1
|
YAVAS E, FANSELOW MS. Defensive behaviors and c-fos expression in the midbrain. Integr Zool 2025; 20:394-406. [PMID: 39218997 PMCID: PMC11871047 DOI: 10.1111/1749-4877.12892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Pavlovian fear conditioning serves as a valuable method for investigating species-specific defensive reactions (SSDRs) such as freezing and flight responses. The present study examines the role of white noise under different experimental conditions. Given that white noise has been shown to elicit both conditional (associative) and unconditional (nonassociative) defensive responses, we compared the response to noise following three separate training conditions: shock-only, white noise paired with shock, and context-only. Results showed that baseline freezing level significantly changed across groups: Both the shock-only group and the white noise paired with shock group froze more than the context-only group on the test day. White noise evoked differential freezing between groups on day 2: The shock-only group froze more than the context-only group although both groups were never exposed to white noise during training. Further, an activity burst triggered by white noise was similar for the shock-only and white noise paired with shock groups during testing, although shock-only group was never exposed to white noise stimuli during training. This aligned with c-fos data, indicating similar c-fos activity levels across different periaqueductal gray (PAG) regions for both shock-only and white noise paired with shock groups. However, the driving force behind c-fos activation-whether freezing, activity burst, or a combination of both-remains uncertain, warranting further analysis to explore specific correlations between SSDRs and c-fos activity within the PAG and related brain areas.
Collapse
Affiliation(s)
- Ersin YAVAS
- Department of PsychologyBartın UniversityBartınTurkey
| | - Michael S. FANSELOW
- Staglin Center for Brain and Behavioral Health, Department of Psychology, UCLALos AngelesCaliforniaUSA
- Department of Psychiatry and Biobehavioral SciencesUCLALos AngelesCaliforniaUSA
| |
Collapse
|
2
|
Cherait A, Banks WA, Vaudry D. The Potential of the Nose-to-Brain Delivery of PACAP for the Treatment of Neuronal Disease. Pharmaceutics 2023; 15:2032. [PMID: 37631246 PMCID: PMC10459484 DOI: 10.3390/pharmaceutics15082032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Research on the neuroprotective effect of pituitary adenylate cyclase-activating polypeptide (PACAP) and its use as a therapeutic agent has grown over the past 30 years. Both in vitro and in vivo experiments have shown that PACAP exerts a strong neuroprotective effect in many central and peripheral neuronal diseases. Various delivery routes have been employed from intravenous (IV) injections to intracerebroventricular (ICV) administration, leading either to systemic or topical delivery of the peptide. Over the last decade, a growing interest in the use of intranasal (IN) administration of PACAP and other therapeutic agents has emerged as an alternative delivery route to target the brain. The aim of this review is to summarize the findings on the neuroprotective effect of PACAP and to discuss how the IN administration of PACAP could contribute to target the effects of this pleiotropic peptide.
Collapse
Affiliation(s)
- Asma Cherait
- Univ Rouen Normandie, Inserm U1245, Medical Faculty, Normandie Univ, F-76000 Rouen, France;
- Department of Second Cycle, Higher School of Agronomy Mostaganem, Mostaganem 27000, Algeria
- Laboratory of Cellular Toxicology, Department of Biology, Faculty of Sciences, University of Badji Mokhtar Annaba, B.P. 12, Annaba 23000, Algeria
| | - William A. Banks
- Geriatric Research Educational and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA
| | - David Vaudry
- Univ Rouen Normandie, Inserm U1245, Medical Faculty, Normandie Univ, F-76000 Rouen, France;
- Univ Rouen Normandie, Inserm US51, Regional Cell Imaging Platform of Normandy (PRIMACEN), Sciences and Technologies Faculty, Normandie Univ, F-76000 Rouen, France
| |
Collapse
|
3
|
Fehér M, Márton Z, Szabó Á, Kocsa J, Kormos V, Hunyady Á, Kovács LÁ, Ujvári B, Berta G, Farkas J, Füredi N, Gaszner T, Pytel B, Reglődi D, Gaszner B. Downregulation of PACAP and the PAC1 Receptor in the Basal Ganglia, Substantia Nigra and Centrally Projecting Edinger-Westphal Nucleus in the Rotenone model of Parkinson's Disease. Int J Mol Sci 2023; 24:11843. [PMID: 37511603 PMCID: PMC10380602 DOI: 10.3390/ijms241411843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/19/2023] [Accepted: 07/22/2023] [Indexed: 07/30/2023] Open
Abstract
