1
|
Farnham MMJ, Tallapragada VJ, O'Connor ET, Nedoboy PE, Dempsey B, Mohammed S, Fong AY, Lung MSY, Derakhshan F, Wilson RJA, Pilowsky PM. PACAP-PAC1 Receptor Activation Is Necessary for the Sympathetic Response to Acute Intermittent Hypoxia. Front Neurosci 2019; 13:881. [PMID: 31496933 PMCID: PMC6712064 DOI: 10.3389/fnins.2019.00881] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 08/05/2019] [Indexed: 01/29/2023] Open
Abstract
Repetitive hypoxia is a key feature of obstructive sleep apnoea (OSA), a condition characterized by intermittent airways obstruction. Patients with OSA present with persistent increases in sympathetic activity and commonly develop hypertension. The objectives of this study were to determine if the persistent increases in sympathetic nerve activity, known to be induced by acute intermittent hypoxia (AIH), are mediated through activation of the pituitary adenylate cyclase activating polypeptide (PACAP) signaling system. Here, we show that the excitatory neuropeptide PACAP, acting in the spinal cord, is important for generating the sympathetic response seen following AIH. Using PACAP receptor knockout mice, and pharmacological agents in Sprague Dawley rats, we measured blood pressure, heart rate, pH, PaCO2, and splanchnic sympathetic nerve activity, under anaesthesia, to demonstrate that the sympathetic response to AIH is mediated via the PAC1 receptor, in a cAMP-dependent manner. We also report that both intermittent microinjection of glutamate into the rostroventrolateral medulla (RVLM) and intermittent infusion of a sub-threshold dose of PACAP into the subarachnoid space can mimic the sympathetic response to AIH. All the sympathetic responses are independent of blood pressure, pH or PaCO2 changes. Our results show that in AIH, PACAP signaling in the spinal cord helps drive persistent increases in sympathetic nerve activity. This mechanism may be a precursor to the development of hypertension in conditions of chronic intermittent hypoxia, such as OSA.
Collapse
Affiliation(s)
- Melissa M J Farnham
- The Heart Research Institute, Newtown, NSW, Australia.,Faculty of Medicine, Macquarie University, North Ryde, NSW, Australia.,Department of Physiology, Faculty of Medicine, University of Sydney, Sydney, NSW, Australia
| | | | - Edward T O'Connor
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Polina E Nedoboy
- The Heart Research Institute, Newtown, NSW, Australia.,Department of Physiology, Faculty of Medicine, University of Sydney, Sydney, NSW, Australia
| | - Bowen Dempsey
- Faculty of Medicine, Macquarie University, North Ryde, NSW, Australia
| | - Suja Mohammed
- The Heart Research Institute, Newtown, NSW, Australia.,Faculty of Medicine, Macquarie University, North Ryde, NSW, Australia.,Department of Physiology, Faculty of Medicine, University of Sydney, Sydney, NSW, Australia
| | - Angelina Y Fong
- Faculty of Medicine, Macquarie University, North Ryde, NSW, Australia.,Department of Physiology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Mandy S Y Lung
- Faculty of Medicine, Macquarie University, North Ryde, NSW, Australia
| | - Fatemeh Derakhshan
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Richard J A Wilson
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Paul M Pilowsky
- The Heart Research Institute, Newtown, NSW, Australia.,Department of Physiology, Faculty of Medicine, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
2
|
Stroth N, Kuri BA, Mustafa T, Chan SA, Smith CB, Eiden LE. PACAP controls adrenomedullary catecholamine secretion and expression of catecholamine biosynthetic enzymes at high splanchnic nerve firing rates characteristic of stress transduction in male mice. Endocrinology 2013; 154:330-9. [PMID: 23221599 PMCID: PMC3529367 DOI: 10.1210/en.2012-1829] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The neuropeptide PACAP (pituitary adenylate cyclase-activating polypeptide) is a cotransmitter of acetylcholine at the adrenomedullary synapse, where autonomic regulation of hormone secretion occurs. We have previously reported that survival of prolonged metabolic stress in mice requires PACAP-dependent biosynthesis and secretion of adrenomedullary catecholamines (CAs). In the present experiments, we show that CA secretion evoked by direct high-frequency stimulation of the splanchnic nerve is abolished in native adrenal slices from male PACAP-deficient mice. Further, we demonstrate that PACAP is both necessary and sufficient for CA secretion ex vivo during stimulation protocols designed to mimic stress. In vivo, up-regulation of transcripts encoding adrenomedullary CA-synthesizing enzymes (tyrosine hydroxylase, phenylethanolamine N-methyltransferase) in response to both psychogenic and metabolic stressors (restraint and hypoglycemia) is PACAP-dependent. Stressor-induced alteration of the adrenomedullary secretory cocktail also appears to require PACAP, because up-regulation of galanin mRNA is abrogated in male PACAP-deficient mice. We further show that hypoglycemia-induced corticosterone secretion is not PACAP-dependent, ruling out the possibility that glucocorticoids are the main mediators of the aforementioned effects. Instead, experiments with bovine chromaffin cells suggest that PACAP acts directly at the level of the adrenal medulla. By integrating prolonged CA secretion, expression of biosynthetic enzymes and production of modulatory neuropeptides such as galanin, PACAP is crucial for adrenomedullary function. Importantly, our results show that PACAP is the dominant adrenomedullary neurotransmitter during conditions of enhanced secretory demand.
