1
|
Sayson LV, Lee HJ, Ortiz DM, Kim M, Custodio RJP, Lee CH, Lee YS, Cheong JH, Kim HJ. The differential vulnerabilities of Per2 knockout mice to the addictive properties of methamphetamine and cocaine. Prog Neuropsychopharmacol Biol Psychiatry 2023; 126:110782. [PMID: 37141987 DOI: 10.1016/j.pnpbp.2023.110782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/18/2023] [Accepted: 04/30/2023] [Indexed: 05/06/2023]
Abstract
With the pervasive occurrence of substance abuse worldwide, unraveling the neuropharmacology of drugs of abuse, such as psychostimulants, is undeniably essential. Mice lacking Period 2 (Per2), a gene associated with the biological time-regulating system or circadian rhythm, have been proposed as a potential animal model for drug abuse vulnerability, demonstrating a greater preference for methamphetamine (METH) reward than wild-type (WT) mice. However, the responses of Per2 knockout (KO) mice to the reinforcing effects of METH or other psychostimulants are yet to be established. In this study, the responses of WT and Per2 KO mice to various psychostimulants via intravenous self-administration were determined, along with their behaviors in METH- or cocaine (COC)-induced conditioned place preference and spontaneous locomotion in the open-field test. Per2 KO mice exhibited greater addiction-like responses to METH and 5-EAPB (1-(1-benzofuran-5-yl)-N-ethylpropan-2-amine), but their responses to COC and dimethocaine were comparable to WT mice, indicating a divergent influence of Per2 deficiency on abuse susceptibility to specific psychostimulants. To potentially define the underlying mechanism for this phenotype, 19 differentially expressed genes were identified, through RNA sequencing, which might respond specifically to repeated METH, but not COC, administration in the mouse striatum and were narrowed down to those previously associated with immediate early genes or synaptic plasticity. The correlation between locomotor activity and mRNA expression levels revealed a moderate correlation between METH-induced behavior and Arc or Junb expression in Per2 KO mice only, suggesting their essential role that may lead to the higher vulnerability of Per2 KO mice to METH, but not COC. These findings indicate a potentially unique effect of Per2 expression level on the involvement of Arc and Junb in determining specific vulnerabilities to drugs, and possibly including abuse potential.
Collapse
Affiliation(s)
- Leandro Val Sayson
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, 815 Hwarang-ro, Nowon-gu, Seoul 01795, Republic of Korea
| | - Hyun Jun Lee
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, 815 Hwarang-ro, Nowon-gu, Seoul 01795, Republic of Korea
| | - Darlene Mae Ortiz
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, 815 Hwarang-ro, Nowon-gu, Seoul 01795, Republic of Korea
| | - Mikyung Kim
- Department of Chemistry & Life Science, Sahmyook University, 815 Hwarang-ro, Nowon-gu, Seoul 01795, Republic of Korea
| | - Raly James Perez Custodio
- Department of Ergonomics, Leibniz Research Centre for Working Environment and Human Factors - IfADo, Ardeystr. 67, 44139 Dortmund, Germany
| | - Chae Hyeon Lee
- Medicinal Chemistry Laboratory, Department of Fundamental Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yong Sup Lee
- Medicinal Chemistry Laboratory, Department of Fundamental Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jae Hoon Cheong
- Institute for New Drug Development, School of Pharmacy, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju, Jeollabuk-do 54896, Republic of Korea.
| | - Hee Jin Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, 815 Hwarang-ro, Nowon-gu, Seoul 01795, Republic of Korea.
| |
Collapse
|
2
|
Fan Y, Zeng F, Brown RW, Price JB, Jones TC, Zhu MY. Transcription Factors Phox2a/2b Upregulate Expression of Noradrenergic and Dopaminergic Phenotypes in Aged Rat Brains. Neurotox Res 2020; 38:793-807. [PMID: 32617854 PMCID: PMC7484387 DOI: 10.1007/s12640-020-00250-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/30/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022]
Abstract
The present study investigated the effects of forced overexpression of Phox2a/2b, two transcription factors, in the locus coeruleus (LC) of aged rats on noradrenergic and dopaminergic phenotypes in brains. Results showed that a significant increase in Phox2a/2b mRNA levels in the LC region was paralleled by marked enhancement in expression of DBH and TH per se. Furthermore, similar increases in TH protein levels were observed in the substantial nigra and striatum, as well as in the hippocampus and frontal cortex. Overexpression of Phox2 genes also significantly increased BrdU-positive cells in the hippocampal dentate gyrus and NE levels in the striatum. Moreover, this manipulation significantly improved the cognition behavior. The in vitro experiments revealed that norepinephrine treatments may increase the transcription of TH gene through the epigenetic action on the TH promoter. The results indicate that Phox2 genes may play an important role in improving the function of the noradrenergic and dopaminergic neurons in aged animals, 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 Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
- Department of Biochemistry, Nantong University College of Medicine, Nantong, China
| | - Fei Zeng
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
- Department of Neurology, Remin Hospital of the Wuhan University, Wuhan, China
| | - Russell W Brown
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Jennifer B Price
- Department of Biological Sciences, College of Arts and Sciences, East Tennessee State University, Johnson City, TN, USA
| | - Thomas C Jones
- Department of Biological Sciences, College of Arts and Sciences, East Tennessee State University, Johnson City, TN, USA
| | - Meng-Yang Zhu
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.
| |
Collapse
|
3
|
Díaz HS, Toledo C, Andrade DC, Marcus NJ, Del Rio R. Neuroinflammation in heart failure: new insights for an old disease. J Physiol 2020; 598:33-59. [PMID: 31671478 DOI: 10.1113/jp278864] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 09/09/2019] [Indexed: 08/25/2023] Open
Abstract
Heart failure (HF) is a complex clinical syndrome affecting roughly 26 million people worldwide. Increased sympathetic drive is a hallmark of HF and is associated with disease progression and higher mortality risk. Several mechanisms contribute to enhanced sympathetic activity in HF, but these pathways are still incompletely understood. Previous work suggests that inflammation and activation of the renin-angiotensin system (RAS) increases sympathetic drive. Importantly, chronic inflammation in several brain regions is commonly observed in aged populations, and a growing body of evidence suggests neuroinflammation plays a crucial role in HF. In animal models of HF, central inhibition of RAS and pro-inflammatory cytokines normalizes sympathetic drive and improves cardiac function. The precise molecular and cellular mechanisms that lead to neuroinflammation and its effect on HF progression remain undetermined. This review summarizes the most recent advances in the field of neuroinflammation and autonomic control in HF. In addition, it focuses on cellular and molecular mediators of neuroinflammation in HF and in particular on brain regions involved in sympathetic control. Finally, we will comment on what is known about neuroinflammation in the context of preserved vs. reduced ejection fraction HF.
Collapse
Affiliation(s)
- Hugo S Díaz
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camilo Toledo
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - David C Andrade
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Noah J Marcus
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA, USA
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Envejecimiento y Regeneración (CARE-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
4
|
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: 3.8] [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
|
5
|
Byrne CJ, Khurana S, Kumar A, Tai TC. Inflammatory Signaling in Hypertension: Regulation of Adrenal Catecholamine Biosynthesis. Front Endocrinol (Lausanne) 2018; 9:343. [PMID: 30013513 PMCID: PMC6036303 DOI: 10.3389/fendo.2018.00343] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/07/2018] [Indexed: 12/24/2022] Open
Abstract
The immune system is increasingly recognized for its role in the genesis and progression of hypertension. The adrenal gland is a major site that coordinates the stress response via the hypothalamic-pituitary-adrenal axis and the sympathetic-adrenal system. Catecholamines released from the adrenal medulla function in the neuro-hormonal regulation of blood pressure and have a well-established link to hypertension. The immune system has an active role in the progression of hypertension and cytokines are powerful modulators of adrenal cell function. Adrenal medullary cells integrate neural, hormonal, and immune signals. Changes in adrenal cytokines during the progression of hypertension may promote blood pressure elevation by influencing catecholamine biosynthesis. This review highlights the potential interactions of cytokine signaling networks with those of catecholamine biosynthesis within the adrenal, and discusses the role of cytokines in the coordination of blood pressure regulation and the stress response.
Collapse
Affiliation(s)
- Collin J. Byrne
- Department of Biology, Laurentian University, Sudbury, ON, Canada
| | - Sandhya Khurana
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON, Canada
| | - Aseem Kumar
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON, Canada
- Biomolecular Sciences Program, Laurentian University, Sudbury, ON, Canada
| | - T. C. Tai
- Department of Biology, Laurentian University, Sudbury, ON, Canada
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON, Canada
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON, Canada
- Biomolecular Sciences Program, Laurentian University, Sudbury, ON, Canada
| |
Collapse
|
6
|
Agarwal D, Dange RB, Raizada MK, Francis J. Angiotensin II causes imbalance between pro- and anti-inflammatory cytokines by modulating GSK-3β in neuronal culture. Br J Pharmacol 2014; 169:860-74. [PMID: 23516971 DOI: 10.1111/bph.12177] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 01/10/2013] [Accepted: 02/11/2013] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Emerging evidence indicates that the balance between pro-inflammatory cytokines (PICs) and anti-inflammatory cytokines (AICs) within the brain is an important determinant in the outcome of hypertension. However, the mechanism by which this dysregulation occurs is not known. We aimed to investigate whether AngII induces imbalance between PIC and AIC by modulating downstream transcription factors, NFκB and cyclic AMP response element-binding protein (CREB), and whether AngII-induced effects are mediated by glycogen synthase kinase-3β (GSK-3β). EXPERIMENTAL APPROACH CATH.a neurons were exposed to AngII (10 nM-1 μM) over a preset time course. In another set of experiments, GSK-3β was knock down by using lentivirus containing short hairpin RNA targeting GSK-3β (L-sh-GSK3β) before AngII exposure. Cell extracts were subjected to RT-PCR, immunoblot and immunoprecipitation. KEY RESULTS AngII caused time-dependent increase in PICs (TNF-α and IL-1β) and reduction in AIC (IL-10). AngII exposure caused reduced phosphorylated CREB(Ser-133) and increased p-NFκB(Ser-276) levels, leading to reduced CREB-CBP and increased NFκB-CBP binding. These results were accompanied by increased activation of GSK-3β, as indicated by increased p-GSK3(Tyr-216) to p-GSK3(Ser-9) ratio. In a subsequent study, pretreatment with L-sh-GSK3β attenuated AngII-induced alterations in PICs and IL-10 by augmenting CREB-CBP and attenuating NFκB-CBP binding. CONCLUSIONS AND IMPLICATIONS Collectively, these findings are the first to provide direct evidence that AngII-induced dysregulation in cytokines is mediated by GSK-3β-mediated alterations in downstream transcription factors in neuronal cells. Our data also reveal that AngII-induced effects could be alleviated by GSK-3β inhibition, suggesting GSK-3β as an important therapeutic target for hypertension that is characterized by increased PICs and NFκB activation.
