1
|
Hong XY, Li S, Li T, Chen W, Li Y, Wang Z, Luo Y. Differential involvement of central and peripheral catecholamines between Alzheimer's disease and vascular dementia. Heliyon 2024; 10:e38843. [PMID: 39398044 PMCID: PMC11471233 DOI: 10.1016/j.heliyon.2024.e38843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024] Open
Abstract
Background and aim The important role of catecholamines has been gradually emphasized in the pathogenesis of neurodegenerative process. As the most prevalent form of cognitive dysfunction, Alzheimer's disease (AD) and vascular dementia (VaD) have the distinct pathological features and pathogenic mechanisms, however, the differential involvement of central and peripheral catecholamines between AD and VaD was still unclear. Methods Triple-transgenic AD (3 × Tg-AD) mice and chronic cerebral hypoperfusion (CCH) in rats induced by two-vessel occlusion (2VO) were used as the AD and VaD model in this study, respectively. The concentrations of catecholamines (dopamine, epinephrine and norepinephrine) and their metabolites (3-methoxytyramine, metanephrine and normetanephrine) in serum and five brain regions (hippocampus, cortex, corpus striatum, thalamus and pons) from 3 × Tg-AD mice and 2VO rats were quantitatively determined by liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay. Results High expression and distribution of hippocampal dopamine, and epinephrine and norepinephrine in the cortex and thalamus were found in the early 3 × Tg-AD model, whereas chronic cerebral hypoperfusion induced by 2VO mainly affected the central noradrenergic and noradrenergic system, but not dopaminergic system. The increased serum levels of catecholamines were investigated in the 2VO rats, but not in the 3 × Tg-AD mice. Conclusion The differential expression and distribution of central catecholamines and their metabolites suggests the distinct catecholamines-related pathogenesis between AD and VaD. Peripheral catecholamine surge may be involved in the development of VaD, and the treatment strategy to prevent or reverse the effects of peripheral catecholamines may be protective for VaD.
Collapse
Affiliation(s)
- Xiao-Yue Hong
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
| | - Siwei Li
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
| | - Tian Li
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
| | - Wei Chen
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
| | - Yirong Li
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
| | - Zhuo Wang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
| | - Yi Luo
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
| |
Collapse
|
2
|
Wimalasena K, Adetuyi O, Eldani M. Metabolic energy decline coupled dysregulation of catecholamine metabolism in physiologically highly active neurons: implications for selective neuronal death in Parkinson's disease. Front Aging Neurosci 2024; 16:1339295. [PMID: 38450382 PMCID: PMC10914975 DOI: 10.3389/fnagi.2024.1339295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/09/2024] [Indexed: 03/08/2024] Open
Abstract
Parkinson's disease (PD) is an age-related irreversible neurodegenerative disease which is characterized as a progressively worsening involuntary movement disorder caused by the loss of dopaminergic (DA) neurons in substantia nigra pars compacta (SNpc). Two main pathophysiological features of PD are the accumulation of inclusion bodies in the affected neurons and the predominant loss of neuromelanin-containing DA neurons in substantia nigra pars compacta (SNpc) and noradrenergic (NE) neurons in locus coeruleus (LC). The inclusion bodies contain misfolded and aggregated α-synuclein (α-Syn) fibrils known as Lewy bodies. The etiology and pathogenic mechanisms of PD are complex, multi-dimensional and associated with a combination of environmental, genetic, and other age-related factors. Although individual factors associated with the pathogenic mechanisms of PD have been widely investigated, an integration of the findings to a unified causative mechanism has not been envisioned. Here we propose an integrated mechanism for the degeneration of DA neurons in SNpc and NE neurons in LC in PD, based on their unique high metabolic activity coupled elevated energy demand, using currently available experimental data. The proposed hypothetical mechanism is primarily based on the unique high metabolic activity coupled elevated energy demand of these neurons. We reason that the high vulnerability of a selective group of DA neurons in SNpc and NE neurons in LC in PD could be due to the cellular energy modulations. Such cellular energy modulations could induce dysregulation of DA and NE metabolism and perturbation of the redox active metal homeostasis (especially copper and iron) in these neurons.
Collapse
Affiliation(s)
- Kandatege Wimalasena
- Department of Chemistry and Biochemistry, Wichita State University, Wichita, KS, United States
| | | | | |
Collapse
|
3
|
Mahmoodi N, Minnow YVT, Harijan RK, Bedard GT, Schramm VL. Cell-Effective Transition-State Analogue of Phenylethanolamine N-Methyltransferase. Biochemistry 2023; 62:2257-2268. [PMID: 37467463 PMCID: PMC10646973 DOI: 10.1021/acs.biochem.3c00103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
Phenylethanolamine N-methyltransferase (PNMT) catalyzes the S-adenosyl-l-methionine (SAM)-dependent methylation of norepinephrine to form epinephrine. Epinephrine is implicated in the regulation of blood pressure, respiration, Alzheimer's disease, and post-traumatic stress disorder (PTSD). Transition-state (TS) analogues bind their target enzymes orders of magnitude more tightly than their substrates. A synthetic strategy for first-generation TS analogues of human PNMT (hPNMT) permitted structural analysis of hPNMT and revealed potential for second-generation inhibitors [Mahmoodi, N.; J. Am. Chem. Soc. 2020, 142, 14222-14233]. A second-generation TS analogue inhibitor of PNMT was designed, synthesized, and characterized to yield a Ki value of 1.2 nM. PNMT isothermal titration calorimetry (ITC) measurements of inhibitor 4 indicated a negative cooperative binding mechanism driven by large favorable entropic contributions and smaller enthalpic contributions. Cell-based assays with HEK293T cells expressing PNMT revealed a cell permeable, intracellular PNMT inhibitor with an IC50 value of 81 nM. Structural analysis demonstrated inhibitor 4 filling catalytic site regions to recapitulate both norepinephrine and SAM interactions. Conformation of the second-generation inhibitor in the catalytic site of PNMT improves contacts relative to those from the first-generation inhibitors. Inhibitor 4 demonstrates up to 51,000-fold specificity for PNMT relative to DNA and protein methyltransferases. Inhibitor 4 also exhibits a 12,000-fold specificity for PNMT over the α2-adrenoceptor.
Collapse
Affiliation(s)
- Niusha Mahmoodi
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461, United States
| | - Yacoba V T Minnow
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461, United States
| | - Rajesh K Harijan
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461, United States
| | - Gabriel T Bedard
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461, United States
| | - Vern L Schramm
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461, United States
| |
Collapse
|
4
|
Neti SS, Wang B, Iwig DF, Onderko EL, Booker SJ. Enzymatic Fluoromethylation Enabled by the S-Adenosylmethionine Analog Te-Adenosyl- L-(fluoromethyl)homotellurocysteine. ACS CENTRAL SCIENCE 2023; 9:905-914. [PMID: 37252363 PMCID: PMC10214534 DOI: 10.1021/acscentsci.2c01385] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Indexed: 05/31/2023]
Abstract
Fluoromethyl, difluoromethyl, and trifluoromethyl groups are present in numerous pharmaceuticals and agrochemicals, where they play critical roles in the efficacy and metabolic stability of these molecules. Strategies for late-stage incorporation of fluorine-containing atoms in molecules have become an important area of organic and medicinal chemistry as well as synthetic biology. Herein, we describe the synthesis and use of Te-adenosyl-L-(fluoromethyl)homotellurocysteine (FMeTeSAM), a novel and biologically relevant fluoromethylating agent. FMeTeSAM is structurally and chemically related to the universal cellular methyl donor S-adenosyl-L-methionine (SAM) and supports the robust transfer of fluoromethyl groups to oxygen, nitrogen, sulfur, and some carbon nucleophiles. FMeTeSAM is also used to fluoromethylate precursors to oxaline and daunorubicin, two complex natural products that exhibit antitumor properties.