Collapse
Affiliation(s)
- N Stroth
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
3
|
Arata S, Nakamachi T, Onimaru H, Hashimoto H, Shioda S. Impaired response to hypoxia in the respiratory center is a major cause of neonatal death of the PACAP-knockout mouse. Eur J Neurosci 2012; 37:407-16. [PMID: 23136967 DOI: 10.1111/ejn.12054] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 10/09/2012] [Accepted: 10/10/2012] [Indexed: 01/19/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide expressed widely in nervous tissues. PACAP-knockout ((-/-)) mice display a sudden infant death syndrome (SIDS)-like phenotype, although the underlying physiological mechanism to explain this remains unclear. Here, we report on the presence of abnormal respiratory activity in PACAP(-/-) mice under hypoxic conditions, which provides a basis for the SIDS-like phenotype. PACAP(-/-) mice display a lowered baseline respiratory activity compared with wild-type animals, and an abnormal response to hypoxia. More specifically, PACAP(-/-) mice at postnatal day 7 showed respiratory arrest in response to hypoxia. In contrast, their response to hypercapnic conditions was the same as that of wild-type mice. Histological and real-time PCR analyses indicated that the catecholaminergic system in the medulla oblongata was impaired in PACAP(-/-) mice, suggesting that endogenous PACAP affects respiratory centers in the medulla oblongata via its action on the catecholaminergic system. We propose that disruption of this system is involved in the SIDS-like phenotype of PACAP(-/-) mice. Thus, disorders of the catecholaminergic system involved with O(2) sensing could be implicated in underlying neuronal mechanisms responsible for SIDS.
Collapse
Affiliation(s)
- Satoru Arata
- Center for Biotechnology, Showa University, Shinagawa-ku, Tokyo, Japan
| | | | | | | | | |
Collapse
|
4
|
Role of reactive oxygen species in the neural and hormonal regulation of the PNMT gene in PC12 cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2011; 2011:756938. [PMID: 22007271 PMCID: PMC3189585 DOI: 10.1155/2011/756938] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 07/12/2011] [Indexed: 01/10/2023]
Abstract
The stress hormone, epinephrine, is produced predominantly by adrenal chromaffin cells and its biosynthesis is regulated by the enzyme phenylethanolamine N-methyltransferase (PNMT). Studies have demonstrated that PNMT may be regulated hormonally via the hypothalamic-pituitary-adrenal axis and neurally via the stimulation of the splanchnic nerve. Additionally, hypoxia has been shown to play a key role in the regulation of PNMT. The purpose of this study was to examine the impact of reactive oxygen species (ROS) produced by the hypoxia mimetic agent CoCl2, on the hormonal and neural stimulation of PNMT in an in vitro cell culture model, utilizing the rat pheochromocytoma (PC12) cell line. RT-PCR analyses show inductions of the PNMT intron-retaining and intronless mRNA splice variants by CoCl2 (3.0- and 1.76-fold, respectively). Transient transfection assays of cells treated simultaneously with CoCl2 and the synthetic glucocorticoid, dexamethasone, show increased promoter activity (18.5-fold), while mRNA levels of both splice variants do not demonstrate synergistic effects. Similar results were observed when investigating the effects of CoCl2-induced ROS on the neural stimulation of PNMT via forskolin. Our findings demonstrate that CoCl2-induced ROS have synergistic effects on hormonal and neural activation of the PNMT promoter.
Collapse
|
5
|
Mahata M, Zhang K, Gayen JR, Nandi S, Brar BK, Ghosh S, Mahapatra NR, Taupenot L, O'Connor DT, Mahata SK. Catecholamine biosynthesis and secretion: physiological and pharmacological effects of secretin. Cell Tissue Res 2011; 345:87-102. [PMID: 21597914 PMCID: PMC10843894 DOI: 10.1007/s00441-011-1177-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Accepted: 04/15/2011] [Indexed: 11/25/2022]
Abstract
Pituitary adenylyl cyclase activating polypeptide (PACAP) and vasoactive intestinal polypeptide (VIP) augment the biosynthesis of tyrosine hydroxylase (TH). We tested whether secretin belonging to the glucagon/PACAP/VIP superfamily would increase transcription of the tyrosine hydroxylase (Th) gene and modulate catecholamine secretion. Secretin activated transcription of the endogenous Th gene and its transfected promoter (EC(50) ∼4.6 nM) in pheochromocytoma (PC12) cells. This was abolished by pre-treatment with a secretin receptor (SCTR) antagonist and by inhibition of protein kinase A (PKA), mitogen-activated protein kinase, or CREB (cAMP response element-binding protein). In agreement, secretin increased PKA activity and induced phosphorylation of CREB and binding to Th CRE, suggesting secretin signaling to transcription via a PKA-CREB pathway. Secretin stimulated catecholamine secretion (EC(50) ∼3.5 μM) from PC12 cells, but this was inhibited by pre-treatment with VIP-preferring receptor (VPAC1)/PACAP-preferring receptor (PAC1) antagonists. Secretin-evoked secretion occurred without extracellular Ca(2+) and was abolished by intracellular Ca(2+) chelation. Secretin augmented phospholipase C (PLC) activity and increased inositol-1,4,5-triphosphate (IP(3)) levels in PC12 cells; PLC-β inhibition blocked secretin-induced catecholamine secretion, indicating the participation of intracellular Ca(2+) from a phospholipase pathway in secretion. Like PACAP, secretin evoked long-lasting catecholamine secretion, even after only a transient exposure. Thus, transcription is triggered by nanomolar concentrations of the peptide through SCTR, with signaling along the cAMP-PKA and extracellular-signal-regulated kinase 1/2 pathways and through CREB. By contrast, secretion is triggered only by micromolar concentrations of peptide through PAC1/VPAC receptors and by utilizing a PLC/intracellular Ca(2+) pathway.