Collapse
Affiliation(s)
- Deepmala Agarwal
- Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | | | | | | |
Collapse
|
7
|
Bucharles C, Bizet P, Arthaud S, Arabo A, Leprince J, Lefranc B, Cartier D, Anouar Y, Lihrmann I. Concordant localization of functional urotensin II and urotensin II-related peptide binding sites in the rat brain: Atypical occurrence close to the fourth ventricle. J Comp Neurol 2014; 522:2634-49. [DOI: 10.1002/cne.23553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 01/23/2014] [Accepted: 01/23/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Christine Bucharles
- Inserm, U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine; University of Rouen; Mont-Saint-Aignan France
- Normandy University, University of Rouen; Mont-Saint-Aignan France
| | - Patrice Bizet
- Inserm, U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine; University of Rouen; Mont-Saint-Aignan France
- Normandy University, University of Rouen; Mont-Saint-Aignan France
| | - Sébastien Arthaud
- Inserm, U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine; University of Rouen; Mont-Saint-Aignan France
- Normandy University, University of Rouen; Mont-Saint-Aignan France
| | - Arnaud Arabo
- Normandy University, University of Rouen; Mont-Saint-Aignan France
- Faculty of Sciences; University of Rouen; Mont-Saint-Aignan France
| | - Jérôme Leprince
- Inserm, U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine; University of Rouen; Mont-Saint-Aignan France
- Normandy University, University of Rouen; Mont-Saint-Aignan France
| | - Benjamin Lefranc
- Inserm, U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine; University of Rouen; Mont-Saint-Aignan France
- Normandy University, University of Rouen; Mont-Saint-Aignan France
| | - Dorthe Cartier
- Inserm, U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine; University of Rouen; Mont-Saint-Aignan France
- Normandy University, University of Rouen; Mont-Saint-Aignan France
| | - Youssef Anouar
- Inserm, U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine; University of Rouen; Mont-Saint-Aignan France
- Normandy University, University of Rouen; Mont-Saint-Aignan France
| | - Isabelle Lihrmann
- Inserm, U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine; University of Rouen; Mont-Saint-Aignan France
- Normandy University, University of Rouen; Mont-Saint-Aignan France
| |
Collapse
|
8
|
Haack KKV, Mitra AK, Zucker IH. NF-κB and CREB are required for angiotensin II type 1 receptor upregulation in neurons. PLoS One 2013; 8:e78695. [PMID: 24244341 PMCID: PMC3823855 DOI: 10.1371/journal.pone.0078695] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 09/12/2013] [Indexed: 01/14/2023] Open
Abstract
Nuclear factor kappa B (NF-κB) and the Ets like gene-1 (Elk-1) are two transcription factors that have been previously established to contribute to the Angiotensin II mediated upregulation of Angiotensin II type 1 receptor (AT1R) in neurons. The cAMP response element binding protein (CREB) is another transcription factor that has also been implicated in AT1R gene transcription. The goal of the current study was to determine if NF-κB and CREB association was required for AT1R upregulation. We hypothesized that the transcription of the AT1R gene occurs via an orchestration of transcription factor interactions including NF-κB, CREB, and Elk-1. The synergistic role of CREB and NFκB in promoting AT1R gene expression was determined using siRNA-mediated silencing of CREB. Electrophorectic Mobility Shift Assay studies employing CREB and NF-κB demonstrated increased protein - DNA binding as a result of Ang II stimulation which was blunted by siRNA silencing of CREB. Upstream inhibition of p38 mitogen activated protein kinase (p38 MAPK) with SB203580 or inhibition of the calmodulin kinase (CAMK) pathway using KN-62 blunted changes in CREB and NF-κB expression. These findings suggest that Ang II may activate multiple signaling pathways involving p38 MAPK leading to the activation of NF-κB and CREB, which feed back to upregulate the AT1R gene. This study provides insight into the molecular mechanisms involving multiple transcription factor activation in a coordinated fashion which may be partially responsible for sympathoexcitation in clinical conditions associated with increased activation of the renin angiotensin system.
Collapse
Affiliation(s)
- Karla K. V. Haack
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Amit K. Mitra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Irving H. Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
9
|
Distribution and neurochemical characterization of neurons in the rat ventrolateral medulla activated by glucoprivation. Brain Struct Funct 2013; 220:117-34. [DOI: 10.1007/s00429-013-0642-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 09/18/2013] [Indexed: 12/14/2022]
|
10
|
Parker LM, Kumar NN, Lonergan T, Goodchild AK. Neurochemical codes of sympathetic preganglionic neurons activated by glucoprivation. J Comp Neurol 2013; 521:2703-18. [DOI: 10.1002/cne.23310] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 09/10/2012] [Accepted: 01/15/2013] [Indexed: 11/10/2022]
Affiliation(s)
- Lindsay M. Parker
- The Australian School of Advanced Medicine; Macquarie University; Macquarie Park; 2109 New South Wales; Australia
| | - Natasha N. Kumar
- The Australian School of Advanced Medicine; Macquarie University; Macquarie Park; 2109 New South Wales; Australia
| | - Tina Lonergan
- The Australian School of Advanced Medicine; Macquarie University; Macquarie Park; 2109 New South Wales; Australia
| | - Ann K. Goodchild
- The Australian School of Advanced Medicine; Macquarie University; Macquarie Park; 2109 New South Wales; Australia
| |
Collapse
|
11
|
Amano A, Tsunoda M, Aigaki T, Maruyama N, Ishigami A. Age-related changes of dopamine, noradrenaline and adrenaline in adrenal glands of mice. Geriatr Gerontol Int 2012; 13:490-6. [PMID: 22934574 DOI: 10.1111/j.1447-0594.2012.00929.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AIM Catecholamines, which are physiologically important neurotransmitters and hormones, apparently decrease in the brain and plasma as some species age. Because this observation has engendered controversy, we used mice to investigate whether age-related changes occur in adrenal catecholamine levels and in the expression of catecholamine synthetic enzymes. METHODS Adrenal glands were collected from male C57BL/6NCr mice at the ages of 6, 12 and 24 months. Catecholamines, such as dopamine (DA), noradrenaline (NA) and adrenaline (AD) from those glands, were measured by using a highly sensitive liquid chromatographic method with peroxyoxalate chemiluminescence reaction detection. Tyrosine hydroxylase (TH), dopa decarboxylase, dopamine beta hydroxylase (DBH) and phenylethanolamine N-methyltransferase (PNMT) mRNA expression levels were measured by quantitative real-time polymerase chain reaction. RESULTS Although DA levels in the adrenals of 24-month-old mice were higher than in 6- and 12-month-old mice, the AD content decreased with age. In such mice, the ratio of DA to NA at 24 months was lower than at 12 months, and the ratio of NA to AD content at 24 months was significantly lower than at 6 months. The mRNA expression ratios in TH, DBH and PNMT in 24-month-old mice were all lower than in 12-month-old mice. CONCLUSIONS These results strongly suggest that catecholamine synthesis, in general, declines with aging in the adrenal glands of mice and that AD, in particular, undergoes a significant decrease with advancing age.
Collapse
Affiliation(s)
- Akiko Amano
- Molecular Regulation of Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | | | | | | | | |
Collapse
|
12
|
Richter AM, Walesch SK, Würl P, Taubert H, Dammann RH. The tumor suppressor RASSF10 is upregulated upon contact inhibition and frequently epigenetically silenced in cancer. Oncogenesis 2012; 1:e18. [PMID: 23552700 PMCID: PMC3412644 DOI: 10.1038/oncsis.2012.18] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 05/10/2012] [Accepted: 05/16/2012] [Indexed: 12/11/2022] Open
Abstract
The Ras association domain family (RASSF) comprises a group of tumor suppressors that are frequently epigenetically inactivated in various tumor entities and linked to apoptosis, cell cycle control and microtubule stability. In this work, we concentrated on the newly identified putative tumor suppressor RASSF10. Methylation analysis reveals RASSF10 promoter hypermethylation in lung cancer, head and neck (HN) cancer, sarcoma and pancreatic cancer. An increase in RASSF10 methylation from normal tissues, primary tumors to cancer cell lines was observed. Methylation was reversed by 5-aza-2'-deoxycytidine treatment leading to reexpression of RASSF10. We further show that overexpression of RASSF10 suppresses colony formation in cancer cell lines. In addition, RASSF10 is upregulated by cell-cell contact and regulated on promoter level as well as endogenously by forskolin, protein kinase A (PKA) and activator Protein 1 (AP-1), linking RASSF10 to the cAMP signaling pathway. Knockdown of the AP-1 member JunD interfered with contact inhibition induced RASSF10 expression. In summary, we found RASSF10 to be epigenetically inactivated by hypermethylation of its CpG island promoter in lung, HN, sarcoma and pancreatic cancer. Furthermore, our novel findings suggest that tumor suppressor RASSF10 is upregulated by PKA and JunD signaling upon contact inhibition and that RASSF10 suppresses growth of cancer cells.
Collapse
Affiliation(s)
- A M Richter
- Institute for Genetics, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
| | - S K Walesch
- Institute for Genetics, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
| | - P Würl
- Department of General and Visceral Surgery, Diakoniekrankenhaus Halle, Halle, Germany
| | - H Taubert
- University Clinic of Urology, Division of Molecular Urology, Friedrich-Alexander-University, Erlangen-Nürnberg, Germany
| | - R H Dammann
- Institute for Genetics, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
| |
Collapse
|
13
|
Haque R, Chong NW, Ali F, Chaurasia SS, Sengupta T, Chun E, Howell JC, Klein DC, Iuvone PM. Melatonin synthesis in retina: cAMP-dependent transcriptional regulation of chicken arylalkylamine N-acetyltransferase by a CRE-like sequence and a TTATT repeat motif in the proximal promoter. J Neurochem 2011; 119:6-17. [PMID: 21790603 DOI: 10.1111/j.1471-4159.2011.07397.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Arylalkylamine N-acetyltransferase (AANAT) is the key regulatory enzyme controlling the daily rhythm of melatonin biosynthesis. In chicken retinal photoreceptor cells, Aanat transcription and AANAT activity are regulated in part by cAMP-dependent mechanisms. The purpose of this study was to identify regulatory elements within the chicken Aanat promoter responsible for cAMP-dependent induction. Photoreceptor-enriched retinal cell cultures were transfected with a luciferase reporter construct containing up to 4 kb of 5'-flanking region and the first exon of Aanat. Forskolin treatment stimulated luciferase activity driven by the ∼4 kb promoter construct and by all 5'-deletion constructs except the smallest, Aanat (-217 to +120)luc. Maximal basal and forskolin-stimulated expression levels were generated by the Aanat (-484 to +120)luc construct. This construct lacks a canonical cyclic AMP-response element (CRE), but contains two other potentially important elements in its sequence: an eight times TTATT repeat (TTATT₈) and a CRE-like sequence. Electrophoretic mobility shift assays, luciferase reporter assays, chromatin immunoprecipitation, and siRNA experiments provide evidence that these elements bind c-Fos, JunD, and CREB to enhance basal and forskolin-stimulated Aanat transcription. We propose that the CRE-like sequence and TTATT₈ elements in the 484 bp proximal promoter interact to mediate cAMP-dependent transcriptional regulation of Aanat.
