1
|
Hashmi AN, Ahmed Dharejo R, Zubair UB, Khan N, Kashif I, Ajmal M, Taj R, Qamar R, Azam M. Association of dopamine β-hydroxylase polymorphism rs1611115 and serum levels with psychiatric disorders in Pakistani population. Int J Neurosci 2024; 134:551-559. [PMID: 36120985 DOI: 10.1080/00207454.2022.2126774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/06/2022] [Accepted: 08/26/2022] [Indexed: 10/14/2022]
Abstract
AIM Dopamine β-hydroxylase (DBH) is a copper-containing enzyme that has an important role in maintaining the cellular homeostasis between the two neurotransmitters, dopamine (DA) and nor-adrenaline (NA). DBH functional polymorphisms are associated with multiple neuro-psychiatric conditions and are found to alter the DBH protein levels in serum affecting DBH enzymatic activity. The current study was conducted to determine the genetic and serum levels association of DBH rs1611115 functional polymorphism with major depressive disorder (MDD), bipolar disorder (BD) and schizophrenia (SHZ) in the Pakistani population. METHODS In total n = 1097 subjects including MDD (n = 427), BD (n = 204), SHZ (n = 134) and healthy controls (n = 332), were screened for the functional polymorphism by polymerase chain reaction-restriction fragment length polymorphism. Univariate logistic regression analysis was applied and the results were adjusted for age and sex. The DBH levels in serum were determined through enzyme-linked immunosorbent assay (ELISA) and the Mann Whitney U test was applied. RESULTS The minor allele (-1021 C > T) was found to be significantly associated with a higher risk of developing BD and SHZ in both univariable and multivariable analyses. The overall total serum concentration of DBH was comparatively raised in MDD, however, in cross-comparison DBH serum levels were found markedly higher in CC homozygotes compared to TT homozygotes within the BD group. CONCLUSION The present study suggested a significant association of DBH rs1611115 with BD and SHZ and also the effect of rs1611115 on DBH serum levels in MDD and BD for the first time in the Pakistani population.
Collapse
Affiliation(s)
- Aisha Nasir Hashmi
- Translational Genomics Laboratory, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Raees Ahmed Dharejo
- Department of Psychiatry, Pakistan Institute of Medical Sciences, Islamabad, Pakistan
- WAPDA Administrative Staff College, Islamabad, Pakistan
| | - Usama Bin Zubair
- Department of Psychiatry, Pakistan Institute of Medical Sciences, Islamabad, Pakistan
| | - Netasha Khan
- Translational Genomics Laboratory, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Iqra Kashif
- Translational Genomics Laboratory, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Muhammad Ajmal
- Translational Genomics Laboratory, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Rizwan Taj
- Department of Psychiatry, Pakistan Institute of Medical Sciences, Islamabad, Pakistan
| | - Raheel Qamar
- Pakistan Academy of Sciences, Islamabad, Pakistan
- Science and Technology Sector, ICESCO, Rabat, Morocco
| | - Maleeha Azam
- Translational Genomics Laboratory, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| |
Collapse
|
2
|
The regulatory role of AP-2β in monoaminergic neurotransmitter systems: insights on its signalling pathway, linked disorders and theragnostic potential. Cell Biosci 2022; 12:151. [PMID: 36076256 PMCID: PMC9461128 DOI: 10.1186/s13578-022-00891-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/28/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractMonoaminergic neurotransmitter systems play a central role in neuronal function and behaviour. Dysregulation of these systems gives rise to neuropsychiatric and neurodegenerative disorders with high prevalence and societal burden, collectively termed monoamine neurotransmitter disorders (MNDs). Despite extensive research, the transcriptional regulation of monoaminergic neurotransmitter systems is not fully explored. Interestingly, certain drugs that act on these systems have been shown to modulate central levels of the transcription factor AP-2 beta (AP-2β, gene: TFAP2Β). AP-2β regulates multiple key genes within these systems and thereby its levels correlate with monoamine neurotransmitters measures; yet, its signalling pathways are not well understood. Moreover, although dysregulation of TFAP2Β has been associated with MNDs, the underlying mechanisms for these associations remain elusive. In this context, this review addresses AP-2β, considering its basic structural aspects, regulation and signalling pathways in the controlling of monoaminergic neurotransmitter systems, and possible mechanisms underpinning associated MNDS. It also underscores the significance of AP-2β as a potential diagnostic biomarker and its potential and limitations as a therapeutic target for specific MNDs as well as possible pharmaceutical interventions for targeting it. In essence, this review emphasizes the role of AP-2β as a key regulator of the monoaminergic neurotransmitter systems and its importance for understanding the pathogenesis and improving the management of MNDs.
Collapse
|
3
|
Gonzalez‐Lopez E, Vrana KE. Dopamine beta‐hydroxylase and its genetic variants in human health and disease. J Neurochem 2019; 152:157-181. [DOI: 10.1111/jnc.14893] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/18/2019] [Accepted: 09/26/2019] [Indexed: 12/12/2022]
Affiliation(s)
| | - Kent E. Vrana
- Department of Pharmacology Penn State College of Medicine Hershey PA USA
| |
Collapse
|
4
|
Ohkubo N, Aoto M, Kon K, Mitsuda N. Lack of zinc finger protein 521 upregulates dopamine β-hydroxylase expression in the mouse brain, leading to abnormal behavior. Life Sci 2019; 231:116559. [PMID: 31200001 DOI: 10.1016/j.lfs.2019.116559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/22/2019] [Accepted: 06/10/2019] [Indexed: 10/26/2022]
Abstract
AIM Previously, we reported that mice deficient in most of the Zfp521 coding region (Zfp521Δ/Δ mice) displayed abnormal behaviors, including hyperlocomotion and lower anxiety. In this study, we aimed to elucidate the involvement and mechanisms of monoamine variation. MAIN METHODS First, we compared the levels of dopamine (DA), noradrenaline (NA), and serotonin in the brains of Zfp521Δ/Δ and Zfp521+/+ mice using enzyme-linked immunosorbent assay. Next, we elucidated the mechanisms using quantitative PCR and Western Blotting. Additionally, we administered inhibitory drug to the mice and performed behavioral tests. KEY FINDINGS Our results showed that the DA level decreased and the NA level increased in Zfp521Δ/Δ mice. We found that ZFP521 suppresses the expression of dopamine β-hydroxylase (DBH), which converts DA into NA. We also demonstrated that paired homeodomain transcription factor 2 and early growth response protein-1, which are the transcription factors for Dbh, were involved in the upregulation of Dbh by ZFP521. The administration of nepicastat, a specific inhibitor of DBH, attenuated the abnormal behaviors of Zfp521Δ/Δ mice. SIGNIFICANCE These results suggest that the lack of ZFP521 upregulates the expression of DBH, which leads to a decrease in the DA level and an increase in the NA level in the brain, resulting in abnormal behaviors.
Collapse
Affiliation(s)
- Nobutaka Ohkubo
- Department of Circulatory Physiology, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan..
| | - Mamoru Aoto
- Department of Circulatory Physiology, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Kazunori Kon
- Department of Circulatory Physiology, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Noriaki Mitsuda
- Department of Circulatory Physiology, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan
| |
Collapse
|
5
|
Sarnat HB, Flores-Sarnat L, Boltshauser E. Area Postrema: Fetal Maturation, Tumors, Vomiting Center, Growth, Role in Neuromyelitis Optica. Pediatr Neurol 2019; 94:21-31. [PMID: 30797593 DOI: 10.1016/j.pediatrneurol.2018.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/12/2018] [Accepted: 12/17/2018] [Indexed: 01/17/2023]
Abstract
INTRODUCTION The area postrema in the caudal fourth ventricular floor is highly vascular without blood-brain or blood-cerebrospinal fluid barrier. In addition to its function as vomiting center, several others are part of the circumventricular organs for vasomotor/angiotensin II regulation, role in neuromyelitis optica related to aquaporin-4, and somatic growth and appetite regulation. Functions are immature at birth. The purpose was to demonstrate neuronal, synaptic, glial, or ependymal maturation in the area postrema of normal fetuses. We describe three area postrema tumors. METHODS Sections of caudal fourth ventricle of 12 normal human fetal brains at autopsy aged six to 40 weeks and three infants aged three to 18 months were examined. Immunocytochemical neuronal and glial markers were applied to paraffin sections. Two infants with area postrema tumors and another with neurocutaneous melanocytosis and pernicious vomiting also studied. RESULTS Area postrema neurons exhibited cytologic maturity and synaptic circuitry by 14 weeks'. Astrocytes coexpressed vimentin, glial fibrillary acidic protein, and S-100β protein. The ependyma is thin over area postrema, with fetal ependymocytic basal processes. A glial layer separates area postrema from medullary tegmentum. Melanocytes infiltrated area postrema in the toddler with pernicious vomiting; two children had primary area postrema pilocytic astrocytomas. CONCLUSIONS Although area postrema is cytologically mature by 14 weeks, growth increases and functions mature during postnatal months. We recommend neuroimaging for patients with unexplained vomiting and that area postrema neuropathology includes synaptophysin and microtubule-associated protein-2 in patients with suspected dysfunction.
Collapse
Affiliation(s)
- Harvey B Sarnat
- Departments of Paediatrics, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada; Pathology (Neuropathology), University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada; Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada.
| | - Laura Flores-Sarnat
- Departments of Paediatrics, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada; Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Eugen Boltshauser
- Department of Paediatric Neurology, Children's University Hospital, Zürich, Switzerland
| |
Collapse
|
6
|
Arige V, Agarwal A, Khan AA, Kalyani A, Natarajan B, Gupta V, Reddy SS, Barthwal MK, Mahapatra NR. Regulation of Monoamine Oxidase B Gene Expression: Key Roles for Transcription Factors Sp1, Egr1 and CREB, and microRNAs miR-300 and miR-1224. J Mol Biol 2019; 431:1127-1147. [PMID: 30738894 DOI: 10.1016/j.jmb.2019.01.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/18/2019] [Accepted: 01/29/2019] [Indexed: 11/15/2022]
Abstract
Monoamine oxidase B (MAO-B), a flavoenzyme located in the outer mitochondrial membrane, is involved in the catabolism of monoamines. Altered levels of MAO-B are associated with cardiovascular/neuronal diseases. However, molecular mechanisms of MAO-B gene regulation are partially understood. We undertook a systematic analysis of the MAO-B gene to identify the key transcriptional/post-transcriptional regulatory molecules. Expression of MAO-B promoter-reporter constructs in cultured cells identified the -144/+25-bp domain as the core promoter region. Stringent in silico analysis of this core promoter predicted binding sites for several transcription factors. Over-expression/down-regulation of transcription factors Sp1/Egr1/CREB increased/decreased the MAO-B promoter-reporter activity and endogenous MAO-B protein level. Electrophoretic mobility shift assays and ChIP assays provided evidence for interactions of Sp1/Egr1/CREB with the MAO-B promoter. MAOB transcript level also positively correlated with the transcript level of Sp1/Egr1/CREB in various human tissue samples. Computational predictions using multiple algorithms coupled with systematic functional analysis revealed direct interactions of the microRNAs miR-1224 and miR-300 with MAO-B 3'-UTR. Dopamine dose-dependently enhanced MAO-B transcript and protein levels via increased binding of CREB to MAO-B promoter and reduced miR-1224/miR-300 levels. 8-Bromo-cAMP and forskolin augmented MAO-B expression, whereas inhibition of PKA diminished the gene expression suggesting involvement of cAMP-PKA axis. Interestingly, Sp1/Egr1/CREB/miR-1224 levels correlate with MAO-B expression in rodent models of hypertension/MPTP-induced neurodegeneration, indicating their roles in governing MAO-B gene expression in these disease states. Taken together, this study elucidates the previously unknown roles of the transcription factors Sp1/Egr1/CREB and microRNAs miR-1224/miR-300 in regulating MAO-B gene expression under basal/disease states involving dysregulated catecholamine levels.