Collapse
Affiliation(s)
- Manjula Mahata
- Department of Medicine, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0838, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Human dopamine β-hydroxylase promoter variant alters transcription in chromaffin cells, enzyme secretion, and blood pressure. Am J Hypertens 2011; 24:24-32. [PMID: 20814407 DOI: 10.1038/ajh.2010.186] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Dopamine β-hydroxylase (DBH) plays an indispensable role in catecholamine synthesis by converting dopamine into norepinephrine. Here, we characterized a DBH promoter polymorphism (C-2073T; rs1989787; minor allele frequency ~16%) that influences not only gene transcription but also enzyme secretion and blood pressure (BP) in vivo. METHODS Plasma DBH activity was measured spectrophotometrically. DBH genetic effects on BP were tested in subjects with the most extreme BP values in a large primary care population. Functional effects of promoter variants were studied by site-directed mutagenesis in DBH promoter haplotype/luciferase reporter plasmids transfected into chromaffin cells. Sequence motifs were predicted from position weight matrices, and endogenous transcription factor binding was probed by Chromatin ImmunoPrecipitation (ChIP). RESULTS The T-allele of common promoter variant C-2073T was contained in a promoter haplotype that associated with plasma DBH activity, a trait also predicted by that variant itself. Promoter haplotypes including C-2073T predicted BP in the population, and the effect was also referable to C-2073T itself. Computationally, C-2073 disrupted a predicted match for transcription factor c-FOS. Site-directed mutagenesis at C-2073T altered not only basal promoter activity, but also transactivation by c-FOS, as well as the chromaffin cell secretory stimuli nicotine or pituitary adenylate cyclase-activating polypeptide (PACAP). Endogenous c-FOS bound to the motif in chromatin. CONCLUSIONS These results suggest that DBH promoter variant C-2073T is functional in vivo: this promoter variant seems to initiate a cascade of transcriptional and biochemical changes including augmented DBH secretion, eventuating in elevation of basal BP, and hence cardiovascular risk. The observations suggest new strategies for probing the pathophysiology, risk, and treatment of hypertension.
Collapse
|
7
|
The role of PACAP in central cardiorespiratory regulation. Respir Physiol Neurobiol 2010; 174:65-75. [PMID: 20470908 DOI: 10.1016/j.resp.2010.05.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 05/03/2010] [Accepted: 05/03/2010] [Indexed: 11/22/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) plays a role in almost every biological process from reproduction to hippocampal function. One area where a role for PACAP is not clearly delineated is central cardiorespiratory regulation. PACAP and its receptors (PAC1, VPAC1 and VPAC2) are present in cardiovascular areas of the ventral medulla and spinal cord and in the periphery. Central administration of PACAP generally increases arterial pressure. Knowledge about the role of PACAP in central cardiovascular regulation is growing, but even less is known about PACAP in central respiratory regulation. No specific data is currently available regarding the presence of PACAP or receptors in key respiratory centers, although it is known that neonatal PACAP knock-out mice die suddenly in a manner similar to sudden infant death syndrome (SIDS). Future studies in mature preparations investigating the role of PACAP in the physiology and integration of central cardiorespiratory reflexes are clearly essential for a full understanding of this important neuropeptide in breathing.
