 10.1093/cvr/cvaa159] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/15/2020] [Accepted: 06/02/2020] [Indexed: 12/13/2022] Open
Abstract
Knowledge about the molecular mechanisms regulating cardiomyocyte (CM) proliferation and differentiation has increased exponentially in recent years. Such insights together with the availability of more efficient protocols for generation of CMs from induced pluripotent stem cells (iPSCs) have raised expectations for new therapeutic strategies to treat congenital and non-congenital heart diseases. However, the poor regenerative potential of the postnatal heart and the incomplete maturation of iPSC-derived CMs represent important bottlenecks for such therapies in future years. CMs undergo dramatic changes at the doorstep between prenatal and postnatal life, including terminal cell cycle withdrawal, change in metabolism, and further specialization of the cellular machinery required for high-performance contraction. Here, we review recent insights into pre- and early postnatal developmental processes that regulate CM maturation, laying specific focus on genetic and metabolic pathways that control transition of CMs from the embryonic and perinatal to the fully mature adult CM state. We recapitulate the intrinsic features of CM maturation and highlight the importance of external factors, such as energy substrate availability and endocrine regulation in shaping postnatal CM development. We also address recent approaches to enhance maturation of iPSC-derived CMs in vitro, and summarize new discoveries that might provide useful tools for translational research on repair of the injured human heart.
Collapse
Affiliation(s)
- Giovanni Maroli
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Rhein-Main, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
| |
Collapse
|
4
|
Agnew EJ, Garcia-Burgos A, Richardson RV, Manos H, Thomson AJW, Sooy K, Just G, Homer NZM, Moran CM, Brunton PJ, Gray GA, Chapman KE. Antenatal dexamethasone treatment transiently alters diastolic function in the mouse fetal heart. J Endocrinol 2019; 241:279-292. [PMID: 31013474 PMCID: PMC6541236 DOI: 10.1530/joe-18-0666] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 04/23/2019] [Indexed: 12/27/2022]
Abstract
Endogenous glucocorticoid action is important in the structural and functional maturation of the fetal heart. In fetal mice, although glucocorticoid concentrations are extremely low before E14.5, glucocorticoid receptor (GR) is expressed in the heart from E10.5. To investigate whether activation of cardiac GR prior to E14.5 induces precocious fetal heart maturation, we administered dexamethasone in the drinking water of pregnant dams from E12.5 to E15.5. To test the direct effects of glucocorticoids upon the cardiovascular system we used SMGRKO mice, with Sm22-Cre-mediated disruption of GR in cardiomyocytes and vascular smooth muscle. Contrary to expectations, echocardiography showed no advancement of functional maturation of the fetal heart. Moreover, litter size was decreased 2 days following cessation of antenatal glucocorticoid exposure, irrespective of fetal genotype. The myocardial performance index and E/A wave ratio, markers of fetal heart maturation, were not significantly affected by dexamethasone treatment in either genotype. Dexamethasone treatment transiently decreased the myocardial deceleration index (MDI; a marker of diastolic function), in control fetuses at E15.5, with recovery by E17.5, 2 days after cessation of treatment. MDI was lower in SMGRKO than in control fetuses and was unaffected by dexamethasone. The transient decrease in MDI was associated with repression of cardiac GR in control fetuses following dexamethasone treatment. Measurement of glucocorticoid levels in fetal tissue and hypothalamic corticotropin-releasing hormone (Crh) mRNA levels suggest complex and differential effects of dexamethasone treatment upon the hypothalamic-pituitary-adrenal axis between genotypes. These data suggest potentially detrimental and direct effects of antenatal glucocorticoid treatment upon fetal heart function.
Collapse
Affiliation(s)
- E J Agnew
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - A Garcia-Burgos
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - R V Richardson
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - H Manos
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - A J W Thomson
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - K Sooy
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - G Just
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - N Z M Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - C M Moran
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - P J Brunton
- Centre for Discovery Brain Sciences, The University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, UK
| | - G A Gray
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - K E Chapman
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
- Correspondence should be addressed to K E Chapman:
| |
Collapse
|
5
|
Agnew EJ, Ivy JR, Stock SJ, Chapman KE. Glucocorticoids, antenatal corticosteroid therapy and fetal heart maturation. J Mol Endocrinol 2018; 61:R61-R73. [PMID: 29720513 PMCID: PMC5976079 DOI: 10.1530/jme-18-0077] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 05/02/2018] [Indexed: 01/08/2023]
Abstract
Glucocorticoids are essential in mammals to mature fetal organs and tissues in order to survive after birth. Hence, antenatal glucocorticoid treatment (termed antenatal corticosteroid therapy) can be life-saving in preterm babies and is commonly used in women at risk of preterm birth. While the effects of glucocorticoids on lung maturation have been well described, the effects on the fetal heart remain less clear. Experiments in mice have shown that endogenous glucocorticoid action is required to mature the fetal heart. However, whether the potent synthetic glucocorticoids used in antenatal corticosteroid therapy have similar maturational effects on the fetal heart is less clear. Moreover, antenatal corticosteroid therapy may increase the risk of cardiovascular disease in adulthood. Here, we present a narrative review of the evidence relating to the effects of antenatal glucocorticoid action on the fetal heart and discuss the implications for antenatal corticosteroid therapy.
Collapse
Affiliation(s)
- Emma J Agnew
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - Jessica R Ivy
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - Sarah J Stock
- MRC Centre for Reproductive HealthUniversity of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - Karen E Chapman
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
- Correspondence should be addressed to K E Chapman:
| |
Collapse
|
6
|
Pravastatin ameliorates placental vascular defects, fetal growth, and cardiac function in a model of glucocorticoid excess. Proc Natl Acad Sci U S A 2016; 113:6265-70. [PMID: 27185937 DOI: 10.1073/pnas.1520356113] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Fetoplacental glucocorticoid overexposure is a significant mechanism underlying fetal growth restriction and the programming of adverse health outcomes in the adult. Placental glucocorticoid inactivation by 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2) plays a key role. We previously discovered that Hsd11b2(-/-) mice, lacking 11β-HSD2, show marked underdevelopment of the placental vasculature. We now explore the consequences for fetal cardiovascular development and whether this is reversible. We studied Hsd11b2(+/+), Hsd11b2(+/-), and Hsd11b2(-/-) littermates from heterozygous (Hsd11b(+/-)) matings at embryonic day (E)14.5 and E17.5, where all three genotypes were present to control for maternal effects. Using high-resolution ultrasound, we found that umbilical vein blood velocity in Hsd11b2(-/-) fetuses did not undergo the normal gestational increase seen in Hsd11b2(+/+) littermates. Similarly, the resistance index in the umbilical artery did not show the normal gestational decline. Surprisingly, given that 11β-HSD2 absence is predicted to initiate early maturation, the E/A wave ratio was reduced at E17.5 in Hsd11b2(-/-) fetuses, suggesting impaired cardiac function. Pravastatin administration from E6.5, which increases placental vascular endothelial growth factor A and, thus, vascularization, increased placental fetal capillary volume, ameliorated the aberrant umbilical cord velocity, normalized fetal weight, and improved the cardiac function of Hsd11b2(-/-) fetuses. This improved cardiac function occurred despite persisting indications of increased glucocorticoid exposure in the Hsd11b2(-/-) fetal heart. Thus, the pravastatin-induced enhancement of fetal capillaries within the placenta and the resultant hemodynamic changes correspond with restored fetal cardiac function. Statins may represent a useful therapeutic approach to intrauterine growth retardation due to placental vascular hypofunction.
Collapse
|
7
|
Laryea G, Muglia L, Arnett M, Muglia LJ. Dissection of glucocorticoid receptor-mediated inhibition of the hypothalamic-pituitary-adrenal axis by gene targeting in mice. Front Neuroendocrinol 2015; 36:150-64. [PMID: 25256348 PMCID: PMC4342273 DOI: 10.1016/j.yfrne.2014.09.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 08/05/2014] [Accepted: 09/11/2014] [Indexed: 12/17/2022]
Abstract
Negative feedback regulation of glucocorticoid (GC) synthesis and secretion occurs through the function of glucocorticoid receptor (GR) at sites in the hypothalamic-pituitary-adrenal (HPA) axis, as well as in brain regions such as the hippocampus, prefrontal cortex, and sympathetic nervous system. This function of GRs in negative feedback coordinates basal glucocorticoid secretion and stress-induced increases in secretion that integrate GC production with the magnitude and duration of the stressor. This review describes the effects of GR loss along major sites of negative feedback including the entire brain, the paraventricular nucleus of the hypothalamus (PVN), and the pituitary. In genetic mouse models, we evaluate circadian regulation of the HPA axis, stress-stimulated neuroendocrine response and behavioral activity, as well as the integrated response of organism metabolism. Our analysis provides information on contributions of region-specific GR-mediated negative feedback to provide insight in understanding HPA axis dysregulation and the pathogenesis of psychiatric and metabolic disorders.