Collapse
Affiliation(s)
- Vikas Arige
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Anshu Agarwal
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Abrar A Khan
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Ananthamohan Kalyani
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Bhargavi Natarajan
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Vinayak Gupta
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - S Santosh Reddy
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi 110025, India
| | - Manoj K Barthwal
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Nitish R Mahapatra
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India.
| |
Collapse
|
7
|
Fan Y, Chen P, Raza MU, Szebeni A, Szebeni K, Ordway GA, Stockmeier CA, Zhu MY. Altered Expression of Phox2 Transcription Factors in the Locus Coeruleus in Major Depressive Disorder Mimicked by Chronic Stress and Corticosterone Treatment In Vivo and In Vitro. Neuroscience 2018; 393:123-137. [PMID: 30315878 DOI: 10.1016/j.neuroscience.2018.09.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/21/2018] [Accepted: 09/26/2018] [Indexed: 12/16/2022]
Abstract
Phox2a and Phox2b are two homeodomain transcription factors playing a pivotal role in the development of noradrenergic neurons during the embryonic period. However, their expression and function in adulthood remain to be elucidated. Using human postmortem brain tissues, rat stress models and cultured cells, this study aimed to examine the alteration of Phox2a and Phox2b expression. The results show that Phox2a and Phox2b are normally expressed in the human locus coeruleus (LC) in adulthood. Furthermore, the levels of Phox2a protein and mRNA and protein levels of Phox2b were significantly elevated in the LC of brain donors that suffered from the major depressive disorder, as compared to age-matched and psychiatrically normal control donors. Fischer 344 rats subjected to chronic social defeat showed higher mRNA and protein levels of Phox2a and Phox2b in the LC, as compared to non-stressed control rats. In rats chronically administered oral corticosterone, mRNA and protein levels of Phox2b, but not Phox2a, in the LC were significantly increased. In addition, the corticosterone-induced increase in Phox2b protein was reversed by simultaneous treatment with either mifepristone or spironolactone. Exposing SH-SY5Y cells to corticosterone significantly increased expression of Phox2a and Phox2b, which was blocked by corticosteroid receptor antagonists. Taken together, these experiments reveal that Phox2 genes are expressed throughout the lifetime in the LC of humans and Fischer 344 rats. Alterations in their expression may play a role in major depressive disorder and possibly other stress-related disorders through their modulatory effects on the noradrenergic phenotype.
Collapse
Affiliation(s)
- Yan Fan
- Department of Biochemistry, Nantong University College of Medicine, Nantong, China
| | - Ping Chen
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Muhammad U Raza
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Attila Szebeni
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Katalin Szebeni
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Gregory A Ordway
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Craig A Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Meng-Yang Zhu
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.
| |
Collapse
|
8
|
Gai Y, Zhang J, Wei C, Cao W, Cui Y, Cui S. miR-375 negatively regulates the synthesis and secretion of catecholamines by targeting Sp1 in rat adrenal medulla. Am J Physiol Cell Physiol 2017; 312:C663-C672. [PMID: 28356269 DOI: 10.1152/ajpcell.00345.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 03/13/2017] [Accepted: 03/13/2017] [Indexed: 02/08/2023]
Abstract
The adrenal gland is an important endocrine gland in balancing homeostasis and the response to stress by synthesizing and secreting catecholamines (CATs), and it has been confirmed that microRNA-375 (miR-375) is highly expressed in adrenal medulla. However, up to now there are few reports about the functions and related mechanisms in adrenal medulla. The present study was thus designed to study the roles and related mechanisms in rat adrenal medulla. Our results showed that miR-375 was specifically expressed in rat adrenal medulla chromaffin cells, and its expression was downregulated when rats were exposed to stress. The further functional studies demonstrated that the inhibition of endogenous miR-375 induced the secretion of CATs in primary rat medulla chromaffin cells and PC12 cells, whereas miR-375 overexpression resulted in a decline of CAT secretion. In addition, our results showed that miR-375 negatively regulated tyrosine hydroxylase (TH) and dopamine-β-hydroxylase (DBH) and mediated adrenomedullary CAT biosynthesis. These functions of miR-375 were accomplished by its binding to the 3'-untranslated region of Sp1, which was involved in the regulation of TH and DBH expressions. These novel findings suggest that miR-375 acts as a potent negative mediator in regulating the synthesis and secretion of CATs in the adrenal medulla during the maintenance of homeostasis under stress.
Collapse
Affiliation(s)
- Yedan Gai
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China; and
| | - Jinglin Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China; and
| | - Chao Wei
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China; and
| | - Wei Cao
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China; and
| | - Yan Cui
- The 306th Hospital of People's Liberation Army, Beijing, People's Republic of China
| | - Sheng Cui
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China; and
| |
Collapse
|
9
|
Zhang R, Piao MJ, Oh MC, Park JE, Shilnikova K, Moon YJ, Kim DH, Jung U, Kim IG, Hyun JW. Protective Effect of an Isoflavone, Tectorigenin, Against Oxidative Stress-induced Cell Death via Catalase Activation. J Cancer Prev 2016; 21:257-263. [PMID: 28053960 PMCID: PMC5207610 DOI: 10.15430/jcp.2016.21.4.257] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 10/11/2016] [Accepted: 10/15/2016] [Indexed: 12/20/2022] Open
Abstract
Background Isoflavones are biologically active compounds that occur naturally in a variety of plants, with relatively high levels in soybean. Tectorigenin, an isoflavone, protects against hydrogen peroxide (H2O2)-induced cell damage. However, the underlying mechanism is unknown. Methods The MTT assay was performed to determine cell viability. Catalase activity was assessed by determining the amount of enzyme required to degrade 1 μM H2O2. Protein expression of catalase, phospho-extracellular signal-regulated kinase (ERK), IκB-α, and NF-κB were evaluated by Western blot analysis. A mobility shift assay was performed to assess the DNA-binding ability of NF-κB. Transient transfection and a NF-κB luciferase assay were performed to assess transcriptional activity. Results Tectorigenin reduced H2O2-induced death of Chinese hamster lung fibroblasts (V79-4). In addition, tectorigenin increased the activity and protein expression of catalase. Blockade of catalase activity attenuated the protective effect of tectorigenin against oxidative stress. Furthermore, tectorigenin enhanced phosphorylation of ERK and nuclear expression of NF-κB, while inhibition of ERK and NF-κB attenuated the protective effect of tectorigenin against oxidative stress. Conclusions Tectorigenin protects cells against oxidative damage by activating catalase and modulating the ERK and NF-κB signaling pathway.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Biochemistry, Jeju National University School of Medicine, Jeju, Korea; School of Medical Science and Laboratory Medicine, Jiangsu University, Jiangsu, China
| | - Mei Jing Piao
- Department of Biochemistry, Jeju National University School of Medicine, Jeju, Korea
| | - Min Chang Oh
- Department of Biochemistry, Jeju National University School of Medicine, Jeju, Korea
| | - Jeong Eon Park
- Department of Biochemistry, Jeju National University School of Medicine, Jeju, Korea
| | - Kristina Shilnikova
- Department of Biochemistry, Jeju National University School of Medicine, Jeju, Korea
| | - Yu Jin Moon
- Department of Biochemistry, Jeju National University School of Medicine, Jeju, Korea
| | - Dong Hyun Kim
- Department of Microbial Chemistry, College of Pharmacy, Kyung Hee University, Seoul, Korea
| | - Uhee Jung
- Radiation Biotechnology Research Division, Korea Atomic Energy Research Institute, Jeongeup, Korea
| | - In Gyu Kim
- Department of Radiation Biology, Environmental Radiation Research Group, Korea Atomic Energy Research Institute, Daejeon, Korea; Department of Radiation Biotechnology and Applied Radioisotope, Korea University of Science and Technology, Daejeon, Korea
| | - Jin Won Hyun
- Department of Biochemistry, Jeju National University School of Medicine, Jeju, Korea
| |
Collapse
|
10
|
Di Lascio S, Belperio D, Benfante R, Fornasari D. Alanine Expansions Associated with Congenital Central Hypoventilation Syndrome Impair PHOX2B Homeodomain-mediated Dimerization and Nuclear Import. J Biol Chem 2016; 291:13375-93. [PMID: 27129232 PMCID: PMC4933246 DOI: 10.1074/jbc.m115.679027] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Indexed: 11/30/2022] Open
Abstract
Heterozygous mutations of the human PHOX2B gene, a key regulator of autonomic nervous system development, lead to congenital central hypoventilation syndrome (CCHS), a neurodevelopmental disorder characterized by a failure in the autonomic control of breathing. Polyalanine expansions in the 20-residues region of the C terminus of PHOX2B are the major mutations responsible for CCHS. Elongation of the alanine stretch in PHOX2B leads to a protein with altered DNA binding, transcriptional activity, and nuclear localization and the possible formation of cytoplasmic aggregates; furthermore, the findings of various studies support the idea that CCHS is not due to a pure loss of function mechanism but also involves a dominant negative effect and/or toxic gain of function for PHOX2B mutations. Because PHOX2B forms homodimers and heterodimers with its paralogue PHOX2A in vitro, we tested the hypothesis that the dominant negative effects of the mutated proteins are due to non-functional interactions with the wild-type protein or PHOX2A using a co-immunoprecipitation assay and the mammalian two-hybrid system. Our findings show that PHOX2B forms homodimers and heterodimerizes weakly with mutated proteins, exclude the direct involvement of the polyalanine tract in dimer formation, and indicate that mutated proteins retain partial ability to form heterodimers with PHOX2A. Moreover, in this study, we investigated the effects of the longest polyalanine expansions on the homeodomain-mediated nuclear import, and our data clearly show that the expanded C terminus interferes with this process. These results provide novel insights into the effects of the alanine tract expansion on PHOX2B folding and activity.