Collapse
|
8
|
Vaudry D, Falluel-Morel A, Bourgault S, Basille M, Burel D, Wurtz O, Fournier A, Chow BKC, Hashimoto H, Galas L, Vaudry H. Pituitary Adenylate Cyclase-Activating Polypeptide and Its Receptors: 20 Years after the Discovery. Pharmacol Rev 2009; 61:283-357. [DOI: 10.1124/pr.109.001370] [Citation(s) in RCA: 829] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
9
|
Bobrovskaya L, Gelain DP, Gilligan C, Dickson PW, Dunkley PR. PACAP stimulates the sustained phosphorylation of tyrosine hydroxylase at serine 40. Cell Signal 2007; 19:1141-9. [PMID: 17261361 DOI: 10.1016/j.cellsig.2006.12.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2006] [Revised: 12/11/2006] [Accepted: 12/14/2006] [Indexed: 11/17/2022]
Abstract
Tyrosine hydroxylase (TH) is the rate-limiting enzyme in catecholamine synthesis. Its activity is controlled by PACAP, acutely by phosphorylation at Ser40 and chronically by protein synthesis. Using bovine adrenal chromaffin cells we found that PACAP, acting via the continuous activation of PACAP 1 receptors, sustained the phosphorylation of TH at Ser40 and led to TH activation for up to 24 h in the absence of TH protein synthesis. The sustained phosphorylation of TH at Ser40 was not mediated by hierarchical phosphorylation of TH at either Ser19 or Ser31. PACAP caused sustained activation of PKA, but did not sustain activation of other protein kinases including ERK, p38 kinase, PKC, MAPKAPK2 and MSK1. The PKA inhibitor H89 substantially inhibited the acute and the sustained phosphorylation of TH mediated by PACAP. PACAP also inhibited the activity of PP2A and PP2C at 24 h. PACAP therefore sustained TH phosphorylation at Ser40 for 24 h by sustaining the activation of PKA and causing inactivation of Ser40 phosphatases. The PKA activator 8-CPT-6Phe-cAMP also caused sustained phosphorylation of TH at Ser40 that was inhibited by the PKA inhibitor H89. Using cyclic AMP agonist pairs we found that sustained phosphorylation of TH was due to both the RI and the RII isotypes of PKA. The sustained activation of TH that occurred as a result of TH phosphorylation at Ser40 could maintain the synthesis of catecholamines without the need for further stimulus of the adrenal cells or increased TH protein synthesis.
Collapse
Affiliation(s)
- Larisa Bobrovskaya
- The School of Biomedical Sciences and Hunter Medical Research Institute, Faculty of Health, The University of Newcastle, Callaghan, NSW 2308, Australia
| | | | | | | | | |
Collapse
|
10
|
Tai TC, Wong-Faull DC, Claycomb R, Wong DL. Nerve Growth Factor Regulates Adrenergic Expression. Mol Pharmacol 2006; 70:1792-801. [PMID: 16926281 DOI: 10.1124/mol.106.026237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The mechanism by which nerve growth factor (NGF) regulates adrenergic expression was examined in PC-12 cells transfected with a rat phenylethanolamine N-methyl-transferase (PNMT) promoter-luciferase reporter gene construct pGL3RP893. NGF treatment increased PNMT promoter-driven luciferase activity in a dose- and time-dependent manner. Induction was attenuated by inhibition of the extracellular signal-regulated kinase mitogen-activated protein kinase (MAPK) pathway ( approximately 60%) but not by inhibition of the protein kinase A (PKA), protein kinase C, phosphoinositol kinase, or p38 MAPK pathways. Deletion PNMT promoter-luciferase reporter gene constructs showed that the NGF-responsive sequences lay within the proximal -392 base pairs (bp) of PNMT promoter, wherein binding elements for Egr-1 (-165 bp) and Sp1 (-48 bp) reside. Western analysis further showed that NGF increased nuclear levels of Egr-1, but not Sp1 or the catalytic subunit of PKA. Gel mobility shift assays showed increased potential for Egr-1, but not Sp1, protein-DNA binding complex formation. Mutation of either the Egr-1 or Sp1 binding sites in the PNMT promoter attenuated NGF activation. NGF, combined with pituitary adenylyl cyclase-activating protein (PACAP), another PNMT transcriptional activator, cooperatively stimulated PNMT promoter driven-luciferase activity beyond levels observed with either neurotrophin alone. Finally, post-transcriptional control seems to be another important mechanism by which neurotrophins regulate the adrenergic phenotype. NGF, PACAP, and a combination of the two stimulated both intron-retaining and intronless PNMT mRNA and PNMT protein, but to different extents.
Collapse
Affiliation(s)
- T C Tai
- Laboratory of Molecular and Developmental Neurobiology, Department of Psychiatry, McLean Hospital, Harvard Medical School, 115 Mill St., MRC 116, Belmont, MA 02478, USA
| | | | | | | |
Collapse
|
11
|
Reglödi D, Lubics A, Kiss P, Lengvári I, Gaszner B, Tóth G, Hegyi O, Tamás A. Effect of PACAP in 6-OHDA-induced injury of the substantia nigra in intact young and ovariectomized female rats. Neuropeptides 2006; 40:265-74. [PMID: 16904179 DOI: 10.1016/j.npep.2006.06.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2006] [Revised: 06/06/2006] [Accepted: 06/10/2006] [Indexed: 11/30/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) has neuroprotective effects in various neuronal cultures and in models of brain pathologies in vivo. Among others, it protects dopaminergic neurons in vitro, against 6-OHDA- and rotenone-induced injury. Recently, we have shown that PACAP reduces dopaminergic cell loss and ameliorates behavioral outcome following unilateral 6-OHDA-induced injury of the substantia nigra in male rats. However, after castration, PACAP led only to a slight amelioration of the behavioral symptoms. The aim of the present study was to investigate the degree of neuroprotection exerted by PACAP in female rats, using the same model. It was found that PACAP had no effect on the dopaminergic cell loss in intact female rats, only caused amelioration of certain acute behavioral signs. In contrast, PACAP effectively increased dopaminergic cell survival and decreased behavioral deficits in ovariectomized females. These results indicate that the neuroprotective effect of PACAP in a rat model of Parkinson's disease is gender-specific.