Collapse
Affiliation(s)
- Gloria Laryea
- Neuroscience Graduate Program, School of Medicine, Vanderbilt University, Nashville, TN, United States; Center for Preterm Birth Research, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, ML 7009, Cincinnati, OH 45229, United States.
| | - Lisa Muglia
- Center for Preterm Birth Research, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, ML 7009, Cincinnati, OH 45229, United States.
| | - Melinda Arnett
- Center for Preterm Birth Research, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, ML 7009, Cincinnati, OH 45229, United States.
| | - Louis J Muglia
- Center for Preterm Birth Research, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, ML 7009, Cincinnati, OH 45229, United States; Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, ML 7009, Cincinnati, OH 45229, United States.
| |
Collapse
|
8
|
Glucocorticoids promote structural and functional maturation of foetal cardiomyocytes: a role for PGC-1α. Cell Death Differ 2014; 22:1106-16. [PMID: 25361084 PMCID: PMC4572859 DOI: 10.1038/cdd.2014.181] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 09/08/2014] [Accepted: 09/22/2014] [Indexed: 01/05/2023] Open
Abstract
Glucocorticoid levels rise dramatically in late gestation to mature foetal organs in readiness for postnatal life. Immature heart function may compromise survival. Cardiomyocyte glucocorticoid receptor (GR) is required for the structural and functional maturation of the foetal heart in vivo, yet the molecular mechanisms are largely unknown. Here we asked if GR activation in foetal cardiomyocytes in vitro elicits similar maturational changes. We show that physiologically relevant glucocorticoid levels improve contractility of primary-mouse-foetal cardiomyocytes, promote Z-disc assembly and the appearance of mature myofibrils, and increase mitochondrial activity. Genes induced in vitro mimic those induced in vivo and include PGC-1α, a critical regulator of cardiac mitochondrial capacity. SiRNA-mediated abrogation of the glucocorticoid induction of PGC-1α in vitro abolished the effect of glucocorticoid on myofibril structure and mitochondrial oxygen consumption. Using RNA sequencing we identified a number of transcriptional regulators, including PGC-1α, induced as primary targets of GR in foetal cardiomyocytes. These data demonstrate that PGC-1α is a key mediator of glucocorticoid-induced maturation of foetal cardiomyocyte structure and identify other candidate transcriptional regulators that may play critical roles in the transition of the foetal to neonatal heart.
Collapse
|
9
|
Gahring LC, Myers E, Palumbos S, Rogers SW. Nicotinic receptor Alpha7 expression during mouse adrenal gland development. PLoS One 2014; 9:e103861. [PMID: 25093893 PMCID: PMC4122369 DOI: 10.1371/journal.pone.0103861] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 07/07/2014] [Indexed: 11/23/2022] Open
Abstract
The nicotinic acetylcholine receptor alpha 7 (α7) is a ligand-activated ion channel that contributes to a diversity of cellular processes involved in development, neurotransmission and inflammation. In this report the expression of α7 was examined in the mouse developing and adult adrenal gland that expresses a green fluorescent protein (GFP) reporter as a bi-cistronic extension of the endogenous α7 transcript (α7(G)). At embryonic day 12.5 (E12.5) α7(G) expression was associated with the suprarenal ganglion and precursor cells of the adrenal gland. The α7(G) cells are catecholaminergic chromaffin cells as reflected by their progressive increase in the co-expression of tyrosine hydroxylase (TH) and dopamine-beta-hydroxylase (DBH) that is complete by E18.5. In the adult, α7(G) expression is limited to a subset of chromaffin cells in the adrenal medulla that cluster near the border with the adrenal cortex. These chromaffin cells co-express α7(G), TH and DBH, but they lack phenylethanolamine N-methyltransferase (PNMT) consistent with only norepinephrine (NE) synthesis. These cell groups appear to be preferentially innervated by pre-ganglionic afferents identified by the neurotrophin receptor p75. No afferents identified by beta-III tubulin, neurofilament proteins or p75 co-expressed α7(G). Occasional α7(G) cells in the pre-E14.5 embryos express neuronal markers consistent with intrinsic ganglion cells and in the adult some α7(G) cells co-express glutamic acid decarboxylase. The transient expression of α7 during adrenal gland development and its prominent co-expression by a subset of NE chromaffin cells in the adult suggests that the α7 receptor contributes to multiple aspects of adrenal gland development and function that persist into adulthood.
Collapse
Affiliation(s)
- Lorise C. Gahring
- Salt Lake City VA Geriatric Research, Education and Clinical Center, Salt Lake City, Utah, United States of America
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah, United States of America
| | - Elizabeth Myers
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah, United States of America
| | - Sierra Palumbos
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah, United States of America
| | - Scott W. Rogers
- Salt Lake City VA Geriatric Research, Education and Clinical Center, Salt Lake City, Utah, United States of America
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah, United States of America
| |
Collapse
|
10
|
Cuffe JSM, Walton SL, Singh RR, Spiers JG, Bielefeldt-Ohmann H, Wilkinson L, Little MH, Moritz KM. Mid- to late term hypoxia in the mouse alters placental morphology, glucocorticoid regulatory pathways and nutrient transporters in a sex-specific manner. J Physiol 2014; 592:3127-41. [PMID: 24801305 DOI: 10.1113/jphysiol.2014.272856] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Maternal hypoxia is a common perturbation that can disrupt placental and thus fetal development, contributing to neonatal impairments. Recently, evidence has suggested that physiological outcomes are dependent upon the sex of the fetus, with males more susceptible to hypoxic insults than females. This study investigated the effects of maternal hypoxia during mid- to late gestation on fetal growth and placental development and determined if responses were sex specific. CD1 mice were housed under 21% or 12% oxygen from embryonic day (E) 14.5 until tissue collection at E18.5. Fetuses and placentas were weighed before collection for gene and protein expression and morphological analysis. Hypoxia reduced fetal weight in both sexes at E18.5 by 7% but did not affect placental weight. Hypoxia reduced placental mRNA levels of the mineralocorticoid and glucocorticoid receptors and reduced the gene and protein expression of the glucocorticoid metabolizing enzyme HSD11B2. However, placentas of female fetuses responded differently to maternal hypoxia than did placentas of male fetuses. Notably, morphology was significantly altered in placentas from hypoxic female fetuses, with a reduction in placental labyrinth blood spaces. In addition mRNA expression of Glut1, Igf2 and Igf1r were reduced in placentas of female fetuses only. In summary, maternal hypoxia altered placental formation in a sex specific manner through mechanisms involving placental vascular development, growth factor and nutrient transporter expression and placental glucocorticoid signalling. This study provides insight into how sex differences in offspring disease development may be due to sex specific placental adaptations to maternal insults.
Collapse
Affiliation(s)
- J S M Cuffe
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - S L Walton
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - R R Singh
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - J G Spiers
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - H Bielefeldt-Ohmann
- School of Veterinary Science, The University of Queensland, Gatton, Queensland, Australia
| | - L Wilkinson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - M H Little
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - K M Moritz
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
11
|
Rog-Zielinska EA, Richardson RV, Denvir MA, Chapman KE. Glucocorticoids and foetal heart maturation; implications for prematurity and foetal programming. J Mol Endocrinol 2014; 52:R125-35. [PMID: 24299741 DOI: 10.1530/jme-13-0204] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Glucocorticoids are steroid hormones, essential in mammals to prepare for life after birth. Blood levels of glucocorticoids (cortisol in most mammals including humans; corticosterone in rats and mice) rise dramatically shortly before birth. This is mimicked clinically in the routine administration of synthetic glucocorticoids to pregnant women threatened by a preterm birth or to preterm infants to improve neonatal survival. Whilst effects on lung are well documented and essential for postnatal survival, those on heart are less well known. In this study, we review recent evidence for a crucial role of glucocorticoids in late gestational heart maturation. Either insufficient or excessive glucocorticoid exposure before birth may alter the normal glucocorticoid-regulated trajectory of heart maturation with potential life-long consequences.
Collapse
Affiliation(s)
- Eva A Rog-Zielinska
- Queen's Medical Research Institute, Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | | | | | | |
Collapse
|
12
|
Shtukmaster S, Schier MC, Huber K, Krispin S, Kalcheim C, Unsicker K. Sympathetic neurons and chromaffin cells share a common progenitor in the neural crest in vivo. Neural Dev 2013; 8:12. [PMID: 23777568 PMCID: PMC3693940 DOI: 10.1186/1749-8104-8-12] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 05/17/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The neural crest (NC) is a transient embryonic structure unique to vertebrates, which generates peripheral sensory and autonomic neurons, glia, neuroendocrine chromaffin and thyroid C-cells, melanocytes, and mesenchymal derivatives such as parts of the skull, heart, and meninges. The sympathoadrenal (SA) cell lineage is one major sub-lineage of the NC that gives rise to sympathetic neurons, chromaffin cells, and the intermediate small intensely fluorescent (SIF) cells. A key question is when during NC ontogeny do multipotent progenitors segregate into the different NC-derived lineages. Recent evidence suggested that sympathetic, sensory, and melanocyte progenitors delaminate from the thoracic neural tube (NT) in successive, largely non-overlapping waves and that at least certain NC progenitors are already fate-restricted within the NT. Whether sympathetic neurons and chromaffin cells, suggested by cell culture studies to share a common progenitor, are also fate segregated in ovo prior to emigration, is not known. RESULTS We have conducted single cell electroporations of a GFP-encoding plasmid into the dorsal midline of E2 chick NTs at the adrenomedullary level of the NC. Analysis of their derivatives, performed at E6, revealed that in most cases, labelled progeny was detected in both sympathetic ganglia and adrenal glands, where cells co-expressed characteristic marker combinations. CONCLUSIONS Our results show that sympathetic neurons and adrenal chromaffin cells share a common progenitor in the NT. Together with previous findings we suggest that phenotypic diversification of these sublineages is likely to occur after delamination from the NT and prior to target encounter.