Collapse
Affiliation(s)
- Rashidul Haque
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Jagalur NB, Ghazvini M, Mandemakers W, Driegen S, Maas A, Jones EA, Jaegle M, Grosveld F, Svaren J, Meijer D. Functional dissection of the Oct6 Schwann cell enhancer reveals an essential role for dimeric Sox10 binding. J Neurosci 2011; 31:8585-94. [PMID: 21653862 PMCID: PMC3137940 DOI: 10.1523/jneurosci.0659-11.2011] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Revised: 03/10/2011] [Accepted: 03/23/2011] [Indexed: 01/28/2023] Open
Abstract
The POU domain transcription factor Pou3f1 (Oct6/Scip/Tst1) initiates the transition from ensheathing, promyelinating Schwann cells to myelinating cells. Axonal and other extracellular signals regulate Oct6 expression through the Oct6 Schwann cell enhancer (SCE), which is both required and sufficient to drive all aspects of Oct6 expression in Schwann cells. Thus, the Oct6 SCE is pivotal in the gene regulatory network that governs the onset of myelin formation in Schwann cells and provides a link between myelin promoting signaling and activation of a myelin-related transcriptional network. In this study, we define the relevant cis-acting elements within the SCE and identify the transcription factors that mediate Oct6 regulation. On the basis of phylogenetic comparisons and functional in vivo assays, we identify a number of highly conserved core elements within the mouse SCE. We show that core element 1 is absolutely required for full enhancer function and that it contains closely spaced inverted binding sites for Sox proteins. For the first time in vivo, the dimeric Sox10 binding to this element is shown to be essential for enhancer activity, whereas monomeric Sox10 binding is nonfunctional. As Oct6 and Sox10 synergize to activate the expression of the major myelin-related transcription factor Krox20, we propose that Sox10-dependent activation of Oct6 defines a feedforward regulatory module that serves to time and amplify the onset of myelination in the peripheral nervous system.
Collapse
Affiliation(s)
- Noorjahan B Jagalur
- Department of Cell Biology and Genetics, Erasmus University Medical Center, 3000 DR Rotterdam, Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Serova LI, Nostramo R, Veerasirikul M, Cappell DB, Sabban EL. Varied mechanisms of oestradiol-mediated regulation of dopamine β-hydroxylase transcription. J Neuroendocrinol 2011; 23:168-76. [PMID: 21062376 PMCID: PMC3077094 DOI: 10.1111/j.1365-2826.2010.02086.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Experiments performed in vivo and in cell culture have demonstrated that oestradiol induces dopamine β-hydroxylase (DBH) gene transcription. In the present study, we examined oestrogen-responsive elements of the rat DBH gene promoter aiming to characterise the mechanisms of oestradiol-induced DBH transcription. Various mutations and deletions of DBH promoter reporter constructs were tested for responsiveness to 17β-oestradiol (E(2) ). Mutation of the half palindromic oestrogen response element (ERE) at position -759 reduced the response to E(2) in PC12 cells co-transfected with oestrogen receptor (ER) α, indicating a functional role for this motif. In cells co-transfected with ERβ, mutations at the -759 site were unresponsive to E(2) . To characterise the additional E(2) responsive elements, mediated by ERα, the DBH promoter was truncated to the proximal 249 or 200 nucleotides upstream of the transcription start site. Despite either truncation, 10 nm E(2) still elicited an approximately two-fold induction of DBH promoter activity. Mutation of a possible ERE-like sequence at -59 had no effect. The lack of a functional ERE in the proximal region of the rat DBH promoter despite E(2) -mediated DBH promoter activity, suggests regulation by a nonclassical mechanism, such as a membrane-initiated signalling pathway. Moreover, the induction of DBH promoter activity and the rise in DBH mRNA levels were observed within hours. To determine whether membrane-initiated E(2) signalling is involved in rat DBH gene transcription, a membrane impermeable E(2) conjugate, β-oestradiol-6-(O-carboxy-methyl) oxime-bovine serum albumin (E(2) BSA), was used. Incubation with E(2) -BSA induced luciferase activity and elicited a significant rise in DBH mRNA levels in the ERα transfected cells. The findings indicate two different mechanisms whereby DBH transcription is regulated by E(2) in the presence of ERα. The results implicate both genomic and membrane-initiated mechanisms, mediated by ERα, in E(2) -induced DBH gene transcription.
Collapse
Affiliation(s)
- Lidia I Serova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York 10595 USA
| | - Regina Nostramo
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York 10595 USA
| | - Mary Veerasirikul
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York 10595 USA
| | - Deborah B. Cappell
- Department of Pulmonary Medicine, Mt Sinai Hospital, New York, NY 10128 USA
| | - Esther L. Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York 10595 USA
| |
Collapse
|
16
|
Mitra AK, Gao L, Zucker IH. Angiotensin II-induced upregulation of AT(1) receptor expression: sequential activation of NF-kappaB and Elk-1 in neurons. Am J Physiol Cell Physiol 2010; 299:C561-9. [PMID: 20554912 PMCID: PMC2944315 DOI: 10.1152/ajpcell.00127.2010] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Accepted: 06/10/2010] [Indexed: 01/16/2023]
Abstract
It has been clearly established that increased circulating angiotensin II (ANG II) with concurrent upregulation of brain and peripheral ANG II type 1 receptors (AT(1)R) are important mediators in the pathophysiology of several diseases characterized by sympatho-excitation. In an effort to further understand the regulation of AT(1)R expression in neurons, we determined the role of sequential activation of the transcription factors nuclear factor-kappaB (NF-kappaB) and Ets-like protein 1 (Elk-1) in AT(1)R upregulation. We used CATH.a neurons as our neuronal cell model. Cells were treated with ANG II (100 nM) over a preset time course. Following ANG II activation, there was a temporal increase in the p65 subunit of NF-kappaB that was observed at 30 min, peaked at 1 h, and was sustained up to 24 h. There was a concomitant decrease of IkappaB and increased IkappaK expression. We also observed an increase in AT(1)R expression which followed the temporal increase of NF-kappaB. The activation of NF-kappaB was blocked by using the inhibitors parthenolide or p65 small interfering RNA (siRNA) which both led to a decrease in AT(1)R expression. The expression of Elk-1 was upregulated over a time period following ANG II activation and was decreased following NF-kappaB inhibition. p65-DNA binding was assessed using electrophoretic mobility shift assay, and it was shown that there was a time-dependent increased binding that was inhibited by means of parthenolide pretreatment or siRNA-mediated p65 gene silencing. Therefore, our results suggest a combined role for the transcription factors NF-kappaB and Elk-1 in the upregulation of AT(1)R in the CATH.a cell neuronal model. These data imply a positive feedback mechanism that may impact neuronal discharge sensitivity in response to ANG II.
Collapse
Affiliation(s)
- Amit K Mitra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | | |
Collapse
|
17
|
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: 363] [Impact Index Per Article: 22.7] [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
|
18
|
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
|
19
|
Das P, Ezashi T, Gupta R, Roberts RM. Combinatorial roles of protein kinase A, Ets2, and 3',5'-cyclic-adenosine monophosphate response element-binding protein-binding protein/p300 in the transcriptional control of interferon-tau expression in a trophoblast cell line. Mol Endocrinol 2007; 22:331-43. [PMID: 17975022 DOI: 10.1210/me.2007-0300] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In ruminants, conceptus interferon-tau (IFNT) production is necessary for maintenance of pregnancy. We examined the role of protein kinase A (PKA) in regulating IFNT expression through the activation of Ets2 in JAr choriocarcinoma cells. Although overexpression of the catalytic subunit of PKA or the addition of 8-bromo-cAMP had little ability to up-regulate boIFNT1 reporter constructs on their own, coexpression with Ets2 led to a large increase in gene expression. Progressive truncation of reporter constructs indicated that the site of PKA/Ets2 responsiveness lay in a region of the promoter between -126 and -67, which lacks a cAMP response element but contains the functional Ets2-binding site and an activator protein 1 (AP1) site. Specific mutation of the former reduced the PKA/Ets2 effects by more than 98%, whereas mutation of an AP1-binding site adjacent to the Ets2 site or pharmacological inhibition of MAPK kinase 2 led to a doubling of the combined Ets2/PKA effects, suggesting there is antagonism between the Ras/MAPK pathway and the PKA signal transduction pathway. Although Ets2 is not a substrate for PKA, lowering the effective concentrations of the coactivators, cAMP response element-binding protein-binding protein (CBP)/p300, known PKA targets, reduced the ability of PKA to synergize with Ets2, suggesting that PKA effects on IFNT regulation might be mediated through CBP/p300 coactivation, particularly as CBP and Ets2 occupy the proximal promoter region of IFNT in bovine trophoblast CT-1 cells. The up-regulation of IFNT in the elongating bovine conceptus is likely due to the combinatorial effects of PKA, Ets2, and CBP/p300 and triggered via growth factors released from maternal endometrium.
Collapse
Affiliation(s)
- Padmalaya Das
- Christopher S. Bond Life Sciences Center, University of Missouri-Columbia, 1201 East Rollins Street, Columbia, MO 65211-7310, USA
| | | | | | | |
Collapse
|
20
|
Page ME, Oropeza VC, Sparks SE, Qian Y, Menko AS, Van Bockstaele EJ. Repeated cannabinoid administration increases indices of noradrenergic activity in rats. Pharmacol Biochem Behav 2007; 86:162-8. [PMID: 17275893 PMCID: PMC1941574 DOI: 10.1016/j.pbb.2006.12.020] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2006] [Revised: 12/19/2006] [Accepted: 12/21/2006] [Indexed: 11/24/2022]
Abstract
The present study examined the impact of repeated administration of a synthetic cannabinoid agonist, WIN 55,212-2 on the coeruleo-cortical pathway, a circuit implicated in anxiety. Male Sprague-Dawley rats received repeated systemic injections of WIN 55,212-2 (3.0 mg/kg). A separate group of rats received repeated WIN 55,212-2 injections followed by a period of abstinence. Control animals received vehicle injections. Ninety minutes following the last injection on day 8, anxiety-related behavior was assessed using the elevated plus maze. The abstinent group was tested after another 8 days. Following behavioral testing, brain tissue was extracted from the locus coeruleus (LC) and probed for tyrosine hydroxylase (TH) expression. In a separate group of animals, in vivo microdialysis was used to monitor extracellular norepinephrine efflux in the frontal cortex following repeated WIN 55,212-2 administration and following a period of abstinence. Repeated administration of WIN 55,212-2 evoked an anxiogenic-like response that was accompanied by an increase in TH protein expression in the LC. A similar neurochemical profile was observed using in vivo microdialysis where an augmented increase in cortical norepinephrine efflux was identified in response to a systemic injection of WIN 55,212-2 on day 8. Anxiety-like behavior, catecholamine synthesizing enzyme levels and NE efflux returned to control values after 8 days of abstinence. The present findings indicate that repeated administration of a synthetic cannabinoid receptor agonist induces transient anxiety-like behaviors that correlate with increases in catecholamine synthesizing enzyme expression in the LC and augmented norepinephrine efflux in response to a challenge injection of WIN 55,212-2.