Numerous in vitro and in vivo models of Parkinson's disease (PD) demonstrate that pituitary adenylate cyclase-activating polypeptide (PACAP) conveys its strong neuroprotective actions mainly via its specific PAC1 receptor (PAC1R) in models of PD. We recently described the decrease in PAC1R protein content in the basal ganglia of macaques in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of PD that was partially reversed by levodopa therapy. In this work, we tested whether these observations occur also in the rotenone model of PD in the rat. The rotarod test revealed motor skill deterioration upon rotenone administration, which was reversed by benserazide/levodopa (B/L) treatment. The sucrose preference test suggested increased depression level while the open field test showed increased anxiety in rats rendered parkinsonian, regardless of the received B/L therapy. Reduced dopaminergic cell count in the substantia nigra pars compacta (SNpc) diminished the dopaminergic fiber density in the caudate-putamen (CPu) and decreased the peptidergic cell count in the centrally projecting Edinger-Westphal nucleus (EWcp), supporting the efficacy of rotenone treatment. RNAscope in situ hybridization revealed decreased PACAP mRNA (Adcyap1) and PAC1R mRNA (Adcyap1r1) expression in the CPu, globus pallidus, dopaminergic SNpc and peptidergic EWcp of rotenone-treated rats, but no remarkable downregulation occurred in the insular cortex. In the entopeduncular nucleus, only the Adcyap1r1 mRNA was downregulated in parkinsonian animals. B/L therapy attenuated the downregulation of Adcyap1 in the CPu only. Our current results further support the evolutionarily conserved role of the PACAP/PAC1R system in neuroprotection and its recruitment in the development/progression of neurodegenerative states such as PD.
Collapse
Affiliation(s)
- Máté Fehér
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Department of Neurosurgery, Kaposi Mór Teaching Hospital, Tallián Gy. u. 20-32, H-7400 Kaposvár, Hungary
| | - Zsombor Márton
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Ákos Szabó
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - János Kocsa
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Viktória Kormos
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Ágnes Hunyady
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - László Ákos Kovács
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Balázs Ujvári
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Gergely Berta
- Department of Medical Biology and Central Electron Microscopic Laboratory, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - József Farkas
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Nóra Füredi
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Tamás Gaszner
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Bence Pytel
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Dóra Reglődi
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- ELKH-PTE PACAP Research Group, Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Balázs Gaszner
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| |
Collapse
|
4
|
Solés-Tarrés I, Cabezas-Llobet N, Lefranc B, Leprince J, Alberch J, Vaudry D, Xifró X. Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) Protects Striatal Cells and Improves Motor Function in Huntington’s Disease Models: Role of PAC1 Receptor. Front Pharmacol 2022; 12:797541. [PMID: 35153755 PMCID: PMC8832515 DOI: 10.3389/fphar.2021.797541] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/31/2021] [Indexed: 12/21/2022] Open
Abstract
Huntington’s disease (HD) is a hereditary neurodegenerative disorder caused by the expression of mutant huntingtin (mHtt). One of the main features of HD is the degeneration of the striatum that leads to motor discoordination. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide that acts through three receptors named PAC1R, VPAC1R, and VPAC2R. In the present study, we first investigated the effect of PACAP on STHdhQ7/Q7 and STHdhQ111/Q111 cells that express wild-type Htt with 7 and mHtt with 111 glutamines, respectively. Then we explored the capacity of PACAP to rescue motor symptoms in the R6/1, a murine model of HD. We found that PACAP treatment (10–7 M) for 24 h protects STHdhQ111/Q111 cells from mHtt-induced apoptosis. This effect is associated with an increase in PAC1R transcription, phosphorylation of ERK and Akt, and an increase of intracellular c-fos, egr1, CBP, and BDNF protein content. Moreover, the use of pharmacological inhibitors revealed that activation of ERK and Akt mediates these antiapoptotic and neurotrophic effects of PACAP. To find out PAC1R implication, we treated STHdh cells with vasoactive intestinal peptide (VIP), which exhibits equal affinity for VPAC1R and VPAC2R, but lower affinity for PAC1R, in contrast to PACAP which has same affinity for the three receptors. VIP reduced cleaved caspase-3 protein level, without promoting the expression of c-fos, egr1, CBP, and the neurotrophin BDNF. We next measured the protein level of PACAP receptors in the striatum and cortex of R6/1 mice. We observed a specific reduction of PAC1R at the onset of motor symptoms. Importantly, the intranasal administration of PACAP to R6/1 animals restored the motor function and increased the striatal levels of PAC1R, CBP, and BDNF. In conclusion, PACAP exerts antiapoptotic and neurotrophic effects in striatal neurons mainly through PAC1R. This effect in HD striatum allows the recovery of motor function and point out PAC1R as a therapeutic target for treatment of HD.