Collapse
Affiliation(s)
- D Reglödi
- Department of Anatomy, Neurohumoral Research Group of the Hungarian Academy of Sciences, University of Pécs, Medical School, 7624 Pécs, Szigeti u 12, Hungary.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Wong DL. Epinephrine biosynthesis: hormonal and neural control during stress. Cell Mol Neurobiol 2006; 26:891-900. [PMID: 16645894 DOI: 10.1007/s10571-006-9056-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2005] [Accepted: 03/14/2006] [Indexed: 10/24/2022]
Abstract
1. Stress contributes to the pathophysiology of many diseases, including psychiatric disorders, immune dysfunction, nicotine addiction and cardiovascular illness. Epinephrine and the glucocorticoids, cortisol and corticosterone, are major stress hormones. 2. Release of epinephrine from the adrenal medulla and glucocorticoids from the adrenal cortex initiate the biological responses permitting the organism to cope with adverse psychological, physiological and environmental stressors. Following its massive release during stress, epinephrine must be restored to replenish cellular pools and sustain release to maintain the heightened awareness and sequelae of responses to re-establish homeostasis and ensure survival. 3. Epinephrine is regulated in part through its biosynthesis catalyzed by the final enzyme in the catecholamine pathway, phenylethanolamine N-methyltransferase (E.C. 2.1.1.28, PNMT). PNMT expression, in turn, is controlled through hormonal and neural stimuli, which exert their effects on gene transcription through protein stability. 4. The pioneering work of Julius Axelrod forged the path to our present understanding of how the stress hormone and neurotransmitter epinephrine, is regulated, in particular via its biosynthesis by PNMT.
Collapse
Affiliation(s)
- Dona Lee Wong
- Department of Psychiatry, Harvard Medical School, Belmont, Massachusetts, USA.
| |
Collapse
|
13
|
Conconi MT, Spinazzi R, Nussdorfer GG. Endogenous Ligands of PACAP/VIP Receptors in the Autocrine–Paracrine Regulation of the Adrenal Gland. INTERNATIONAL REVIEW OF CYTOLOGY 2006; 249:1-51. [PMID: 16697281 DOI: 10.1016/s0074-7696(06)49001-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) are the main endogenous ligands of a class of G protein-coupled receptors (Rs). Three subtypes of PACAP/VIP Rs have been identified and named PAC(1)-Rs, VPAC(1)-Rs, and VPAC(2)-Rs. The PAC(1)-R almost exclusively binds PACAP, while the other two subtypes bind with about equal efficiency VIP and PACAP. VIP, PACAP, and their receptors are widely distributed in the body tissues, including the adrenal gland. VIP and PACAP are synthesized in adrenomedullary chromaffin cells, and are released in the adrenal cortex and medulla by VIPergic and PACAPergic nerve fibers. PAC(1)-Rs are almost exclusively present in the adrenal medulla, while VPAC(1)-Rs and VPAC(2)-Rs are expressed in both the adrenal cortex and medulla. Evidence indicates that VIP and PACAP, acting via VPAC(1)-Rs and VPAC(2)-Rs coupled to adenylate cyclase (AC)- and phospholipase C (PLC)-dependent cascades, stimulate aldosterone secretion from zona glomerulosa (ZG) cells. There is also proof that they can also enhance aldosterone secretion indirectly, by eliciting the release from medullary chromaffin cells of catecholamines and adrenocorticotropic hormone (ACTH), which in turn may act on the cortical cells in a paracrine manner. The involvement of VIP and PACAP in the regulation of glucocorticoid secretion from inner adrenocortical cells is doubtful and surely of minor relevance. VIP and PACAP stimulate the synthesis and release of adrenomedullary catecholamines, and all three subtypes of PACAP/VIP Rs mediate this effect, PAC(1)-Rs being coupled to AC, VPAC(1)-Rs to both AC and PLC, and VPAC(2)-Rs only to PLC. A privotal role in the catecholamine secretagogue action of VIP and PACAP is played by Ca(2+). VIP and PACAP may also modulate the growth of the adrenal cortex and medulla. The concentrations attained by VIP and PACAP in the blood rule out the possibility that they act as true circulating hormones. Conversely, their adrenal content is consistent with a local autocrine-paracrine mechanism of action.