Collapse
Affiliation(s)
- Stella Shtukmaster
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology University of Freiburg, Albertstr, 17, Freiburg D-79104, Germany
| | | | | | | | | | | |
Collapse
|
13
|
Rog-Zielinska EA, Thomson A, Kenyon CJ, Brownstein DG, Moran CM, Szumska D, Michailidou Z, Richardson J, Owen E, Watt A, Morrison H, Forrester LM, Bhattacharya S, Holmes MC, Chapman KE. Glucocorticoid receptor is required for foetal heart maturation. Hum Mol Genet 2013; 22:3269-82. [PMID: 23595884 DOI: 10.1093/hmg/ddt182] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Glucocorticoids are vital for the structural and functional maturation of foetal organs, yet excessive foetal exposure is detrimental to adult cardiovascular health. To elucidate the role of glucocorticoid signalling in late-gestation cardiovascular maturation, we have generated mice with conditional disruption of glucocorticoid receptor (GR) in cardiomyocytes and vascular smooth muscle cells using smooth muscle protein 22-driven Cre recombinase (SMGRKO mice) and compared them with mice with global deficiency in GR (GR(-/-)). Echocardiography shows impaired heart function in both SMGRKO and GR(-/-) mice at embryonic day (E)17.5, associated with generalized oedema. Cardiac ultrastructure is markedly disrupted in both SMGRKO and GR(-/-) mice at E17.5, with short, disorganized myofibrils and cardiomyocytes that fail to align in the compact myocardium. Failure to induce critical genes involved in contractile function, calcium handling and energy metabolism underpins this common phenotype. However, although hearts of GR(-/-) mice are smaller, with 22% reduced ventricular volume at E17.5, SMGRKO hearts are normally sized. Moreover, while levels of mRNA encoding atrial natriuretic peptide are reduced in E17.5 GR(-/-) hearts, they are normal in foetal SMGRKO hearts. These data demonstrate that structural, functional and biochemical maturation of the foetal heart is dependent on glucocorticoid signalling within cardiomyocytes and vascular smooth muscle, though some aspects of heart maturation (size, ANP expression) are independent of GR at these key sites.
Collapse
Affiliation(s)
- Eva A Rog-Zielinska
- Centre for Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh EH16 4TJ, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Unsicker K, Huber K, Schober A, Kalcheim C. Resolved and open issues in chromaffin cell development. Mech Dev 2012; 130:324-9. [PMID: 23220335 DOI: 10.1016/j.mod.2012.11.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 11/14/2012] [Accepted: 11/16/2012] [Indexed: 11/19/2022]
Abstract
Ten years of research within the DFG-funded Collaborative Research Grant SFB 488 at the University of Heidelberg have added many new facets to our understanding of chromaffin cell development. Glucocorticoid signaling is no longer the key for understanding the determination of the chromaffin phenotype, yet a novel role has been attributed to glucocorticoids: they are essential for the postnatal maintenance of adrenal and extra-adrenal chromaffin cells. Transcription factors, as, e.g. MASH1 and Phox2B, have similar, but also distinct functions in chromaffin and sympathetic neuronal development, and BMP-4 not only induces sympathoadrenal (SA) cells at the dorsal aorta and within the adrenal gland, but also promotes chromaffin cell maturation. Chromaffin cells and sympathetic neurons share a common progenitor in the dorsal neural tube (NT) in vivo, as revealed by single cell electroporations into the dorsal NT. Thus, specification of chromaffin cells is likely to occur after cell emigration either during migration or close to colonization of the target regions. Mechanisms underlying the specification of chromaffin cells vs. sympathetic neurons are currently being explored.
Collapse
Affiliation(s)
- Klaus Unsicker
- Dept. of Molecular Embryology, University of Freiburg, Albertstr. 17, D-79104 Freiburg, Germany.
| | | | | | | |
Collapse
|
15
|
Tai TC, Wong-Faull DC, Claycomb R, Aborn JL, Wong DL. PACAP-regulated phenylethanolamine N-methyltransferase gene expression. J Neurochem 2010; 115:1195-205. [DOI: 10.1111/j.1471-4159.2010.07005.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
16
|
|
17
|
Martinez-Finley EJ, Ali AMS, Allan AM. Learning deficits in C57BL/6J mice following perinatal arsenic exposure: consequence of lower corticosterone receptor levels? Pharmacol Biochem Behav 2009; 94:271-7. [PMID: 19751756 DOI: 10.1016/j.pbb.2009.09.006] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Revised: 09/02/2009] [Accepted: 09/03/2009] [Indexed: 01/31/2023]
Abstract
Most studies on arsenic as a drinking water contaminant have focused on its carcinogenic potential but a few suggest that arsenic can adversely affect cognitive development. One parameter of the hypothalamic-pituitary-adrenal axis, the corticosterone receptor (CR) has been shown to be altered by arsenic. These receptors are found throughout the central nervous system, particularly in the hippocampus, an area of the brain of central importance for learning and memory. We examined the impact of perinatal exposure to 50 parts per billion (ppb) sodium arsenate on CRs and learning and memory behavior in the C57BL/6J mouse. Measurements of CRs revealed that arsenic-exposed offspring have significantly lower levels of these receptors in the nucleus than controls. Exposed offspring showed longer latency to approach a novel object than controls in an object recognition task. In the 8-way radial arm maze, arsenic offspring had a significant increase in the number of entry errors compared to controls. Results suggest that moderate exposures to perinatal arsenic can significantly reduce CR levels in the hippocampus and can have adverse effects on learning and memory behavior.
Collapse
Affiliation(s)
- Ebany J Martinez-Finley
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131, United States.
| | | | | |
Collapse
|
18
|
Parlato R, Otto C, Tuckermann J, Stotz S, Kaden S, Gröne HJ, Unsicker K, Schütz G. Conditional inactivation of glucocorticoid receptor gene in dopamine-beta-hydroxylase cells impairs chromaffin cell survival. Endocrinology 2009; 150:1775-81. [PMID: 19036879 DOI: 10.1210/en.2008-1107] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Glucocorticoid hormones (GCs) have been thought to determine the fate of chromaffin cells from sympathoadrenal progenitor cells. The analysis of mice carrying a germ line deletion of the glucocorticoid receptor (GR) gene has challenged these previous results because the embryonic development of adrenal chromaffin cells is largely unaltered. In the present study, we have analyzed the role of GC-dependent signaling in the postnatal development of adrenal chromaffin cells by conditional inactivation of the GR gene in cells expressing dopamine-beta-hydroxylase, an enzyme required for the synthesis of noradrenaline and adrenaline. These mutant mice are viable, allowing to study whether in the absence of GC signaling further development of the adrenal medulla is affected. Our analysis shows that the loss of GR leads not only to the loss of phenylethanolamine-N-methyl-transferase expression and, therefore, to inhibition of adrenaline synthesis, but also to a dramatic reduction in the number of adrenal chromaffin cells. We provide evidence that increased apoptotic cell death is the main consequence of GR loss. These findings define the essential role of GCs for survival of chromaffin cells and underscore the specific requirement of GCs for adrenergic chromaffin cell differentiation and maintenance.
Collapse
Affiliation(s)
- Rosanna Parlato
- Department of Molecular Biology of the Cell I, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Murata T, Tsuboi M, Koide N, Hikita K, Kohno S, Kaneda N. Neuronal differentiation elicited by glial cell line-derived neurotrophic factor and ciliary neurotrophic factor in adrenal chromaffin cell line tsAM5D immortalized with temperature-sensitive SV40 T-antigen. J Neurosci Res 2008; 86:1694-710. [PMID: 18293415 DOI: 10.1002/jnr.21632] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To understand the characteristics of tsAM5D cells immortalized with the temperature-sensitive simian virus 40 large T-antigen, we first examined the responsiveness of the cells to ligands of the glial cell line-derived neurotrophic factor (GDNF) family. tsAM5D cells proliferated at the permissive temperature of 33 degrees C in response to either GDNF or neurturin, but not persephin or artemin. At the nonpermissive temperature of 39 degrees C, GDNF or neurturin caused tsAM5D cells to differentiate into neuron-like cells; however, the differentiated cells died in a time-dependent manner. Interestingly, ciliary neurotrophic factor (CNTF) did not affect the GDNF-mediated cell proliferation at 33 degrees C but promoted the survival and differentiation of GDNF-treated cells at 39 degrees C. In the presence of GDNF plus CNTF, the morphological change induced by the temperature shift was associated with up-regulated expression of various neuronal marker genes, indicating that the cells had undergone neuronal differentiation. In addition, tsAM5D cells caused to differentiate by GDNF plus CNTF at 39 degrees C became dependent solely on nerve growth factor (NGF) for their survival and neurite outgrowth. Moreover, upon treatment with GDNF plus CNTF, the dopaminergic phenotype was suppressed by the temperature shift. Thus, we demonstrated that tsAM5D cells had the capacity to differentiate terminally into neuron-like cells in response to GDNF plus CNTF when the oncogene was inactivated by the temperature shift. This cell line provides a useful model system for studying the role of a variety of signaling molecules for GDNF/CNTF-induced neuronal differentiation.