Collapse
Affiliation(s)
- Irene Solés-Tarrés
- New Therapeutic Targets Group, Department of Medical Science, Faculty of Medicine, University of Girona, Girona, Spain
| | - Núria Cabezas-Llobet
- New Therapeutic Targets Group, Department of Medical Science, Faculty of Medicine, University of Girona, Girona, Spain
| | - Benjamin Lefranc
- Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, UNIROUEN, Inserm, Normandie University, Rouen, France
- Regional Cell Imaging Platform of Normandy (PRIMACEN), UNIROUEN, Normandie University, Rouen, France
| | - Jérôme Leprince
- Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, UNIROUEN, Inserm, Normandie University, Rouen, France
- Regional Cell Imaging Platform of Normandy (PRIMACEN), UNIROUEN, Normandie University, Rouen, France
| | - Jordi Alberch
- Departament de Biomedicina, Institut de Neurociències, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
- Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - David Vaudry
- Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, UNIROUEN, Inserm, Normandie University, Rouen, France
- Regional Cell Imaging Platform of Normandy (PRIMACEN), UNIROUEN, Normandie University, Rouen, France
| | - Xavier Xifró
- New Therapeutic Targets Group, Department of Medical Science, Faculty of Medicine, University of Girona, Girona, Spain
- *Correspondence: Xavier Xifró,
| |
Collapse
|
5
|
Hayata-Takano A, Shintani Y, Moriguchi K, Encho N, Kitagawa K, Nakazawa T, Hashimoto H. PACAP-PAC1 Signaling Regulates Serotonin 2A Receptor Internalization. Front Endocrinol (Lausanne) 2021; 12:732456. [PMID: 34759890 PMCID: PMC8574227 DOI: 10.3389/fendo.2021.732456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/27/2021] [Indexed: 11/26/2022] Open
Abstract
Mice lacking pituitary adenylate cyclase-activating polypeptide (PACAP) display psychomotor abnormalities, most of which are ameliorated by atypical antipsychotics with serotonin (5-HT) 2A receptor (5-HT2A) antagonism. Heterozygous Pacap mutant mice show a significantly higher hallucinogenic response than wild-type mice to a 5-HT2A agonist. Endogenous PACAP may, therefore, affect 5-HT2A signaling; however, the underlying neurobiological mechanism for this remains unclear. Here, we examined whether PACAP modulates 5-HT2A signaling by addressing cellular protein localization. PACAP induced an increase in internalization of 5-HT2A but not 5-HT1A, 5-HT2C, dopamine D2 receptors or metabotropic glutamate receptor 2 in HEK293T cells. This PACAP action was inhibited by protein kinase C inhibitors, β-arrestin2 silencing, the PACAP receptor PAC1 antagonist PACAP6-38, and PAC1 silencing. In addition, the levels of endogenous 5-HT2A were decreased on the cell surface of primary cultured cortical neurons after PACAP stimulation and were increased in frontal cortex cell membranes of Pacap-/- mice. Finally, intracerebroventricular PACAP administration suppressed 5-HT2A agonist-induced head twitch responses in mice. These results suggest that PACAP-PAC1 signaling increases 5-HT2A internalization resulting in attenuation of 5-HT2A-mediated signaling, although further study is necessary to determine the relationship between behavioral abnormalities in Pacap-/- mice and PACAP-induced 5-HT2A internalization.