Collapse
Affiliation(s)
- Maria Teresa Conconi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Padua, I-35121 Padua, Italy
| | | | | |
Collapse
|
14
|
Ogawa T, Nakamachi T, Ohtaki H, Hashimoto H, N S, Baba A, Watanabe J, Kikuyama S, Shioda S. Monoaminergic neuronal development is not affected in PACAP-gene-deficient mice. ACTA ACUST UNITED AC 2005; 126:103-8. [PMID: 15620422 DOI: 10.1016/j.regpep.2004.08.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) has been implicated in several physiological functions. Several lines of evidence from in vitro studies have shown that PACAP plays some important roles in development of nervous system such as neural proliferation and differentiation. Recently, mice lacking PACAP have been reported to show a higher mortality shortly after birth, impaired thermal adaptation, and altered psychomotor behaviors. Inasmuch as monoaminergic nervous systems are implicated in these phenotypes and a quite few data have been reported on the role of this peptide in nervous development in vitro, we studied early development [embryonic days 10.5 (E10.5) and 12.5 (E12.5)] of monoaminergic nervous systems in mice lacking PACAP. The fetuses lacking PACAP showed immunoreactivities (IRs) for tyrosine hydroxylase (TH) and serotonin (5-HT) similarly to the wild type. We observed TH-IR in the forebrain [striatal differentiating zone (dz) and hypothalamic dz], midbrain, hindbrain, neural-crest-derived sympathetic ganglionic primordia, ventral spinal cord dz, and bowel at E10.5 in both PACAP null and wild type with no difference. At E12.5, in the wild-type- and PACAP-gene-deficient mice, no differences of 5-HT- and TH-IRs were observed in several brain regions, including brainstem (midbrain and pons). Thus, the depletion of PACAP does not affect monoaminergic nervous systems in the early development.
Collapse
Affiliation(s)
- Tetsuo Ogawa
- Department of Anatomy, Showa University School of Medicine, Tokyo 142-8555, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Yu Y, Jawa A, Pan W, Kastin AJ. Effects of peptides, with emphasis on feeding, pain, and behavior A 5-year (1999-2003) review of publications in Peptides. Peptides 2004; 25:2257-89. [PMID: 15572212 DOI: 10.1016/j.peptides.2004.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2004] [Accepted: 09/21/2004] [Indexed: 11/28/2022]
Abstract
Novel effects of naturally occurring peptides are continuing to be discovered, and their mechanisms of actions as well as interactions with other substances, organs, and systems have been elucidated. Synthetic analogs may have actions similar or antagonistic to the endogenous peptides, and both the native peptides and analogs have potential as drugs or drug targets. The journal Peptides publishes many leading articles on the structure-activity relationship of peptides as well as outstanding reviews on some families of peptides. Complementary to the reviews, here we extract information from the original papers published during the past five years in Peptides (1999-2003) to summarize the effects of different classes of peptides, their modulation by other chemicals and various pathophysiological states, and the mechanisms by which the effects are exerted. Special attention is given to peptides related to feeding, pain, and other behaviors. By presenting in condensed form the effects of peptides which are essential for systems biology, we hope that this summary of existing knowledge will encourage additional novel research to be presented in Peptides.
Collapse
Affiliation(s)
- Yongmei Yu
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | | | | | | |
Collapse
|
16
|
Wong DL, Tai TC, Wong-Faull DC, Claycomb R, Kvetnansky R. Genetic mechanisms for adrenergic control during stress. Ann N Y Acad Sci 2004; 1018:387-97. [PMID: 15240394 DOI: 10.1196/annals.1296.048] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Cortisol and epinephrine released in response to stress are replenished via activation of the hypothalamic-pituitary-adrenal (HPA or stress) axis. Immobilization (IMMO) stress in rats stimulates epinephrine production in part via the gene encoding the epinephrine-synthesizing enzyme phenylethanolamine N-methyltransferase (PNMT). PNMT mRNA rose up to 7.0-fold with acute or chronic stress. Two transcription factors mediating stress induction of the PNMT gene are the glucocorticoid receptor (GR) and Egr-1, which interact with -533, -759, and -773 bp, and -165 bp binding sites in the rat PNMT promoter, respectively. To identify molecular mechanisms involved, effects of hypoxic stress on PNMT promoter activity were examined in PC12 cells transfected with the PNMT promoter-luciferase reporter gene construct pGL3RP893. Oxygen reduction to 5% increased PNMT promoter-driven luciferase expression, with maximum activity at 6 h. Pretreatment of the cells with protein kinase A (PKA) and protein kinase C (PKC) inhibitors, H-89 and GF109203X, respectively, attenuated the rise in luciferase. Similarly, PKA-deficient PC12 cells transfected with pGL3RP893 and exposed to hypoxia also showed attenuated PNMT promoter-driven luciferase expression. Mutation of the Egr-1 binding site completely prevented PNMT promoter activation, indicating that Egr-1 is essential to the stress response. Consistent with this result, hypoxia increased Egr-1 protein. Hypoxia also increased endogenous PNMT mRNA. However, a shift to intron-retaining mRNA from which truncated, nonfunctional protein is produced, occurred, suggesting that posttranscriptional regulation may be an important genetic mechanism controlling adrenergic expression and hence, epinephrine, during stress.