Collapse
Affiliation(s)
- Simona Di Lascio
- From the Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20129 Milan, Italy and
| | - Debora Belperio
- From the Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20129 Milan, Italy and
| | - Roberta Benfante
- From the Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20129 Milan, Italy and the National Research Council (CNR) Neuroscience Institute, 20129 Milan, Italy
| | - Diego Fornasari
- From the Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20129 Milan, Italy and the National Research Council (CNR) Neuroscience Institute, 20129 Milan, Italy
| |
Collapse
|
11
|
Gokozan HN, Baig F, Corcoran S, Catacutan FP, Gygli PE, Takakura AC, Moreira TS, Czeisler C, Otero JJ. Area postrema undergoes dynamic postnatal changes in mice and humans. J Comp Neurol 2015; 524:1259-69. [PMID: 26400711 DOI: 10.1002/cne.23903] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 09/15/2015] [Accepted: 09/15/2015] [Indexed: 12/29/2022]
Abstract
The postnatal period in mammals represents a developmental epoch of significant change in the autonomic nervous system (ANS). This study focuses on postnatal development of the area postrema, a crucial ANS structure that regulates temperature, breathing, and satiety, among other activities. We find that the human area postrema undergoes significant developmental changes during postnatal development. To characterize these changes further, we used transgenic mouse reagents to delineate neuronal circuitry. We discovered that, although a well-formed ANS scaffold exists early in embryonic development, the area postrema shows a delayed maturation. Specifically, postnatal days 0-7 in mice show no significant change in area postrema volume or synaptic input from PHOX2B-derived neurons. In contrast, postnatal days 7-20 show a significant increase in volume and synaptic input from PHOX2B-derived neurons. We conclude that key ANS structures show unexpected dynamic developmental changes during postnatal development. These data provide a basis for understanding ANS dysfunction and disease predisposition in premature and postnatal humans.
Collapse
Affiliation(s)
- Hamza Numan Gokozan
- The Ohio State University, College of Medicine, Department of Pathology, Division of Neuropathology, Columbus, Ohio, 43210
| | - Faisal Baig
- The Ohio State University, College of Medicine, Department of Pathology, Division of Neuropathology, Columbus, Ohio, 43210
| | - Sarah Corcoran
- The Ohio State University, College of Medicine, Department of Pathology, Division of Neuropathology, Columbus, Ohio, 43210
| | - Fay Patsy Catacutan
- The Ohio State University, College of Medicine, Department of Pathology, Division of Neuropathology, Columbus, Ohio, 43210
| | - Patrick Edwin Gygli
- The Ohio State University, College of Medicine, Department of Pathology, Division of Neuropathology, Columbus, Ohio, 43210
| | - Ana C Takakura
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, 05508-900, São Paulo, Brazil
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo, 05508-900, São Paulo, Brazil
| | - Catherine Czeisler
- The Ohio State University, College of Medicine, Department of Pathology, Division of Neuropathology, Columbus, Ohio, 43210
| | - José J Otero
- The Ohio State University, College of Medicine, Department of Pathology, Division of Neuropathology, Columbus, Ohio, 43210
| |
Collapse
|
12
|
Living without DAT: Loss and compensation of the dopamine transporter gene in sauropsids (birds and reptiles). Sci Rep 2015; 5:14093. [PMID: 26364979 PMCID: PMC4894405 DOI: 10.1038/srep14093] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 08/18/2015] [Indexed: 11/17/2022] Open
Abstract
The dopamine transporter (DAT) is a major regulator of synaptic dopamine (DA) availability. It plays key roles in motor control and motor learning, memory formation, and reward-seeking behavior, is a major target of cocaine and methamphetamines, and has been assumed to be conserved among vertebrates. We have found, however, that birds, crocodiles, and lizards lack the DAT gene. We also found that the unprecedented loss of this important gene is compensated for by the expression of the noradrenaline transporter (NAT) gene, and not the serotonin transporter genes, in dopaminergic cells, which explains the peculiar pharmacology of the DA reuptake activity previously noted in bird striatum. This unexpected pattern contrasts with that of ancestral vertebrates (e.g. fish) and mammals, where the NAT gene is selectively expressed in noradrenergic cells. DA circuits in birds/reptiles and mammals thus operate with an analogous reuptake mechanism exerted by different genes, bringing new insights into gene expression regulation in dopaminergic cells and the evolution of a key molecular player in reward and addiction pathways.
Collapse
|
13
|
Burbach JPH, Hellemons AJCGM, Grant P, Pant HC. The homeodomain transcription factor Phox2 in the stellate ganglion of the squid Loligo pealei. Biol Open 2015; 4:954-60. [PMID: 26116657 PMCID: PMC4542286 DOI: 10.1242/bio.012476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Homeodomain transcription factors regulate development of embryos and cellular physiology in adult systems. Paired-type homeodomain genes constitute a subclass that has been particularly implicated in establishment of neuronal identity in the mammalian nervous system. We isolated fragments of eight homeodomain genes of this subclass expressed in the stellate ganglion of the North Atlantic long finned squid Loligo pealei (lp) [Note: Loligo pealei has been officially renamed Doryteuthis pealei. For reasons of uniformity and clarity Loligo pealei (lp) is used here]. Of the most abundant ones, we cloned a full length cDNA which encoded the squid ortholog of the paired-type homeodomain proteins Phox2a/b. The homology of lpPhox2 to invertebrate and mammalian Phox2 was limited to the homeodomain. In contrast to mouse Phox2b, lpPhox2 was unable to transactivate the dopamine beta-hydroxylase (DBH) promoter in a heterologous mammalian transfection system. In vivo, lpPhox2 was expressed in the developing stellate ganglion of stage 27 squid embryos and continued to be expressed in the adult stellate neurons where expression was confined to the giant fiber lobe containing the neurons that form the giant axons. The expression of lpPhox was similarly timed and distributed as the Fmrf gene. Furthermore, the Fmrf upstream region contained putative Phox2a/b binding sites. These results suggest a role of lpPhox2 in the developmental specification of neuronal identity and regulation of neurons of the squid giant axon.
Collapse
Affiliation(s)
- J. Peter H. Burbach
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht University, Utrecht 3584CG, The Netherlands
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Anita J. C. G. M. Hellemons
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht University, Utrecht 3584CG, The Netherlands
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Philip Grant
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
- Laboratory of Neurochemistry, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Harish C. Pant
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
- Laboratory of Neurochemistry, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| |
Collapse
|
14
|
Harrison C, Shepherd IT. Choices choices: regulation of precursor differentiation during enteric nervous system development. Neurogastroenterol Motil 2013; 25:554-62. [PMID: 23634805 PMCID: PMC4062358 DOI: 10.1111/nmo.12142] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 03/30/2013] [Indexed: 02/08/2023]
Abstract
Background The enteric nervous system (ENS) is the largest subdivision of the peripheral nervous system and forms a complex circuit of neurons and glia that controls the function of the gastrointestinal (GI) tract. Within this circuit, there are multiple subtypes of neurons and glia. Appropriate differentiation of these various cell subtypes is vital for normal ENS and GI function. Studies of the pediatric disorder Hirschprung's Disease (HSCR) have provided a number of important insights into the mechanisms and molecules involved in ENS development; however, there are numerous other GI disorders that potentially may result from defects in development/differentiation of only a subset of ENS neurons or glia. Purpose Our understanding of the mechanisms and molecules involved in enteric nervous system differentiation is far from complete. Critically, it remains unclear at what point the fates of enteric neural crest cells (ENCCs) become committed to a specific subtype cell fate and how these cell fate choices are made. We will review our current understanding of ENS differentiation and highlight key questions that need to be addressed to gain a more complete understanding of this biological process.
Collapse
Affiliation(s)
- Colin Harrison
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta GA 30322, USA
| | - Iain T. Shepherd
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta GA 30322, USA
| |
Collapse
|
15
|
Farina MI, Scarani R, Po' C, Agosto C, Ottonello G, Benini F. Congenital central hypoventilation syndrome and hypoglycaemia. Acta Paediatr 2012; 101:e92-6. [PMID: 22103583 DOI: 10.1111/j.1651-2227.2011.02533.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
UNLABELLED Congenital central hypoventilation syndrome (CCHS) is a rare genetic disorder typically presenting in infants with an impaired automatic control of breathing, particularly during sleep, and often associated with variable patterns of autonomic nervous system dysregulations. We studied three children who had CCHS associated with episodes of severe hypoglycaemia and hyperinsulinaemia; we discuss the possible relationship with impaired dopamine-beta-hydroxylase function. CONCLUSION Hypoglycaemia and hyperinsulinaemia might be suspected in children with CCHS presenting with seizures and hyperhydrosis; though, further studies are needed to confirm this association.
Collapse
Affiliation(s)
- Maria I Farina
- Pediatric Pain and Palliative Care Service, Department of Pediatrics, University of Padua, Italy.
| | | | | | | | | | | |
Collapse
|
16
|
Fan Y, Huang J, Duffourc M, Kao RL, Ordway GA, Huang R, Zhu MY. Transcription factor Phox2 upregulates expression of norepinephrine transporter and dopamine β-hydroxylase in adult rat brains. Neuroscience 2011; 192:37-53. [PMID: 21763404 PMCID: PMC3166407 DOI: 10.1016/j.neuroscience.2011.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 07/05/2011] [Accepted: 07/06/2011] [Indexed: 01/22/2023]
Abstract
Degeneration of the noradrenergic locus coeruleus (LC) in aging and neurodegenerative diseases is well documented. Slowing or reversing this effect may have therapeutic implications. Phox2a and Phox2b are homeodomain transcriptional factors that function as determinants of the noradrenergic phenotype during embryogenesis. In the present study, recombinant lentiviral eGFP-Phox2a and -Phox2b (vPhox2a and vPhox2b) were constructed to study the effects of Phox2a/2b over-expression on dopamine β-hydroxylase (DBH) and norepinephrine transporter (NET) levels in central noradrenergic neurons. Microinjection of vPhox2 into the LC of adult rats significantly increased Phox2 mRNA levels in the LC region. Over-expression of either Phox2a or Phox2b in the LC was paralleled by significant increases in mRNA and protein levels of DBH and NET in the LC. Similar increases in DBH and NET protein levels were observed in the hippocampus following vPhox2 microinjection. In the frontal cortex, only NET protein levels were significantly increased by vPhox2 microinjection. Over-expression of Phox2 genes resulted in a significant increase in BrdU-positive cells in the hippocampal dentate gyrus. The present study demonstrates an upregulatory effect of Phox2a and Phox2b on the expression of DBH and NET in noradrenergic neurons of rat brains, an effect not previously shown in adult animals. Phox2 genes may play an important role in maintaining the function of the noradrenergic neurons after birth, and regulation of Phox2 gene expression may have therapeutic utility in aging or disorders involving degeneration of noradrenergic neurons.