Collapse
Affiliation(s)
- Atsuko Hayata-Takano
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
- Molecular Research Center for Children’s Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Japan
- *Correspondence: Hitoshi Hashimoto, ; Atsuko Hayata-Takano,
| | - Yusuke Shintani
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Keita Moriguchi
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Naoki Encho
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Kohei Kitagawa
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Takanobu Nakazawa
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
- Department of Bioscience, Tokyo University of Agriculture, Setagaya-ku, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
- Molecular Research Center for Children’s Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Japan
- Division of Bioscience, Institute for Datability Science, Osaka University, Suita, Japan
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Department of Molecular Pharmaceutical Science, Graduate School of Medicine, Osaka University, Suita, Japan
- *Correspondence: Hitoshi Hashimoto, ; Atsuko Hayata-Takano,
| |
Collapse
|
6
|
Winters SJ, Moore JP. PACAP: A regulator of mammalian reproductive function. Mol Cell Endocrinol 2020; 518:110912. [PMID: 32561449 PMCID: PMC7606562 DOI: 10.1016/j.mce.2020.110912] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/14/2020] [Accepted: 06/06/2020] [Indexed: 12/19/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is an ancestral molecule that was isolated from sheep hypothalamic extracts based on its action to stimulate cAMP production by pituitary cell cultures. PACAP is one of a number of ligands that coordinate with GnRH to control reproduction. While initially viewed as a hypothalamic releasing factor, PACAP and its receptors are widely distributed, and there is growing evidence that PACAP functions as a paracrine/autocrine regulator in the CNS, pituitary, gonads and placenta, among other tissues. This review will summarize current knowledge concerning the expression and function of PACAP in the hypothalamic-pituitary-gonadal axis with special emphasis on its role in pituitary function in the fetus and newborn.
Collapse
Affiliation(s)
- Stephen J Winters
- Division of Endocrinology, Metabolism and Diabetes, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| | - Joseph P Moore
- Division of Endocrinology, Metabolism and Diabetes, University of Louisville School of Medicine, Louisville, KY, 40202, USA; Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| |
Collapse
|
7
|
Cabezas-Llobet N, Vidal-Sancho L, Masana M, Fournier A, Alberch J, Vaudry D, Xifró X. Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) Enhances Hippocampal Synaptic Plasticity and Improves Memory Performance in Huntington's Disease. Mol Neurobiol 2018. [PMID: 29526016 DOI: 10.1007/s12035-018-0972-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Deficits in hippocampal synaptic plasticity result in cognitive impairment in Huntington's disease (HD). Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide that exerts neuroprotective actions, mainly through the PAC1 receptor. However, the role of PACAP in cognition is poorly understood, and no data exists in the context of Huntington's disease (HD). Here, we investigated the ability of PACAP receptor stimulation to enhance memory development in HD. First, we observed a hippocampal decline of all three PACAP receptor expressions, i.e., PAC1, VPAC1, and VPAC2, in two different HD mouse models, R6/1 and HdhQ7/Q111, from the onset of cognitive dysfunction. In hippocampal post-mortem human samples, we found a specific decrease of PAC1, without changes in VPAC1 and VPAC2 receptors. To determine whether activation of PACAP receptors could contribute to improve memory performance, we conducted daily intranasal administration of PACAP38 to R6/1 mice at the onset of cognitive impairment for seven days. We found that PACAP treatment rescued PAC1 level in R6/1 mice, promoted expression of the hippocampal brain-derived neurotrophic factor, and reduced the formation of mutant huntingtin aggregates. Furthermore, PACAP administration counteracted R6/1 mice memory deficits as analyzed by the novel object recognition test and the T-maze spontaneous alternation task. Importantly, the effect of PACAP on cognitive performance was associated with an increase of VGlut-1 and PSD95 immunolabeling in hippocampus of R6/1 mice. Taken together, these results suggest that PACAP, acting through stimulation of PAC1 receptor, may have a therapeutic potential to counteract cognitive deficits induced in HD.
Collapse
Affiliation(s)
- N Cabezas-Llobet
- New Therapeutic Targets Group, Department of Medical Science, School of Medicine, Universitat de Girona, Girona, Spain
| | - L Vidal-Sancho
- New Therapeutic Targets Group, Department of Medical Science, School of Medicine, Universitat de Girona, Girona, Spain
- Departament de Biomedicina, Institut de Neurociències, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - M Masana
- Departament de Biomedicina, Institut de Neurociències, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - A Fournier
- INRS-Institut Armand-Frappier, 531 boul. des Prairies, Laval, QC, H7V 1B7, Canada
- International Associate Laboratory Samuel de Champlain, 531 boul. des Prairies, Laval, QC, H7 1B7, Canada
| | - J Alberch
- Departament de Biomedicina, Institut de Neurociències, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - D Vaudry
- Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, Normandie Univ, UNIROUEN, Inserm, 76000, Rouen, France
| | - X Xifró
- New Therapeutic Targets Group, Department of Medical Science, School of Medicine, Universitat de Girona, Girona, Spain.