Collapse
Affiliation(s)
- Dona L Wong
- Department of Psychiatry, Harvard Medical School, Laboratory of Molecular and Developmental Neurobiology, McLean Hospital 115 Mill Street, Mailman Research Center Rm 116, Belmont, MA 02478, USA.
| | | | | | | | | |
Collapse
|
17
|
Tai TC, Wong DL. Protein kinase A and protein kinase C signaling pathway interaction in phenylethanolamine N-methyltransferase gene regulation. J Neurochem 2003; 85:816-29. [PMID: 12694408 DOI: 10.1046/j.1471-4159.2003.01728.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The protein kinase A (PKA) and protein kinase C (PKC) signaling pathways appear to interact in regulating phenylethanolamine N-methyltransferase (PNMT) promoter-driven gene transcription in PC12 cells. Forskolin treatment of cells transfected with the rat PNMT promoter-luciferase reporter gene construct pGL3RP893 increased promoter activity approximately two-fold whereas phorbol-12-myristate-13 acetate (PMA) treatment had no effect. However, simultaneous forskolin and PMA treatment synergistically activated the PNMT promoter approximately four-fold, suggesting that PKC stimulation requires prior induction of the PKA pathway. Consistent with this possibility the adenylate cyclase inhibitor MDL12,330A, and the PKA inhibitor H-89 prevented PNMT promoter stimulation by the combination of forskolin and PMA. PKA and PKC regulation seems to be mediated in part by Egr-1 and Sp1 through their consensus elements in the PNMT promoter. Forskolin and PMA treatment of PC12 cells increased Egr-1 protein and phosphorylated Egr-1/DNA-binding complex formation to the same extent but only increased phosphorylated Sp1/DNA binding complex formation without altering Sp1 protein levels. Mutation of the - 165 bp Egr-1 and - 48 bp Sp1 sites, respectively, attenuated and abolished combined forskolin and PMA-mediated promoter activation. PNMT promoter analysis further showed that synergistic stimulation by PKA and PKC involves DNA sequences between - 442 and - 392 bp, and potentially a GCM binding element lying within this region.
Collapse
Affiliation(s)
- T C Tai
- Department of Psychiatry, Harvard Medical School, Laboratory of Molecular and Developmental Neurobiology, McLean Hospital, Belmont, Massachusetts 02478, USA
| | | |
Collapse
|
18
|
Kim D, Choi HJ, Kim SW, Cho SW, Hwang O. Upregulation of catecholamine biosynthetic enzymes by nitric oxide. J Neurosci Res 2003; 72:98-104. [PMID: 12645083 DOI: 10.1002/jnr.10557] [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] [Indexed: 11/07/2022]
Abstract
Nitric oxide (NO) is recognized as an essential intercellular messenger in central and peripheral nervous systems. In the present study, whether NO exerts effects on catecholamine (CA) biosynthetic enzymes was determined in primary cultured bovine chromaffin cells. The NO generators sodium nitroprusside (SNP) and S-nitroso-N-acetyl-D,L-penicillamine, in a dose-dependent manner, upregulated transcript levels of tyrosine hydroxylase, dopamine beta-hydroxylase, and phenylethanolamine N-methyltransferase, accompanied by long-term increases in their enzyme activities and the intracellular CA levels. The SNP effect was diminished by co-treatment with LY83583, an inhibitor of soluble guanylate cyclase, or H-8, a cyclic GMP (cGMP)-dependent protein kinase inhibitor. Co-treatment with 8-Br-cGMP did not increase further the expression of these enzyme genes induced by SNP. Taken together, the data suggest that NO leads to long-term upregulation of the CA system via induction of the genes involved and that this is mediated by cGMP-dependent signaling pathway.
Collapse
Affiliation(s)
- Donghou Kim
- Department of Cell and Anatomy, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | | | | |
Collapse
|
19
|
Wong DL, Anderson LJ, Tai TC. Cholinergic and peptidergic regulation of phenylethanolamine N-methyltransferase gene expression. Ann N Y Acad Sci 2002; 971:19-26. [PMID: 12438084 DOI: 10.1111/j.1749-6632.2002.tb04428.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The splanchnic nerve, innervating the adrenal medulla, releases a variety of neurotransmitters that stimulate genes involved in catecholamine biosynthesis. In particular, cholinergic agonists have been shown to induce phenylethanolamine N-methyltransferase (PNMT) gene expression through activation of both nicotinic and muscarinic receptors in vivo and in vitro. By contrast, the role of peptidergic neurotransmitters in adrenal medullary PNMT gene expression remains unclear. Using transient transfection assays, we demonstrate that rat PNMT promoter-luciferase reporter gene constructs are markedly activated by 10 nM PACAP when expressed in PC12 cells. PACAP appears to mediate its effects primarily through PAC1 receptors and, subsequently, cAMP-protein kinase A (PKA) and extracellular Ca(2+) signaling mechanisms. Activation of these signal transduction pathways markedly increases nuclear levels of the immediately early gene transcription factor Egr-1 and the developmental factor AP2. A slight decrease in Sp1 expression may also occur, whereas MAZ and glucocorticoid receptor expression remains unaltered. Although PACAP stimulates rapid changes in transcription factor expression and PNMT promoter activity, its effects are long lasting. PNMT promoter induction continues to rise and is sustained for > or=48 hours. By contrast, while muscarine, nicotine, or carbachol (100 micro M) also evoke rapid increases in rat PNMT promoter activity, peak activity is observed at 6 hours, followed by a decline and restoration to basal levels by 24 hours. Cholinergic activation of the PNMT promoter also seems to involve the cAMP-PKA signaling mechanism. However, the magnitude of stimulation and antagonist blockade with H-89 or the polypeptide inhibitor PKI suggests that the extent of activation is much less than that with PACAP.