Collapse
Affiliation(s)
- Tomiyasu Murata
- Department of Analytical Neurobiology, Faculty of Pharmacy, Meijo University, Tempaku, Nagoya, Japan
| | | | | | | | | | | |
Collapse
|
20
|
Huber K, Franke A, Brühl B, Krispin S, Ernsberger U, Schober A, von Bohlen und Halbach O, Rohrer H, Kalcheim C, Unsicker K. Persistent expression of BMP-4 in embryonic chick adrenal cortical cells and its role in chromaffin cell development. Neural Dev 2008; 3:28. [PMID: 18945349 PMCID: PMC2582231 DOI: 10.1186/1749-8104-3-28] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2008] [Accepted: 10/22/2008] [Indexed: 11/29/2022] Open
Abstract
Background Adrenal chromaffin cells and sympathetic neurons both originate from the neural crest, yet signals that trigger chromaffin development remain elusive. Bone morphogenetic proteins (BMPs) emanating from the dorsal aorta are important signals for the induction of a sympathoadrenal catecholaminergic cell fate. Results We report here that BMP-4 is also expressed by adrenal cortical cells throughout chick embryonic development, suggesting a putative role in chromaffin cell development. Moreover, bone morphogenetic protein receptor IA is expressed by both cortical and chromaffin cells. Inhibiting BMP-4 with noggin prevents the increase in the number of tyrosine hydroxylase positive cells in adrenal explants without affecting cell proliferation. Hence, adrenal BMP-4 is likely to induce tyrosine hydroxylase in sympathoadrenal progenitors. To investigate whether persistent BMP-4 exposure is able to induce chromaffin traits in sympathetic ganglia, we locally grafted BMP-4 overexpressing cells next to sympathetic ganglia. Embryonic day 8 chick sympathetic ganglia, in addition to principal neurons, contain about 25% chromaffin-like cells. Ectopic BMP-4 did not increase this proportion, yet numbers and sizes of 'chromaffin' granules were significantly increased. Conclusion BMP-4 may serve to promote specific chromaffin traits, but is not sufficient to convert sympathetic neurons into a chromaffin phenotype.
Collapse
Affiliation(s)
- Katrin Huber
- Neuroanatomy, Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
In the brain, glucocorticoids exert functions in neurogenesis, synaptic plasticity and behavioural responses, as well as in the control of hypothalamic-pituitary-adrenal axis activity. The generation of mice harbouring germline mutations that result either in loss or in gain of glucocorticoid receptor function provided a useful tool for understanding the role of glucocorticoids in the brain in vivo. The improvement of genomic technologies additionally allowed the establishment of mouse models with function-selective point mutations of the receptor as well as the generation of mice harbouring spatially and/or temporally restricted loss of glucocorticoid receptor, specifically within the brain. These models will provide the opportunity to better understand the mechanisms involved in glucocorticoid signalling within the nervous system.
Collapse
Affiliation(s)
- G Erdmann
- German Cancer Research Center, Division Molecular Biology of the Cell I, Im Neuenheimer Feld 280, Heidelberg, Germany
| | | | | |
Collapse
|
22
|
Levitt P, Ferri R, Eagleson K. Molecular contributions to cerebral cortical specification. CIBA FOUNDATION SYMPOSIUM 2007; 193:200-13; discussion 251-7. [PMID: 8727493 DOI: 10.1002/9780470514795.ch10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Evidence is accumulating that decisions of cell fate and commitment to specific regional phenotypes in the cerebral cortex occur through cell interactions that likely begin early in development, perhaps in the proliferative zone. We have focused on the development of the limbic cortex in rats, which includes areas involved in both cognitive and autonomic functions and is marked by expression of the limbic system-associated membrane protein. Transplantation studies show that precursor cells are sensitive to environmental cues which can control expression of area-specific phenotypes, including limbic system-associated protein synthesis and connectivity patterns, but early postmitotic neurons faithfully express traits based on their origin in the donor. We have studied this sensitive period of decision making in vitro. Molecules from the epidermal growth factor family influence dramatically the fate of precursor cells, but only in the presence of matrix molecules. In vivo, both the epidermal growth factor receptor and collagen type IV are expressed in the progenitor cell pool indicating that they can directly affect the initial decisions in differentiation. We suggest that early patterns of gene expression, influenced by environmental cues, are likely to provide a specific framework for subsequent decisions that lead to establishing cortical areas.
Collapse
Affiliation(s)
- P Levitt
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School-UMDNJ, Piscataway 08854, USA
| | | | | |
Collapse
|
23
|
Lohr J, Gut P, Karch N, Unsicker K, Huber K. Development of adrenal chromaffin cells in Sf1 heterozygous mice. Cell Tissue Res 2006; 325:437-44. [PMID: 16685530 DOI: 10.1007/s00441-006-0213-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Accepted: 04/06/2006] [Indexed: 10/24/2022]
Abstract
Adrenal medullary chromaffin cells are derivatives of the neural crest and are widely believed to share a common sympathoadrenal (SA) progenitor with sympathetic neurons. For decades, the adrenal cortical environment was assumed to be essential for channelling SA progenitors towards an endocrine chromaffin cell fate. Our recent analysis of steroidogenic factor 1(Sf1) -/- mice, which lack an adrenal cortex, has challenged this view: in Sf1 -/- mice chromaffin cells migrate to the correct "adrenal" location and undergo largely normal differentiation. In contrast to Sf1 homozygous mutants, heterozygous animals have an adrenal cortex, which, however, is smaller than in wildtype littermates. We show here that the Sf1 +/- adrenal cortical anlagen attract normal numbers of chromaffin progenitor cells into their vicinity by embryonic day 13.5 (E13.5). Two days later, however, only a few scattered cells with highly immature features have immigrated into the adrenal cortex, whereas the remainder form a coherent cell assembly ectopically located at the medial surface of the gland. These cells appear more mature than the scattered intracortical chromaffin progenitors and express the adrenaline synthesizing enzyme PNMT with a delay of 1 day in comparison with wildtype littermates. Nevertheless, chromaffin progenitor cells undergo a numerical reduction of approximately 30% by E17.5. Together, our data suggest that normal adrenocortical development is critical for the correct immigration of chromaffin progenitors into the cortical anlagen, for the timing of PNMT expression and for the regulation of chromaffin cell numbers.
Collapse
Affiliation(s)
- Jennifer Lohr
- Neuroanatomy and Interdisciplinary Centre for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
24
|
Gut P, Huber K, Lohr J, Brühl B, Oberle S, Treier M, Ernsberger U, Kalcheim C, Unsicker K. Lack of an adrenal cortex in Sf1 mutant mice is compatible with the generation and differentiation of chromaffin cells. Development 2005; 132:4611-9. [PMID: 16176945 DOI: 10.1242/dev.02052] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The diversification of neural-crest-derived sympathoadrenal (SA) progenitor cells into sympathetic neurons and neuroendocrine adrenal chromaffin cells was thought to be largely understood. In-vitro studies with isolated SA progenitor cells had suggested that chromaffin cell differentiation depends crucially on glucocorticoids provided by adrenal cortical cells. However, analysis of mice lacking the glucocorticoid receptor gene had revealed that adrenal chromaffin cells develop mostly normally in these mice. Alternative cues from the adrenal cortex that may promote chromaffin cell determination and differentiation have not been identified. We therefore investigated whether the chromaffin cell phenotype can develop in the absence of an adrenal cortex, using mice deficient for the nuclear orphan receptor steroidogenic factor-1 (SF1), which lack adrenal cortical cells and gonads. We show that in Sf1–/– mice typical chromaffin cells assemble correctly in the suprarenal region adjacent to the suprarenal sympathetic ganglion. The cells display most features of chromaffin cells, including the typical large chromaffin granules. Sf1–/–chromaffin cells are numerically reduced by about 50% compared with the wild type at embryonic day (E) 13.5 and E17.5. This phenotype is not accounted for by reduced survival or cell proliferation beyond E12.5. However, already at E12.5 the `adrenal' region in Sf1–/– mice is occupied by fewer PHOX2B+ and TH+ SA cells as well as SOX10+ neural crest cells. Our results suggest that cortical cues are not essential for determining chromaffin cell fate, but may be required for proper migration of SA progenitors to and/or colonization of the adrenal anlage.
Collapse
Affiliation(s)
- Philipp Gut
- Neuroanatomy and Interdisciplinary Center for Neurosciences (IZN) of Heidelberg, INF 307, D-69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Unsicker K, Huber K, Schütz G, Kalcheim C. The Chromaffin Cell and its Development. Neurochem Res 2005; 30:921-5. [PMID: 16187226 DOI: 10.1007/s11064-005-6966-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2005] [Indexed: 11/29/2022]
Abstract
This article summarizes some of the recent progress in understanding the development of chromaffin cells. These cells are derivatives of the neural crest and are intimately associated with the sympathetic nervous system. Although a common sympathoadrenal (SA) progenitor cell for chromaffin cells and sympathetic neurons has been postulated, there is evidence to suggest that chromaffin progenitors are already distinct, at least in part, from neuronal SA progenitors prior to invading the adrenal gland. The concept of an essential role of glucocorticoid signalling for chromaffin cell development has been shaken by the observation that chromaffin cells in mice lacking the glucocorticoid receptor develop largely normal. Distinct developmental requirements of chromaffin cells and sympathetic neurons must also be assumed based on the analyses of mice carrying targeted mutations of the genes for two transcription factors, MASH1 and Phox2B. Both genes are expressed by SA progenitors, but are distinctly required for the development of chromaffin cells and sympathetic neurons. There is an ongoing search for molecules selectively operating at the sites, where chromaffin cells develop. Such molecules may be candidates for triggering the distinct developmental pathway of chromaffin cells, as opposed to sympathetic neurons.