Collapse
Affiliation(s)
- Yan Fan
- Department of Pharmacology, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
- Department of Biochemistry and Molecular Biology, Soochow University School of Medicine, Suzhou, China
| | - Jingjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Michelle Duffourc
- Department of Pharmacology, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Race L. Kao
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Gregory A. Ordway
- Department of Pharmacology, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Rui Huang
- Department of Biochemistry and Molecular Biology, Soochow University School of Medicine, Suzhou, China
| | - Meng-Yang Zhu
- Department of Pharmacology, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| |
Collapse
|
17
|
Flames N, Hobert O. Transcriptional Control of the Terminal Fate of Monoaminergic Neurons. Annu Rev Neurosci 2011; 34:153-84. [DOI: 10.1146/annurev-neuro-061010-113824] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Nuria Flames
- Department of Biochemistry and Molecular Biophysics, Howard Hughes Medical Institute, Columbia University Medical Center, New York, New York 10032;
- Genes & Disease Program, Center for Genomic Regulation (CRG), Barcelona, Spain E-08003;
- Present address: Instituto de Biomedicina de Valencia IBV-CSIC, E-46010 Valencia, Spain
| | - Oliver Hobert
- Department of Biochemistry and Molecular Biophysics, Howard Hughes Medical Institute, Columbia University Medical Center, New York, New York 10032;
| |
Collapse
|
18
|
Cytokines inhibit norepinephrine transporter expression by decreasing Hand2. Mol Cell Neurosci 2011; 46:671-80. [PMID: 21241805 DOI: 10.1016/j.mcn.2011.01.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 12/16/2010] [Accepted: 01/08/2011] [Indexed: 11/22/2022] Open
Abstract
Functional noradrenergic transmission requires the coordinate expression of enzymes involved in norepinephrine (NE) synthesis, as well as the norepinephrine transporter (NET) which removes NE from the synapse. Inflammatory cytokines acting through gp130 can suppress the noradrenergic phenotype in sympathetic neurons. This occurs in a subset of sympathetic neurons during development and also occurs in adult neurons after injury. For example, cytokines suppress noradrenergic function in sympathetic neurons after axotomy and during heart failure. The molecular basis for suppression of noradrenergic genes is not well understood, but previous studies implicated a reduction of Phox2a in cytokine suppression of dopamine beta hydroxylase. We used sympathetic neurons and neuroblastoma cells to investigate the role of Phox2a in cytokine suppression of NET transcription. Chromatin immunoprecipitation experiments revealed that Phox2a did not bind the NET promoter, and overexpression of Phox2a did not prevent cytokine suppression of NET transcription. Hand2 and Gata3 are transcription factors that induce noradrenergic genes during development and are present in mature sympathetic neurons. Both Hand2 and Gata3 were decreased by cytokines in sympathetic neurons and neuroblastoma cells. Overexpression of either Hand2 or Gata3 was sufficient to rescue NET transcription following suppression by cytokines. We examined expression of these genes following axotomy to determine if their expression was altered following nerve injury. NET and Hand2 mRNAs decreased significantly in sympathetic neurons 48 h after axotomy, but Gata3 mRNA was unchanged. These data suggest that cytokines can inhibit NET expression through downregulation of Hand2 or Gata3 in cultured sympathetic neurons, but axotomy in adult animals selectively suppresses Hand2 expression.
Collapse
|
19
|
Apostolova G, Dechant G. Development of neurotransmitter phenotypes in sympathetic neurons. Auton Neurosci 2009; 151:30-8. [DOI: 10.1016/j.autneu.2009.08.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
20
|
Generating diversity: Mechanisms regulating the differentiation of autonomic neuron phenotypes. Auton Neurosci 2009; 151:17-29. [PMID: 19819195 DOI: 10.1016/j.autneu.2009.08.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sympathetic and parasympathetic postganglionic neurons innervate a wide range of target tissues. The subpopulation of neurons innervating each target tissue can express unique combinations of neurotransmitters, neuropeptides, ion channels and receptors, which together comprise the chemical phenotype of the neurons. The target-specific chemical phenotype shown by autonomic postganglionic neurons arises during development. In this review, we examine the different mechanisms that generate such a diversity of neuronal phenotypes from the pool of apparently homogenous neural crest progenitor cells that form the sympathetic ganglia. There is evidence that the final chemical phenotype of autonomic postganglionic neurons is generated by both signals at the level of the cell body that trigger cell-autonomous programs, as well as signals from the target tissues they innervate.
Collapse
|
21
|
Fan Y, Huang J, Kieran N, Zhu MY. Effects of transcription factors Phox2 on expression of norepinephrine transporter and dopamine beta-hydroxylase in SK-N-BE(2)C cells. J Neurochem 2009; 110:1502-13. [PMID: 19573018 DOI: 10.1111/j.1471-4159.2009.06260.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Phox2a and Phox2b are two homeodomain proteins that control the differentiation of noradrenergic neurons during embryogenesis. In the present study, we examined the possible effect of Phox2a/2b on the in vitro expression of the norepinephrine transporter (NET) and dopamine beta-hydroxylase (DBH), two important markers of the noradrenergic system. SK-N-BE(2)C cells were transfected with cDNAs or short hairpin RNAs specific to the human Phox2a and Phox2b genes. Transfection of 0.1 to 5 mug of cDNAs of Phox2a or Phox2b significantly increased mRNA and protein levels of NET and DBH in a concentration-dependent manner. As a consequence of the enhanced expression of NET after transfection, there was a parallel increase in the uptake of [(3)H]norepinephrine. Co-transfection of Phox2a and Phox2b did not further increase the expression of noradrenergic markers when compared with transfection of either Phox2a or Phox2b alone. Transfection of shRNAs specific to Phox2a or Phox2b genes significantly reduced mRNA and protein levels of NET and DBH after shutdown of endogenous Phox2, which was accompanied by a decreased [(3)H]norepinephrine uptake. Furthermore, there was an additive effect after cotransfection with both shRNAs specific to Phox2a or Phox2b genes on NET mRNA levels. Finally, the reduced DBH expression caused by the shRNA specific to Phox2a could be reversed by transfection with Phox2b cDNA and vice versa. The present findings verify the determinant role of Phox2a and Phox2b on the expression and function of NET and DBH in vitro. Further clarifying the regulatory role of these two transcription factors on key proteins of the noradrenergic system may open a new avenue for therapeutics of aging-caused dysfunction of the noradrenergic system.
Collapse
Affiliation(s)
- Yan Fan
- Department of Biochemistry and Molecular Biology, School of Medicine, Soochow University Suzhou, China
| | | | | | | |
Collapse
|
22
|
Rout UK, Clausen P. Common increase of GATA-3 level in PC-12 cells by three teratogens causing autism spectrum disorders. Neurosci Res 2009; 64:162-9. [DOI: 10.1016/j.neures.2009.02.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2008] [Revised: 02/19/2009] [Accepted: 02/23/2009] [Indexed: 01/17/2023]
|
23
|
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: 360] [Impact Index Per Article: 24.0] [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
|
24
|
Benfante R, Antonini RA, Kuster N, Schuderer J, Maercker C, Adlkofer F, Clementi F, Fornasari D. The expression of PHOX2A, PHOX2B and of their target gene dopamine-β-hydroxylase (DβH) is not modified by exposure to extremely-low-frequency electromagnetic field (ELF-EMF) in a human neuronal model. Toxicol In Vitro 2008; 22:1489-95. [DOI: 10.1016/j.tiv.2008.05.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2007] [Revised: 04/16/2008] [Accepted: 05/12/2008] [Indexed: 11/25/2022]
|
25
|
Hong SJ, Choi HJ, Hong S, Huh Y, Chae H, Kim KS. Transcription factor GATA-3 regulates the transcriptional activity of dopamine beta-hydroxylase by interacting with Sp1 and AP4. Neurochem Res 2008; 33:1821-31. [PMID: 18338249 DOI: 10.1007/s11064-008-9639-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Accepted: 02/20/2008] [Indexed: 11/28/2022]
Abstract
GATA-3 is a zinc finger transcription factor that is expressed in T cell lineages as well as in the nervous system during development. In this study, we report that forced expression of GATA-3 resulted in an increased number of dopamine beta-hydroxylase (DBH)-expressing neurons in primary neural crest stem cell (NCSC) culture, suggesting that the DBH gene may be a downstream target gene of GATA-3. GATA-3 robustly transactivates the promoter function of the noradrenaline (NA)-synthesizing DBH gene, via two specific upstream promoter domains; one at -62 to -32 bp and the other at -891 to -853 bp. Surprisingly, none of these domains contain GATA-3 binding sites but encompass binding motifs for transcription factors Sp1 and AP4, respectively. Protein-protein interaction analyses both in vitro and in vivo and chromatin immunoprecipitation (ChIP) assays showed that GATA-3 effects its transcriptional regulatory function through physical interactions with these transcription factors.
Collapse
Affiliation(s)
- Seok Jong Hong
- Molecular Neurobiology Laboratory, MRC215, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, USA
| | | | | | | | | | | |
Collapse
|
26
|
Hong SJ, Chae H, Lardaro T, Hong S, Kim KS. Trim11 increases expression of dopamine beta-hydroxylase gene by interacting with Phox2b. Biochem Biophys Res Commun 2008; 368:650-5. [PMID: 18275850 DOI: 10.1016/j.bbrc.2008.01.165] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Accepted: 01/25/2008] [Indexed: 11/26/2022]
Abstract
The homeodomain transcription factor Phox2b is one of the key determinants involved in the development of noradrenergic (NA) neurons in both the central nervous system (CNS) and the peripheral nervous system (PNS). Using yeast two-hybrid screening, we isolated a Phox2b interacting protein, Trim11, which belongs to TRIM (Tripartite motif) or RBCC proteins family, and contains a RING domain, B-boxes, a coiled-coil domain, and the B30.2/SPRY domain. Protein-protein interaction assays showed that Phox2b was able to physically interact with Trim11. The B30.2/SPRY domain of Trim11 was required for the interaction with Phox2b. Expression of Phox2b and Trim11 was detected in the sympathetic ganglia (SG) of mouse embryos. Forced expression of Trim11 with Phox2b further increased mRNA levels of dopamine beta-hydroxylase (DBH) gene in primary avian neural crest stem cell (NCSC) culture. This study suggests a potential role for Trim11 in the specification of NA phenotype by interaction with Phox2b.
Collapse
Affiliation(s)
- Seok Jong Hong
- Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA 02478, USA
| | | | | | | | | |
Collapse
|
27
|
Regulation of rat dopamine beta-hydroxylase gene transcription by early growth response gene 1 (Egr1). Brain Res 2007; 1193:1-11. [PMID: 18190898 DOI: 10.1016/j.brainres.2007.11.055] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2007] [Revised: 10/30/2007] [Accepted: 11/26/2007] [Indexed: 01/13/2023]
Abstract
Egr1, a transcription factor rapidly induced by various stimuli including stress, can elevate transcription of genes for the catecholamine biosynthetic enzymes TH and PNMT. To examine if Egr1 also regulates dopamine beta-hydroxylase (DBH) gene expression, PC12 cells were transfected with expression vector for full length or truncated inactive Egr1 and various DBH promoter-driven luciferase constructs. While Egr1 elevated TH promoter activity, DBH promoter activity was reduced. The reduction occurred as early as 4 h and reached maximal inhibition 16-40 h after transfection. Egr1 also reduced the expression of endogenous DBH mRNA and the induction of DBH promoter activity by cAMP. These effects were not observed with truncated Egr1 lacking the DNA binding domain. The first 247, but not 200, nucleotides of DBH promoter are sufficient for this suppression. Several putative Egr1 motifs were identified, and mutagenesis showed that the motif at -227/-224 is required. Binding of Egr1 to this region of the DBH promoter was verified by chromatin immunoprecipitation and electrophoretic mobility shift assays. This study demonstrates that DBH promoter contains at least one functional Egr1 motif; and indicates, for the first time, that Egr1 can play an inhibitory role in regulation of DBH gene transcription.