Collapse
Affiliation(s)
- Syam Sundar Neti
- Department
of Chemistry, Department of Biochemistry and Molecular Biology, and Howard Hughes
Medical Institute, The Pennsylvania State
University, University
Park, Pennsylvania 16802, United States
| | - Bo Wang
- Department
of Chemistry, Department of Biochemistry and Molecular Biology, and Howard Hughes
Medical Institute, The Pennsylvania State
University, University
Park, Pennsylvania 16802, United States
| | - David F. Iwig
- Department
of Chemistry, Department of Biochemistry and Molecular Biology, and Howard Hughes
Medical Institute, The Pennsylvania State
University, University
Park, Pennsylvania 16802, United States
| | - Elizabeth L. Onderko
- Department
of Chemistry, Department of Biochemistry and Molecular Biology, and Howard Hughes
Medical Institute, The Pennsylvania State
University, University
Park, Pennsylvania 16802, United States
| | - Squire J. Booker
- Department
of Chemistry, Department of Biochemistry and Molecular Biology, and Howard Hughes
Medical Institute, The Pennsylvania State
University, University
Park, Pennsylvania 16802, United States
| |
Collapse
|
5
|
Tsujimoto Y, Yamamoto M, Nishikage S, Kanie K, Kanzawa M, Bando H, Yoshino K, Hirota Y, Fukuoka H, Ogawa W. Successful diagnosis and treatment of pheochromocytoma during severe coronavirus disease 2019 (COVID-19): a case report. Endocr J 2023; 70:259-265. [PMID: 36384707 DOI: 10.1507/endocrj.ej22-0446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Pheochromocytoma is a rare but life-threatening condition due to catecholamine release induced by drug treatments such as β-blockers or glucocorticoids. We present a case of hypertensive crisis due to pheochromocytoma, induced after the initiation of dexamethasone and landiolol during intensive care for severe coronavirus disease 2019 (COVID-19). Based on a detailed medical history review, the patient was previously diagnosed with primary aldosteronism by confirmatory tests, moreover, an abdominal computed tomography scan identified an adrenal tumor 2 years before current admission. We tentatively diagnosed the patient with pheochromocytoma and initiated α-blockers without conducting a catecholamine report, leading to stable hemodynamics. We present a successfully managed case of pheochromocytoma concomitant with COVID-19, which has become a global crisis.
Collapse
Affiliation(s)
- Yasutaka Tsujimoto
- Division of Diabetes and Endocrinology, Kobe University Hospital, Kobe 650-0017, Japan
| | - Masaaki Yamamoto
- Division of Diabetes and Endocrinology, Kobe University Hospital, Kobe 650-0017, Japan
| | - Seiji Nishikage
- Division of Diabetes and Endocrinology, Kobe University Hospital, Kobe 650-0017, Japan
| | - Keitaro Kanie
- Division of Diabetes and Endocrinology, Kobe University Hospital, Kobe 650-0017, Japan
| | - Maki Kanzawa
- Department of Diagnostic Pathology, Kobe University Hospital, Kobe 650-0017, Japan
| | - Hironori Bando
- Division of Diabetes and Endocrinology, Kobe University Hospital, Kobe 650-0017, Japan
| | - Kei Yoshino
- Division of Diabetes and Endocrinology, Kobe University Hospital, Kobe 650-0017, Japan
| | - Yushi Hirota
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Hidenori Fukuoka
- Division of Diabetes and Endocrinology, Kobe University Hospital, Kobe 650-0017, Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| |
Collapse
|
6
|
Fernandez CJ, Hanna FW, Pacak K, Nazari MA. Catecholamines and blood pressure regulation. ENDOCRINE HYPERTENSION 2023:19-34. [DOI: 10.1016/b978-0-323-96120-2.00010-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
7
|
Jensterle M, Lezaic L, Jeruc J, Janez A. A rare metanephrine-predominant secretion phenotype of paraganglioma challenged an interpretation of imaging localization. Endocrine 2021; 74:432-434. [PMID: 34075540 DOI: 10.1007/s12020-021-02772-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/19/2021] [Indexed: 10/21/2022]
Affiliation(s)
- Mojca Jensterle
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| | - Luka Lezaic
- Department for Nuclear Medicine, University Medical Centre, Faculty of Medicine Ljubljana, Ljubljana, Slovenia
| | - Jera Jeruc
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Andrej Janez
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
8
|
Parsons RB, Facey PD. Nicotinamide N-Methyltransferase: An Emerging Protagonist in Cancer Macro(r)evolution. Biomolecules 2021; 11:1418. [PMID: 34680055 PMCID: PMC8533529 DOI: 10.3390/biom11101418] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 12/15/2022] Open
Abstract
Nicotinamide N-methyltransferase (NNMT) has progressed from being considered merely a Phase II metabolic enzyme to one with a central role in cell function and energy metabolism. Over the last three decades, a significant body of evidence has accumulated which clearly demonstrates a central role for NNMT in cancer survival, metastasis, and drug resistance. In this review, we discuss the evidence supporting a role for NNMT in the progression of the cancer phenotype and how it achieves this by driving the activity of pro-oncogenic NAD+-consuming enzymes. We also describe how increased NNMT activity supports the Warburg effect and how it promotes oncogenic changes in gene expression. We discuss the regulation of NNMT activity in cancer cells by both post-translational modification of the enzyme and transcription factor binding to the NNMT gene, and describe for the first time three long non-coding RNAs which may play a role in the regulation of NNMT transcription. We complete the review by discussing the development of novel anti-cancer therapeutics which target NNMT and provide insight into how NNMT-based therapies may be best employed clinically.
Collapse
Affiliation(s)
- Richard B. Parsons
- Institute of Pharmaceutical Science, King’s College London, 150 Stamford Street, London SE1 9NH, UK
| | - Paul D. Facey
- Singleton Park Campus, Swansea University Medical School, Swansea University, Swansea SA2 8PP, UK;
| |
Collapse
|
9
|
Giatti S, Di Domizio A, Diviccaro S, Falvo E, Caruso D, Contini A, Melcangi RC. Three-Dimensional Proteome-Wide Scale Screening for the 5-Alpha Reductase Inhibitor Finasteride: Identification of a Novel Off-Target. J Med Chem 2021; 64:4553-4566. [PMID: 33843213 PMCID: PMC8154553 DOI: 10.1021/acs.jmedchem.0c02039] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Indexed: 12/15/2022]
Abstract
Finasteride, a 5-alpha reductase (5α-R) inhibitor, is a widely used drug for treating androgen-dependent conditions. However, its use is associated with sexual, psychological, and physical complaints, suggesting that other mechanisms, in addition to 5α-R inhibition, may be involved. Here, a multidisciplinary approach has been used to identify potential finasteride off-target proteins. SPILLO-PBSS software suggests an additional inhibitory activity of finasteride on phenylethanolamine N-methyltransferase (PNMT), the limiting enzyme in formation of the stress hormone epinephrine. The interaction of finasteride with PNMT was supported by docking and molecular dynamics analysis and by in vitro assay, confirming the inhibitory nature of the binding. Finally, this inhibition was also confirmed in an in vivo rat model. Literature data indicate that PNMT activity perturbation may be correlated with sexual and psychological side effects. Therefore, results here obtained suggest that the binding of finasteride to PNMT might have a role in producing the side effects exerted by finasteride treatment.
Collapse
Affiliation(s)
- Silvia Giatti
- Department
of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milano, Italy
| | - Alessandro Di Domizio
- Department
of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milano, Italy
- SPILLOproject, via Stradivari
17, Paderno Dugnano, 20037 Milano, Italy
| | - Silvia Diviccaro
- Department
of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milano, Italy
| | - Eva Falvo
- Department
of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milano, Italy
| | - Donatella Caruso
- Department
of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milano, Italy
| | - Alessandro Contini
- Dipartimento
Di Scienze Farmaceutiche, Università
degli Studi di Milano, 20133 Milano, Italy
| | - Roberto Cosimo Melcangi
- Department
of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milano, Italy
| |
Collapse
|
10
|
Lu J, Bart AG, Wu Q, Criscione KR, McLeish MJ, Scott EE, Grunewald GL. Structure-Based Drug Design of Bisubstrate Inhibitors of Phenylethanolamine N-Methyltransferase Possessing Low Nanomolar Affinity at Both Substrate Binding Domains 1. J Med Chem 2020; 63:13878-13898. [PMID: 33147410 DOI: 10.1021/acs.jmedchem.0c01475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The enzyme phenylethanolamine N-methyltransferase (PNMT, EC 2.1.1.28) catalyzes the final step in the biosynthesis of epinephrine and is a potential drug target, primarily for the control of hypertension. Unfortunately, many potent PNMT inhibitors also possess significant affinity for the a2-adrenoceptor, which complicates the interpretation of their pharmacology. A bisubstrate analogue approach offers the potential for development of highly selective inhibitors of PNMT. This paper documents the design, synthesis, and evaluation of such analogues, several of which were found to possess human PNMT (hPNMT) inhibitory potency <5 nM versus AdoMet. Site-directed mutagenesis studies were consistent with bisubstrate binding. Two of these compounds (19 and 29) were co-crystallized with hPNMT and the resulting structures revealed both compounds bound as predicted, simultaneously occupying both substrate binding domains. This bisubstrate inhibitor approach has resulted in one of the most potent (20) and selective (vs the a2-adrenoceptor) inhibitors of hPNMT yet reported.