Collapse
Affiliation(s)
- M E Page
- Thomas Jefferson University, Department of Neurosurgery, Farber Institute for Neurosciences, Philadelphia, PA 19107, United States.
| | | | | | | | | | | |
Collapse
|
21
|
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: 0.8] [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
|
22
|
Wang J, Shannon MF, Young IG. A role for Ets1, synergizing with AP-1 and GATA-3 in the regulation of IL-5 transcription in mouse Th2 lymphocytes. Int Immunol 2005; 18:313-23. [PMID: 16373364 DOI: 10.1093/intimm/dxh370] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
IL-5 is a key regulator of eosinophilic inflammation and is selectively expressed by antigen-activated Th2 lymphocytes. An important role for the proximal AP-1 and GATA sites in regulating IL-5 transcription is generally accepted but the significance of an adjacent Ets/NFAT site has remained unclear. We have investigated its role using the mouse Th2 clone D10.G4.1. Transcription of IL-5 reporter gene plasmids could be induced in D10 cells by phorbol myristate acetate/cyclic adenosine monophosphate (PMA/cAMP) stimulation and significantly further enhanced by activation of the mitogen-activated protein (MAP) kinase pathways. Strong induction of IL-5 mRNA was also induced by PMA/cAMP. Mutagenesis showed that the Ets/NFAT site is of critical importance along with the AP-1 and GATA sites in regulating IL-5 transcription stimulated by PMA/cAMP and MAP kinase activation. Transactivation was used to investigate the transcription factors which could function at the three sites and possible synergistic interactions. AP-1 (c-Fos/c-Jun) strongly induced IL-5 transcription and dominant negative AP-1 constructs confirmed that AP-1 plays an important role in regulating IL-5 expression. Ets1, unlike other members of the Ets/NFAT family, synergized strongly with AP-1 suggesting that Ets1 is the family member which functions at the Ets/NFAT site. AP-1/Ets1 transactivation also stimulated IL-5 mRNA expression. Ets1 binding to the proximal promoter region, demonstrated by chromatin immunoprecipitation, was stimulated by PMA/cAMP. The absolute dependence on the binding sites for Ets1, AP-1 and GATA-3 together with the strong synergy between Ets1 and AP-1 suggest close cooperative interactions between the three transcription factors in the regulation of IL-5 expression in mouse T cells.
Collapse
Affiliation(s)
- Jun Wang
- Division of Molecular Bioscience, John Curtin School of Medical Research, Australian National University, Mills Road, Acton, ACT 0200 Australia
| | | | | |
Collapse
|
23
|
Hsieh MM, Lupas G, Rychlik J, Dziennis S, Habecker BA, Lewis EJ. ERK1/2 is a negative regulator of homeodomain protein Arix/Phox2a. J Neurochem 2005; 94:1719-27. [PMID: 16156742 DOI: 10.1111/j.1471-4159.2005.03333.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The homeodomain protein Arix/Phox2a plays a role in the development and maintenance of the noradrenergic cell type by regulating the transcription of genes involved in the biosynthesis and metabolism of noradrenaline. Previous work has shown that Arix/Phox2a is a phosphoprotein, and the phosphorylated form of Arix/Phox2a exhibits poorer DNA-binding activity than does the dephosphorylated form. Here, we demonstrate that Arix/Phox2a is phosphorylated by extracellular signal-related kinase (ERK)1/2 at two sites within the N-terminal transactivation domain. The phosphorylation level of Arix in cultured SH-SY5Y neuroblastoma cells is reduced when cells are treated with the mitogen activated protein kinase kinase 1 (MEK1) inhibitor UO126. Treatment of sympathetic neurons with the MEK1 inhibitor, PD98059, results in an elevation of mRNAs encoding noradrenergic proteins, dopamine beta-hydroxylase (DBH) and norepinephrine transporter (NET), but not tyrosine hydroyxlase (TH). Treatment of neuroblastoma cultures with PD98059 increases the interaction of Arix with DBH and NET genes, but not the TH gene. Together, these results suggest that phosphorylation of Arix by ERK1/2 inhibits its ability to interact with target genes, and that both specificity of expression and modulation by external stimuli are monitored through the same transcription factor.
Collapse
Affiliation(s)
- Marlene M Hsieh
- Department of Biochemistry and Molecular Biology, Oregon Health Sciences University, Portland, Oregon 97239, USA
| | | | | | | | | | | |
Collapse
|
24
|
Liu H, Margiotta JF, Howard MJ. BMP4 supports noradrenergic differentiation by a PKA-dependent mechanism. Dev Biol 2005; 286:521-36. [PMID: 16165122 DOI: 10.1016/j.ydbio.2005.08.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2005] [Revised: 08/10/2005] [Accepted: 08/11/2005] [Indexed: 01/13/2023]
Abstract
Differentiation of neural crest-derived noradrenergic neurons depends upon signaling mediated downstream of BMP binding to cognate receptors and involving cAMP. Compiled data from many groups suggest that neurogenesis and cell type-specific noradrenergic marker gene regulation is coordinated through the expression and function of the basic helix-loop-helix DNA binding protein HAND2 and the homeodomain DNA binding protein Phox2a. However, information detailing how BMP-mediated signaling and signaling through cAMP are coordinated has been lacking. We now provide compelling data suggesting that differentiation of noradrenergic sympathetic ganglion neurons depends upon both canonical and non-canonical pathways of BMP-mediated signaling. The non-canonical pathway involves the activation of protein kinase A (PKA) independent of cAMP. This is a novel mechanism in neural crest-derived cells and is necessary to support neurogenesis as well as aspects of DBH promoter regulation involving HAND2 phosphorylation and dimerization. The expression of transcripts encoding HAND2 and Phox2a is regulated via canonical BMP signaling and thus affects both neurogenesis and cell type-specific gene expression. Interestingly, cAMP- and MapK-mediated signaling modulate specific target sites in both the canonical and non-canonical BMP pathways. Activity of MapK is required for HAND2 transcription and thus affects neurogenesis. Signaling affected by cAMP is necessary for the transcription of Phox2a as well as regulation of DBH promoter transactivation by Phox2a and HAND2. We suggest a comprehensive model that shows how BMP- and cAMP-mediated intracellular signaling integrate neurogenesis and cell type-specific noradrenergic marker gene expression and function.
Collapse
Affiliation(s)
- Hongbin Liu
- Department of Neurosciences, Program in Molecular and Cellular Neuroscience, Medical University of Ohio, 3000 Arlington Ave., Toledo, OH 43614, USA
| | | | | |
Collapse
|
25
|
Kaufman GD. Fos expression in the vestibular brainstem: what one marker can tell us about the network. ACTA ACUST UNITED AC 2005; 50:200-11. [PMID: 16039721 DOI: 10.1016/j.brainresrev.2005.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2004] [Revised: 05/12/2005] [Accepted: 06/09/2005] [Indexed: 11/28/2022]
Abstract
Fos inducible transcription factor expression in rodent brains (rats and gerbils) during manipulations of vestibular input is reviewed. Stimuli included centripetal hypergravity, unilateral labyrinth lesion or semicircular canal plugging, rotational axis cross-coupling (Coriolis forces), high and low rotational vestibulo-ocular reflex gain adaptation, translabyrinth galvanic stimulation, pharmacological manipulation, and combinations thereof. Each type of stimulation elicited unique but partially redundant response patterns in the vestibulo-olivo-cerebellar (VOC) network that reflect the origin and interaction of the labyrinth inputs. On the basis of these patterns, a trained observer can predict what the animal experienced during testing; the patterns of VOC Fos expression reveal a trace of recent genomic activity. Based on principal component analysis, VOC network modules associated with lesion recovery, spatial representation and the calibration of gravity, and optokinetic influences are proposed. Probable and possible gene targets of the Fos protein are also reviewed.
Collapse
Affiliation(s)
- Galen D Kaufman
- 7.102 Medical Research Building, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-1063, USA.
| |
Collapse
|
26
|
Chen WK, Chang NCA, Chang YH, Chang KL, Wu SC, Yang TS, Wu SM, Chang AC. Characterization of the regulatory region of Adra2c, the gene encoding the murine alpha2C adrenoceptor subtype. J Biomed Sci 2005; 11:886-901. [PMID: 15591786 DOI: 10.1007/bf02254374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2004] [Accepted: 06/29/2004] [Indexed: 01/17/2023] Open
Abstract
The 5' flanking sequence (3,227 base pairs, bp) of the mouse Adra2c subtype gene was determined and characterized. The transcription start site was mapped to nucleotide 'A' of two initiator motifs in tandem array, i.e. 1,159 and 1,153 bp upstream from the initiation codon of the open reading frame (ORF) of Adra2c, respectively. One structural feature salient to the 5' regulatory region of Adra2c is present in the sequence 1 kb immediately upstream from the receptor ORF, which is highly enriched in GC content (76%) and CpG island counts (i.e. CpG/GpC, 146:177), and thus rich in Sp1-binding motifs. At the 3' flanking region, the polyadenylation signal was mapped to 481 bp downstream from the termination codon. The transcript defined by sequence data thereby is consistent with a size of 3 kb (brain form) determined by Northern blot analysis. The transgene, Adra2c-NN- lacZ, which links the promoter region of Adra2c to the lacZ reporter gene, was constructed in order to evaluate the functional capacity of the promoter and the putative motifs residing within the defined regulatory region (1.9 kb upstream from the ORF) in directing the reporter gene expression in vitro in transiently transfected cells and in vivo in transgenic (Tg) mice. Permissive cell types to Adra2c-NN include those derived from neural and kidney lineages. Significant Adra2c-NN-driven reporter expression in Tg mice established suggests that alpha2C adrenoceptor expression is permissive under Adra2c-NN in central (cerebral cortex, hippocampus, subthalamus, hypothalamus, superior colliculus, cerebellum, and brain stem) and peripheral (pancreatic beta-islets) tissues.