Collapse
|
28
|
The Drosophila basic helix-loop-helix protein DIMMED directly activates PHM, a gene encoding a neuropeptide-amidating enzyme. Mol Cell Biol 2007; 28:410-21. [PMID: 17967878 DOI: 10.1128/mcb.01104-07] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The basic helix-loop-helix (bHLH) protein DIMMED (DIMM) supports the differentiation of secretory properties in numerous peptidergic cells of Drosophila melanogaster. DIMM is coexpressed with diverse amidated neuropeptides and with the amidating enzyme peptidylglycine alpha-hydroxylating monooxygenase (PHM) in approximately 300 cells of the late embryo. Here we confirm that DIMM has transcription factor activity in transfected HEK 293 cells and that the PHM gene is a direct target. The mammalian DIMM orthologue MIST1 also transactivated the PHM gene. DIMM activity was dependent on the basic region of the protein and on the sequences of three E-box sites within PHM's first intron; the sites make different contributions to the total activity. These data suggest a model whereby the three E boxes interact cooperatively and independently to produce high PHM transcriptional activation. This DIMM-controlled PHM regulatory region displayed similar properties in vivo. Spatially, its expression mirrored that of the DIMM protein, and its activity was largely dependent on dimm. Further, in vivo expression was highly dependent on the sequences of the same three E boxes. This study supports the hypothesis that DIMM is a master regulator of a peptidergic cell fate in Drosophila and provides a detailed transcriptional mechanism of DIMM action on a defined target gene.
Collapse
|
29
|
Kang KA, Lee KH, Zhang R, Piao MJ, Kang MY, Kwak YS, Yoo BS, You HJ, Hyun JW. Protective effects of Castanopsis cuspidate through activation of ERK and NF-kappaB on oxidative cell death induced by hydrogen peroxide. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2007; 70:1319-28. [PMID: 17654250 DOI: 10.1080/15287390701429315] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The protective effect of Castanopsis cuspidate var. sieboldii was examined on H2O2-induced cell damage. The ethanol extract of Castanopsis cuspidate was found to scavenge 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical and reduce intracellular reactive oxygen species (ROS) generation, and thus prevent lipid peroxidation and cellular DNA damage induced by H2O2. As a result, Castanopsis extract reduced H2O2-induced cell death of V79-4 cells via inhibition of apoptosis. Castanopsis extract was also found to increase catalase activity and its protein expression. Further molecular mechanistic studies revealed that Castanopsis extract enhanced phosphorylation of extracellular signal regulated kinase (ERK) and activity of nuclear factor kappa B (NF-kappaB). Taken together, the results suggest that Castanopsis extract protects V79-4 cells against oxidative damage by enhancing catalase activity and modulating the ERK and NF-kappaB signal pathway.
Collapse
Affiliation(s)
- Kyoung Ah Kang
- Department of Biochemistry, College of Medicine and Applied Radiological Science Research Institute, Cheju National University, Jeju-si, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Zhang R, Kang KA, Piao MJ, Lee KH, Jang HS, Park MJ, Kim BJ, Kim JS, Kim YS, Ryu SY, Hyun JW. Rhapontigenin from Rheum undulatum protects against oxidative-stress-induced cell damage through antioxidant activity. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2007; 70:1155-66. [PMID: 17558811 DOI: 10.1080/15287390701252766] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
The antioxidant properties of rhapontigenin and rhaponticin isolated from Rheum undulatum were investigated. Rhapontigenin was found to scavenge intracellular reactive oxygen species (ROS), the 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical, and hydrogen peroxide (H2O2). The radical scavenging effect of rhapontigenin was more effective than rhaponticin. Rhapontigenin protected against H2O2-induced membrane lipid peroxidation and cellular DNA damage, which are the main targets of oxidative stress-induced cellular damage. The radical scavenging activity of rhapontigenin protected Chinese hamster lung fibroblast (V79-4) cells exposed to H2O2 by inhibiting apoptosis. Rhapontigenin inhibited cell damage induced by serum starvation and was also found to increase the activity of catalase and its protein expression. Further, rhapontigenin increased phosphorylation of extracellular signal-regulated kinase (ERK) and inhibited the activity of activator protein 1 (AP-1), a redox-sensitive transcription factor. In summary, these results suggest that rhapontigenin protects V79-4 cells against oxidative damage by enhancing the cellular antioxidant activity and modulating cellular signal pathways.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Biochemistry, College of Medicine and Applied Radiological Science Research Institute, Cheju National University, Jeju-si, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Wen G, Wessel J, Zhou W, Ehret GB, Rao F, Stridsberg M, Mahata SK, Gent PM, Das M, Cooper RS, Chakravarti A, Zhou H, Schork NJ, O’Connor DT, Hamilton BA. An ancestral variant of Secretogranin II confers regulation by PHOX2 transcription factors and association with hypertension. Hum Mol Genet 2007; 16:1752-64. [PMID: 17584765 PMCID: PMC2695823 DOI: 10.1093/hmg/ddm123] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Granins regulate secretory vesicle formation in neuroendocrine cells and granin-derived peptides are co-released with neurotransmitters as modulatory signals at sympathetic sites. We report evidence for association between a regulatory polymorphism in Secretogranin II (SCG2) and hypertension in African-American subjects. The minor allele is ancestral in the human lineage and is associated with disease risk in two case-control studies and with elevated blood pressure in a separate familial study. Mechanistically, the ancestral allele acts as a transcriptional enhancer in cells that express endogenous Scg2, whereas the derived allele does not. ARIX (PHOX2A) and PHOX2B are identified as potential transactivating factors by oligonucleotide affinity chromatography and mass spectrometry and confirmed by chromatin immunoprecipitation. Each of these transcription factors preferentially binds the risk allele, both in vitro and in vivo. Population genetic considerations suggest positive selection of the protective allele within the human lineage. These results identify a common regulatory variation in SCG2 and implicate granin gene expression in the control of human blood pressure and susceptibility to hypertension.
Collapse
Affiliation(s)
- Gen Wen
- Department of Medicine, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
| | - Jennifer Wessel
- Department of Psychiatry, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
- Department of Family and Preventive Medicine, UCSD and Graduate School of Public Health, San Diego State University Joint Doctoral Program in Public Health Epidemiology
- Scripps Genomic Medicine and The Scripps Research Institute, La Jolla, CA
| | - Weidong Zhou
- Ludwig Institute for Cancer Research, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
| | - Georg B. Ehret
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Fangwen Rao
- Department of Medicine, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
| | - Mats Stridsberg
- Department of Medical Sciences, University Hospital, Uppsala, Sweden
| | - Sushil K. Mahata
- Department of Medicine, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
- VA San Diego Healthcare System
| | - Peter M. Gent
- Biomedical Sciences Graduate Program, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
| | - Madhusudan Das
- Department of Medicine, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
| | - Richard S. Cooper
- Department of Preventive Medicine and Epidemiology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois 60153
| | - Aravinda Chakravarti
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Huilin Zhou
- Ludwig Institute for Cancer Research, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
- Biomedical Sciences Graduate Program, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
- Moores UCSD Cancer Center, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
| | - Nicholas J. Schork
- Department of Psychiatry, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
- Biomedical Sciences Graduate Program, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
- Center for Human Genetics and Genomics, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
- Scripps Genomic Medicine and The Scripps Research Institute, La Jolla, CA
| | - Daniel T. O’Connor
- Department of Medicine, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
- Biomedical Sciences Graduate Program, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
- Center for Human Genetics and Genomics, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
- Moores UCSD Cancer Center, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
| | - Bruce A. Hamilton
- Department of Medicine, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
- Biomedical Sciences Graduate Program, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
- Moores UCSD Cancer Center, University of California, San Diego School of Medicine 9500 Gilman Drive, La Jolla, California 92093
- author for correspondence Telephone: (858) 822-1055
| |
Collapse
|
32
|
Borghini S, Di Duca M, Santamaria G, Vargiolu M, Bachetti T, Cargnin F, Pini Prato A, De Giorgio R, Lerone M, Stanghellini V, Jasonni V, Fornasari D, Ravazzolo R, Ceccherini I. Transcriptional regulation of TLX2 and impaired intestinal innervation: possible role of the PHOX2A and PHOX2B genes. Eur J Hum Genet 2007; 15:848-55. [PMID: 17505528 DOI: 10.1038/sj.ejhg.5201852] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
TLX2 (also known as HOX11L1, Ncx and Enx) is a transcription factor playing a crucial role in the development of the enteric nervous system, as confirmed by mice models exhibiting intestinal hyperganglionosis and pseudo-obstruction. However, congenital defects of TLX2 have been excluded as a major cause of intestinal motility disorders in patients affected with intestinal neuronal dysplasia (IND) or pseudo-obstruction. After demonstrating the direct regulation of TLX2 expression by the homeoprotein PHOX2B, in the present work, we have focused on its paralogue PHOX2A. By co-transfections, electrophoretic mobility shift assays and chromatin immunoprecipitation, we have demonstrated that PHOX2A, like PHOX2B, is involved in the cascade leading to TLX2 transactivation and presumably in the intestinal neuronal differentiation. Based on the hypothesis that missed activation of the TLX2 gene induces the development of enteric nervous system defects, PHOX2A and PHOX2B have been regarded as novel candidate genes involved in IND and pseudo-obstruction and consequently analyzed for mutations in a specific set of 26 patients. We have identified one still unreported PHOX2A variant; however, absence of any functional effect on TLX2 transactivation suggests that regulators or effectors other than the PHOX2 genes must act in the same pathway, likely playing a non redundant and direct role in the pathogenesis of such enteric disorders.
Collapse
Affiliation(s)
- Silvia Borghini
- Laboratorio di Genetica Molecolare, Istituto G Gaslini, Genova, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Benfante R, Flora A, Di Lascio S, Cargnin F, Longhi R, Colombo S, Clementi F, Fornasari D. Transcription Factor PHOX2A Regulates the Human α3 Nicotinic Receptor Subunit Gene Promoter. J Biol Chem 2007; 282:13290-302. [PMID: 17344216 DOI: 10.1074/jbc.m608616200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
PHOX2A is a paired-like homeodomain transcription factor that participates in specifying the autonomic nervous system. It is also involved in the transcriptional control of the noradrenergic neurotransmitter phenotype as it regulates the gene expression of tyrosine hydroxylase and dopamine-beta-hydroxylase. The results of this study show that the human orthologue of PHOX2A is also capable of regulating the transcription of the human alpha3 nicotinic acetylcholine receptor gene, which encodes the ligand-binding subunit of the ganglionic type nicotinic receptor. In particular, we demonstrated by chromatin immunoprecipitation and DNA pulldown assays that PHOX2A assembles on the SacI-NcoI region of alpha3 promoter and, by co-transfection experiments, that it exerts its transcriptional effects by acting through the 60-bp minimal promoter. PHOX2A does not seem to bind to DNA directly, and its DNA binding domain seems to be partially dispensable for the regulation of alpha3 gene transcription. However, as suggested by the findings of our co-immunoprecipitation assays, it may establish direct or indirect protein-protein interactions with Sp1, thus regulating the expression of alpha3 through a DNA-independent mechanism. As the alpha3 subunit is expressed in every terminally differentiated ganglionic cell, this is the first example of a "pan-autonomic" gene whose expression is regulated by PHOX2 proteins.