Collapse
Affiliation(s)
- Jian Lu
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
| | - Aaron G Bart
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
| | - Qian Wu
- Department of Chemistry and Chemical Biology, Purdue School of Science, IUPUI, Indianapolis, Indiana 46202, United States
| | - Kevin R Criscione
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
| | - Michael J McLeish
- Department of Chemistry and Chemical Biology, Purdue School of Science, IUPUI, Indianapolis, Indiana 46202, United States
| | - Emily E Scott
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
| | - Gary L Grunewald
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
| |
Collapse
|
11
|
Mahmoodi N, Harijan RK, Schramm VL. Transition-State Analogues of Phenylethanolamine N-Methyltransferase. J Am Chem Soc 2020; 142:14222-14233. [PMID: 32702980 DOI: 10.1021/jacs.0c05446] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Phenylethanolamine N-methyltransferase (PNMT) is a critical enzyme in catecholamine synthesis. It transfers the methyl group of S-adenosylmethionine (SAM) to catalyze the synthesis of epinephrine from norepinephrine. Epinephrine has been associated with diverse human processes, including the regulation of blood pressure and respiration, as well as neurodegeneration found in Alzheimer's disease. Human PNMT (hPNMT) proceeds through an SN2 transition state (TS) in which the transfer of the methyl group is rate limiting. TS analogue enzyme inhibitors are specific for their target and bind orders of magnitude more tightly than their substrates. Molecules resembling the TS of hPNMT were designed, synthesized, and kinetically characterized. This new inhibitory scaffold was designed to mimic the geometry and electronic properties of the hPNMT TS. Synthetic efforts resulted in a tight-binding inhibitor with a Ki value of 12.0 nM. This is among the first of the TS analogue inhibitors of methyltransferase enzymes to show an affinity in the nanomolar range. Isothermal titration calorimetry (ITC) measurements indicated negative cooperative binding of inhibitor to the dimeric protein, driven by favorable entropic contributions. Structural analysis revealed that inhibitor 3 binds to hPNMT by filling the catalytic binding pockets for the cofactor (SAM) and the substrate (norepinephrine) binding sites.
Collapse
Affiliation(s)
- Niusha Mahmoodi
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York 10461, United States
| | - Rajesh K Harijan
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York 10461, United States
| | - Vern L Schramm
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York 10461, United States
| |
Collapse
|
12
|
Grassam-Rowe A, Ou X, Lei M. Novel cardiac cell subpopulations: Pnmt-derived cardiomyocytes. Open Biol 2020; 10:200095. [PMID: 32810421 PMCID: PMC7479933 DOI: 10.1098/rsob.200095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/29/2020] [Indexed: 11/12/2022] Open
Abstract
Diversity among highly specialized cells underlies the fundamental biology of complex multi-cellular organisms. One of the essential scientific questions in cardiac biology has been to define subpopulations within the heart. The heart parenchyma comprises specialized cardiomyocytes (CMs). CMs have been canonically classified into a few phenotypically diverse subpopulations largely based on their function and anatomic localization. However, there is growing evidence that CM subpopulations are in fact numerous, with a diversity of genetic origin and putatively different roles in physiology and pathophysiology. In this chapter, we introduce a recently discovered CM subpopulation: phenylethanolamine-N-methyl transferase (Pnmt)-derived cardiomyocytes (PdCMs). We discuss: (i) canonical classifications of CM subpopulations; (ii) discovery of PdCMs; (iii) Pnmt and the role of catecholamines in the heart; similarities and dissimilarities of PdCMs and canonical CMs; and (iv) putative functions of PdCMs in both physiological and pathological states and future directions, such as in intra-cardiac adrenergic signalling.
Collapse
Affiliation(s)
| | - Xianghong Ou
- Key Laboratory of Medical Electrophysiology of the Ministry of Education and Institute of Cardiovascular Research, Southwest Medical University, Luzhou 6400, People's Republic of China
| | - Ming Lei
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
- Key Laboratory of Medical Electrophysiology of the Ministry of Education and Institute of Cardiovascular Research, Southwest Medical University, Luzhou 6400, People's Republic of China
| |
Collapse
|
13
|
Sadhu N, Jhun EH, Posen A, Yao Y, He Y, Molokie RE, Wilkie DJ, Wang ZJ. Phenylethanolamine N-methyltransferase gene polymorphisms associate with crisis pain in sickle cell disease patients. Pharmacogenomics 2020; 21:269-278. [PMID: 32162598 DOI: 10.2217/pgs-2019-0096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Phenylethanolamine N-methyltransferase (PNMT) catalyzes the conversion of sympathetic neurotransmitter norepinephrine to epinephrine. We examined the association of PNMT polymorphisms with acute and chronic pain in sickle cell disease (SCD). Methods: Utilization of emergency care owing to painful crisis was used as a marker for acute pain in 131 patients with SCD. Results: rs876493 A allele, rs2934965 T allele and rs2941523 G allele were significantly associated with decreased utilization (p ≤ 0.05). rs876493 A allele showed association with utilization in females (p = 0.003), not males (p = 0.803). rs2934965 T allele and rs2941523 G allele were predicted to cause loss of putative transcription factor binding sites. This is the first report of the association of PNMT polymorphisms with acute crisis pain in SCD. Together with our previous findings in catechol-o-methyltransferase, polymorphisms in catecholamine metabolizing enzymes appear to primarily influence acute pain in SCD.
Collapse
Affiliation(s)
- Nilanjana Sadhu
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago College of Pharmacy, Chicago, IL 60607, USA
| | - Ellie H Jhun
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago College of Pharmacy, Chicago, IL 60607, USA
| | - Andrew Posen
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago College of Pharmacy, Chicago, IL 60607, USA
| | - Yingwei Yao
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing, Gainesville, FL, USA
| | - Ying He
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago College of Pharmacy, Chicago, IL 60607, USA.,Comprehensive Sickle Cell Center, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Robert E Molokie
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago College of Pharmacy, Chicago, IL 60607, USA.,Comprehensive Sickle Cell Center, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Jesse Brown Veteran's Administration Medical Center, Chicago, IL, USA.,Division of Hematology/Oncology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Diana J Wilkie
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing, Gainesville, FL, USA
| | - Zaijie J Wang
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago College of Pharmacy, Chicago, IL 60607, USA.,Comprehensive Sickle Cell Center, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
14
|
Fan X, Sun T, Crawford W, Tan X, Ou X, Terrar DA, Ebert SN, Lei M. Pnmt-Derived Cardiomyocytes: Anatomical Localization, Function and Future Perspectives. Front Physiol 2019; 10:713. [PMID: 31354504 PMCID: PMC6635595 DOI: 10.3389/fphys.2019.00713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 05/21/2019] [Indexed: 12/15/2022] Open
Abstract
In this mini-review, we provide an overview of phenylethanolamine-N-methyl transferase (Pnmt)-derived cardiomyocytes (PdCMs), a recently discovered cardiomyocyte subpopulation. We discuss their anatomical localization, physiological characteristics, possible function, and future perspectives. Their unique distribution in the heart, electrical activity, Ca2+ transient properties, and potential role in localized adrenergic signaling are discussed.
Collapse
Affiliation(s)
- Xuehui Fan
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Tianyi Sun
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - William Crawford
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Xiaoqiu Tan
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xianhong Ou
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Derek A. Terrar
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Steven N. Ebert
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Ming Lei
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
15
|
Elkhatib SK, Case AJ. Autonomic regulation of T-lymphocytes: Implications in cardiovascular disease. Pharmacol Res 2019; 146:104293. [PMID: 31176794 DOI: 10.1016/j.phrs.2019.104293] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/22/2019] [Accepted: 05/31/2019] [Indexed: 12/20/2022]
Abstract
The nervous and immune systems both serve as essential assessors and regulators of physiological function. Recently, there has been a great interest in how the nervous and immune systems interact to modulate both physiological and pathological states. In particular, the autonomic nervous system has a direct line of communication with immune cells anatomically, and moreover, immune cells possess receptors for autonomic neurotransmitters. This circumstantial evidence is suggestive of a functional interplay between the two systems, and extensive research over the past few decades has demonstrated neurotransmitters such as the catecholamines (i.e. dopamine, norepinephrine, and epinephrine) and acetylcholine have potent immunomodulating properties. Furthermore, immune cells, particularly T-lymphocytes, have now been found to express the cellular machinery for both the synthesis and degradation of neurotransmitters, which suggests the ability for both autocrine and paracrine signaling from these cells independent of the nervous system. The details underlying the functional interplay of this complex network of neuroimmune communication are still unclear, but this crosstalk is suggestive of significant implications on the pathogenesis of a number of autonomic-dysregulated and inflammation-mediated diseases. In particular, it is widely accepted that numerous forms of cardiovascular diseases possess imbalanced autonomic tone as well as altered T-lymphocyte function, but a paucity of literature exists discussing the direct role of neurotransmitters in shaping the inflammatory microenvironment during the progression or therapeutic management of these diseases. This review seeks to provide a fundamental framework for this autonomic neuroimmune interaction within T-lymphocytes, as well as the implications this may have in cardiovascular diseases.