Collapse
Affiliation(s)
- Dona Lee Wong
- Department of Psychiatry, Harvard Medical School and Laboratory of Molecular and Developmental Neurobiology, McLean Hospital, Belmont, Massachusetts 02478, USA.
| | | | | |
Collapse
|
20
|
Shizukuda Y, Buttrick PM. Subtype specific roles of beta-adrenergic receptors in apoptosis of adult rat ventricular myocytes. J Mol Cell Cardiol 2002; 34:823-31. [PMID: 12099721 DOI: 10.1006/jmcc.2002.2020] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have previously reported that beta-adrenergic receptor (beta-AR) stimulation promotes apoptosis in adult ventricular myocytes through PKCepsilon-mediated suppression of ERK. In this study, we investigated differential effects of beta-AR subtypes on this signal pathway. The apoptosis induced by the non-specific beta-AR agonist isoproterenol was largely blocked by the beta(1)-selective antagonist CGP 20712A, but not by the beta(2)-selective antagonist ICI 118551. A pro-apoptotic effect of beta(1)-AR was also blocked by the PKA inhibitor H89, while the protein kinase A (PKA) activators forskolin and dibutyryl-cAMP both induced apoptosis. These results indicate that beta(1)-AR-mediated PKA activation is largely responsible for the apoptosis induced by beta-AR in adult rat cardiac myocytes. This conclusion was also supported by the finding that PKA was preferentially activated by beta(1)-AR over beta(2)-AR. beta(2)-AR selectively induced anti-apoptotic ERK activation in the presence of PKCepsilon suppression, and this ERK activation was sensitive to pertussis toxin. PKCepsilon itself as well as Akt, the other anti-apoptotic factor were activated by both beta-AR subtypes. Thus, beta(1)-AR induces pro-apoptotic signals mainly through PKA activation. In contrast, beta(2)-AR is linked to Gi-mediated ERK activation, which is involved in the anti-apoptotic pathway, and is regulated by PKCepsilon. Therefore, our findings suggest a rather complex role for beta-AR subtypes in the regulation of apoptosis in adult ventricular myocytes.
Collapse
Affiliation(s)
- Yukitaka Shizukuda
- Section of Cardiology, Department of Medicine, University of Illinois at Chicago, Illinois 60612, USA.
| | | |
Collapse
|
21
|
|
22
|
Turquier V, Yon L, Grumolato L, Alexandre D, Fournier A, Vaudry H, Anouar Y. Pituitary adenylate cyclase-activating polypeptide stimulates secretoneurin release and secretogranin II gene transcription in bovine adrenochromaffin cells through multiple signaling pathways and increased binding of pre-existing activator protein-1-like transcription factors. Mol Pharmacol 2001; 60:42-52. [PMID: 11408599 DOI: 10.1124/mol.60.1.42] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Secretoneurin (SN) is a novel bioactive peptide that derives from the neuroendocrine protein secretogranin II (SgII) by proteolytic processing and participates in neuro-immune communication. The neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP-38) dose-dependently stimulates (EC(50) approximately 3 nM) SN release (up to 4-fold) and SgII gene expression (up to 60-fold) in cultured bovine adrenochromaffin cells. The effect of PACAP on both SN secretion and SgII mRNA levels is rapid and long lasting. We analyzed in this neuroendocrine cell model the transduction pathways involved in both SN secretion and SgII gene transcription in response to PACAP. The cytosolic calcium chelator BAPTA-AM and the nonselective calcium channel antagonist NiCl(2) equally inhibited both secretion of the peptide and transcription of the SgII gene, indicating a major contribution of calcium influx in PACAP-induced SN biosynthesis and release in chromaffin cells. Inhibition of protein kinase A (PKA) or C (PKC) also reduced PACAP-evoked SN release but did not alter the stimulatory effect of PACAP on SgII mRNA levels. Conversely, application of mitogen-activated protein kinase inhibitors suppressed PACAP-induced SgII gene expression. The effect of PACAP on SgII mRNA levels, like the effect of the PKC stimulator 12-O-tetradecanoylphorbol-13-acetate (TPA), was not affected by cycloheximide, whereas the effects of the PKA stimulator forskolin or cell-depolarization by high K(+) were significantly reduced by the protein synthesis inhibitor. PACAP and TPA both increased the binding activity of the SgII cAMP response element to trans-acting factors present in chromaffin cell nuclear extracts, which are recognized by antibodies to activator protein-1-related proteins. These data indicate that SN biosynthesis is regulated by PACAP in chromaffin cells through complex signaling cascades, suggesting that SN may play a function during trans-synaptic stimulation of the adrenal medulla.
Collapse
Affiliation(s)
- V Turquier
- Institut Fédératif de Recherches Multidisciplinaires sur les Peptides (IFRMP 23), Laboratoire de Neuroendocrinologie Cellulaire et Moléculaire, INSERM U413, UA CNRS, Université de Rouen, Mont-Saint-Aignan, France
| | | | | | | | | | | | | |
Collapse
|