Collapse
MESH Headings
- Amino Acid Motifs
- Animals
- Base Sequence
- Blotting, Northern
- Blotting, Southern
- Brain/metabolism
- Central Nervous System/metabolism
- Cloning, Molecular
- Codon
- Computational Biology
- CpG Islands
- Gene Library
- Gene Transfer Techniques
- Genes, Reporter
- Humans
- Islets of Langerhans/metabolism
- Lac Operon
- Mice
- Mice, Transgenic
- Models, Genetic
- Molecular Sequence Data
- Nucleic Acid Hybridization
- Open Reading Frames
- Polymerase Chain Reaction
- Promoter Regions, Genetic
- Receptors, Adrenergic, alpha-2/chemistry
- Receptors, Adrenergic, alpha-2/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Tissue Distribution
- Transcription Factors
- Transcription, Genetic
- Transfection
- Transgenes
- beta-Galactosidase/metabolism
Collapse
Affiliation(s)
- Wen-Kwei Chen
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, ROC
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Hwang DY, Hwang MM, Kim HS, Kim KS. Genetically engineered dopamine beta-hydroxylase gene promoters with better PHOX2-binding sites drive significantly enhanced transgene expression in a noradrenergic cell-specific manner. Mol Ther 2005; 11:132-41. [PMID: 15585414 DOI: 10.1016/j.ymthe.2004.08.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2004] [Accepted: 08/24/2004] [Indexed: 11/24/2022] Open
Abstract
A continuously growing body of evidence suggests that dysregulation of noradrenergic (NA) neurons is implicated in the etiology and pathophysiology of various human diseases such as depression, drug addiction, and autonomic dysfunction. An efficient NA neuron-specific promoter is potentially valuable to investigate the precise role of NA neurons in normal as well as in diseased brain and to treat the associated disorders by gene therapy. In this study, we tested a novel strategy to modify genetically the promoter of the human dopamine beta-hydroxylase (hDBH) gene to overcome its inherent weakness while maintaining its cell-type specificity. We optimized the nucleotide sequence motifs of PHOX2-binding sites (PRS2 and PRS3) residing within the hDBH promoter. Optimization of both PRS2 and PRS3 motifs significantly increased their binding affinities to PHOX2A, leading to a dramatic increase in the promoter strength (>20-fold). More importantly, these modifications do not alter the level of transgene expression in non-NA cells either in vitro or in vivo, demonstrating tight cell-type specificity. This work shows that a cellular gene promoter can be genetically modified to strengthen its promoter activity without losing cell-type specificity by optimizing critical cis-regulatory elements. Our genetically engineered promoter may be useful for cell-type-specific gene targeting as well as for generating in vivo animal models with altered gene expression in a specific cell type.
Collapse
Affiliation(s)
- Dong-Youn Hwang
- Molecular Neurobiology Laboratory, McLean Hospital, and Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA
| | | | | | | |
Collapse
|
28
|
Howard MJ. Mechanisms and perspectives on differentiation of autonomic neurons. Dev Biol 2005; 277:271-86. [PMID: 15617674 DOI: 10.1016/j.ydbio.2004.09.034] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2004] [Revised: 09/22/2004] [Accepted: 09/27/2004] [Indexed: 01/17/2023]
Abstract
Neurons share many features in common but are distinguished by expression of phenotypic characteristics that define their specific function, location, or connectivity. One aspect of neuronal fate determination that has been extensively studied is that of neurotransmitter choice. The generation of diversity of neuronal subtypes within the developing nervous system involves integration of extrinsic and intrinsic instructive cues resulting in the expression of a core set of regulatory molecules. This review focuses on mechanisms of growth and transcription factor regulation in the generation of peripheral neural crest-derived neurons. Although the specification and differentiation of noradrenergic neurons are the focus, I have tried to integrate these into a larger picture providing a general roadmap for development of autonomic neurons. There is a core of DNA binding proteins required for the development of sympathetic, parasympathetic, and enteric neurons, including Phox2 and MASH1, whose specificity is regulated by the recruitment of additional transcriptional regulators in a subtype-specific manner. For noradrenergic neurons, the basic helix-loop-helix DNA binding protein HAND2 (dHAND) appears to serve this function. The studies reviewed here support the notion that neurotransmitter identity is closely linked to other aspects of neurogenesis and reveal a molecular mechanism to coordinate expression of pan-neuronal genes with cell type-specific genes.
Collapse
Affiliation(s)
- Marthe J Howard
- Department of Neurosciences, Medical College of Ohio, Toledo, OH 43614, USA.
| |
Collapse
|
29
|
Kim JS, Coon SL, Blackshaw S, Cepko CL, Møller M, Mukda S, Zhao WQ, Charlton CG, Klein DC. Methionine adenosyltransferase:adrenergic-cAMP mechanism regulates a daily rhythm in pineal expression. J Biol Chem 2004; 280:677-84. [PMID: 15504733 DOI: 10.1074/jbc.m408438200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
(S)-adenosylmethionine (SAM) is a critical element of melatonin synthesis as the methyl donor in the last step of the pathway, the O-methylation of N-acetyl 5-hydroxytryptamine by hydroxyindole-O-methyltransferase. The activity of the enzyme that synthesizes SAM, methionine adenosyltransferase (MAT), increases 2.5-fold at night in the pineal gland. In this study, we found that pineal MAT2A mRNA and the protein it encodes, MAT II, also increase at night, suggesting that the increase in MAT activity is caused by an increase in MAT II gene products. The night levels of MAT2A mRNA in the pineal gland were severalfold higher than in other neural and non-neural tissues examined, consistent with the requirement for SAM in melatonin synthesis. Related studies indicate that the nocturnal increase in MAT2A mRNA is caused by activation of a well described neural pathway that mediates photoneural-circadian regulation of the pineal gland. MAT2A mRNA and MAT II protein were increased in organ culture by treatment with norepinephrine (NE), the sympathetic neurotransmitter that stimulates the pineal gland at night. NE is known to markedly elevate pineal cAMP, and here it was found that cAMP agonists elevate MAT2A mRNA levels by increasing MAT2A mRNA synthesis and that drugs that block cAMP activation of cAMP dependent protein kinase block effects of NE. Therefore, the NE-cAMP dependent increase in pineal MAT activity seems to reflect an increase in MAT II protein, which occurs in response to cAMP-->protein kinase-dependent increased MAT2A expression. The existence of this MAT regulatory system underscores the importance that MAT plays in melatonin biogenesis. These studies also point to the possibility that SAM production in other tissues might be regulated through cAMP.
Collapse
Affiliation(s)
- Jong-So Kim
- Section on Neuroendocrinology, Laboratory of Developmental Neurobiology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Chun JT, Di Dato V, D'Andrea B, Zannini M, Di Lauro R. The CRE-like element inside the 5'-upstream region of the rat sodium/iodide symporter gene interacts with diverse classes of b-Zip molecules that regulate transcriptional activities through strong synergy with Pax-8. Mol Endocrinol 2004; 18:2817-29. [PMID: 15319451 DOI: 10.1210/me.2004-0020] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We previously demonstrated that transcription of the rat sodium/iodide symporter (NIS) gene is regulated by NUE, an upstream enhancer located between nucleotides -2264 and -2495 of the 5'-flanking region. To elucidate the mechanism of TSH/cAMP-mediated regulation of NIS gene expression, we have characterized the putative cAMP response element (CRE)/activator protein (AP)-1 site (termed NUC) that is closely located between the two Pax-8 (paired box domain transcription factor-8) binding sites within NUE. In two different approaches using either gel supershift analyses or dominant-negative inhibitors of b-Zip molecules, we have shown that NUC can be recognized by several members of the AP-1 and CREB family transcription factors that modulate the transcriptional activity of NUE. Using tethered dimers of b-Zip molecules, we have also demonstrated that specific homo- or heterodimers of AP-1 can synergistically stimulate NUE activity in concert with Pax-8. To demonstrate further that NUC is a bona fide CRE, we made an artificial promoter with the five-time tandem repeat of this sequence (5xNUC). In comparison to the canonical CRE (5xCRE), 5xNUC manifested greater transcriptional activity and broader response to cAMP signaling. Hence, we postulate that the significance of this evolutionally conserved CRE-like site may lie in its broader cell type specificity.
Collapse
Affiliation(s)
- J T Chun
- Department of Biochemistry and Molecular Biology, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Napoli, Italy
| | | | | | | | | |
Collapse
|
31
|
Naville D, Bordet E, Berthelon MC, Durand P, Bégeot M. Activator protein-1 is necessary for angiotensin-II stimulation of human adrenocorticotropin receptor gene transcription. ACTA ACUST UNITED AC 2003. [DOI: 10.1046/j.1432-1327.2001.02055.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
32
|
Xu H, Firulli AB, Zhang X, Howard MJ. HAND2 synergistically enhances transcription of dopamine-beta-hydroxylase in the presence of Phox2a. Dev Biol 2003; 262:183-93. [PMID: 14512028 DOI: 10.1016/s0012-1606(03)00361-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Noradrenergic neuronal identity and differentiation are controlled by cascades of transcription factors acting downstream of BMP4, including the basic helix-loop-helix DNA binding protein HAND2 and the homeodomain factor Phox2a. Dopamine-beta-hydroxylase (DBH) is the penultimate enzyme required for synthesis of norepinephrine and is thus a noradrenergic cell type-specific marker. We have examined the interaction of HAND2 and Phox2a at the DBH promoter. Using transient transfection of P19 or NT-2 cells, HAND2 is shown to synergistically enhance Phox2a-driven transcriptional activity at the DBH promoter, an effect that is enhanced by cAMP. While mutation of the Phox2a homeodomain binding sites HD1, HD2, and HD3 results in the loss of HAND2/Phox2a transactivation of DBH, it is the interaction of HAND2/Phox2a at the CRE/AP1-HD1/2 domains in the DBH enhancer that are required for synergistic activation by HAND2. We find that HAND2 functions as a transcriptional activator without directly binding to E-box sequences in the DBH promoter, suggesting that HAND2-mediated DBH activity occurs by protein-protein interactions with other transcriptional regulators. Although we were unable to detect interaction of HAND2 and Phox2a in IP/Western blots, HAND2 synergistic activation of DBH is blocked by E1A, suggesting that HAND2 interacts with CBP (cAMP response element binding protein) in this transcriptional complex. In the presence of the putative HAND2 dimerization partner, E12, synergistic activation of DBH transcription is titrated away, suggesting that HAND2 does not functionally dimerize with E12 in the DBH transcription complex. Our data suggest that HAND2 regulates cell type-specific expression of norepinephrine in concert with Phox2a by a novel mechanism.