- Departament de Biomedicina, Institut de Neurociències, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
- Departament de Ciències Mèdiques, Facultat de Medicina, Universitat de Girona, C/ Emili Grahit 77, E-17003, Girona, Spain.
| |
Collapse
|
8
|
Georg B, Falktoft B, Fahrenkrug J. PKA, novel PKC isoforms, and ERK is mediating PACAP auto-regulation via PAC 1R in human neuroblastoma NB-1 cells. Neuropeptides 2016; 60:83-89. [PMID: 27745706 DOI: 10.1016/j.npep.2016.09.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/30/2016] [Accepted: 09/25/2016] [Indexed: 11/22/2022]
Abstract
The neuropeptide PACAP is expressed throughout the central and peripheral nervous system where it modulates diverse physiological functions including neuropeptide gene expression. We here report that in human neuroblastoma NB-1 cells PACAP transiently induces its own expression. Maximal PACAP mRNA expression was found after stimulation with PACAP for 3h. PACAP auto-regulation was found to be mediated by activation of PACAP specific PAC1Rs as PACAP had >100-fold higher efficacy than VIP, and the PAC1R selective agonist Maxadilan potently induced PACAP gene expression. Experiments with pharmacological kinase inhibitors revealed that both PKA and novel but not conventional PKC isozymes were involved in the PACAP auto-regulation. Inhibition of MAPK/ERK kinase (MEK) also impeded the induction, and we found that PKA, novel PKC and ERK acted in parallel and were thus not part of the same pathways. The expression of the transcription factor EGR1 previously ascribed as target of PACAP signalling was found to be transiently induced by PACAP and pharmacological inhibition of either PKC or MEK1/2 abolished PACAP mediated EGR1 induction. In contrast, inhibition of PKA mediated increased PACAP mediated EGR1 induction. Experiments using siRNA against EGR1 to lower the expression did however not affect the PACAP auto-regulation indicating that this immediate early gene product is not part of PACAP auto-regulation in NB-1 cells. We here reveal that in NB-1 neuroblastoma cells, PACAP induces its own expression by activation of PAC1R, and that the signalling is different from the PAC1R signalling mediating induction of VIP in the same cells. PACAP auto-regulation depends on parallel activation of PKA, novel PKC isoforms, and ERK, while EGR1 does not seem to be part of the PACAP auto-regulation.
Collapse
Affiliation(s)
- Birgitte Georg
- Department of Clinical Biochemistry, Faculty of Health and Medical Sciences, Bispebjerg Hospital, University of Copenhagen, Denmark.
| | - Birgitte Falktoft
- Department of Clinical Biochemistry, Faculty of Health and Medical Sciences, Bispebjerg Hospital, University of Copenhagen, Denmark
| | - Jan Fahrenkrug
- Department of Clinical Biochemistry, Faculty of Health and Medical Sciences, Bispebjerg Hospital, University of Copenhagen, Denmark
| |
Collapse
|
9
|
Jóźwiak-Bębenista M, Kowalczyk E. Neuroleptic Drugs and PACAP Differentially Affect the mRNA Expression of Genes Encoding PAC1/VPAC Type Receptors. Neurochem Res 2016; 42:943-952. [PMID: 27900577 PMCID: PMC5375968 DOI: 10.1007/s11064-016-2127-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/17/2016] [Accepted: 11/25/2016] [Indexed: 01/19/2023]
Abstract
Several lines of evidence suggest that pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide playing an important role as a neuromodulator. It has been indicated that PACAP is associated with mental diseases, and that regulation of the PACAPergic signals could be a potential target for the treatment of such psychiatric states as schizophrenia. Recent studies have suggested that action of neuroleptic drugs is mediated not only by dopaminergic and serotonergic neurotransmission, but also via neuropeptides which may act both as neurotransmitters and as neuromodulators. The present study examines whether currently-used neuroleptics influence the action of PACAP receptors, whose expression is altered in a schizophrenic patient. Real-time polymerase chain reaction (PCR) was used to examine the effects of haloperidol, olanzapine and amisulpride on the expression of genes coding PAC1/VPAC type receptors in the T98G glioblastoma cell line, as an example of an in vitro model of glial cells. PAC1 mRNA expression fell after 24-h incubation with haloperidol or olanzapine; however the effect was not maintained after 72 h, and haloperidol even up-regulated PAC1 mRNA expression in a dose-dependent manner. All the examined drugs decreased VPAC2 mRNA expression, especially after 72-h incubation. Haloperidol (typical neuroleptic) was distinctly more potent than atypical neuroleptic drugs (olanzapine and amisulpride). In addition, PACAP increased PAC1 and VPAC2 mRNA expression. In conclusion, our findings suggest PACAP receptors may be involved in the mechanism of typical and atypical neuroleptic drugs.