Collapse
Affiliation(s)
- Klaus Unsicker
- Interdisciplinary Center for Neurosciences (IZN) & Neuroanatomy, University of Heidelberg, INF 307, D-69120, Heidelberg, Germany.
| | | | | | | |
Collapse
|
26
|
Eaton MJ, Duplan H. Useful cell lines derived from the adrenal medulla. Mol Cell Endocrinol 2004; 228:39-52. [PMID: 15541571 DOI: 10.1016/j.mce.2003.02.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2002] [Accepted: 02/06/2003] [Indexed: 01/07/2023]
Abstract
Five approaches for the preparation of adrenal chromaffin cell lines have been developed. Initially, continuous chromaffin lines were derived from spontaneous pheochromocytoma tumors of the medulla, either from murine or human sources, such as the rat PC12 cell line and the human KNA and KAT45 cell lines. Over the last few decades, more sophisticated molecular methods have allowed for induced tumorigenesis and targeted oncogenesis in vivo, where isolation of specific populations of mouse cell lines of endocrine origin have resulted in model cells to examine a variety of regulatory pathways in the chromaffin phenotype. As well, conditional immortalization with retroviral infection of chromaffin precursors has provided homogeneous and expandable chromaffin cells for transplant studies in animal models of pain. This same strategy of immortalization with conditionally expressed oncogenes has been expanded recently to create the first disimmortalizable chromaffin cells, with an excisable oncogenic cassette, as might be envisioned for the creation of human chromaffin cell lines. Eventually, as we increase our understanding of regulating the phenotypic fate of chromaffin cells in vitro, stem or progenitor adrenal medullary cell lines will be derived as an alternative source for expansion and clinical use.
Collapse
Affiliation(s)
- M J Eaton
- The Miami Project To Cure Paralysis, University of Miami School of Medicine, 1095 N.W. 14th Terrace (R-48), Miami, FL 33136, USA.
| | | |
Collapse
|
27
|
Leret ML, Peinado V, González JC, Suárez LM, Rúa C. Maternal adrenalectomy affects development of adrenal medulla. Life Sci 2004; 74:1861-7. [PMID: 14761667 DOI: 10.1016/j.lfs.2003.07.050] [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] [Received: 05/22/2003] [Accepted: 07/31/2003] [Indexed: 10/26/2022]
Abstract
This work investigates the effects of maternal adrenalectomy (ADX) on the development of the adrenal medulla. Adrenal catecholamines (AC) were measured at postnatal day (PN) 1, 8, 12 and 22 in rat offspring of ADX dams and in pups of control dams. The pups of ADX rats showed a reduction in AC concentrations in the adrenal medulla at PN 1, 12 and 22, although these were higher than in the pups of sham dams at PN 8. Further, in the pups of control mothers, there was an increase in ACs during the first two weeks of life whereas pups of ADX mothers only showed increases in noradrenaline, dopamine and adrenaline levels at day 8. These results suggest that maternal absence of corticosterone affects the medulla catecholamine content during development. These data support the idea that a maternal glucocorticoids are involved in the differentiation or/and maturation of the adrenal medulla.
Collapse
Affiliation(s)
- M L Leret
- Department of Animal Biology-II (Animal Physiology), Faculty of Biological Sciences, Complutense University of Madrid, Madrid, Spain
| | | | | | | | | |
Collapse
|
28
|
Pei Y, He X, Xie Z. Dopaminergic neuron differentiation of ventral mesencephalic progenitors regulated by developmental signals in vitro. Neuroreport 2003; 14:1567-70. [PMID: 14502077 DOI: 10.1097/00001756-200308260-00005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Extrinsic signals play an important role in the differentiation of neural progenitor cells, but little is known about the underlying mechanism. In order to investigate how extrinsic signals influence the fate switch of progenitors during development, we cultured ventral mesencephalic progenitors from E13 and E16 rats in the presence of soluble factors. Here we report that soluble factors in early developmental stages can induce the production of dopaminergic neurons. E16 may be an important developmental stage in which the responsiveness of the progenitors to the soluble factors is much more sensitive. Our results indicate that a combination of cell-intrinsic changes and extrinsic cues controls the competence of mesencephalic progenitors to produce dopaminergic neurons. Cell fate restriction may result from a series of extrinsic factors acting on a multipotent progenitor progressively.
Collapse
Affiliation(s)
- Yanxin Pei
- Department of Biological Science and Biotechnology, State Key Laboratory and Biomembrane and Membrane Biotechnology, Tsinghua University, Beijing, China
| | | | | |
Collapse
|
29
|
Murata T, Hikita K, Tsuboi M, Niwa K, Suzuki M, Kaneda N. Temperature-dependent, neurotrophic factor-elicited, neuronal differentiation in adrenal chromaffin cell line immortalized with temperature-sensitive SV40 T-antigen. J Neurochem 2003; 85:1126-38. [PMID: 12753072 DOI: 10.1046/j.1471-4159.2003.01765.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We established adrenal medullary cell lines from transgenic mice expressing an oncogene, the temperature-sensitive simian virus 40 large T-antigen, under the control of the tyrosine hydroxylase promoter. A clonal cell line, named tsAM5D, conditionally grew at a permissive temperature of 33 degrees C and exhibited the dopaminergic chromaffin cell phenotype as exemplified by the expression pattern of mRNA for catecholamine-synthesizing enzymes and secretory vesicle-associated proteins. tsAM5D cells proliferated at the permissive temperature in response to basic fibroblast growth factor (bFGF) and ciliary neurotrophic factor (CNTF). At a non-permissive temperature of 39 degrees C, bFGF and CNTF acted synergistically to differentiate tsAM5D cells into neuron-like cells. In addition, tsAM5D cells caused to differentiate by bFGF plus CNTF at 39 degrees C became dependent solely on nerve growth factor for their survival and showed markedly enhanced neurite outgrowth. In the presence of bFGF and CNTF, the morphological change induced by the temperature shift was associated with up-regulated expression of neuronal marker genes including neuron-specific enolase, growth-associated protein-43, microtubule-associated protein 2, neurofilament, and p75 neurotrophin receptor, indicating that the cells underwent neuronal differentiation. Thus, we demonstrated that tsAM5D cells could proliferate at permissive 33 degrees C, and also had the capacity to terminally differentiate into neuron-like cells in response to bFGF and CNTF when the oncogene was inactivated by shifting the temperature to non-permissive 39 degrees C. These results suggest that tsAM5D cells should be a good tool to allow a detailed study of mechanisms regulating neuronal differentiation.
Collapse
Affiliation(s)
- Tomiyasu Murata
- Department of Analytical Neurobiology, Faculty of Pharmacy, Meijo University, Tempaku, Nagoya, Japan
| | | | | | | | | | | |
Collapse
|
30
|
Payet MD, Bilodeau L, Breault L, Fournier A, Yon L, Vaudry H, Gallo-Payet N. PAC1 receptor activation by PACAP-38 mediates Ca2+ release from a cAMP-dependent pool in human fetal adrenal gland chromaffin cells. J Biol Chem 2003; 278:1663-70. [PMID: 12429744 DOI: 10.1074/jbc.m206470200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previous studies have shown that human fetal adrenal gland from 17- to 20-week-old fetuses expressed pituitary adenylate cyclase-activating polypeptide (PACAP) receptors, which were localized on chromaffin cells. The aim of the present study was to identify PACAP receptor isoforms and to determine whether PACAP can affect intracellular calcium concentration ([Ca(2+)](i)) and catecholamine secretion. Using primary cultures and specific stimulation of chromaffin cells, we demonstrate that PACAP-38 induced an increase in [Ca(2+)](i) that was blocked by PACAP (6-38), was independent of external Ca(2+), and originated from thapsigargin-insensitive internal stores. The PACAP-triggered Ca(2+) increase was not affected by inhibition of PLC beta (preincubation with U-73122) or by pretreatment of cells with Xestospongin C, indicating that the inositol 1,4,5-triphosphate-sensitive stores were not mobilized. However, forskolin (FSK), which raises cytosolic cAMP, induced an increase in Ca(2+) similar to that recorded with PACAP-38. Blockage of PKA by H-89 or (R(p))-cAMPS suppressed both PACAP-38 and FSK calcium responses. The effect of PACAP-38 was also abolished by emptying the caffeine/ryanodine-sensitive Ca(2+) stores. Furthermore, treatment of cells with orthovanadate (100 microm) impaired Ca(2+) reloading of PACAP-sensitive stores indicating that PACAP-38 can mobilize Ca(2+) from secretory vesicles. Moreover, PACAP induced catecholamine secretion by chromaffin cells. It is concluded that PACAP-38, through the PAC(1) receptor, acts as a neurotransmitter in human fetal chromaffin cells inducing catecholamine secretion, through nonclassical, recently described, ryanodine/caffeine-sensitive pools, involving a cAMP- and PKA-dependent phosphorylation mechanism.
Collapse
Affiliation(s)
- Marcel D Payet
- Department of Physiology and Biophysics, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Quebec J1H 5N4, Canada.
| | | | | | | | | | | | | |
Collapse
|
31
|
Gartner H, Graul MC, Oesterreicher TJ, Finegold MJ, Henning SJ. Development of the fetal intestine in mice lacking the glucocorticoid receptor (GR). J Cell Physiol 2003; 194:80-7. [PMID: 12447992 DOI: 10.1002/jcp.10189] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
During rodent development there are two surges of circulating corticosterone: one just prior to birth and then one in the third postnatal week. Prior studies have shown that the latter controls the rate of intestinal development in the postnatal period. To date, a role for the earlier surge in the prenatal phase of intestinal development has not been investigated. We hypothesized that the late fetal surge of circulating corticosterone is involved in both morphologic and functional maturation of the intestinal epithelium, and thus that such maturation would be delayed if glucocorticoid action was abrogated. The hypothesis was tested by studying intestinal development in mice lacking a functional glucocorticoid receptor (GR). After GR+/- mice were bred onto a C57Bl/6 background, heterozygote matings yielded the expected ratios of -/-, +/-, and +/+ offspring. Analysis of GR mRNA in intestines of +/+ and -/- fetuses confirmed expression in wild-type mice but not in the GR-null mice. Intestinal histology of GR+/+ and -/- littermates at E13.5, E15.5, and E18.5 showed no effect of GR genotype on morphologic development. Further studies at E18.5 showed that GR-/- mice have normal functional maturation of the intestinal epithelium as assessed by: lactase activity in the enterocyte lineage, normal numbers of goblet and enteroendocrine cells, and normal numbers of proliferating cells in the intestinal crypts. Neither the minerolocorticoid receptor (MR) nor the pregnane X receptor (PXR) showed compensatory up-regulation in GR-/- mice. We conclude that, in contrast to our original hypothesis, the rodent intestine passes through a phase of glucocorticoid independence (late fetal) prior to becoming responsive to glucocorticoids in the postnatal period. These findings have implications for the clinical use of corticosteroids to enhance intestinal maturation in preterm infants.