Collapse
Affiliation(s)
- Haiming Xu
- Department of Anatomy and Neurobiology, Medical College of Ohio, 3000 Arlington Ave., Toledo, OH 43614, USA
| | | | | | | |
Collapse
|
33
|
Rychlik JL, Gerbasi V, Lewis EJ. The interaction between dHAND and Arix at the dopamine beta-hydroxylase promoter region is independent of direct dHAND binding to DNA. J Biol Chem 2003; 278:49652-60. [PMID: 14506227 DOI: 10.1074/jbc.m308577200] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Dopamine beta-hydroxylase (DBH) catalyzes the production of norepinephrine, and its expression defines the noradrenergic phenotype. Transcription factors dHAND, a basic helix-loop-helix protein, and Arix/Phox2a, a homeoprotein, have been demonstrated to play a role in the differentiation and maintenance of catecholaminergic neurons. Three Arix regulatory sites have been identified in the DBH promoter proximal region, but there is no such evidence for dHAND. Cotransfection with a DBH promoter-luciferase reporter construct plus dHAND or dHAND-E12 expression plasmids did not alter luciferase activity, whereas transfection with Arix resulted in a 2.5-fold stimulation of luciferase activity. However, a 5.5-fold increase was observed when Arix and dHAND were combined, and an 8-fold level of expression was observed when Arix was transfected with a dHAND mutant lacking the basic DNA-binding domain. When the homeodomain sites in the DBH promoter proximal region were mutated, all activity was lost, demonstrating dependence upon Arix-DNA interaction for transcriptional activation. In electrophoretic mobility shift assays, the addition of dHAND decreased the amount of Arix needed to elicit a mobility shift with the DBH homeodomain sites, and the dHAND basic mutant potentiated Arix binding in a manner similar to wild-type dHAND. The dHAND-Arix complex was dissociated upon the addition of an unlabeled competitor containing a homeodomain, but not upon the addition of a competitor containing E-boxes. Arix coprecipitated with antisera directed against recombinant dHAND, demonstrating direct protein-protein interactions. These results indicate that the activation of the DBH promoter by Arix is potentiated by dHAND via a mechanism independent of a direct interaction of dHAND with DNA.
Collapse
Affiliation(s)
- Jennifer L Rychlik
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon 97239, USA.
| | | | | |
Collapse
|
34
|
Sun B, Tank AW. c-Fos is essential for the response of the tyrosine hydroxylase gene to depolarization or phorbol ester. J Neurochem 2003; 85:1421-30. [PMID: 12787062 DOI: 10.1046/j.1471-4159.2003.01789.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Tyrosine hydroxylase (TH) gene transcription rate increases in response to numerous pharmacological and physiological stimuli. The AP1 site within the TH gene proximal promoter is thought to play an important role in mediating many of these responses; however, it is unclear which AP1 factors are required. To investigate whether c-Fos is essential for the response of the TH gene to different stimuli, c-Fos-deficient PC12 cell lines were produced utilizing an antisense RNA strategy. In these cell lines, stimulus-induced increases in c-Fos protein levels were dramatically attenuated, while c-Jun and CREB levels remained unchanged. TH gene transcription rate increased from four- to eight-fold in control cells after treatment with either 50 mM KCl or TPA. These responses were dramatically decreased in the c-Fos-deficient cell lines. In contrast, c-Fos down-regulation had little effect on the response of the TH gene to forskolin. Stimulation of TH gene promoter activity, which was observed in control cell lines treated with either 50 mm KCl or TPA was also dramatically inhibited in the c-Fos-deficient cells. These results suggest that c-Fos induction is essential for maximal stimulation of the TH gene in response to either depolarization or PKC activation in PC12 cells.
Collapse
Affiliation(s)
- Baoyong Sun
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York 14642, USA.
| | | |
Collapse
|
35
|
van den Munckhof P, Luk KC, Ste-Marie L, Montgomery J, Blanchet PJ, Sadikot AF, Drouin J. Pitx3 is required for motor activity and for survival of a subset of midbrain dopaminergic neurons. Development 2003; 130:2535-42. [PMID: 12702666 DOI: 10.1242/dev.00464] [Citation(s) in RCA: 229] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mesencephalic dopaminergic (MesDA) neurons play crucial roles in motor and behavioral processes; their loss in Parkinson's disease (PD) results in striatal dopamine (DA) deficiency and hypokinetic movement disorder. The Pitx3 homeobox gene is expressed in the MesDA system. We now show that only a subset of MesDA neurons express Pitx3 and that in Pitx3-deficient aphakia mice, this subset is progressively lost by apoptosis during fetal (substantia nigra, SN) and postnatal (ventral tegmental area) development, resulting in very low striatal DA and akinesia. Similar to human PD, dorsal SN neurons (which are Pitx3 negative) are spared in mutant mice. Thus, Pitx3 defines a pathway for survival of neurons that are implicated in PD and that are required for spontaneous locomotor activity.
Collapse
Affiliation(s)
- Pepijn van den Munckhof
- Unité de recherche en génétique moléculaire, Institut de recherches cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada
| | | | | | | | | | | | | |
Collapse
|
36
|
Tai TC, Wong DL. Protein kinase A and protein kinase C signaling pathway interaction in phenylethanolamine N-methyltransferase gene regulation. J Neurochem 2003; 85:816-29. [PMID: 12694408 DOI: 10.1046/j.1471-4159.2003.01728.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The protein kinase A (PKA) and protein kinase C (PKC) signaling pathways appear to interact in regulating phenylethanolamine N-methyltransferase (PNMT) promoter-driven gene transcription in PC12 cells. Forskolin treatment of cells transfected with the rat PNMT promoter-luciferase reporter gene construct pGL3RP893 increased promoter activity approximately two-fold whereas phorbol-12-myristate-13 acetate (PMA) treatment had no effect. However, simultaneous forskolin and PMA treatment synergistically activated the PNMT promoter approximately four-fold, suggesting that PKC stimulation requires prior induction of the PKA pathway. Consistent with this possibility the adenylate cyclase inhibitor MDL12,330A, and the PKA inhibitor H-89 prevented PNMT promoter stimulation by the combination of forskolin and PMA. PKA and PKC regulation seems to be mediated in part by Egr-1 and Sp1 through their consensus elements in the PNMT promoter. Forskolin and PMA treatment of PC12 cells increased Egr-1 protein and phosphorylated Egr-1/DNA-binding complex formation to the same extent but only increased phosphorylated Sp1/DNA binding complex formation without altering Sp1 protein levels. Mutation of the - 165 bp Egr-1 and - 48 bp Sp1 sites, respectively, attenuated and abolished combined forskolin and PMA-mediated promoter activation. PNMT promoter analysis further showed that synergistic stimulation by PKA and PKC involves DNA sequences between - 442 and - 392 bp, and potentially a GCM binding element lying within this region.
Collapse
Affiliation(s)
- T C Tai
- Department of Psychiatry, Harvard Medical School, Laboratory of Molecular and Developmental Neurobiology, McLean Hospital, Belmont, Massachusetts 02478, USA
| | | |
Collapse
|
37
|
Brooks HL, Ageloff S, Kwon TH, Brandt W, Terris JM, Seth A, Michea L, Nielsen S, Fenton R, Knepper MA. cDNA array identification of genes regulated in rat renal medulla in response to vasopressin infusion. Am J Physiol Renal Physiol 2003; 284:F218-28. [PMID: 12388413 DOI: 10.1152/ajprenal.00054.2002] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
With the aim of identifying possible gene targets for direct or indirect regulation by vasopressin in the renal medulla, we have carried out cDNA array experiments in inner medullas of Brattleboro rats infused with the V(2) receptor-selective vasopressin analog desamino-Cys1,d-Arg8 vasopressin (dDAVP) for 72 h. Of the 1,176 genes on the array, 137 transcripts were increased by 2-fold or more, and 10 transcripts were decreased to 0.5-fold or less. Quantitative, real-time RT-PCR measurements confirmed increases seen for six selected transcripts (Wilms' tumor protein, beta-arrestin 2, neurofibromin, casein kinase IIbeta, aquaporin-3, and aquaporin-4). To correlate changes in mRNA expression with changes in protein expression, we carried out quantitative immunoblotting for 28 of the proteins whose cDNAs were on the array. For several targets including aquaporin-2, transcript abundance and protein abundance changes did not correlate. However, for most genes examined, changes in mRNA abundances were associated with concomitant protein abundance changes. Targets with demonstrated increases in both protein and mRNA abundances included neurofibromin, casein kinase IIbeta, the beta-subunit of the epithelial Na channel (beta-ENaC), 11beta-hydroxysteroid dehydrogenase type 2, and c-Fos. Additional cDNA arrays revealed that several transcripts that were increased in abundance after 72 h of dDAVP were also increased after 4 h, including casein kinase IIbeta, beta-ENaC, aquaporin-3, UT-A, and syntaxin 2. These studies have identified several transcripts whose abundances are regulated in the inner medulla in response to infusion of dDAVP and that could play roles in the regulation of salt and water excretion.
Collapse
Affiliation(s)
- Heddwen L Brooks
- Laboratory of Kidney and Electrolyte Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Fralix KD, Zhao S, Venkatasubbarao K, Freeman JW. Rap1 reverses transcriptional repression of TGF-beta type II receptor by a mechanism involving AP-1 in the human pancreatic cancer cell line, UK Pan-1. J Cell Physiol 2003; 194:88-99. [PMID: 12447993 DOI: 10.1002/jcp.10192] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The TGF-beta signaling pathway has potent anti-mitogenic effects in epithelial cells and loss of negative growth regulation is often associated with increased tumorigenicity. The human pancreatic ductal adenocarcinoma cell line, UK Pan-1, which expresses DPC4, is not highly responsive to TGF-beta due to transcriptional repression of TGF-beta type II receptor (RII). Here, we show that UK Pan-1 cells transfected with a plasmid to overexpress rap1 protein (UK/rap1) causes an increase in RII transcription and restores sensitivity to TGF-beta growth inhibition. The overexpression of rap1 was associated with diminished ras signaling as measured by ras binding domain (RBD)-binding assays. Electrophoretic mobility shift assays (EMSA) analysis revealed increased binding of nuclear proteins to a previously identified positive regulatory element (PRE1) of the RII promoter in rap1 transfected cells. Competition with an oligo containing the AP-1 consensus site was able to inhibit this binding of nuclear proteins to the PRE1 region. Further EMSA analysis using antibodies to various AP-1 components revealed that junB antibodies partially depleted the increase in binding to the PRE1 seen in UK/rap1 cells while antibodies to other AP-1 constituents such as c-jun, c-fos, and ATF-1 had no effect on binding. Consistent with this data, transient transfection of UK Pan-1 cells with junB resulted in greater RII transcription (twofold) as measured by RII-luciferase assay. Mutation of the AP-1 site inhibited junB-mediated or rap1-mediated increases in RII promoter activity. These data suggest that rap1 signaling may mediate an increase in RII transcription via increased binding of nuclear factors including junB to the PRE1 region of the RII promoter.