Collapse
Affiliation(s)
- Safwan K Elkhatib
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Adam J Case
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States.
| |
Collapse
|
16
|
Vaitkus JA, Smith SC, Boikos SA, Celi FS. Unusual Case of Autoimmune Diabetes Mellitus in the Setting of Extra-Adrenal Paraganglioma with Loss of Succinate Dehydrogenase Expression. AACE Clin Case Rep 2018. [DOI: 10.4158/accr-2018-0072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
17
|
Okoshi MP, Cezar MD, Okoshi K. Adrenaline: More than a century after its discovery and still a mystery. Int J Cardiol 2018; 253:124-125. [DOI: 10.1016/j.ijcard.2017.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 11/02/2017] [Indexed: 11/29/2022]
|
18
|
Stratton CF, Poulin MB, Du Q, Schramm VL. Kinetic Isotope Effects and Transition State Structure for Human Phenylethanolamine N-Methyltransferase. ACS Chem Biol 2017; 12:342-346. [PMID: 27997103 PMCID: PMC5553282 DOI: 10.1021/acschembio.6b00922] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Phenylethanolamine N-methyltransferase (PNMT) catalyzes the S-adenosyl-l-methionine (SAM)-dependent conversion of norepinephrine to epinephrine. Epinephrine has been associated with critical processes in humans including the control of respiration and blood pressure. Additionally, PNMT activity has been suggested to play a role in hypertension and Alzheimer's disease. In the current study, labeled SAM substrates were used to measure primary methyl-14C and 36S and secondary methyl-3H, 5'-3H, and 5'-14C intrinsic kinetic isotope effects for human PNMT. The transition state of human PNMT was modeled by matching kinetic isotope effects predicted via quantum chemical calculations to intrinsic values. The model provides information on the geometry and electrostatics of the human PNMT transition state structure and indicates that human PNMT catalyzes the formation of epinephrine through an early SN2 transition state in which methyl transfer is rate-limiting.
Collapse
Affiliation(s)
- Christopher F. Stratton
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | | | - Quan Du
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | | |
Collapse
|
19
|
New Insights Into the Roles of Retinoic Acid Signaling in Nervous System Development and the Establishment of Neurotransmitter Systems. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 330:1-84. [PMID: 28215529 DOI: 10.1016/bs.ircmb.2016.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Secreted chiefly from the underlying mesoderm, the morphogen retinoic acid (RA) is well known to contribute to the specification, patterning, and differentiation of neural progenitors in the developing vertebrate nervous system. Furthermore, RA influences the subtype identity and neurotransmitter phenotype of subsets of maturing neurons, although relatively little is known about how these functions are mediated. This review provides a comprehensive overview of the roles played by RA signaling during the formation of the central and peripheral nervous systems of vertebrates and highlights its effects on the differentiation of several neurotransmitter systems. In addition, the evolutionary history of the RA signaling system is discussed, revealing both conserved properties and alternate modes of RA action. It is proposed that comparative approaches should be employed systematically to expand our knowledge of the context-dependent cellular mechanisms controlled by the multifunctional signaling molecule RA.
Collapse
|
20
|
Peltsch H, Khurana S, Byrne CJ, Nguyen P, Khaper N, Kumar A, Tai TC. Cardiac phenylethanolamine N-methyltransferase: localization and regulation of gene expression in the spontaneously hypertensive rat. Can J Physiol Pharmacol 2015; 94:363-72. [PMID: 26761434 DOI: 10.1139/cjpp-2015-0303] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Phenylethanolamine N-methyltransferase (PNMT) is the terminal enzyme in the catecholamine biosynthetic pathway responsible for adrenaline biosynthesis. Adrenaline is involved in the sympathetic control of blood pressure; it augments cardiac function by increasing stroke volume and cardiac output. Genetic mapping studies have linked the PNMT gene to hypertension. This study examined the expression of cardiac PNMT and changes in its transcriptional regulators in the spontaneously hypertensive (SHR) and wild type Wistar-Kyoto (WKY) rats. SHR exhibit elevated levels of corticosterone, and lower levels of the cytokine IL-1β, revealing systemic differences between SHR and WKY. PNMT mRNA was significantly increased in all chambers of the heart in the SHR, with the greatest increase in the right atrium. Transcriptional regulators of the PNMT promoter show elevated expression of Egr-1, Sp1, AP-2, and GR mRNA in all chambers of the SHR heart, while protein levels of Sp1, Egr-1, and GR were elevated only in the right atrium. Interestingly, only AP-2 protein-DNA binding was increased, suggesting it may be a key regulator of cardiac PNMT in SHR. This study provides the first insights into the molecular mechanisms involved in the dysregulation of cardiac PNMT in a genetic model of hypertension.
Collapse
Affiliation(s)
- Heather Peltsch
- a Department of Biology, Laurentian University, Sudbury, ON, Canada
| | - Sandhya Khurana
- e Medical Sciences Division, Northern Ontario School of Medicine, East Campus, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada
| | - Collin J Byrne
- a Department of Biology, Laurentian University, Sudbury, ON, Canada
| | - Phong Nguyen
- a Department of Biology, Laurentian University, Sudbury, ON, Canada
| | - Neelam Khaper
- d Medical Sciences Division, Northern Ontario School of Medicine, Thunder Bay, ON, Canada
| | - Aseem Kumar
- b Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON, Canada.,c Biomolecular Sciences, Laurentian University, Sudbury, ON, Canada
| | - T C Tai
- a Department of Biology, Laurentian University, Sudbury, ON, Canada.,b Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON, Canada.,c Biomolecular Sciences, Laurentian University, Sudbury, ON, Canada.,e Medical Sciences Division, Northern Ontario School of Medicine, East Campus, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
21
|
Xia J, Varudkar N, Baker CN, Abukenda I, Martinez C, Natarajan A, Grinberg A, Pfeifer K, Ebert SN. Targeting of the enhanced green fluorescent protein reporter to adrenergic cells in mice. Mol Biotechnol 2013; 54:350-60. [PMID: 22706789 PMCID: PMC11104505 DOI: 10.1007/s12033-012-9570-3] [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] [Indexed: 10/28/2022]
Abstract
Adrenaline and noradrenaline are important neurotransmitter hormones that mediate physiological stress responses in adult mammals, and are essential for cardiovascular function during a critical period of embryonic/fetal development. In this study, we describe a novel mouse model system for identifying and characterizing adrenergic cells. Specifically, we generated a reporter mouse strain in which a nuclear-localized enhanced green fluorescent protein gene (nEGFP) was inserted into exon 1 of the gene encoding Phenylethanolamine n-methyltransferase (Pnmt), the enzyme responsible for production of adrenaline from noradrenaline. Our analysis demonstrates that this knock-in mutation effectively marks adrenergic cells in embryonic and adult mice. We see expression of nEGFP in Pnmt-expressing cells of the adrenal medulla in adult animals. We also note that nEGFP expression recapitulates the restricted expression of Pnmt in the embryonic heart. Finally, we show that nEGFP and Pnmt expressions are each induced in parallel during the in vitro differentiation of pluripotent mouse embryonic stem cells into beating cardiomyocytes. Thus, this new mouse genetic model should be useful for the identification and functional characterization of adrenergic cells in vitro and in vivo.
Collapse
Affiliation(s)
- Jixiang Xia
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Baba Y, Nakajo M, Hayashi S. Adrenal venous catecholamine concentrations in patients with adrenal masses other than pheochromocytoma. Endocrine 2013; 43:219-24. [PMID: 22971951 DOI: 10.1007/s12020-012-9792-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 09/04/2012] [Indexed: 11/25/2022]
Abstract
The aim of this study was to retrospectively examine adrenal venous catecholamine concentrations and related indices in non-pheochromocytoma patients and to estimate from the obtained results whether measurements of adrenal venous catecholamine concentrations by adrenal venous sampling (AVS) are useful for localizing adrenal pheochromocytoma. The study population comprised 15 patients in whom AVS was performed for evaluation of adrenal non-pheochromocytoma masses (primary aldosteronism, n = 8; Cushing syndrome, n = 5; non-hyperfunctioning tumor, n = 2) without hormonal intervention and was successful in bilaterally judging adrenal vein to infra-renal inferior vena cava cortisol ratios as >3.0. Wide ranges of catecholamine concentrations were seen for both right (epinephrine, 35-175,821 pg/ml; norepinephrine, 115-32,102 pg/ml; dopamine, 9-232 pg/ml) and left (epinephrine, 16-27,251 pg/ml; norepinephrine, 155-9,267 pg/ml; dopamine, 5-234 pg/ml) adrenal veins. High- to low-side adrenal vein concentration ratios also showed wide ranges of up to 779 for epinephrine, 22.5 for norepinephrine, and 7.8 for dopamine. Adrenal venous catecholamine concentrations obtained by AVS and simple comparisons between bilateral adrenal veins might not be useful to localize adrenal pheochromocytoma, as wide variations in concentrations and high- to low-side adrenal vein concentration ratios were noted in patients with adrenal non-pheochromocytoma.