Collapse
MESH Headings
- Animals
- Cell Differentiation/genetics
- Cell Lineage/genetics
- Corticosterone/metabolism
- Female
- Fetus
- Genotype
- Goblet Cells/cytology
- Goblet Cells/metabolism
- Intestinal Mucosa/cytology
- Intestinal Mucosa/embryology
- Intestinal Mucosa/metabolism
- Intestine, Small/cytology
- Intestine, Small/embryology
- Intestine, Small/metabolism
- Ki-67 Antigen/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mutation/genetics
- Paneth Cells/cytology
- Paneth Cells/metabolism
- Pregnane X Receptor
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Glucocorticoid/deficiency
- Receptors, Glucocorticoid/genetics
- Receptors, Mineralocorticoid/genetics
- Receptors, Steroid/genetics
Collapse
Affiliation(s)
- Hans Gartner
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
32
|
Huber K, Brühl B, Guillemot F, Olson EN, Ernsberger U, Unsicker K. Development of chromaffin cells depends on MASH1 function. Development 2002; 129:4729-38. [PMID: 12361965 DOI: 10.1242/dev.129.20.4729] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The sympathoadrenal (SA) cell lineage is a derivative of the neural crest (NC), which gives rise to sympathetic neurons and neuroendocrine chromaffin cells. Signals that are important for specification of these two types of cells are largely unknown. MASH1 plays an important role for neuronal as well as catecholaminergic differentiation. Mash1 knockout mice display severe deficits in sympathetic ganglia, yet their adrenal medulla has been reported to be largely normal suggesting that MASH1 is essential for neuronal but not for neuroendocrine differentiation. We show now that MASH1 function is necessary for the development of the vast majority of chromaffin cells. Most adrenal medullary cells in Mash1–/– mice identified by Phox2b immunoreactivity, lack the catecholaminergic marker tyrosine hydroxylase. Mash1 mutant and wild-type mice have almost identical numbers of Phox2b-positive cells in their adrenal glands at embryonic day (E) 13.5; however, only one-third of the Phox2b-positive adrenal cell population seen in Mash1+/+ mice is maintained in Mash1–/– mice at birth. Similar to Phox2b, cells expressing Phox2a and Hand2 (dHand) clearly outnumber TH-positive cells. Most cells in the adrenal medulla of Mash1–/– mice do not contain chromaffin granules, display a very immature, neuroblast-like phenotype, and, unlike wild-type adrenal chromaffin cells, show prolonged expression of neurofilament and Ret comparable with that observed in wild-type sympathetic ganglia. However, few chromaffin cells in Mash1–/– mice become PNMT positive and downregulate neurofilament and Ret expression. Together, these findings suggest that the development of chomaffin cells does depend on MASH1 function not only for catecholaminergic differentiation but also for general chromaffin cell differentiation.
Collapse
Affiliation(s)
- Katrin Huber
- Neuroanatomy, Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, INF 307, D-69120 Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
33
|
Tai TC, Claycomb R, Her S, Bloom AK, Wong DL. Glucocorticoid responsiveness of the rat phenylethanolamine N-methyltransferase gene. Mol Pharmacol 2002; 61:1385-92. [PMID: 12021400 DOI: 10.1124/mol.61.6.1385] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Two newly identified, overlapping (1 bp) glucocorticoid response elements (GREs) at -759 and -773 bp in the promoter of the rat phenylethanolamine N-methyltransferase (PNMT; EC 2.1.1.28) gene are primarily responsible for its glucocorticoid sensitivity, rather than the originally identified -533-bp GRE. A dose-dependent increase in PNMT promoter activity was observed in RS1 cells transfected with a wild-type PNMT promoter-luciferase reporter gene construct and treated with dexamethasone (maximum activation at 0.1 microM). The type II glucocorticoid receptor antagonist RU38486 (10 microM) fully inhibited dexamethasone (1 microM) activation of the PNMT promoter, consistent with classical glucocorticoid receptors mediating corticosteroid-stimulated transcriptional activity. Relative IC(50) values from gel mobility shift competition assays showed that the -759-bp GRE has a 2-fold greater affinity for the glucocorticoid receptor than the -773-bp GRE. Site-directed mutation of the -533-, -759-, and -773-bp GREs alone or in tandem demonstrated that the -759-bp GRE was also functionally more important, but both the -759- and -773-bp GREs are required for maximum glucocorticoid responses. Moreover, the -533-bp GRE, rather than increasing glucocorticoid sensitivity of the promoter, may limit corticosteroid responsiveness mediated via the -759- and -773-bp GREs. Finally, the glucocorticoid receptor bound to the -759- and -773-bp GREs interacts cooperatively with Egr-1 and/or AP-2 to stimulate PNMT promoter activity in RS1 cells treated with dexamethasone. In contrast, glucocorticoid receptors bound to the -533-bp GRE only seem to participate in synergistic activation of the PNMT promoter through interaction with activator protein 2.
Collapse
Affiliation(s)
- T C Tai
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | |
Collapse
|
34
|
Zammit DJ, Berzins SP, Gill JW, Randle-Barrett ES, Barnett L, Koentgen F, Lambert GW, Harvey RP, Boyd RL, Classon BJ. Essential role for the lymphostromal plasma membrane Ly-6 superfamily molecule thymic shared antigen 1 in development of the embryonic adrenal gland. Mol Cell Biol 2002; 22:946-52. [PMID: 11784869 PMCID: PMC133557 DOI: 10.1128/mcb.22.3.946-952.2002] [Citation(s) in RCA: 14] [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
Thymic shared antigen 1 (TSA-1) is a plasma membrane protein of the Ly-6 superfamily expressed on thymocytes, thymic stromal cells, and other cells of the hematopoietic system. TSA-1 is also expressed in other nonhematopoietic tissues, in particular, embryonic and adult adrenal glands. To address the function of TSA-1, we generated mutant mice in which TSA-1 expression was inactivated by gene targeting. Here we show that deletion of both TSA-1 alleles results in abnormal adrenal gland development and midgestational lethality due to cardiac abnormalities. We also report that TSA-1-deficient adrenal glands have significantly reduced levels of the catecholamines noradrenaline and adrenaline. We conclude that TSA-1 is required for normal embryonic development but that deletion of its expression does not obviously impair lymphoid development.
Collapse
Affiliation(s)
- David J Zammit
- Department of Pathology and Immunology, Monash Medical School, Prahran, Victoria, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Young JB, Landsberg L. Synthesis, Storage, and Secretion of Adrenal Medullary Hormones: Physiology and Pathophysiology. Compr Physiol 2001. [DOI: 10.1002/cphy.cp070401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
36
|
Abstract
Glucocorticoid receptor activation in the fetal lung triggers maturation necessary for extra-uterine life. Antenatal treatment with betamethasone and dexamethasone has lowered severity of respiratory distress in very low birth weight infants, and dexamethasone given postnatally has resulted in short-term improvement in chronic lung disease. Recently, however, surfactant therapy has diminished the differential benefit of antenatal glucocorticoid treatment, and it has been difficult to show that postnatal dexamethasone therapy improves survival. Treated infants may have reduced weight gain, adrenal suppression, increased incidence of intestinal perforation and infection, and long-term developmental and metabolic problems. Recent data suggest that the fetal hypothalamic/pituitary/adrenal axis is active early and is precisely structured for an intricate sequence of specifically fetal developmental events, which may be deranged by dexamethasone therapy. We consider data suggesting that persistence of the fetal pattern in some premature infants constitutes adrenal insufficiency, and that therapy at stress replacement doses with less potent glucocorticoids might avoid side effects seen with traditional regimens.