Collapse
Affiliation(s)
- Marta Jóźwiak-Bębenista
- Department of Pharmacology and Toxicology, The Interfaculty Chair of Basic and Clinical Pharmacology and Toxicology, Medical University of Lodz, Zeligowskiego 7/9, 90-752, Lodz, Poland.
| | - Edward Kowalczyk
- Department of Pharmacology and Toxicology, The Interfaculty Chair of Basic and Clinical Pharmacology and Toxicology, Medical University of Lodz, Zeligowskiego 7/9, 90-752, Lodz, Poland
| |
Collapse
|
10
|
Fukuchi M, Kuwana Y, Tabuchi A, Tsuda M. Balance between cAMP and Ca(2+) signals regulates expression levels of pituitary adenylate cyclase-activating polypeptide gene in neurons. Genes Cells 2016; 21:921-9. [PMID: 27383213 DOI: 10.1111/gtc.12393] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/09/2016] [Indexed: 11/30/2022]
Abstract
Mice lacking the gene encoding pituitary adenylate cyclase-activating polypeptide (PACAP) or its specific receptor, PAC1, show abnormal behaviors related to schizophrenia. However, the regulation of PACAP expression in neurons remains unclear. Here, we report that Pacap mRNA levels are regulated transcriptionally and post-transcriptionally by cAMP and Ca(2+) signals in cultured rat cortical cells. Pacap mRNA levels decreased proportionately with the intensity of cAMP signaling, and this decrease was accelerated by N-methyl-D-aspartate (NMDA) receptor blockade, suggesting that cAMP signaling enhances the degradation of Pacap mRNA, whereas NMDA receptor-mediated signals inhibit its degradation. However, depolarization (which produced a robust increase in Ca(2+) signals) together with cAMP signaling resulted in a synergistic induction of Pacap mRNA through calcineurin and its substrate, cAMP-response element-binding protein (CREB)-regulated transcription coactivator 1. These results strongly support the concept that while cAMP signaling can accelerate the degradation of Pacap mRNA, it can also synergistically enhance Ca(2+) signaling-induced transcriptional activation of Pacap. Taken together, our findings suggest that a balance between Ca(2+) and cAMP signals regulates PACAP levels in neurons and that a perturbation of this balance may result in psychiatric disorders, such as schizophrenia.
Collapse
Affiliation(s)
- Mamoru Fukuchi
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Yuki Kuwana
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Akiko Tabuchi
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Masaaki Tsuda
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| |
Collapse
|
11
|
PACAP enhances axon outgrowth in cultured hippocampal neurons to a comparable extent as BDNF. PLoS One 2015; 10:e0120526. [PMID: 25807538 PMCID: PMC4373823 DOI: 10.1371/journal.pone.0120526] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Accepted: 01/26/2015] [Indexed: 11/19/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) exerts neurotrophic activities including modulation of synaptic plasticity and memory, hippocampal neurogenesis, and neuroprotection, most of which are shared with brain-derived neurotrophic factor (BDNF). Therefore, the aim of this study was to compare morphological effects of PACAP and BDNF on primary cultured hippocampal neurons. At days in vitro (DIV) 3, PACAP increased neurite length and number to similar levels by BDNF, but vasoactive intestinal polypeptide showed much lower effects. In addition, PACAP increased axon, but not dendrite, length, and soma size at DIV 3 similarly to BDNF. The PACAP antagonist PACAP6–38 completely blocked the PACAP-induced increase in axon, but not dendrite, length. Interestingly, the BDNF-induced increase in axon length was also inhibited by PACAP6–38, suggesting a mechanism involving PACAP signaling. K252a, a TrkB receptor inhibitor, inhibited axon outgrowth induced by PACAP and BDNF without affecting dendrite length. These results indicate that in primary cultured hippocampal neurons, PACAP shows morphological actions via its cognate receptor PAC1, stimulating neurite length and number, and soma size to a comparable extent as BDNF, and that the increase in total neurite length is ascribed to axon outgrowth.