Collapse
Affiliation(s)
- Roberta Benfante
- Department of Pharmacology, School of Medicine, University of Milan, 20129 Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Morikawa Y, D’Autréaux F, Gershon MD, Cserjesi P. Hand2 determines the noradrenergic phenotype in the mouse sympathetic nervous system. Dev Biol 2007; 307:114-26. [PMID: 17531968 PMCID: PMC1952239 DOI: 10.1016/j.ydbio.2007.04.027] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2007] [Revised: 04/11/2007] [Accepted: 04/23/2007] [Indexed: 12/20/2022]
Abstract
The basic helix-loop-helix (bHLH) transcription factor Hand2 has been shown to play a role in the development of the mammalian sympathetic nervous system (SNS); however, its precise role could not be uncovered because Hand2 is required for early embryonic survival. We therefore generated a conditional Hand2 knockout mouse line by excising Hand2 in Wnt1-Cre-expressing neural crest-derived cells. These mice die at 12.5 dpc with embryos showing severe cardiovascular and facial defects. Crest-derived cells, however, populate sites of SNS development and proliferate normally. Sympathetic precursors differentiate into neurons and express the pan-neuronal markers, beta3-tubulin (Tuj1) and Hu showing that Hand2 is not essential for SNS neuronal differentiation. To determine whether Hand2 regulates noradrenergic differentiation, the levels of the norepinephrine biosynthetic enzymes, tyrosine hydroxylase (TH) and dopamine beta-hydroxylase (DBH) was examined. Both enzymes were dramatically reduced in mutant embryos suggesting that the primary role of Hand2 in the SNS is determination of neuronal phenotype. Loss of Hand2 did not affect the expression of other members of the transcriptional circuit regulating SNS development, including Phox2a/b, Mash1 and Gata2/3; however, Hand2 was required for Hand1 expression. Our data suggest that the major role of Hand2 during SNS development is to permit sympathetic neurons to acquire a catecholaminergic phenotype.
Collapse
Affiliation(s)
- Yuka Morikawa
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118
| | - Fabien D’Autréaux
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Michael D. Gershon
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Peter Cserjesi
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118
- * To whom correspondence should be addressed: Department of Cell and Molecular Biology, Tulane University, 2000 Percival Stern Hall, New Orleans, LA 70118. Ph. (504) 862-8081 Fax (504) 862-8081. E-mail:
| |
Collapse
|
35
|
Lucas ME, Müller F, Rüdiger R, Henion PD, Rohrer H. The bHLH transcription factor hand2 is essential for noradrenergic differentiation of sympathetic neurons. Development 2007; 133:4015-24. [PMID: 17008447 DOI: 10.1242/dev.02574] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The basic helix-loop-helix transcription factor Hand2, together with Ascl1, Phox2a, Phox2b and Gata2/Gata3, is induced by bone morphogenetic proteins in neural crest-derived precursor cells during sympathetic neuron generation. Hand2 overexpression experiments and the analysis of its function at the Dbh promotor implicated Hand2 in the control of noradrenergic gene expression. Using the zebrafish hand2 deletion mutant hands off, we have now investigated the physiological role of hand2 in the development of sympathetic ganglia. In hands off mutant embryos, sympathetic precursor cells aggregate to form normal sympathetic ganglion primordia characterized by the expression of phox2b, phox2a and the achaete-scute family member zash1a/ascl1. The expression of the noradrenergic marker genes th and dbh is strongly reduced, as well as the transcription factors gata2 and tfap2a (Ap-2alpha). By contrast, generic neuronal differentiation seems to be unaffected, as the expression of elavl3 (HuC) is not reduced in hands off sympathetic ganglia. These results demonstrate in vivo an essential and selective function of hand2 for the noradrenergic differentiation of sympathetic neurons, and implicates tfap2a and gata2 as downstream effectors.
Collapse
Affiliation(s)
- Marsha E Lucas
- Center for Molecular Neurobiology, Molecular, Cellular and Developmental Biology Program, Department of Neuroscience, The Ohio State University, 105 Rightmire Hall, 1060 Carmack Road, Columbus, OH 43210, USA
| | | | | | | | | |
Collapse
|
36
|
Wrobel LJ, Ogier M, Chatonnet F, Autran S, Mézières V, Thoby-Brisson M, McLean H, Taeron C, Champagnat J. Abnormal inspiratory depth in Phox2a haploinsufficient mice. Neuroscience 2007; 145:384-92. [PMID: 17218061 DOI: 10.1016/j.neuroscience.2006.11.055] [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] [Received: 06/29/2006] [Revised: 11/21/2006] [Accepted: 11/28/2006] [Indexed: 10/23/2022]
Abstract
Mutations of genes encoding Phox2a or Phox2b transcription factors induce modifications of different brainstem neuronal networks. Such modifications are associated with defects in breathing behavior at birth. In particular, an abnormal breathing frequency is observed in Phox2a-/- mutant mice, resulting from abnormal development of the locus coeruleus (LC) nucleus. However, the role of Phox2a proteins in the establishment of respiratory neuronal pathways is unknown, largely because mutants die shortly after birth. In the present study, we examined the effects of a haploinsufficiency of the Phox2a gene. Phox2a heterozygotes survive and exhibit a significantly larger inspiratory volume both during normoxic breathing and in response to hypoxia and a delayed maturation of inspiratory duration compared to wild-type animals. This phenotype accompanied by an unaltered frequency is evident at birth and persists until at least postnatal day 10. Morphological analyses of Phox2a+/- animals revealed no anomaly in the LC region, but highlighted an increase in the number of cells expressing tyrosine hydroxylase enzyme, a marker of chemoafferent neurons, in the petrosal sensory ganglion. These data indicate that Phox2a plays a critical role in the ontogeny of the reflex control of inspiration.
Collapse
Affiliation(s)
- L J Wrobel
- Neurobiologie Génétique et Intégrative, UPR2216 CNRS, 1 Avenue de la Terrasse, 91198 Gif sur Yvette, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Mahapatra NR, Mahata M, Ghosh S, Gayen JR, O'Connor DT, Mahata SK. Molecular basis of neuroendocrine cell type-specific expression of the chromogranin B gene: Crucial role of the transcription factors CREB, AP-2, Egr-1 and Sp1. J Neurochem 2006; 99:119-33. [PMID: 16987240 DOI: 10.1111/j.1471-4159.2006.04128.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The molecular basis of neuroendocrine-specific expression of chromogranin B gene (Chgb) has remained elusive. Utilizing wild-type and mutant Chgb promoter/luciferase reporter constructs, this study established a crucial role for the cAMP response element (CRE) box at -102/-95 bp in endocrine [rat pheochromocytoma (chromaffin) cell line (PC12) and rat pituitary somatotrope cell line (GC)] and neuronal [rat dorsal root ganglion/mouse neuroblastoma hybrid cell line (F-11), cortical and hippocampal primary neurons] cells. Additionally, G/C-rich domains at -134/-127, -125/-117 and -115/-110 bp played especially important roles for endocrine-specific expression of the Chgb gene. Co-transfection of expression plasmids for CREB, activator protein-2 (transcription factor) (AP-2), early growth response protein (transcription factor) (Egr-1) or specificity protein 1 (transcription factor) (Sp1) with the Chgb promoter constructs trans-activated expression of the Chgb gene. Nuclear extracts from either PC12 or F-11 cells formed specific complexes with the Chgb (-110/-87 bp) (CRE) oligonucleotide, which were either supershifted or disrupted by anti-CREB antibodies. In addition PC12 nuclear extracts also formed a specific complex with a Chgb (-140/-104-bp) oligonucleotide containing three G/C-rich regions, which was dose-dependently disrupted by anti-AP-2, anti-Egr-1 or anti-Sp1 antibodies; indeed, any one of these three antibodies completely abolished the complex, suggesting that all three factors bind the region simultaneously, at least in vitro. Chromatin immunoprecipitation assays documented the binding of the transcription factors CREB, AP-2, Egr-1 and Sp1 to the chromosomal Chgb gene promoter in vivo in PC12 cells within the context of chromatin. We conclude that the neuroendocrine-specific expression of Chgb is mediated by the CRE and G/C boxes in cis and the transcription factors CREB, AP-2, Egr-1 and Sp1 in trans.
Collapse
Affiliation(s)
- Nitish R Mahapatra
- Department of Medicine, University of California, California 92093-0838, USA
| | | | | | | | | | | |
Collapse
|
38
|
Hong SJ, Huh Y, Chae H, Hong S, Lardaro T, Kim KS. GATA-3 regulates the transcriptional activity of tyrosine hydroxylase by interacting with CREB. J Neurochem 2006; 98:773-81. [PMID: 16893419 DOI: 10.1111/j.1471-4159.2006.03924.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The zinc finger transcription factor GATA-3 is a master regulator of type 2 T-helper cell development. Interestingly, in GATA-3-/- mice, noradrenaline (NA) deficiency is a proximal cause of embryonic lethality. However, neither the role of GATA-3 nor its target gene(s) in the nervous system were known. Here, we report that forced expression of GATA-3 resulted in an increased number of tyrosine hydroxylase (TH) expressing neurons in primary neural crest stem cell (NCSC) culture. We also found that GATA-3 transactivates the promoter function of TH via specific upstream sequences, a domain of the TH promoter residing at -61 to -39 bp. Surprisingly, this domain does not contain GATA-3 binding sites but possesses a binding motif, a cAMP response element (CRE), for the transcription factor, CREB. In addition, we found that site-directed mutation of this CRE almost completely abolished transactivation of the TH promoter by GATA-3. Furthermore, protein-protein interaction assays showed that GATA-3 is able to physically interact with CREB in vitro as well as in vivo. Based on these results, we propose that GATA-3 may regulate TH gene transcription via a novel and distinct protein-protein interaction, and directly contributes to NA phenotype specification.
Collapse
Affiliation(s)
- Seok Jong Hong
- Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, Massachusetts 02478, USA
| | | | | | | | | | | |
Collapse
|
39
|
Joshi PP, Kulkarni MV, Yu BK, Smith KR, Norton DL, van Veelen W, Höppener JWM, Franklin DS. Simultaneous downregulation of CDK inhibitors p18(Ink4c) and p27(Kip1) is required for MEN2A-RET-mediated mitogenesis. Oncogene 2006; 26:554-70. [PMID: 16953232 DOI: 10.1038/sj.onc.1209811] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Multiple endocrine neoplasia type 2A (MEN2A) is predisposed by mutations in the RET proto-oncogene. Low expression of the cyclin-dependent kinase inhibitor (CDKI) p27(Kip1) is present in thyroid tumors, and recent evidence demonstrates p27 downregulation by the active RET mutant, RET/PTC1, found in papillary thyroid carcinoma. This implicates decreased p27 activity as an important event during thyroid tumorigenesis. However, p27(-/-) mice develop MEN-like tumors only in combination with loss of another CDKI, p18(Ink4c). This suggests that p18 and p27 functionally collaborate in suppression of tumorigenesis, that loss of both is critical in the development of MEN tumors and that both p18 and p27 are regulated by RET. We report that induction of the constitutively active MEN2A-specific RET mutant, RET2A(C634R), correlates with reduced p18/p27, and elevated cyclin D protein levels, leading to increased CDK activity, increased pRb phosphorylation and proliferation under growth arrest conditions. Mechanistically, RET2A represses p18/p27 mRNA levels while elevating cyclin D1 mRNA levels. RET2A expression also correlates with decreased p27 protein stability. RET2A-mediated regulation of p18 and p27, but not of cyclins D1 and D2, requires functional mitogen-activated protein kinase signaling. Additionally, RET2A-dependent p18 repression is required and sufficient to increase cell proliferation. Perhaps most significantly, MEN2A adrenal tumors also display these changes in cell cycle expression profile, demonstrating the biological relevance of our cell culture studies. Our results demonstrate for the first time that RET2A regulates p18, and suggest that loss of not only p27 but also of p18 expression is a key step in MEN tumorigenesis.