Collapse
Affiliation(s)
- P G Brosnan
- Department of Pediatrics, School of Medicine University of Texas Health Science Center, Houston 77030, USA.
| |
Collapse
|
37
|
Combs SE, Ernsberger U, Krieglstein K, Unsicker K. Reduction of endogenous TGF-beta does not affect phenotypic development of sympathoadrenal progenitors into adrenal chromaffin cells. Mech Dev 2001; 109:295-302. [PMID: 11731241 DOI: 10.1016/s0925-4773(01)00553-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Adrenal chromaffin cells and sympathetic neurons are related, but phenotypically distinct derivatives of the neural crest. Molecular cues that determine the chromaffin cell phenotype have not yet been identified; in contrast to a widely held belief, glucocorticoid signaling is apparently not relevant (Development 126 (1999) 2935). Transforming growth factor-betas (TGF-betas) regulate various aspects of embryonic development and are expressed in the environment of sympathoadrenal (SA) progenitor cells. We have previously shown that neutralization of endogenous TGF-beta from E4 to E8 in the quail embryo significantly increases numbers of adrenal tyrosine hydroxylase-positive cells. Whether endogenous TGF-beta may also be involved in influencing phenotypic development of adrenal chromaffin cells and their SA progenitors has not been analyzed. We now demonstrate that neutralization of endogenous TGF-beta1, -beta2 and -beta3 with a pan-anti-TGF-beta antibody in quail embryos during distinct time windows does not alter phenotypic development of chromaffin cells. In situ hybridizations revealed unaltered expression of neurofilament (NF-160), synaptotagmin I and neurexin I in adrenal glands. Likewise, the NF-associated antigen 3A10, and polyphosphorylated NF epitopes (RT 97) were unaltered. Most importantly, the typical ultrastructure of adrenal chromaffin cells including their large chromaffin secretory granules, a hallmark of the neuroendocrine phenotype, which distinguishes them from sympathetic neurons, was not affected. We therefore conclude that neutralization of endogenous TGF-beta influences chromaffin cell proliferation, but does not interfere with the development of the typical chromaffin cell phenotype.
Collapse
Affiliation(s)
- S E Combs
- Neuroanatomy and Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 307, 69120, Heidelberg, Germany
| | | | | | | |
Collapse
|
38
|
Schober A, Krieglstein K, Unsicker K. Molecular cues for the development of adrenal chromaffin cells and their preganglionic innervation. Eur J Clin Invest 2000; 30 Suppl 3:87-90. [PMID: 11281376 DOI: 10.1046/j.1365-2362.2000.0300s3087.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Based on recent evidence from in vitro and gene knockout/insertion studies, this short review summarizes the molecular scenario underlying the development of adrenal chromaffin cells and their preganglionic innervation. During migration of neural crest cells from the dorsal surface of the neural tube to their destinations in the sympathetic primordia and adrenal glands, precursors of the so-called sympathoadrenal (SA) cell lineage are exposed to signals from the notochord and ventral neural tube probably including the protein, Sonic hedgehog. These, and signals in the region of the dorsal aorta (members of the family of bone morphogentic proteins), where SA progenitor cells subsequently assemble, are essential for the induction of the adrenergic phenotype. SA progenitor cells subsequently differentiate into paravertebral and prevertebral sympathetic neurones, intra- and extra-adrenal chromaffin cells and intermediate SIF (small intensely fluorescent) cells. Based on in vitro studies with isolated SA and chromaffin progenitor cells, glucocortiocids have been claimed as essential for suppressing neuronal commitment and for channelling SA cells towards the chromaffin phenotype. However, mice deficient for a functional glucocorticoid receptor possess the full complement of adrenal chromaffin cells at birth, suggesting that signals other than glucocorticoid hormones may be important in triggering chromaffin cell differentiation. The cholinergic neurones that are preganglionic to adrenal chromaffin cells have their cell bodies located in the intermediolateral column (IML) of the spinal cord. For their normal development, these neurones require signals from the adrenal medulla, which include neurotrophin-4, a major neurotrophic factor of adrenal chromaffin cells. Taken together, these data provide a more complete picture of molecular signalling in the development of one of the most important neuroendocrine tissues in vertebrates.
Collapse
Affiliation(s)
- A Schober
- Neuroanatomy, University of Heidelberg, Germany.
| | | | | |
Collapse
|
39
|
Kennedy B, Ziegler MG. Ontogeny of epinephrine metabolic pathways in the rat: role of glucocorticoids. Int J Dev Neurosci 2000; 18:53-9. [PMID: 10708906 DOI: 10.1016/s0736-5748(99)00106-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Recent studies suggest that the initial expression of adrenal phenylethanolamine N-methyltransferase (PNMT) and epinephrine (E) are dependent upon stimulation of adrenal glucocorticoid receptors. However, evidence suggests that the expression of heart and brain PNMT is independent of glucocorticoids. We measured PNMT activity and E levels in adrenal, heart and head over the latter half of gestation in rat fetuses treated chronically with glucocorticoids, and in normal controls. Chronic glucocorticoid treatment ending on embryonic day (e)12 did not affect heart, head or trunk PNMT activity or E levels. In contrast, chronic glucocorticoid exposure ending e19 or e20 resulted in marked increases in both PNMT and E in adrenal, heart and head tissues. The elevation of E in all three tissues was unaffected by maternal adrenalectomy, indicating enhanced fetal E synthesis. In the absence of exogenous glucocorticoid treatment heart PNMT activity peaked on e12, prior to the earliest reported appearance of glucocorticoid receptors. We conclude that expression of PNMT in all three tissues is glucocorticoid independent until the latter part of gestation when it is readily enhanced by glucocorticoids.
Collapse
Affiliation(s)
- B Kennedy
- Department of Medicine, University of California San Diego, Medical Center, 200 West Arbor Drive, 8341, San Diego, CA, USA.
| | | |
Collapse
|
40
|
Eaton MJ, Frydel BR, Lopez TL, Nie XT, Huang J, McKillop J, Sagen J. Generation and initial characterization of conditionally immortalized chromaffin cells. J Cell Biochem 2000. [DOI: 10.1002/1097-4644(2000)79:1<38::aid-jcb50>3.0.co;2-u] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
41
|
Finotto S, Krieglstein K, Schober A, Deimling F, Lindner K, Brühl B, Beier K, Metz J, Garcia-Arraras JE, Roig-Lopez JL, Monaghan P, Schmid W, Cole TJ, Kellendonk C, Tronche F, Schütz G, Unsicker K. Analysis of mice carrying targeted mutations of the glucocorticoid receptor gene argues against an essential role of glucocorticoid signalling for generating adrenal chromaffin cells. Development 1999; 126:2935-44. [PMID: 10357937 DOI: 10.1242/dev.126.13.2935] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Molecular mechanisms underlying the generation of distinct cell phenotypes is a key issue in developmental biology. A major paradigm of determination of neural cell fate concerns the development of sympathetic neurones and neuroendocrine chromaffin cells from a common sympathoadrenal (SA) progenitor cell. Two decades of in vitro experiments have suggested an essential role of glucocorticoid receptor (GR)-mediated signalling in generating chromaffin cells. Targeted mutation of the GR should consequently abolish chromaffin cells. The present analysis of mice lacking GR gene product demonstrates that animals have normal numbers of adrenal chromaffin cells. Moreover, there are no differences in terms of apoptosis and proliferation or in expression of several markers (e.g. GAP43, acetylcholinesterase, adhesion molecule L1) of chromaffin cells in GR-deficient and wild-type mice. However, GR mutant mice lack the adrenaline-synthesizing enzyme PNMT and secretogranin II. Chromaffin cells of GR-deficient mice exhibit the typical ultrastructural features of this cell phenotype, including the large chromaffin granules that distinguish them from sympathetic neurones. Peripherin, an intermediate filament of sympathetic neurones, is undetectable in chromaffin cells of GR mutants. Finally, when stimulated with nerve growth factor in vitro, identical proportions of chromaffin cells from GR-deficient and wild-type mice extend neuritic processes. We conclude that important phenotypic features of chromaffin cells that distinguish them from sympathetic neurones develop normally in the absence of GR-mediated signalling. Most importantly, chromaffin cells in GR-deficient mice do not convert to a neuronal phenotype. These data strongly suggest that the dogma of an essential role of glucocorticoid signalling for the development of chromaffin cells must be abandoned.
Collapse
Affiliation(s)
- S Finotto
- Neuroanatomy, University of Heidelberg, D-69120 Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
The glucocorticoid receptor is an ubiquitously expressed transcription factor involved in the regulation of many different physiological processes. Activated by glucocorticoids the receptor regulates transcription positively or negatively either by direct binding to DNA or by protein protein interactions. In order to define the role of the receptor during development and in physiology several mutations have been generated in the mouse. Mice with a disrupted glucocorticoid receptor gene die shortly after birth due to respiratory failure indicating an important role of the receptor in lung function. Transcription of genes encoding gluconeogenic enzymes in the liver is decreased, proliferation of erythroid progenitors is impaired and the HPA axis is strongly upregulated. To analyze molecular mechanisms of glucocorticoid receptor action in vivo a point mutation has been introduced into the mouse genome which allows to separate DNA-binding-dependent from DNA-binding-independent actions of the receptor. Mice homozygous for the point mutation survive indicating that DNA-binding of the receptor is not required for survival. Induction of glucoconegenic enzymes and proliferation of erythroid progenitors however is impaired. Interestingly, repression of corticotropin releasing factor (CRF) synthesis is maintained, whereas proopiomelanocortin (POMC) expression is upregulated. Since mice with a disrupted glucocorticoid receptor gene die shortly after birth attempts using the Cre/loxP-recombination system are made to bypass early lethality and to study the function of the receptor in defined cell types of adult animals.
Collapse
Affiliation(s)
- C Kellendonk
- Molecular Biology of the Cell I, German Cancer Research Center, Heidelberg
| | | | | | | |
Collapse
|
43
|
Bauer R, Weiss C, Marksteiner J, Doblinger A, Fischer-Colbrie R, Laslop A. The new chromogranin-like protein NESP55 is preferentially localized in adrenaline-synthesizing cells of the bovine and rat adrenal medulla. Neurosci Lett 1999; 263:13-6. [PMID: 10218899 DOI: 10.1016/s0304-3940(99)00091-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The protein NESP55, a new member of the chromogranin family, is present in large dense-core secretory granules of neuroendocrine tissues. We investigated its cellular distribution in adrenal medulla with immunohistochemistry and in situ hybridization. A preferential co-localization of NESP55 with phenylethanolamine-N-methyltransferase in the adrenergic cell population was found by immunolabelling of consecutive sections. Noradrenergic cells also contained small amounts of NESP55, but the levels as measured by radioimmunoassay were five times lower. The distribution of NESP55 mRNA was similar to preproenkephalin mRNA which previously was shown to be confined to adrenaline-producing cells of the adrenal medulla. The present study indicates that stimulation of adrenergic cells will release significantly higher amounts of NESP55. The functional implications of this preferential secretion, however, have yet to be discovered.