Collapse
Affiliation(s)
- Yasutaka Baba
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan.
| | | | | |
Collapse
|
23
|
Stress-triggered changes in peripheral catecholaminergic systems. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2013; 68:359-97. [PMID: 24054153 DOI: 10.1016/b978-0-12-411512-5.00017-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The sympathetic nervous system not only regulates cardiovascular and metabolic responses to stress but also is altered by stress. The sympathoneural and sympathoadrenomedullary systems are modified by different metabolic pathways and have different responses to short- and to long-term stressors. Stress also induces nonneuronal catecholamine enzymes, primarily through corticosteroids. Catecholamine synthetic enzymes are induced by different pathways in response to short- and long-term acting stressors, like cold exposure or immobilization, and differently in the sympathetic ganglia and the adrenal medulla. However, a long-term exposure to one stressor can increase the response to a second, different stressor. Tyrosine hydroxylase gene transcription increases after only 5min of immobilization through phosphorylation of CREB, but this response is short lived. However, repeated stress gives a longer-lived response utilizing transcription factors such as Egr-1 and Fra-2. Glucocorticoids and ACTH also induce sympathoneural enzymes leading to distinct patterns of short-term and long-lived activation of the sympathetic nervous system. Nonneuronal phenylethanolamine N-methyltransferase (PNMT) develops early in the heart and then diminishes. However, intrinsic cardiac adrenergic cells remain and nonneuronal PNMT is present in many cells of the adult organism and increases in response to glucocorticoids. Both stress-induced and administered glucocorticoids induce fetal PNMT and hypertension. Human stressors such as caring for an ill spouse or sleep apnea cause a persistent increase in blood norepinephrine, increased blood pressure, and downregulated catecholamine receptors. Hypertension is associated with a loss of slow-wave sleep, when sympathetic nerve activity is lowest. These findings indicate that stress-induced alteration of the sympathetic nervous system occurs in man as in experimental animals.
Collapse
|
24
|
Marino F, Cosentino M. Adrenergic modulation of immune cells: an update. Amino Acids 2011; 45:55-71. [PMID: 22160285 DOI: 10.1007/s00726-011-1186-6] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 11/23/2011] [Indexed: 12/25/2022]
Abstract
Sympathoadrenergic pathways are crucial to the communication between the nervous system and the immune system. The present review addresses emerging issues in the adrenergic modulation of immune cells, including: the specific pattern of adrenoceptor expression on immune cells and their role and changes upon cell differentiation and activation; the production and utilization of noradrenaline and adrenaline by immune cells themselves; the dysregulation of adrenergic immune mechanisms in disease and their potential as novel therapeutic targets. A wide array of sympathoadrenergic therapeutics is currently used for non-immune indications, and could represent an attractive source of non-conventional immunomodulating agents.
Collapse
Affiliation(s)
- Franca Marino
- Department of Clinical Medicine, Section of Experimental and Clinical Pharmacology, University of Insubria, Via Ottorino Rossi n. 9, 21100 Varese, VA, Italy
| | | |
Collapse
|
25
|
Osuala K, Telusma K, Khan SM, Wu S, Shah M, Baker C, Alam S, Abukenda I, Fuentes A, Seifein HB, Ebert SN. Distinctive left-sided distribution of adrenergic-derived cells in the adult mouse heart. PLoS One 2011; 6:e22811. [PMID: 21818395 PMCID: PMC3144959 DOI: 10.1371/journal.pone.0022811] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2011] [Accepted: 07/06/2011] [Indexed: 11/23/2022] Open
Abstract
Adrenaline and noradrenaline are produced within the heart from neuronal and non-neuronal sources. These adrenergic hormones have profound effects on cardiovascular development and function, yet relatively little information is available about the specific tissue distribution of adrenergic cells within the adult heart. The purpose of the present study was to define the anatomical localization of cells derived from an adrenergic lineage within the adult heart. To accomplish this, we performed genetic fate-mapping experiments where mice with the cre-recombinase (Cre) gene inserted into the phenylethanolamine-n-methyltransferase (Pnmt) locus were cross-mated with homozygous Rosa26 reporter (R26R) mice. Because Pnmt serves as a marker gene for adrenergic cells, offspring from these matings express the β-galactosidase (βGAL) reporter gene in cells of an adrenergic lineage. βGAL expression was found throughout the adult mouse heart, but was predominantly (89%) located in the left atrium (LA) and ventricle (LV) (p<0.001 compared to RA and RV), where many of these cells appeared to have cardiomyocyte-like morphological and structural characteristics. The staining pattern in the LA was diffuse, but the LV free wall displayed intermittent non-random staining that extended from the apex to the base of the heart, including heavy staining of the anterior papillary muscle along its perimeter. Three-dimensional computer-aided reconstruction of XGAL+ staining revealed distribution throughout the LA and LV, with specific finger-like projections apparent near the mid and apical regions of the LV free wall. These data indicate that adrenergic-derived cells display distinctive left-sided distribution patterns in the adult mouse heart.
Collapse
Affiliation(s)
- Kingsley Osuala
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States of America
| | - Kathleen Telusma
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States of America
| | - Saad M. Khan
- School of Electrical Engineering and Computer Science, College of Engineering and Computer Science, University of Central Florida, Orlando, Florida, United States of America
| | - Shandong Wu
- School of Electrical Engineering and Computer Science, College of Engineering and Computer Science, University of Central Florida, Orlando, Florida, United States of America
| | - Mubarak Shah
- School of Electrical Engineering and Computer Science, College of Engineering and Computer Science, University of Central Florida, Orlando, Florida, United States of America
| | - Candice Baker
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States of America
| | - Sabikha Alam
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States of America
| | - Ibrahim Abukenda
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States of America
| | - Aura Fuentes
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States of America
| | - Hani B. Seifein
- Florida Heart Group, Orlando, Florida, United States of America
| | - Steven N. Ebert
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States of America
- * E-mail:
| |
Collapse
|
26
|
Wu XM, Hu CP, Li XZ, Zou YQ, Zou JT, Li YY, Feng JT. Asthma pregnancy alters postnatal development of chromaffin cells in the rat adrenal medulla. PLoS One 2011; 6:e20337. [PMID: 21647384 PMCID: PMC3103586 DOI: 10.1371/journal.pone.0020337] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 04/29/2011] [Indexed: 11/26/2022] Open
Abstract
Background Adrenal neuroendocrine plays an important role in asthma. The activity of the
sympathoadrenal system could be altered by early life events. The effects of
maternal asthma during pregnancy on the adrenal medulla of offspring remain
unknown. Methodology/Principal Findings This study aims to explore the influence of maternal asthma during pregnancy
on the development and function of adrenal medulla in offspring from
postnatal day 3 (P3) to postnatal day 60 (P60). Asthmatic pregnant rats
(AP), nerve growth factor (NGF)-treated pregnant rats (NP) and NGF
antibody-treated pregnant rats (ANP) were sensitized and challenged with
ovalbumin (OVA); NP and ANP were treated with NGF and NGF antibody
respectively. Offspring rats from the maternal group were divided into four
groups: offspring from control pregnant rats (OCP), offspring from AP (OAP),
offspring from NP (ONP), and offspring from ANP (OANP). The expressions of
phenylethanolamine N-methyltransferase (PNMT) protein in adrenal medulla
were analyzed. The concentrations of epinephrine (EPI), corticosterone and
NGF in serum were measured. Adrenal medulla chromaffin cells (AMCC) were
prone to differentiate into sympathetic nerve cells in OAP and ONP. Both EPI
and PNMT were decreased in OAP from P3 to P14, and then reached normal level
gradually from P30 to P60, which were lower from birth to adulthood in ONP.
Corticosterone concentration increased significantly in OAP and ONP. Conclusion/Significance Asthma pregnancy may promote AMCC to differentiate into sympathetic neurons
in offspring rats and inhibit the synthesis of EPI, resulting in dysfunction
of bronchial relaxation.