Collapse
|
12
|
Tsumuraya T, Ohtaki H, Song D, Sato A, Watanabe J, Hiraizumi Y, Nakamachi T, Xu Z, Dohi K, Hashimoto H, Atsumi T, Shioda S. Human mesenchymal stem/stromal cells suppress spinal inflammation in mice with contribution of pituitary adenylate cyclase-activating polypeptide (PACAP). J Neuroinflammation 2015; 12:35. [PMID: 25889720 PMCID: PMC4346126 DOI: 10.1186/s12974-015-0252-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/21/2015] [Indexed: 01/01/2023] Open
Abstract
Background Adult human mesenchymal stem/stromal cells (hMSCs) from bone marrow have been reported to exhibit beneficial effects on spinal cord injury (SCI). A neuropeptide, pituitary adenylate cyclase-activating polypeptide (PACAP) is known to decrease neuronal cell death and inflammatory response after ischemia, SCI, and other neuronal disorders. Recently, we found that expression of the gene for mouse PACAP (Adcyap1) was greater in animals receiving hMSCs with neural injury such as ischemia. However, the association of PACAP with hMSCs to protect nerve cells against neural injuries is still unclear. Methods Wild-type and PACAP-gene-deficient (Adcyap1+/−) mice were subjected to spinal cord transection, and hMSCs (5 × 105 cells) were injected into the intervertebral spinal cord on day 1 post-operation (p.o.). Locomotor activity, injury volume, retention of hMSCs, mouse and human cytokine genes (which contribute to macrophage (MΦ) and microglial activation), and Adcyap1 were evaluated. Results hMSCs injected into wild-type mice improved locomotor activity and injury volume compared with vehicle-treated mice. In contrast, non-viable hMSCs injected into wild-type mice, and viable hMSCs injected into Adcyap1+/− mice, did not. Wild-type mice injected with hMSCs exhibited increased Adcyap1 expression, and observed PACAP immunoreaction in neuron-like cells. Gene expression levels for IL-1, tumor necrosis factor α (TNFα), interleukin-10 (IL-10), and transforming growth factor β (TGFβ) decreased, while that for interleukin-4 (IL-4) increased, in hMSC-injected wild-type mice. In contrast, IL-1, TGFβ, and IL-4 gene expression levels were all abolished in hMSC-injected Adcyap1+/− mice on day 7 post-operation. Moreover, the mice-implanted hMSCs increased an alternative activating macrophage/microglial marker, arginase activity. The human gene profile indicated that hMSCs upregulated the gene of IL-4 and growth factors which were reported to enhance Adcyap1 expression. Finally, we demonstrated that hMSCs express human ADCYAP1 and its receptor gene after the inflammation-related interferon-γ (IFNγ) in vitro. Conclusions These results suggest that hMSCs attenuate the deleterious effects of SCI by reducing associated inflammatory responses and enhancing IL-4 production. This effect could be mediated in part by cell-cell cross-talk involving the neuropeptide PACAP. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0252-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tomomi Tsumuraya
- Department of Anatomy, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan. .,Department of Orthopedic Surgery, Showa University Fujigaoka Hospital, 1-30 Fujigaoka, Aoba-ku, Yokohama, Kanagawa, 227-8501, Japan.
| | - Hirokazu Ohtaki
- Department of Anatomy, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
| | - Dandan Song
- Department of Anatomy, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
| | - Atsushi Sato
- Department of Anatomy, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan. .,Department of Orthopedic Surgery, Showa University Fujigaoka Hospital, 1-30 Fujigaoka, Aoba-ku, Yokohama, Kanagawa, 227-8501, Japan.
| | - Jun Watanabe
- Department of Anatomy, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
| | - Yutaka Hiraizumi
- Department of Orthopedic Surgery, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
| | - Tomoya Nakamachi
- Department of Anatomy, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan. .,Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, 3190 Gofuku, Toyama, Toyama, 930-8555, Japan.
| | - Zhifang Xu
- Department of Anatomy, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
| | - Kenji Dohi
- Department of Anatomy, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan. .,Department of Emergency Medicine, Tokyo Jikei University School of Medicine, 3-25-8 Nishishinnbashi, Minato-ku, Tokyo, 105-8461, Japan.