Collapse
Affiliation(s)
- Kimberly D Fralix
- Department of Pharmacology, University of Texas Health Science Center, San Antonio 78229, USA
| | | | | | | |
Collapse
|
39
|
Adachi M, Lewis EJ. The paired-like homeodomain protein, Arix, mediates protein kinase A-stimulated dopamine beta-hydroxylase gene transcription through its phosphorylation status. J Biol Chem 2002; 277:22915-24. [PMID: 11943777 DOI: 10.1074/jbc.m201695200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The homeodomain transcription factor Arix/Phox2a plays a critical role in the specification of noradrenergic neurons by inducing the expression of dopamine beta-hydroxylase (DBH), the terminal enzyme for noradrenaline biosynthesis. In reporter assays, Arix together with activation of cAMP-dependent protein kinase (PKA) potentiates DBH gene transcription. We have evaluated whether post-translational modification of Arix regulates PKA-mediated DBH gene transcription. We found that Arix is constitutively phosphorylated in vivo at the basal level and that the phosphorylation level is substantially decreased upon stimulation of the PKA pathway. The change in the Arix phosphorylation state coincides with DNA binding activity of Arix. Treatment of cells with forskolin results in a robust enhancement of the DNA binding of Arix, which is reversed by treatment with serine/threonine and tyrosine phosphatase inhibitors. Consistent with the DNA binding activity of Arix, treatment of cultured cells with phosphatase inhibitors diminishes transcriptional activation with Arix plus forskolin. Amino acid analysis demonstrates the presence of phosphoserine within Arix. The results collectively suggest that dephosphorylation of Arix is a necessary event to fully activate PKA-mediated DBH transcription. Thus, the present study demonstrates that Arix can integrate extrinsic signals through post-translational modification, regulating DBH gene transcription in response to activation of the PKA pathway.
Collapse
Affiliation(s)
- Megumi Adachi
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon 97201, USA
| | | |
Collapse
|
40
|
Stork PJS, Schmitt JM. Crosstalk between cAMP and MAP kinase signaling in the regulation of cell proliferation. Trends Cell Biol 2002; 12:258-66. [PMID: 12074885 DOI: 10.1016/s0962-8924(02)02294-8] [Citation(s) in RCA: 704] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Hormonal stimulation of cyclic adenosine monophosphate (cAMP) and the cAMP-dependent protein kinase PKA regulates cell growth by multiple mechanisms. A hallmark of cAMP is its ability to stimulate cell growth in many cell types while inhibiting cell growth in others. In this review, the cell type-specific effects of cAMP on the mitogen-activated protein (MAP) kinase (also called extracellular signal-regulated kinase, or ERK) cascade and cell proliferation are examined. Two basic themes are discussed. First, the capacity of cAMP for either positive or negative regulation of the ERK cascade accounts for many of the cell type-specific actions of cAMP on cell proliferation. Second, there are several specific mechanisms involved in the inhibition or activation of ERKs by cAMP. Emerging new data suggest that one of these mechanisms might involve the activation of the GTPase Rap1, which can activate or inhibit ERK signaling in a cell-specific manner.
Collapse
Affiliation(s)
- Philip J S Stork
- Vollum Institute and the Dept of Cell and Developmental Biology, Oregon Health and Science University, Portland, OR 97201, USA.
| | | |
Collapse
|
41
|
Buono P, Cassano S, Alfieri A, Mancini A, Salvatore F. Human aldolase C gene expression is regulated by adenosine 3',5'-cyclic monophosphate (cAMP) in PC12 cells. Gene 2002; 291:115-21. [PMID: 12095685 DOI: 10.1016/s0378-1119(02)00586-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We have examined the effects of an adenosine 3',5'-cyclic monophosphate (cAMP) analog on human aldolase C gene expression in the rat pheochromocytoma cell line PC12. Incubation for 4 h with 500 microM 8-Br-cAMP increased aldolase C mRNA expression 2.5-fold and the expression was still above basal level 24 h later. Using transient transfection experiments we demonstrate that the distal element D in the promoter region of the human aldolase C gene, which binds a transcriptional activator (NGFI-B), is involved in this regulation. NGFI-B mRNA and protein expression were promptly (15 min) increased after 8-Br-cAMP treatment and precedes aldolase C mRNA increase (30 min). After 4 h of 8-Br-cAMP treatment, the binding of NGFI-B protein to the distal element D in the distal promoter region was increased twofold and this correlates with the increased expression of the clone that contains distal element D. These results indicate that the distal element D in the promoter region of the human aldolase C gene is the target of a cAMP-dependent regulation pathway.
Collapse
Affiliation(s)
- Pasqualina Buono
- Facoltà di Scienze Motorie, Università degli Studi di Napoli Parthenope, via Acton 38, 80125 Naples, Italy
| | | | | | | | | |
Collapse
|
42
|
Serova L, Rivkin M, Nakashima A, Sabban EL. Estradiol stimulates gene expression of norepinephrine biosynthetic enzymes in rat locus coeruleus. Neuroendocrinology 2002; 75:193-200. [PMID: 11914591 DOI: 10.1159/000048237] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Gender-specific differences in susceptibility to a number of disorders related to catecholaminergic systems, including depression and hypertension, have been postulated to be mediated, at least in part, by estrogens. In this study, we examined if estrogens may regulate gene expression of norepinephrine biosynthetic enzymes. Administration of five injections of 15 or 40 microg/kg estradiol benzoate to ovariectomized (OVX) female rats elicited a dose-dependent elevation in mRNA levels of tyrosine hydroxylase (TH) in locus coeruleus, to as great as 3-fold over control. Dopamine beta-hydroxylase (DBH) mRNA levels were also similarly increased. To examine the mechanism, PC12 cells were cotransfected with luciferase reporter constructs under control of DBH or TH promoters [pDBH/Luc(-2,236/+21) or pTH/Luc(-272/+27 or -773/+27)] with an expression vector for estradiol receptor alpha. The cells were treated with 17beta-estradiol (E(2)) for 12-36 h. E(2) triggered a several fold increase in luciferase activity under control of the DBH promoter in a dose-dependent fashion. Omission of estrogen receptor alpha or addition of the estrogen receptor antagonist ICI 182,780 prevented the DBH promoter-driven increase in luciferase. When E(2) was given with 0.2 mM CPT-cAMP, reporter activity with pDBH/Luc(-2,236/+21) was increased greater than with either treatment alone. In contrast, addition of E(2) to cells transfected with pTH/Luc(-272/+27) elicited no change in basal luciferase activity nor in the response to 0.2 mM CPT-cAMP. These findings are the first to reveal that estrogen can stimulate DBH gene expression. Differing mechanisms may underlie the regulation of TH and DBH gene expression by estrogens.
Collapse
Affiliation(s)
- Lidia Serova
- Department of Biochemistry and Molecular Biology, Basic Sciences Building, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | |
Collapse
|
43
|
Hong SJ, Kim CH, Kim KS. Structural and functional characterization of the 5' upstream promoter of the human Phox2a gene: possible direct transactivation by transcription factor Phox2b. J Neurochem 2001; 79:1225-36. [PMID: 11752063 DOI: 10.1046/j.1471-4159.2001.00672.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The specification of neurotransmitter identity is a critical step in neural development. Recent progresses have indicated that the closely related homeodomain factors Phox2a and 2b are essential for development of noradrenergic (NA) neuron differentiation, and may directly determine the neurotransmitter identity. With a long-term goal of understanding the regulatory cascade of NA phenotype determination, we isolated and characterized a hPhox2a genomic clone encompassing approximately 7.5 kb of the 5' upstream promoter region, the entire exon-intron structure, and approximately 4 kb of the 3' flanking region. Using mRNAs isolated from the Phox2a-expressing human cell line, both primer extension and 5'-rapid amplification of cDNA ends analyses identified a single transcription start site that resides 172 nucleotides upstream of the start codon. The transcription start site was preceded by a TATA-like sequence motif and transcripts from this site contained an additional G residue at the 5' position, supporting the authenticity of this site as the transcriptional start site of hPhox2a. We assembled hPhox2a-luciferase reporter constructs containing different lengths of the 5' upstream sequences. Transient transfection assays of these reporter constructs in both hPhox2a-positive and -negative cell lines show that 1.3-kb or longer upstream sequences of the hPhox2a gene may confer NA cell-specific reporter gene expression. Furthermore, cotransfection assays in the Phox2a-negative HeLa cell line show that forced expression of Phox2b, but not that of Phox2a or MASH1, significantly transactivates the transcriptional activity of hPhox2a. This study will provide a frame to further delineate the regulatory cascade of NA neuron differentiation.
Collapse
Affiliation(s)
- S J Hong
- Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, Massachusetts 02478, USA
| | | | | |
Collapse
|
44
|
de Santa Barbara P, Méjean C, Moniot B, Malclès MH, Berta P, Boizet-Bonhoure B. Steroidogenic factor-1 contributes to the cyclic-adenosine monophosphate down-regulation of human SRY gene expression. Biol Reprod 2001; 64:775-83. [PMID: 11207191 DOI: 10.1095/biolreprod64.3.775] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
In mammals, male sex determination is initiated by SRY (sex-determining region of the Y chromosome) gene expression and followed by testicular development. This study describes specific down-regulation of the human SRY gene transcription by cAMP stimulation using reverse transcription-polymerase chain reaction experiments. Using transfection experiments, conserved nuclear hormone receptor (NHR1) and Sp1 consensus binding sites were identified as essential for this cAMP transcriptional response. Steroidogenic factor-1 (SF-1), a component of the sex-determination cascade, binds specifically to the NHR1 site and activates the SRY promoter. Activation of SF-1 was abolished by cAMP pretreatment of the cells, suggesting a possible effect of cAMP on the SF-1 protein itself. Indeed, human SF-1 protein contains at least two in vitro cAMP-dependent protein kinase (PKA) phosphorylation sites, leading after phosphorylation to a modification of both DNA-binding activity and interaction with general transcription factors such as Sp1. Taken together, these data suggest that cAMP responsiveness of human SRY promoter involves both SF-1 and Sp1 sites and could act via PKA phosphorylation of the SF-1 protein itself.