Collapse
Affiliation(s)
- Xiu-Ming Wu
- Department of Respiratory Medicine, Xiangya Hospital, Central South
University, Changsha, Hunan, China
| | - Cheng-Ping Hu
- Department of Respiratory Medicine, Xiangya Hospital, Central South
University, Changsha, Hunan, China
| | - Xiao-Zhao Li
- Department of Respiratory Medicine, Xiangya Hospital, Central South
University, Changsha, Hunan, China
| | - Ye-Qiang Zou
- Department of Respiratory Medicine, Xiangya Hospital, Central South
University, Changsha, Hunan, China
| | - Jun-Tao Zou
- Department of Respiratory Medicine, Xiangya Hospital, Central South
University, Changsha, Hunan, China
| | - Yuan-Yuan Li
- Department of Respiratory Medicine, Xiangya Hospital, Central South
University, Changsha, Hunan, China
| | - Jun-Tao Feng
- Department of Respiratory Medicine, Xiangya Hospital, Central South
University, Changsha, Hunan, China
- * E-mail:
| |
Collapse
|
27
|
Spindler V, Waschke J. Beta-adrenergic stimulation contributes to maintenance of endothelial barrier functions under baseline conditions. Microcirculation 2011; 18:118-27. [PMID: 21166930 DOI: 10.1111/j.1549-8719.2010.00072.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVES cAMP signaling within the endothelium is known to reduce paracellular permeability and to protect against loss of barrier functions under various pathological conditions. Because activation of β-adrenergic receptors elevates cellular cAMP, we tested whether β-adrenergic receptor signaling contributes to the maintenance of baseline endothelial barrier properties. METHODS We compared hydraulic conductivity of rat postcapillary venules in vivo with resistance measurements and with reorganization of endothelial adherens junctions in cultured microvascular endothelial cells downstream of β-adrenergic receptor-mediated changes of cAMP levels. RESULTS Inhibition of β-adrenergic receptors by propranolol increased hydraulic conductivity, reduced both cAMP levels and TER of microvascular endothelial cell monolayers and induced fragmentation of VE-cadherin staining. In contrast, activation by epinephrine both increased cAMP levels and TER and resulted in linearized VE-cadherin distribution, however this was not sufficient to block barrier-destabilization by propranolol. Similarly, PDE inhibition did not prevent propranolol-induced TER reduction and VE-cadherin reorganization whereas increased cAMP formation by AC activation enhanced endothelial barrier functions under baseline conditions and under conditions of propranolol treatment. CONCLUSIONS Our results indicate that generation of cAMP mediated by activation of β-adrenergic receptor signaling contributes to the maintenance of endothelial barrier properties under baseline conditions.
Collapse
Affiliation(s)
- Volker Spindler
- Institute of Anatomy and Cell Biology, University of Würzburg, Koellikerstraße 6, Würzburg, Germany
| | | |
Collapse
|
28
|
Kvetnansky R, Sabban EL, Palkovits M. Catecholaminergic systems in stress: structural and molecular genetic approaches. Physiol Rev 2009; 89:535-606. [PMID: 19342614 DOI: 10.1152/physrev.00042.2006] [Citation(s) in RCA: 375] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Stressful stimuli evoke complex endocrine, autonomic, and behavioral responses that are extremely variable and specific depending on the type and nature of the stressors. We first provide a short overview of physiology, biochemistry, and molecular genetics of sympatho-adrenomedullary, sympatho-neural, and brain catecholaminergic systems. Important processes of catecholamine biosynthesis, storage, release, secretion, uptake, reuptake, degradation, and transporters in acutely or chronically stressed organisms are described. We emphasize the structural variability of catecholamine systems and the molecular genetics of enzymes involved in biosynthesis and degradation of catecholamines and transporters. Characterization of enzyme gene promoters, transcriptional and posttranscriptional mechanisms, transcription factors, gene expression and protein translation, as well as different phases of stress-activated transcription and quantitative determination of mRNA levels in stressed organisms are discussed. Data from catecholamine enzyme gene knockout mice are shown. Interaction of catecholaminergic systems with other neurotransmitter and hormonal systems are discussed. We describe the effects of homotypic and heterotypic stressors, adaptation and maladaptation of the organism, and the specificity of stressors (physical, emotional, metabolic, etc.) on activation of catecholaminergic systems at all levels from plasma catecholamines to gene expression of catecholamine enzymes. We also discuss cross-adaptation and the effect of novel heterotypic stressors on organisms adapted to long-term monotypic stressors. The extra-adrenal nonneuronal adrenergic system is described. Stress-related central neuronal regulatory circuits and central organization of responses to various stressors are presented with selected examples of regulatory molecular mechanisms. Data summarized here indicate that catecholaminergic systems are activated in different ways following exposure to distinct stressful stimuli.
Collapse
Affiliation(s)
- Richard Kvetnansky
- Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | | | | |
Collapse
|
29
|
Sun P, Bao X, Elayan H, Milic M, Liu F, Ziegler MG. Epinephrine regulation of hemodynamics in catecholamine knockouts and the pithed mouse. Ann N Y Acad Sci 2009; 1148:325-30. [PMID: 19120125 DOI: 10.1196/annals.1410.078] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Phenylethanolamine N-methyltransferase (PNMT) catalyzes synthesis of epinephrine (E) and is present in the brain, heart, and adrenal. E is a neurotransmitter and important hormone; however, its role in regulating cardiovascular dynamics is still unclear. We generated an E-deficient mouse model by knocking out the PNMT gene. The PNMT KO mouse had normal resting blood pressure, while treadmill exercise caused hypertension, suggesting an impaired response to stress in the absence of the stress hormone E. As PNMT occurs at a lower concentration in many extra-adrenal tissues including the brain, we set up a pithed mouse model to study the peripheral effects of E on cardiovascular dynamics, using pithing to eliminate central and reflex effects. The pithed mouse requires different surgical techniques and stimulation voltages than rats, and showed voltage- and frequency-dependent blood pressure responses to electrical stimuli. Stimulation with the alpha-adrenergic agonist phenylephrine gave a marked systolic pressor response, while the beta2 agonist salbutamol lowered diastolic blood pressure. The pithed PNMT KO mouse had an exaggerated blood pressure response to salbutamol, suggesting beta2 receptor supersensitivity. A targeted KO of tyrosine hydroxylase in PNMT-producing cells produced a mouse deficient in catecholamines in the adrenal. These targeted KO mice displayed significantly smaller pressor responses than pithed control mice. We find that E release during stress prevents an excessive increase in blood pressure.
Collapse
Affiliation(s)
- Ping Sun
- Department of Medicine, University of California at San Diego, San Diego, California 92103-8341, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Bakos J, Bobryshev P, Tillinger A, Kvet��ansk�� R, Jezova D. PhenylethanolamineN-Methyltransferase Gene Expression in the Heart and Blood Pressure Response to Oxytocin Treatment in Rats Exposed to Voluntary Wheel Running. Ann N Y Acad Sci 2008; 1148:302-7. [DOI: 10.1196/annals.1410.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
31
|
Wong DL, Tai TC, Wong-Faull DC, Claycomb R, Kvetnanský R. Adrenergic responses to stress: transcriptional and post-transcriptional changes. Ann N Y Acad Sci 2008; 1148:249-56. [PMID: 19120117 PMCID: PMC2722431 DOI: 10.1196/annals.1410.048] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stress effects on adrenergic responses in rats were examined in adrenal medulla, the primary source of circulating epinephrine (Epi). Irrespective of duration, immobilization (IMMO) increased adrenal corticosterone to the same extent. In contrast, Epi changed little, suggesting that Epi synthesis replenishes adrenal pools and sustains circulating levels for the heightened alertness and physiological changes required of the "flight or fight" response. IMMO also induced the Epi-synthesizing enzyme, phenylethanolamine N-methyltransferase (PNMT). The rise in its mRNA and protein was preceded by increases in Egr-1 and Sp1 mRNA, protein, and protein-DNA binding complex formation. With repeated and prolonged stress, PNMT protein did not reflect the magnitude of change in mRNA. The latter suggests that post-transcriptional, in addition to transcriptional mechanisms, regulate PNMT responses to stress. To further reveal molecular mechanisms underlying stress-induced changes in adrenergic function, the effects of hypoxia on PNMT promoter-driven gene expression are being examined in adrenal medulla-derived PC12 cells. Hypoxia activates the PNMT promoter to increase PNMT promoter-driven luciferase reporter gene expression and endogenous PNMT in PC12 cells. Induction of both appear mediated via activation of multiple signaling pathways and downstream activation of hypoxia inducible factor and PNMT transcriptional activators, Egr-1 and Sp1. Hypoxia generates both partially and fully processed forms of PNMT mRNA. The former reportedly is translated into a truncated, nonfunctional protein, and the latter into enzymatically active PNMT. Together, findings suggest that stress increases PNMT gene transcriptional activity but post-transcriptional regulatory mechanisms limit the biological end-point of functional PNMT enzyme and, thereby, Epi.
Collapse
Affiliation(s)
- Dona L Wong
- Department of Psychiatry, Harvard Medical School and Laboratory of Molecular and Developmental Neurobiology, McLean Hospital, Belmont, Massachusetts 02478, USA.
| | | | | | | | | |
Collapse
|
32
|
Haavik J, Blau N, Thöny B. Mutations in human monoamine-related neurotransmitter pathway genes. Hum Mutat 2008; 29:891-902. [PMID: 18444257 DOI: 10.1002/humu.20700] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Biosynthesis and metabolism of serotonin and catecholamines involve at least eight individual enzymes that are mainly expressed in tissues derived from the neuroectoderm, e.g., the central nervous system (CNS), pineal gland, adrenal medulla, enterochromaffin tissue, sympathetic nerves, and ganglia. Some of the enzymes appear to have additional biological functions and are also expressed in the heart and various other internal organs. The biosynthetic enzymes are tyrosine hydroxylase (TH), tryptophan hydroxylases type 1 and 2 (TPH1, TPH2), aromatic amino acid decarboxylase (AADC), dopamine beta-hydroxylase (DbetaH), and phenylethanolamine N-methyltransferase (PNMT), and the specific catabolic enzymes are monoamine oxidase A (MAO-A) and catechol O-methyltransferase (COMT). For the TH, DDC, DBH, and MAOA genes, many single nucleotide polymorphisms (SNPs) with unknown function, and small but increasing numbers of cases with autosomal recessive mutations have been recognized. For the remaining genes (TPH1, TPH2, PNMT, and COMT) several different genetic markers have been suggested to be associated with regulation of mood, pain perception, and aggression, as well as psychiatric disturbances such as schizophrenia, depression, suicidality, and attention deficit/hyperactivity disorder. The genetic markers may either have a functional role of their own, or be closely linked to other unknown functional variants. In the future, molecular testing may become important for the diagnosis of such conditions. Here we present an overview on mutations and polymorphisms in the group of genes encoding monoamine neurotransmitter metabolizing enzymes. At the same time we propose a unified nomenclature for the nucleic acid aberrations in these genes. New variations or details on mutations will be updated in the Pediatric Neurotransmitter Disorder Data Base (PNDDB) database (www.bioPKU.org).