Collapse
Affiliation(s)
- P P Joshi
- Department of Biological Sciences, Purdue Cancer Center, Purdue University, West Lafayette, IN, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Huber K. The sympathoadrenal cell lineage: specification, diversification, and new perspectives. Dev Biol 2006; 298:335-43. [PMID: 16928368 DOI: 10.1016/j.ydbio.2006.07.010] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2006] [Revised: 07/08/2006] [Accepted: 07/11/2006] [Indexed: 11/17/2022]
Abstract
During the past years considerable progress has been made in understanding the generation of cell diversity in the neural crest (NC). Sympathoadrenal (SA) cells constitute a major lineage among NC derivatives; they give rise to sympathetic neurons, neuroendocrine chromaffin cells, and the intermediate small intensely fluorescent (SIF) cells. The classic perception of how this diversification is achieved implies that (i) there is a common progenitor cell for sympathetic neurons and chromaffin cells, (ii) NC cells are instructed to a SA cell fate by signals derived from the wall of the dorsal aorta, especially bone morphogenetic proteins (BMP), and (iii) the local environments of secondary sympathetic ganglia and adrenal gland, respectively, are crucial for inducing differentiation of SA cells into sympathetic neurons and adrenal chromaffin cells. However, recent studies have suggested that the adrenal cortex is dispensable for the acquisition of a chromaffin cell fate. This review summarizes the current understanding of the development of SA cells. It covers the specification of SA cells from multipotent NC crest cells, the role of transcription factors during their development, the classic model of their subsequent diversification as well as alternative views for explaining the generation of endocrine versus neuronal SA derivatives.
Collapse
Affiliation(s)
- Katrin Huber
- Department of Neuroanatomy and Interdisciplinary Center for Neurosciences, University of Heidelberg, INF 307, D-69120 Heidelberg, Germany.
| |
Collapse
|
41
|
Sarkar AA, Howard MJ. Perspectives on integration of cell extrinsic and cell intrinsic pathways of signaling required for differentiation of noradrenergic sympathetic ganglion neurons. Auton Neurosci 2006; 126-127:225-31. [PMID: 16647305 DOI: 10.1016/j.autneu.2006.02.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2006] [Revised: 02/21/2006] [Accepted: 02/27/2006] [Indexed: 12/14/2022]
Abstract
This review presents an analysis of current research aimed at deciphering the interplay of cell extrinsic and intrinsic signals required for specification and differentiation of noradrenergic sympathetic ganglion neurons. The development of noradrenergic sympathetic ganglion neurons depends upon expression of a core set of DNA regulatory molecules, including the Phox2 homeodomain proteins and the basic helix-loop-helix proteins, HAND2 and MASH1 whose expression is dependent upon cell extrinsic cues. Both bone morphogenetic protein(s) and cAMP have an integral role in the specification/differentiation of noradrenergic sympathetic ganglion neurons but how signaling downstream of these molecules is integrated and identification of their particular functions is just beginning to be elucidated. Data currently available suggests a model with BMP providing both instructive and permissive cues in a pathway integrated by cAMP and MAPK by activation of both canonical and non-canonical intracellular signaling cascades.
Collapse
Affiliation(s)
- Anjali A Sarkar
- Department of Neurosciences, Program in Cellular and Molecular Neurobiology, Medical University of Ohio, 3000 Arlington Avenue, Toledo, OH 43614, USA
| | | |
Collapse
|
42
|
Borghini S, Bachetti T, Fava M, Di Duca M, Cargnin F, Fornasari D, Ravazzolo R, Ceccherini I. The TLX2 homeobox gene is a transcriptional target of PHOX2B in neural-crest-derived cells. Biochem J 2006; 395:355-61. [PMID: 16402914 PMCID: PMC1422762 DOI: 10.1042/bj20051386] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The TLX2 (HOX11L1, Ncx, Enx) and PHOX2B genes encode transcription factors crucial in the development of neural-crest-derived cells, leading to ANS (autonomic nervous system) specific neuronal lineages. Moreover, they share a similar expression pattern and are both involved in downstream steps of BMP (bone morphogenetic protein) signalling. In an attempt to reconstruct the gene network sustaining the correct development of the ANS, we have undertaken an in vitro experimental strategy to identify direct upstream regulators of the TLX2 gene. After characterizing a sequence displaying enhancer property in its 5' flanking region, we confirmed the functional link between the human PHOX2B and TLX2 genes. Transient transfections and electrophoretic-mobility-shift assays suggested that PHOX2B is able to bind the cell-specific element in the 5' regulatory region of the TLX2 gene, determining its transactivation in neuroblastoma cells. Such interaction was also confirmed in vivo by means of chromatin immunoprecipitation assay and, in addition, up-regulation of endogenous TLX2 mRNA level was demonstrated following PHOX2B over-expression, by quantitative real-time PCR. Finally, PHOX2B proteins carrying mutations responsible for CCHS (congenital central hypoventilation syndrome) development showed a severe impairment in activating TLX2 expression, both in vitro and in vivo. Taken together, these results support the PHOX2B-TLX2 promoter interaction, suggesting a physiological role in the transcription-factor cascade underlying the differentiation of neuronal lineages of the ANS during human embryogenesis.
Collapse
Affiliation(s)
- Silvia Borghini
- *Laboratorio di Genetica Molecolare, Istituto Gerolamo Gaslini, 16148 Genova, Italy
| | - Tiziana Bachetti
- *Laboratorio di Genetica Molecolare, Istituto Gerolamo Gaslini, 16148 Genova, Italy
| | - Monica Fava
- *Laboratorio di Genetica Molecolare, Istituto Gerolamo Gaslini, 16148 Genova, Italy
| | - Marco Di Duca
- †Laboratorio di Fisiopatologia dell'Uremia, Istituto Giannina Gaslini, 16148 Genova, Italy
| | - Francesca Cargnin
- ‡Dipartimento di Farmacologia, Facolta' di Medicina, Universita' di Milano e CNR Istituto di Neuroscienze, Milano, Italy
| | - Diego Fornasari
- ‡Dipartimento di Farmacologia, Facolta' di Medicina, Universita' di Milano e CNR Istituto di Neuroscienze, Milano, Italy
| | - Roberto Ravazzolo
- *Laboratorio di Genetica Molecolare, Istituto Gerolamo Gaslini, 16148 Genova, Italy
- §Dipartimento di Pediatria e CEBR, Università di Genova, Genova, Italy
| | - Isabella Ceccherini
- *Laboratorio di Genetica Molecolare, Istituto Gerolamo Gaslini, 16148 Genova, Italy
- To whom correspondence should be addressed (email )
| |
Collapse
|
43
|
Hyun JW, Yoon SH, Yu Y, Han CS, Park JS, Kim HS, Lee SJ, Lee YS, You HJ, Chung MH. Oh8dG induces G1 arrest in a human acute leukemia cell line by upregulating P21 and blocking the RAS to ERK signaling pathway. Int J Cancer 2006; 118:302-9. [PMID: 16052517 DOI: 10.1002/ijc.21329] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We reported previously that KG-1, a human acute leukemia cell line, has mutational loss of 8-oxoguanine (8-hydroxyguanine; oh8Gua) glycosylase 1 (OGG1) activity and undergoes apoptotic death after treatment with 7,8-dihydro-8-oxo-2'-deoxyguanosine (8-oxodeoxyguanosine, 8-hydroxydeoxyguanosine; oh8dG). In our present study, we further characterized the effects of oh8dG in KG-1 cells and found that, in addition to apoptosis, oh8dG induced the arrest of KG-1 at the G1 phase. Simultaneously, oh8dG-treated KG-1 showed an increase in the oh8Gua content of DNA, upregulation of p21 (an inhibitor of cdk), and Ras inactivation. Moreover, the upregulation of p21 was followed by the inactivations of cdk4 and cdk2, the hypophosphorylation of Rb, and a marked decline in the expression of c-myc (a gene regulated by E2F that is a transcription factor whose activity is suppressed when it is bound to hypophosphorylated Rb). Ras inactivation was also followed by the inactivation of ERK kinase (MEK) and the inactivation of AP-1, a downstream target of the Ras signaling pathway. The specific MEK inhibitors, PD98059 and U0126, also induced G1 arrest. These findings suggest that p21 upregulation and Ras inactivation contribute to G1 arrest. An increase of oh8Gua content in DNA does not seem to be a principal contributor to G1 arrest, however, because the kinetics of increases of oh8Gua content in DNA and of G1 cell number did not coincide. We report that oh8dG induces the arrest of KG-1 growth at the G1 phase mainly by upregulating p21 and inactivating Ras.
Collapse
Affiliation(s)
- Jin Won Hyun
- Department of Biochemistry, College of Medicine and Applied Radiological Science Research Institute, Cheju National University, Jeju, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Morikawa Y, Dai YS, Hao J, Bonin C, Hwang S, Cserjesi P. The basic helix-loop-helix factor Hand 2 regulates autonomic nervous system development. Dev Dyn 2006; 234:613-21. [PMID: 16145670 PMCID: PMC2653092 DOI: 10.1002/dvdy.20544] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Mammalian autonomic nervous system (ANS) development requires the combinatorial action of a number of transcription factors, which include Mash 1, Phox 2b, and GATA 3. Here we show that the bHLH transcription factor, Hand 2 (dHAND), is expressed concurrently with Mash 1 during sympathetic nervous system (SNS) development and that the expression of Hand 2 is not dependent on Mash 1. This suggests that these two bHLH factors work in parallel during SNS development. We also show that ectopic expression of Hand 2 activates the neuronal program and promotes the acquisition of a phenotype corresponding to peripheral neurons including neurons of the SNS lineage in P19 embryonic carcinoma cells. We propose that Hand 2 works in parallel with other members of the transcriptional network to regulate ANS developmental but can ectopically activate the program by a cross-regulatory mechanism that includes the activation of Mash 1. We show that this function is dependent on its interaction with the histone acetyltransferase p300/CBP, indicating that Hand 2 functions to promote ANS development as part of a larger transcriptional complex.