Collapse
Affiliation(s)
- P de Santa Barbara
- Human Molecular Genetics Group, Institut de Génétique Humaine, CNRS UPR1142, 34396 Montpellier Cedex 5, France
| | | | | | | | | | | |
Collapse
|
45
|
Tsygankova OM, Saavedra A, Rebhun JF, Quilliam LA, Meinkoth JL. Coordinated regulation of Rap1 and thyroid differentiation by cyclic AMP and protein kinase A. Mol Cell Biol 2001; 21:1921-9. [PMID: 11238928 PMCID: PMC86776 DOI: 10.1128/mcb.21.6.1921-1929.2001] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Originally identified as an antagonist of Ras action, Rap1 exhibits many Ras-independent effects, including a role in signaling pathways initiated by cyclic AMP (cAMP). Since cAMP is a critical mediator of the effects of thyrotropin (TSH) on cell proliferation and differentiation, we examined the regulation of Rap1 by TSH in a continuous line of rat thyroid-like cells. Both cAMP and protein kinase A (PKA) contribute to the regulation of Rap1 activity and signaling by TSH. TSH activates Rap1 through a cAMP-mediated and PKA-independent mechanism. TSH phosphorylates Rap1 in a PKA-dependent manner. Interference with PKA activity blocked phosphorylation but not the activation of Rap1. Rather, PKA inhibitors prolonged Rap1 activation, as did expression of a Rap1A mutant lacking a PKA phosphorylation site. These results indicate that PKA elicits negative feedback regulation on cAMP-stimulated Rap1 activity in some cells. The dual regulation of Rap1 by cAMP and PKA extends to downstream effectors. The ability of TSH to stimulate Akt phosphorylation was markedly enhanced by the expression of activated Rap1A and was repressed in cells expressing a putative dominant-negative Rap1A mutant. Although the expression of activated Rap1A was sufficient to stimulate wortmannin-sensitive Akt phosphorylation, TSH further increased Akt phosphorylation in a phosphatidylinositol 3-kinase- and PKA-dependent manner. The ability of TSH to phosphorylate Akt was impaired in cells expressing a Rap1A mutant that could be activated but not phosphorylated. These findings indicate that dual signals, Rap1 activation and phosphorylation, contribute to TSH-stimulated Akt phosphorylation. Rap1 plays an essential role in cAMP-regulated differentiation. TSH effects on thyroid-specific gene expression, but not its effects on proliferation, were markedly enhanced in cells expressing activated Rap1A and repressed in cells expressing a dominant-negative Rap1A mutant. These findings reveal complex regulation of Rap1 by cAMP including PKA-independent activation and PKA-dependent negative feedback regulation. Both signals appear to be required for TSH signaling to Akt.
Collapse
Affiliation(s)
- O M Tsygankova
- Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6084, USA
| | | | | | | | | |
Collapse
|
46
|
Nerve growth factor, but not epidermal growth factor, increases Fra-2 expression and alters Fra-2/JunD binding to AP-1 and CREB binding elements in pheochromocytoma (PC12) cells. J Neurosci 2001. [PMID: 11150315 DOI: 10.1523/jneurosci.21-01-00018.2001] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In pheochromocytoma (PC12) cells nerve growth factor (NGF) and epidermal growth factor (EGF) activate similar receptor tyrosine kinase signaling pathways but evoke strikingly different biological outcomes: NGF induces differentiation and EGF acts as a mitogen. A novel approach was developed for identifying transcription factor activities associated with NGF-activated, but not EGF-activated, signaling, using random oligonucleotide clones from a DNA recognition library to isolate specific DNA binding proteins from PC12 nuclear extracts. A protein complex from NGF-treated, but not EGF-treated, cells was identified that exhibits increased mobility and DNA binding activity in gel mobility shift assays. The binding complex was identified in supershift assays as Fra-2/JunD. The clones used as probes contain either AP-1 or cAMP response element binding (CREB) recognition elements. Time course experiments revealed further differences in NGF and EGF signaling in PC12 cells. NGF elicits a more delayed and sustained ERK phosphorylation than EGF, consistent with previous reports. Both growth factors transiently induce c-fos, but NGF evokes a greater response than EGF. NGF specifically increases Fra-1 and Fra-2 levels at 4 and 24 hr. The latter is represented in Western blots by bands in the 40-46 kDa range. NGF, but not EGF, enhances the upper bands, corresponding to phosphorylated Fra-2. These findings suggest that prolonged alterations in Fra-2 and subsequent increases in Fra-2/JunD binding to AP-1 and CREB response elements common among many gene promoters could serve to trigger broadly an NGF-specific program of gene expression.
Collapse
|
47
|
York RD, Molliver DC, Grewal SS, Stenberg PE, McCleskey EW, Stork PJ. Role of phosphoinositide 3-kinase and endocytosis in nerve growth factor-induced extracellular signal-regulated kinase activation via Ras and Rap1. Mol Cell Biol 2000; 20:8069-83. [PMID: 11027277 PMCID: PMC86417 DOI: 10.1128/mcb.20.21.8069-8083.2000] [Citation(s) in RCA: 191] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neurotrophins promote multiple actions on neuronal cells including cell survival and differentiation. The best-studied neurotrophin, nerve growth factor (NGF), is a major survival factor in sympathetic and sensory neurons and promotes differentiation in a well-studied model system, PC12 cells. To mediate these actions, NGF binds to the TrkA receptor to trigger intracellular signaling cascades. Two kinases whose activities mediate these processes include the mitogen-activated protein (MAP) kinase (or extracellular signal-regulated kinase [ERK]) and phosphoinositide 3-kinase (PI3-K). To examine potential interactions between the ERK and PI3-K pathways, we studied the requirement of PI3-K for NGF activation of the ERK signaling cascade in dorsal root ganglion cells and PC12 cells. We show that PI3-K is required for TrkA internalization and participates in NGF signaling to ERKs via distinct actions on the small G proteins Ras and Rap1. In PC12 cells, NGF activates Ras and Rap1 to elicit the rapid and sustained activation of ERKs respectively. We show here that Rap1 activation requires both TrkA internalization and PI3-K, whereas Ras activation requires neither TrkA internalization nor PI3-K. Both inhibitors of PI3-K and inhibitors of endocytosis prevent GTP loading of Rap1 and block sustained ERK activation by NGF. PI3-K and endocytosis may also regulate ERK signaling at a second site downstream of Ras, since both rapid ERK activation and the Ras-dependent activation of the MAP kinase kinase kinase B-Raf are blocked by inhibition of either PI3-K or endocytosis. The results of this study suggest that PI3-K may be required for the signals initiated by TrkA internalization and demonstrate that specific endocytic events may distinguish ERK signaling via Rap1 and Ras.
Collapse
Affiliation(s)
- R D York
- Vollum Institute, Oregon Health Sciences University, Portland, Oregon 97201, USA
| | | | | | | | | | | |
Collapse
|
48
|
Deavall DG, Raychowdhury R, Dockray GJ, Dimaline R. Control of CCK gene transcription by PACAP in STC-1 cells. Am J Physiol Gastrointest Liver Physiol 2000; 279:G605-12. [PMID: 10960361 DOI: 10.1152/ajpgi.2000.279.3.g605] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The mechanisms by which neuroendocrine stimulants regulate CCK gene transcription are unclear. We examined promoter activation by pituitary adenylate cyclase-activating polypeptide (PACAP), a known CCK secretagogue, in the enteroendocrine cell line STC-1. The promoter region from -70 to -87 bp, relative to the transcriptional start site, contains a composite calcium/cyclic AMP response element (CRE)/activator protein 1 (AP1) site that may bind CRE binding protein (CREB) and AP1. PACAP (with IBMX) stimulated expression of an 87-bp construct 3.35+/-0.36-fold but had no effect on a -70 construct. The effect was blocked by the protein kinase A inhibitor H-89 and by a dominant-negative CREB plasmid. Mutation of the CRE/AP1 site to a canonical CRE site did not affect the response to PACAP, but mutation to a canonical AP1 site prevented it. CREB phosphorylation was increased after PACAP treatment. Electrophoretic mobility shift assay and supershift analysis revealed that CREB and not AP1 bound to the CRE/AP1 site and that PACAP increased the proportion of phosphorylated CREB that was bound. We conclude that PACAP increases CCK gene expression via a cAMP-mediated pathway involving CREB phosphorylation by protein kinase A and activation of a composite CRE/AP1 site.
Collapse
Affiliation(s)
- D G Deavall
- Physiological Laboratory, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | | | | | | |
Collapse
|
49
|
Tolliver BK, Sganga MW, Sharp FR. Suppression of c-fos induction in the nucleus accumbens prevents acquisition but not expression of morphine-conditioned place preference. Eur J Neurosci 2000; 12:3399-406. [PMID: 10998122 DOI: 10.1046/j.1460-9568.2000.00214.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The c-fos immediate-early gene is induced by morphine and other drugs of abuse in the nucleus accumbens (NAc), a mesolimbic region implicated in drug abuse and reward. This study examined the role of c-fos in the acquisition and expression of the conditioned place paradigm (CPP) in the rat by suppressing Fos protein expression with c-fos antisense oligodeoxynucleotides (ODNs). CPP was completely prevented by c-fos antisense ODN infused bilaterally into the NAc prior to each systemic morphine injection, whereas sense and missense NAc injections had no effect on CPP. NAc administration of c-fos antisense ODN after the last systemic morphine conditioning session did not affect the expression of morphine-CPP. These results suggest that c-fos expression in NAc is necessary for the acquisition but not expression of morphine-CPP, and they have important implications for understanding conditioned behaviours and drug craving and addiction.
Collapse
Affiliation(s)
- B K Tolliver
- Department of Neurology, University of Cincinnati, Vontz Center for Molecular Studies Rm 2327, 3125 Eden Ave, University of Cincinnati, Cincinnati, OH 45267-0536, USA
| | | | | |
Collapse
|
50
|
Adachi M, Browne D, Lewis EJ. Paired-like homeodomain proteins Phox2a/Arix and Phox2b/NBPhox have similar genetic organization and independently regulate dopamine beta-hydroxylase gene transcription. DNA Cell Biol 2000; 19:539-54. [PMID: 11034547 DOI: 10.1089/104454900439773] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The homeodomain transcription factors Arix/Phox2a and NBPhox/Phox2b play a role in the specification of the noradrenergic phenotype of central and peripheral neurons. To better understand the functions of these two factors, we have compared the genetic organization, chromosomal location, and transcriptional regulatory properties of Arix and NBPhox. The gene structure is very similar, with each gene containing three exons and two introns, extending a total of approximately 5 kb. Arix and NBPhox are unlinked in human and mouse genomes. NBPhox is located on human Chromosome 4p12 and mouse Chromosome 5, while Arix is located on human Chromosome 11q13 and mouse Chromosome 7. Both proteins bind to three sites in the promoter proximal region of the rat dopamine beta-hydroxylase gene (DBH). In vitro, Arix and NBPhox form DNA-independent multimers and exhibit cooperative binding to the DB1 regulatory element, which contains two homeodomain recognition sites. Both proteins regulate transcription from the rat DBH promoter, and transcription is synergistically increased in the presence of the protein kinase A catalytic subunit (PKA) plus either Arix or NBPhox. The transcription factors exhibit similar concentration-dependent efficacies, and when they are coexpressed, transcription is stimulated to a value approximately equal to that seen with either factor alone. The N-terminal segment of Arix is essential for transcriptional regulatory activity, and this region bears 50% identity with NBPhox, suggesting a similar mechanism of transcriptional activation of the DBH gene. We conclude from this study that Arix and NBPhox exhibit indistinguishable and independent transcriptional regulatory properties on the DBH promoter.
Collapse
Affiliation(s)
- M Adachi
- Department of Biochemistry and Molecular Biology, Oregon Health Sciences University, Portland 97201, USA
| | | | | |
Collapse
|