Collapse
Affiliation(s)
- Jan Haavik
- Department of Biomedicine, University of Bergen, Norway
| | | | | |
Collapse
|
33
|
Bao X, Lu CM, Liu F, Gu Y, Dalton ND, Zhu BQ, Foster E, Chen J, Karliner JS, Ross J, Simpson PC, Ziegler MG. Epinephrine Is Required for Normal Cardiovascular Responses to Stress in the Phenylethanolamine N-Methyltransferase Knockout Mouse. Circulation 2007; 116:1024-31. [PMID: 17698731 DOI: 10.1161/circulationaha.107.696005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Epinephrine (EPI) is an important neurotransmitter and hormone. Its role in regulating cardiovascular function at rest and with stress is unclear, however.
Methods and Results—
An epinephrine-deficient mouse model was generated in which the epinephrine-synthesizing enzyme phenylethanolamine N-methyltransferase was knocked out (KO). Blood pressure and heart rate were monitored by telemetry at rest and during graded treadmill exercise. Cardiac structure and function were evaluated by echocardiography in mice under 1 of 2 conditions: unstressed and lightly anesthetized or restrained and awake. In KO mice, resting cardiovascular function, including blood pressure, heart rate, and cardiac output, was the same as that in wild-type mice, and the basal norepinephrine plasma level was normal. However, inhibition of sympathetic innervation with the ganglion blocker hexamethonium caused a 54% smaller decrease in blood pressure in KO mice, and treadmill exercise caused an 11% higher increase in blood pressure, both suggesting impaired vasodilation in KO mice. Interestingly, phenylethanolamine N-methyltransferase KO did not change the heart rate response to ganglionic blockade and exercise. By echocardiography, KO mice had an increased ratio of left ventricular posterior wall thickness to internal dimensions but did not have cardiac hypertrophy, suggesting concentric remodeling in the KO heart. Finally, in restrained, awake KO mice, heart rate and ejection fraction remained normal, but cardiac output was significantly reduced because of diminished end-diastolic volume.
Conclusion—
Our data suggest that epinephrine is required for normal blood pressure and cardiac filling responses to stress but is not required for tachycardia during stress or normal cardiovascular function at rest.
Collapse
Affiliation(s)
- Xuping Bao
- Department of Medicine, University of California San Diego, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Kepp K, Juhanson P, Kozich V, Ots M, Viigimaa M, Laan M. Resequencing PNMT in European hypertensive and normotensive individuals: no common susceptibilily variants for hypertension and purifying selection on intron 1. BMC MEDICAL GENETICS 2007; 8:47. [PMID: 17645789 PMCID: PMC1947951 DOI: 10.1186/1471-2350-8-47] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Accepted: 07/23/2007] [Indexed: 12/29/2022]
Abstract
BACKGROUND Human linkage and animal QTL studies have indicated the contribution of genes on Chr17 into blood pressure regulation. One candidate gene is PNMT, coding for phenylethanolamine-N-methyltransferase, catalyzing the synthesis of epinephrine from norepinephrine. METHODS Fine-scale variation of PNMT was screened by resequencing hypertensive (n = 50) and normotensive (n = 50) individuals from two European populations (Estonians and Czechs). The resulting polymorphism data were analyzed by statistical genetics methods using Genepop 3.4, PHASE 2.1 and DnaSP 4.0 software programs. In silico prediction of transcription factor binding sites for intron 1 was performed with MatInspector 2.2 software. RESULTS PNMT was characterized by minimum variation and excess of rare SNPs in both normo- and hypertensive individuals. None of the SNPs showed significant differences in allelic frequencies among population samples, as well as between screened hypertensives and normotensives. In the joint case-control analysis of the Estonian and the Czech samples, hypertension patients had a significant excess of heterozygotes for two promoter region polymorphisms (SNP-184; SNP-390). The identified variation pattern of PNMT reflects the effect of purifying selection consistent with an important role of PNMT-synthesized epinephrine in the regulation of cardiovascular and metabolic functions, and as a CNS neurotransmitter. A striking feature is the lack of intronic variation. In silico analysis of PNMT intron 1 confirmed the presence of a human-specific putative Glucocorticoid Responsive Element (GRE), inserted by Alu-mediated transfer. Further analysis of intron 1 supported the possible existence of a full Glucocorticoid Responsive Unit (GRU) predicted to consist of multiple gene regulatory elements known to cooperate with GRE in driving transcription. The role of these elements in regulating PNMT expression patterns and thus determining the dynamics of the synthesis of epinephrine is still to be studied. CONCLUSION We suggest that the differences in PNMT expression between normotensives and hypertensives are not determined by the polymorphisms in this gene, but rather by the interplay of gene expression regulators, which may vary among individuals. Understanding the determinants of PNMT expression may assist in developing PNMT inhibitors as potential novel therapeutics.
Collapse
Affiliation(s)
- Katrin Kepp
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Peeter Juhanson
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Viktor Kozich
- Institute of Inherited Metabolic Diseases, Charles University – First Faculty of Medicine, Prague, Czech Republic
| | - Mai Ots
- Department of Internal Medicine, University of Tartu, Tartu, Estonia
| | - Margus Viigimaa
- Division of Cardiology, Northern Estonian Regional Hospital, Tallinn, Estonia
| | - Maris Laan
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| |
Collapse
|
35
|
Kvetnansky R, Kubovcakova L, Tillinger A, Micutkova L, Krizanova O, Sabban EL. Gene expression of phenylethanolamine N-methyltransferase in corticotropin-releasing hormone knockout mice during stress exposure. Cell Mol Neurobiol 2006; 26:735-54. [PMID: 16691441 PMCID: PMC11520757 DOI: 10.1007/s10571-006-9063-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2006] [Accepted: 03/14/2006] [Indexed: 10/24/2022]
Abstract
AIMS Epinephrine (EPI) synthesizing enzyme phenylethanolamine N-methyltransferase (PNMT, EC 2.1.1.28) is primarily localized in the adrenal medulla (AM). We have recently described existence of the PNMT gene expression in cardiac atria and ventricles and in sympathetic ganglia of adult rats and mice. The aim of the present work was to study regulation of the PNMT gene expression in corticotropin-releasing hormone knockout mice (CRH KO) and matched control wild-type mice (WT) under normal and stress conditions. METHODS Levels of the PNMT mRNA were determined by RT-PCR; PNMT immunoprotein and protein of transcription factor EGR-1 by Western Blot. Plasma EPI and corticosterone (CORT) levels were determined by radioenzymatic and RIA methods. Immobilization (IMMO) was used as a stressor. RESULTS Stress-induced increases in the PNMT mRNA and protein levels observed in WT mice were almost completely absent in CRH KO mouse adrenal medulla, stellate ganglia, and cardiac atria, while ventricular PNMT mRNA elevation was not CRH-dependent. Plasma EPI and CORT levels were markedly reduced in CRH KO compared to WT mice both before and after the stress. Levels of EGR-1, crucial transcription factor for regulation of the PNMT were highly increased in stressed WT and CRH KO mice in cardiac areas, but not in the adrenal medulla. CONCLUSIONS Data show that the CRH deficiency can markedly prevent immobilization-triggered induction of the PNMT mRNA and protein levels in the adrenal medulla and stellate ganglia. Reduced plasma epinephrine and corticosterone levels and adrenal medullary EGR-1 protein levels in CRH knockout versus WT mice during stress indicate that the HPA axis plays a crucial role in regulation of the PNMT gene expression in these organs. Cardiac atrial PNMT gene expression with stress is also dependent on intact HPA axis. However, in cardiac ventricles, especially after the single stress exposure, its expression is not impaired by CRH deficiency. Since cardiac EGR-1 protein levels in CRH KO mice are also not affected by the single stress exposure, we propose existence of different regulation of the PNMT gene expression, especially in the cardiac ventricles.Overall, our findings reveal that the PNMT gene expression is regulated through the HPA in both sympathoadrenal system and the heart and also via EGR-1 in the adrenal medulla, but apparently not in the heart. Regulation of the PNMT gene expression in various compartments of heart includes both corticosterone-dependent and independent mechanisms.