Collapse
Affiliation(s)
- R Bauer
- Department of Pharmacology, University of Innsbruck, Austria
| | | | | | | | | | | |
Collapse
|
44
|
Unsworth BR, Hayman GT, Carroll A, Lelkes PI. Tissue-specific alternative mRNA splicing of phenylethanolamine N-methyltransferase (PNMT) during development by intron retention. Int J Dev Neurosci 1999; 17:45-55. [PMID: 10219960 DOI: 10.1016/s0736-5748(98)00058-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
The expression of phenylethanolamine N-methyl transferase (EC 2. 1.1.2.8, PNMT), the final enzyme in the cascade of catecholamine synthesis, is differentially regulated in adrenergic neurons in the brain and in adrenal chromaffin cells. Using reverse transcription-polymerase chain reaction-based techniques, we detected in the prenatal developing rat brainstem, two species of PNMT mRNA which were produced by a rare alternative splicing mechanism known as intron retention. The spliced, intronless message was downregulated postnatally, while the intron-retained mRNA species continued to be constitutively expressed through adulthood. By contrast in the adrenals, at all stages of development examined, only the intronless message was expressed. In line with previous reports on the failure of glucocorticoids to induce PNMT expression in the brain, the pattern of PNMT splicing in brainstem explants was not affected by the presence of the synthetic glucocorticoid dexamethasone. Undifferentiated sympathoadrenal PC12 pheochromocytoma cells expressed very low basal levels of both mRNA variants, accompanied by a very low basal PNMT enzymatic activity. Exposure of PC12 cells to dexamethasone resulted in the upregulation of only the spliced mRNA variant concomitant with a 3-fold increase in PNMT enzymatic activity. In contrast, treatment of PC 12 cells with nerve growth factor (NGF) enhanced the expression of both the intron-retained and the intronless mRNA species without changes in the basal enzyme activity. This latter result suggests that the translation of the intronless mRNA species may be regulated by the intron-retained mRNA species, which by itself may yield a truncated, yet enzymatically functional translational product. Our data suggest that the tissue-specific regulation of PNMT expression is based on a rare alternative splicing mechanism termed intron retention, and that in the adrenal, but not in the brain, this mechanism is sensitive to regulation by glucocorticoids. Thus, this system is uniquely suited for studying the hormonal control of tissue-specific splicing in the nervous system.
Collapse
Affiliation(s)
- B R Unsworth
- Department of Biology, Marquette University, Milwaukee, WI 53233, USA
| | | | | | | |
Collapse
|
45
|
Edlund T, Jessell TM. Progression from extrinsic to intrinsic signaling in cell fate specification: a view from the nervous system. Cell 1999; 96:211-24. [PMID: 9988216 DOI: 10.1016/s0092-8674(00)80561-9] [Citation(s) in RCA: 382] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- T Edlund
- Department of Microbiology, University of Umea, Sweden.
| | | |
Collapse
|
46
|
Affiliation(s)
- A K Groves
- Division of Biology, California Institute of Technology, Pasadena 91125, USA
| | | |
Collapse
|
47
|
Tronche F, Kellendonk C, Reichardt HM, Schütz G. Genetic dissection of glucocorticoid receptor function in mice. Curr Opin Genet Dev 1998; 8:532-8. [PMID: 9794823 DOI: 10.1016/s0959-437x(98)80007-5] [Citation(s) in RCA: 135] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Upon hormone binding, the activated glucocorticoid receptor (GR) functions as a transcription factor via different modes of action to control gene expression. Recent gene-targeting studies in mice provide new insight into the role of GR in vivo and are helping decipher the molecular mechanisms underlying its actions.
Collapse
Affiliation(s)
- F Tronche
- Molecular Biology of the Cell I, Deutsches Krebsforschungzentrum, Im Neuenheimer Feld 280, 69120, Heidelberg, Federal Republic of Germany
| | | | | | | |
Collapse
|
48
|
Manojlivić M, Hristić M, Kalafatić D, Plećas B, Ugresić N. The influence of dexamethasone treatment of pregnant rats on the development of chromaffin tissue in their offspring during the fetal and neonatal period. J Endocrinol Invest 1998; 21:211-8. [PMID: 9624594 DOI: 10.1007/bf03347305] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The aim of these examinations was to determine the influence of dexamethasone (Dx)-treatment of gravid females, on day 16 of gestation on the development of medullary chromaffin tissue of their fetuses and neonatal offspring. In conducting these investigations we used stereological as well as spectrofluorimetric measurements, in 20-day-old fetuses and 1-, 3-, 5-, 7-, 9-, 11-, 13- and 14-day-old neonatal rats. Single Dx-treatment (1.5 mg/kg bw) of the dams led to a significant decrease in body and adrenal weight of their fetuses and neonatal offspring, and also reduction of the medullary volume and the number of chromaffin cells during the entire period examined as a result of decreased cell proliferation in the fetal and early neonatal period (till the 5th day of age). The proliferative activity of the chromaffin cells was evaluated through the mitotic index after applying the cytostatic vincristine-sulphate. During the second neonatal week the mitotic index showed significantly higher values in comparison with the corresponding controls, which indicates that there is regeneration and recovery of the adrenal gland medulla. Adrenaline content in the adrenal gland tissue of offspring of Dx-treated dams was significantly reduced only on the 1st neonatal day. Thus, the change in blood glucocorticoid level of pregnant females after a single Dx injection during the period critical for development of the hypothalamo-pituitary-adrenal system in fetuses affects the development and kinetics of medullar chromaffin cell division.
Collapse
Affiliation(s)
- M Manojlivić
- Department of Cytology, Institute for Biological Research, Belgrade, Yugoslavia
| | | | | | | | | |
Collapse
|
49
|
Lillien L, Wancio D. Changes in Epidermal Growth Factor Receptor Expression and Competence to Generate Glia Regulate Timing and Choice of Differentiation in the Retina. Mol Cell Neurosci 1998; 10:296-308. [PMID: 9618220 DOI: 10.1006/mcne.1997.0659] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previous studies demonstrated that the level of epidermal growth factor receptors (EGF-Rs) expressed by progenitor cells in the newborn (P0) rat retina was limiting for the generation of Muller glial cells but not for proliferation. To determine whether EGF-R signaling biases cells to generate a specific cell type or regulates more general processes during progenitor cell development, we have introduced extra copies of the EGF-R into progenitor cells at earlier stages (E15 and E18), when different cell types are produced. We show that progenitor cells in early embryonic retina (E15) normally express lower levels of EGF-Rs than progenitor cells in later retina (E18 and P0). Whereas lower levels of stimulation of endogenous and virally transduced EGF-Rs enhanced proliferation, higher levels reduced proliferation, resulting in premature differentiation. At E15, very few EGF-R-infected progenitor cells differentiated prematurely into Muller glial cells, unlike E18 and P0 cells, even when they were exposed to an older retinal environment. Higher levels of EGF-R-mediated signaling alone therefore do not specify a glial fate, indicating that competence to generate glia is temporally regulated by additional mechanisms. The differences in EGF-R expression observed among retinal progenitor cells at distinct developmental stages may instead help to define signaling thresholds which delay or accelerate their differentiation. Copyright 1998 Academic Press.
Collapse
Affiliation(s)
- L Lillien
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261
| | | |
Collapse
|
50
|
Unsicker K, Finotto S, Krieglstein K. Generation of cell diversity in the peripheral autonomic nervous system: the sympathoadrenal cell lineage revisited. Ann Anat 1997; 179:495-500. [PMID: 9442255 DOI: 10.1016/s0940-9602(97)80002-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Based on recent evidence from in vitro and gene knock-out/knock-in studies this short review summarizes the molecular scenario underlying the development of autonomic neurons from the neural crest. The focus is on the sympathoadrenal (SA) cell lineage. While migrating ventrally precursors of this cell lineage are exposed to signals from notochord/ventral neural tube probably including the protein sonic hedgehog. These and signals in the region of the dorsal aorta (members of the family of bone morphogenetic proteins), where SA progenitor cells subsequently assemble, are essential for the induction of the adrenergic phenotype. SA progenitor cells subsequently differentiate into paravertebral and prevertebral sympathetic neurons, intra- and extra-adrenal chromaffin cells and intermediate SIF (small intensely fluorescent) cells. Based on in vitro studies with isolated SA and chromaffin progenitor cells glucocorticoids have been claimed as essential for suppressing a neuronal commitment and channeling SA cells towards the chromaffin phenotype. Unexpectedly, mice deficient for a functional glucocorticoid receptor possess the full complement of adrenal chromaffin cells at birth. We present a hypothetical scenario consistent with these data, in which chromaffin cell development would be the default pathway in the SA cell lineage, while development into a neuronal direction requires specific growth factor signaling, which is probably distinct for paravertebral and prevertebral sympathetic neurons.
Collapse
Affiliation(s)
- K Unsicker
- Department of Anatomy and Cell Biology (Neuroanatomy), University of Heidelberg, Germany
| | | | | |
Collapse
|