Collapse
Affiliation(s)
- Yuka Morikawa
- Department of Cell and Molecular Biology, Tulane University, 2000 Percival Stern Hall, New Orleans, LA, 70118
| | - Yan-Shan Dai
- Department of Anatomy and Cell Biology, Columbia University, 604 West 168 Street, New York, NY, 10032
| | - Jianming Hao
- Department of Anatomy and Cell Biology, Columbia University, 604 West 168 Street, New York, NY, 10032
| | - Christopher Bonin
- Department of Anatomy and Cell Biology, Columbia University, 604 West 168 Street, New York, NY, 10032
| | - Sunny Hwang
- Department of Anatomy and Cell Biology, Columbia University, 604 West 168 Street, New York, NY, 10032
| | - Peter Cserjesi
- Department of Cell and Molecular Biology, Tulane University, 2000 Percival Stern Hall, New Orleans, LA, 70118
- Corresponding Author, Ph. (504) 862-8081, Fax (504) 862-8082, E-mail:
| |
Collapse
|
45
|
Benjanirut C, Paris M, Wang WH, Hong SJ, Kim KS, Hullinger RL, Andrisani OM. The cAMP pathway in combination with BMP2 regulates Phox2a transcription via cAMP response element binding sites. J Biol Chem 2005; 281:2969-81. [PMID: 16330553 DOI: 10.1074/jbc.m503939200] [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: 11/06/2022] Open
Abstract
Combined BMP2 and cAMP signaling induces the catechola-minergic lineage in neural crest (NC) cultures by increasing expression of the proneural transcription factor Phox2a, in a cAMP response element (CRE)-binding protein (CREB)-mediated mechanism. To determine whether CREB acts directly on Phox2a transcription induced by BMP2+cAMP-elevating agent IBMX, transient transfections of hPhox2a-reporter constructs were performed in avian NC cultures and murine, catecholaminergic CAD cells. Although BMP2+IBMX increased endogenous Phox2a expression, the 7.5-kb hPhox2a reporters expressing either luciferase or DsRed1-E5 fluorescent protein were unresponsive to BMP2+IBMX, but active in both cell types. Cell sorting of fluorescence-positive NC cells expressing the 7.5-kb hPhox2a fluorescent timer reporter differentiated to equal numbers of catecholaminergic cells as fluorescence-negative cells, suggesting inappropriate transcription from the transfected hPhox2a promoter. NC or CAD cells treated with histone deacetylase inhibitor trichostatin A and BMP2+IBMX display increased endogenous Phox2a transcription and prolonged CREB phosphorylation, indicating Phox2a chromatin remodeling is linked to CREB activation. Chromatin immunoprecipitations employing CREB, CREB-binding protein, and acetylated H4 antibodies identified two CRE half-sites at -5.5 kb in the murine Phox2a promoter, which is also conserved in the human promoter. Proximal to the CRE half-sites, within a 170-bp region, are E-box and CCAAT binding sites, also conserved in mouse and human genes. This 170-bp promoter region confers cAMP, BMP2, and enhanced BMP2+cAMP regulation to Phox2a-luciferase reporters. We conclude these CREs are functional, with CREB directly activating Phox2a transcription. Because the E-box binds bHLH proteins like ASH1 induced in NC cells by BMP2, we propose this novel 170-bp cis-acting element is a composite site, mediating the synergistic regulation by BMP2+cAMP on Phox2a transcription.
Collapse
Affiliation(s)
- Chutamas Benjanirut
- Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana 47907, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Kang KA, Lee KH, Chae S, Zhang R, Jung MS, Kim SY, Kim HS, Kim DH, Hyun JW. Cytoprotective effect of tectorigenin, a metabolite formed by transformation of tectoridin by intestinal microflora, on oxidative stress induced by hydrogen peroxide. Eur J Pharmacol 2005; 519:16-23. [PMID: 16102749 DOI: 10.1016/j.ejphar.2005.06.043] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2005] [Revised: 06/27/2005] [Accepted: 06/28/2005] [Indexed: 10/25/2022]
Abstract
In the present study, the antioxidative properties of tectorigenin, a metabolite formed by transformation of tectoridin by intestinal microflora, were investigated. Tectorigenin was found to scavenge intracellular reactive oxygen species, and 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical, and thus prevented lipid peroxidation. The radical scavenging activity of tectorigenin protected the viability of Chinese hamster lung fibroblast (V79-4) cells exposed to hydrogen peroxide (H2O2) via activation of extracellular signal regulated kinase (ERK) pathway. Furthermore, tectorigenin reduced the apoptotic cells formation and cell cycle arrest at G2/M phase induced by H2O2. Tectorigenin increased the activities of cellular antioxidant enzymes like superoxide dismutase, catalase, glutathione peroxidase, and also increased their protein level. Taken together, these findings suggest that tectorigenin protected V79-4 cells against H2O2 damage, by enhancing the antioxidative activity and by activating ERK pathway.
Collapse
Affiliation(s)
- Kyoung Ah Kang
- Department of Biochemistry, College of Medicine and Applied Radiological Science Research Institute, Cheju National University, Jeju-si 690-756, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Adachi M, Keefer EW, Jones FS. A segment of the Mecp2 promoter is sufficient to drive expression in neurons. Hum Mol Genet 2005; 14:3709-22. [PMID: 16251199 DOI: 10.1093/hmg/ddi402] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Rett syndrome (RTT) is caused by mutations in the gene encoding methyl CpG-binding protein 2 (MeCP2). Although MeCP2 shows widespread expression in both neuronal and non-neuronal tissues, the symptoms of RTT are largely neurological. Herein, we have identified the regulatory region of the mouse Mecp2 gene that is sufficient for its restricted expression in neurons. A segment of the Mecp2 gene (-677/+56) exhibited strong promoter activity in neuronal cell lines and cortical neurons, but was inactive in non-neuronal cells and glia. The region necessary for neuronal-specific promoter activity was located within a 19 bp region (-63/-45). Several nuclear factors were found to bind to this region and some of these factors were enriched in nuclear extracts prepared from the brain. To examine the activity of the Mecp2 promoter in vivo, we generated transgenic mice expressing the LacZ reporter driven by the -677/+56 region of the Mecp2 gene. The transgene was expressed in the mesencephalon as early as embryonic day 10 and in the hindbrain and spinal cord by E12. Interestingly, a marked induction of transgene expression was observed postnatally throughout the brain, similar to that of endogenous MeCP2. However, expression of the transgene was absent in non-neuronal tissues that are known to express Mecp2. Taken together, these data indicate that the -677/+56 region of the Mecp2 promoter partially recapitulates the native expression pattern of the Mecp2 gene, which possesses restricted expression in neurons of the central nervous system.
Collapse
Affiliation(s)
- Megumi Adachi
- The Neurosciences Institute, San Diego, Ca 92121, USA
| | | | | |
Collapse
|
48
|
Trochet D, Hong SJ, Lim JK, Brunet JF, Munnich A, Kim KS, Lyonnet S, Goridis C, Amiel J. Molecular consequences of PHOX2B missense, frameshift and alanine expansion mutations leading to autonomic dysfunction. Hum Mol Genet 2005; 14:3697-708. [PMID: 16249188 DOI: 10.1093/hmg/ddi401] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Heterozygous mutations of the PHOX2B gene account for a broad variety of disorders of the autonomic nervous system, either isolated or combined, including congenital central hypoventilation syndrome (CCHS), tumours of the sympathetic nervous system and Hirschsprung disease. In CCHS, the prevalent mutation is an expansion of a 20-alanine stretch ranging from +5 to +13 alanines, whereas frameshift and missense mutations are found occasionally. To determine the molecular basis of impaired PHOX2B function, we assayed the transactivation and DNA binding properties of wild-type and mutant PHOX2B proteins. Furthermore, we investigated aggregate formation by proteins with polyalanine tract expansions ranging from +5 to +13 alanines using immunofluorescence of transfected cells and gel filtration of in vitro translated proteins. We found that transactivation of the dopamine beta-hydroxylase promoter by PHOX2B proteins with frameshift and missense mutations was abolished or severely curtailed, as was in vitro DNA binding although the proteins localized to the nucleus. The transactivation potential of proteins with polyalanine tract expansions declined with increasing length of the polyalanine stretch, and DNA binding was affected for an expansion of +9 alanines and above. Cytoplasmic aggregation in transfected cells was only observed for the longest expansions, whereas even the short expansion mutants were prone to form multimers in vitro. Such a tendency to protein misfolding could explain loss of transactivation for alanine expansion mutations. However, additional mechanisms such as toxic gain-of-function may play a role in the pathogenic process.
Collapse
Affiliation(s)
- Delphine Trochet
- Université Paris-Descartes, Faculté de Médecine; INSERM; AP-HP, Hôpital Necker-Enfants Malades, INSERM U-393, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Kang KA, Kim YW, Kim SU, Chae S, Koh YS, Kim HS, Choo MK, Kim DH, Hyun JW. G1 phase arrest of the cell cycle by a ginseng metabolite, compound K, in U937 human monocytic leukamia cells. Arch Pharm Res 2005; 28:685-90. [PMID: 16042078 DOI: 10.1007/bf02969359] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We recently reported that the ginseng saponin metabolite, compound K (20-O-beta-D-glucopyranosyl-20(S)-protopanaxadiol, IH901), inhibits the growth of U937 cells through caspase-dependent apoptosis pathway. In this study, we further characterized the effects of compound K on U937 cells and found that, in addition to apoptosis, compound K induced the arrest of the G1 phase. The compound K treated U937 cells showed increased p21 expression; an inhibitory protein of cyclin-cdk complex. The up-regulation of p21 was followed by the inactivation of cyclin D and the cdk4 protein, which act at the early G1 phase, and cyclin E, which acts at the late G1 phase. Furthermore, compound K induced the activation of JNK and the transcription factor AP-1, which is a downstream target of JNK. These findings suggest that the up-regulation of p21 and activation of JNK in the compound K treated cells contribute to the arrest of the G1 phase.
Collapse
Affiliation(s)
- Kyoung Ah Kang
- Department of Biochemistry, College of Medicine and Applied Radiological Science Research Institute, Cheju National University, 66 Jejudaehakno, Jeju-si 690-756 Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Zhu MY, Wang WP, Iyo AH, Ordway GA, Kim KS. Age-associated changes in mRNA levels of Phox2, norepinephrine transporter and dopamine beta-hydroxylase in the locus coeruleus and adrenal glands of rats. J Neurochem 2005; 94:828-38. [PMID: 16033425 PMCID: PMC2923405 DOI: 10.1111/j.1471-4159.2005.03245.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Age-related changes in the gene expression of the transcription factors, Phox2a and 2b, and two marker proteins, norepinephrine transporter (NET) and dopamine beta-hydroxylase (DBH), of noradrenergic neurons were characterized in the locus coeruleus (LC) and adrenal glands using in situ hybridization. Analysis of changes was performed in rats that were 1-23 months of age. Compared to 1-month-old rats, there was a 62% increase of Phox2a messenger RNA (mRNA) in the LC of 3-month-old rats, and a decline of 37% in 23-month-old rats. In contrast, levels of Phox2b mRNA in the LC remained unchanged in 3-month-old rats, but declined to a 30% reduction in 23-month-old rats. Interestingly, mRNA levels of NET in the LC decreased with increasing age to a reduction of 29%, 30% and 43% in 3-, 8- and 23-month-old rats, respectively. Similarly, DBH mRNA in the LC declined with increasing age to a 56% reduction in 23-month-old rats. mRNA levels of Phox2a, Phox2b, NET and DBH in the adrenal medulla of 23-month-old rats were significantly lower than those of 1-month-old rats. Semi-quantitative reverse transcription assays of the same genes yielded data similar to in situ hybridization experiments, with beta-actin mRNA levels being unchanged across the ages. Taken together, these data reveal that reduced Phox2 mRNAs in the LC and adrenal medulla of aging rats are accompanied by a coincidental decline in mRNA levels of NET and DBH and suggest a possible relationship between Phox2 genes and the marker genes in noradrenergic neurons after birth.
Collapse
Affiliation(s)
- Meng-Yang Zhu
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA.
| | | | | | | | | |
Collapse
|