Collapse
Affiliation(s)
- R Kvetnansky
- Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | | | | | | | | | | |
Collapse
|
36
|
Kubovcakova L, Micutkova L, Bartosova Z, Sabban EL, Krizanova O, Kvetnansky R. Identification of phenylethanolamine N-methyltransferase gene expression in stellate ganglia and its modulation by stress. J Neurochem 2006; 97:1419-30. [PMID: 16696852 DOI: 10.1111/j.1471-4159.2006.03832.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Phenylethanolamine N-methyltransferase (PNMT, EC 2.1.1.28) is the terminal enzyme of the catecholaminergic pathway converting noradrenaline to adrenaline. Although preferentially localized in adrenal medulla, evidence exists that PNMT activity and gene expression are also present in the rat heart, kidney, spleen, lung, skeletal muscle, thymus, retina and different parts of the brain. However, data concerning PNMT gene expression in sympathetic ganglia are still missing. In this study, our effort was focused on identification of PNMT mRNA and/or protein in stellate ganglia and, if present, testing the effect of stress on PNMT mRNA and protein levels in this type of ganglia. We identified both PNMT mRNA and protein in stellate ganglia of rats and mice, although in much smaller amounts compared with adrenal medulla. PNMT gene expression and protein levels were also increased after repeated stress exposure in stellate ganglia of rats and wild-type mice. Similarly to adrenal medulla, the immobilization-induced increase was probably regulated by glucocorticoids, as determined indirectly using corticotropin-releasing hormone knockout mice, where immobilization-induced increase of PNMT mRNA was suppressed. Thus, glucocorticoids might play an important role in regulation of PNMT gene expression in stellate ganglia under stress conditions.
Collapse
Affiliation(s)
- L Kubovcakova
- Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | | | | | | | | | | |
Collapse
|
37
|
Czermak C, Lehofer M, Liebmann PM, Traynor J. [35S]GTPgammaS binding at the human dopamine D4 receptor variants hD4.2, hD4.4 and hD4.7 following stimulation by dopamine, epinephrine and norepinephrine. Eur J Pharmacol 2006; 531:20-4. [PMID: 16423344 DOI: 10.1016/j.ejphar.2005.11.063] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2005] [Revised: 11/21/2005] [Accepted: 11/28/2005] [Indexed: 11/13/2022]
Abstract
Aim of the present study was to investigate possible differences between the human dopamine D4 receptor 48 bp polymorphism variants hD4.2, hD.4. and hD4.7 in agonist stimulated [35S]GTPgammaS binding, to investigate dopamine D4 receptor sodium sensitivity and to further characterize norepinephrine and epinephrine agonism at this receptor. G-protein activation at the receptor variants hD4.2, hD4.4 and hD4.7 expressed in CHO-K1 cells, following stimulation by dopamine, norepinephrine and epinephrine, was investigated using the [35S]GTPgammaS assay at experimental conditions of 10 and 100 mM sodium, respectively. Dopamine displayed a 2 fold higher potency of stimulating [35S]GTPgammaS binding at the hD4.2, compared to the hD4.4 and hD4.7 at 10 mM sodium. A significant difference in sodium sensitivity of basal [35S]GTPgammaS binding was found, with the hD4.7 being 1.7 fold more sensitive than the hD4.4 and 2.5 fold more sensitive than the hD4.2. Norepinephrine and epinephrine both produced concentration-dependent increases in [35S]GTPgammaS binding at all three receptor variants, and epinephrine showed only 2 fold less potency than dopamine. The present results are in certain line with previous reports of functional 2-3 fold differences between the dopamine D4 receptor variants. Agonism of norepinephrine and epinephrine at the dopamine D4 receptor may indicate an important way of cross-reactivity among the different monoamine neurotransmitter systems.
Collapse
Affiliation(s)
- Christoph Czermak
- Department of General Psychiatry I, Sigmund Freud Clinics Graz, Wagner Jauregg Platz 1, 8053 Graz, Austria.
| | | | | | | |
Collapse
|
38
|
Hoffman-Goetz L, Quadrilatero J, Boudreau J, Guan J. Adrenalectomy in mice does not prevent loss of intestinal lymphocytes after exercise. J Appl Physiol (1985) 2005; 96:2073-81. [PMID: 15133013 DOI: 10.1152/japplphysiol.01262.2003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Exhaustive exercise is associated with an increase in circulating glucocorticoids (GCs), lymphocyte apoptosis, and a reduction in intestinal lymphocyte number. The present study examined the role of GCs on the numerical changes seen in intestinal lymphocytes after exercise. Female C57BL/6 mice were bilaterally adrenalectomized (ADX; n = 18) or given sham surgery (Sham; n = 18) and assigned to one of three exercise conditions: treadmill running (28 m/min, 90 min, 2 degrees slope) and killed immediately or after 24 h recovery, or not exercised and killed immediately after 90-min exposure to the treadmill environment. Lymphocytes were isolated from the intestines with CD45(+) cells collected by positive selection using magnetic bead separation columns, and lymphocyte subpopulations were analyzed by flow cytometry for CD45(+), CD3alphabeta(+), CD3gammadelta(+), CD8beta(+), CD8alpha(+), CD4(+), and NK(+) phenotypic markers. ADX mice had significantly more intestinal CD45(+) leukocytes (P < 0.05) and CD3alphabeta(+) (P < 0.05), CD3gammadelta(+) (P < 0.01), CD8alpha(+) (P < 0.001), and NK(+) (P < 0.05) intestinal lymphocytes than Sham mice. There was a significant effect of exercise condition on total intestinal CD45(+) leukocytes (P < 0.01) and CD3alphabeta(+) (P < 0.05), CD8alpha(+) (P < 0.001), and CD4(+) (P < 0.05) intestinal lymphocytes, with fewer cells at 24 h postexercise compared with the other treatment conditions. There were no surgical x exercise interaction effects on the CD3 and CD8 phenotype numbers. Plasma corticosterone was virtually nil in ADX mice regardless of exercise condition but was significantly elevated in Sham mice immediately postexercise (P < 0.001). The data indicate that ADX does not prevent the loss of lymphocytes from the intestinal mucosa 24 h after strenuous exercise and GCs are not directly causal in the leukopenia of exercise.
Collapse
Affiliation(s)
- L Hoffman-Goetz
- Department of Health Studies and Gerontology, Faculty of Applied Health Sciences, University of Waterloo, Waterloo, Ontario, Canada N2L 3G1
| | | | | | | |
Collapse
|
39
|
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: 1.4] [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
|
40
|
Her S, Claycomb R, Tai TC, Wong DL. Regulation of the rat phenylethanolamine N-methyltransferase gene by transcription factors Sp1 and MAZ. Mol Pharmacol 2003; 64:1180-8. [PMID: 14573768 DOI: 10.1124/mol.64.5.1180] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The rat phenylethanolamine N-methyltransferase (PNMT) gene promoter contains 1-base pair (bp) overlapping consensus sequences for Sp1 and MAZ transcription factors at -48 and -38 bp, respectively. Gel mobility assays using PC-12-derived RS1 cell nuclear extracts or in vitro translated proteins showed that Sp1 and MAZ specifically bind to these elements, that MAZ displaces/prevents Sp1 binding, and that Sp1 and MAZ binding is mutually exclusive, with occupancy dependent on each factor's concentration and affinity for its consensus element. In transfection assays, PNMT promoter activation by Sp1 and MAZ depends on promoter length, with -893 bp of sequence yielding greatest activation. Although MAZ has higher affinity for its binding element, it is a less effective activator. Changes in PNMT promoter activity for the constructs pGL3RP60 or pGL3RP893 using a fixed amount of MAZ expression construct and a variable amount of Sp1 expression construct or vice versa confirmed the latter. Mutation of the MAZ or Sp1 sites in pGL3RP60 attenuated but did not eliminate PNMT promoter activity, even though the proteins no longer bind to their consensus elements. Phosphatase treatment of RS1 cell nuclear extracts prevented MAZ- and Sp1-DNA binding complex formation. Although MAZ and Sp1 elevate endogenous PNMT mRNA in RS1 cells, MAZ preferentially increases intron-retaining whereas Sp1 preferentially increases intronless mRNA. Thus, expression of the PNMT gene seems to be modulated through competitive binding of phosphorylated Sp1 and MAZ to their consensus elements in the promoter. In addition, post-transcriptional regulation seems to be another important mechanism controlling PNMT expression.
Collapse
Affiliation(s)
- Song Her
- Department of Psychiatry, Harvard Medical School Laboratory of Molecular and Developmental Neurobiology, McLean Hospital, 115 Mill Street, Rm 116, Belmont, MA 02478, USA
| | | | | | | |
Collapse
|