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Takashi Atsumi
- Department of Orthopedic Surgery, Showa University Fujigaoka Hospital, 1-30 Fujigaoka, Aoba-ku, Yokohama, Kanagawa, 227-8501, Japan.
| | - Seiji Shioda
- Department of Anatomy, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
| |
Collapse
|
13
|
Seaborn T, Ravni A, Au R, Chow BKC, Fournier A, Wurtz O, Vaudry H, Eiden LE, Vaudry D. Induction of serpinb1a by PACAP or NGF is required for PC12 cells survival after serum withdrawal. J Neurochem 2014; 131:21-32. [PMID: 24899316 DOI: 10.1111/jnc.12780] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 04/30/2014] [Accepted: 05/13/2014] [Indexed: 01/11/2023]
Abstract
PC12 cells are used to study the signaling mechanisms underlying the neurotrophic and neuroprotective activities of pituitary adenylate cyclase-activating polypeptide (PACAP) and nerve growth factor (NGF). Previous microarray experiments indicated that serpinb1a was the most induced gene after 6 h of treatment with PACAP or NGF. This study confirmed that serpinb1a is strongly activated by PACAP and NGF in a time-dependent manner with a maximum induction (~ 50-fold over control) observed after 6 h of treatment. Co-incubation with PACAP and NGF resulted in a synergistic up-regulation of serpinb1a expression (200-fold over control), suggesting that PACAP and NGF act through complementary mechanisms. Consistently, PACAP-induced serpinb1a expression was not blocked by TrkA receptor inhibition. Nevertheless, the stimulation of serpinb1a expression by PACAP and NGF was significantly reduced in the presence of extracellular signal-regulated kinase, calcineurin, protein kinase A, p38, and PI3K inhibitors, indicating that the two trophic factors share some common pathways in the regulation of serpinb1a. Finally, functional investigations conducted with siRNA revealed that serpinb1a is not involved in the effects of PACAP and NGF on PC12 cell neuritogenesis, proliferation or body cell volume but mediates their ability to block caspases 3/7 activity and to promote PC12 cell survival.
Collapse
Affiliation(s)
- Tommy Seaborn
- Neurotrophic Factor and Neuronal Differentiation Team, Inserm U982, DC2N, Mont-Saint-Aignan, France; International Associated Laboratory Samuel de Champlain, Mont-Saint-Aignan, France; Department of Pediatrics, Hôpital St-François d'Assise, Centre de Recherche du Centre Hospitalier Universitaire de Québec (CRCHUQ), Laval University, Québec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Winters SJ, Ghooray DT, Yang RQ, Holmes JB, O'Brien AR, Morgan J, Moore JP. Dopamine-2 receptor activation suppresses PACAP expression in gonadotrophs. Endocrinology 2014; 155:2647-57. [PMID: 24823390 PMCID: PMC4060190 DOI: 10.1210/en.2013-2147] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is expressed at a high level in the fetal pituitary and decreases profoundly between embryonic day 19 and postnatal day 1 (PN1), with a further decrease from PN1 to PN4. In this series of experiments, we investigated the hypothesis that dopamine 2 receptor (Drd2) activation interrupts a cAMP-dependent feed-forward loop that maintains PACAP expression at a high level in the fetal pituitary. Using single-cell RT-PCR of pituitary cell cultures from newborn rats, Drd2 mRNA was identified in gonadotrophs that were also positive for PACAP mRNA. PACAP expression in pituitary cultures from embryonic day 19 rats was suppressed by the PACAP6-38 antagonist and by the Drd2 agonist bromocriptine. Increasing concentrations of bromocriptine inhibited cAMP production as well as cAMP signaling based on cAMP response element-luciferase activity, decreased PACAP promoter activity, and decreased PACAP mRNA levels in αT3-1 gonadotroph cells. Furthermore, blockade of dopamine receptors by injecting haloperidol into newborn rat pups partially reversed the developmental decline in pituitary PACAP mRNA that occurs between PN1 and PN4. These results provide evidence that dopamine receptor signaling regulates PACAP expression under physiological conditions and lend support to the hypothesis that a rise in hypothalamic dopamine at birth abrogates cAMP signaling in fetal gonadotrophs to interrupt a feed-forward mechanism that maintains PACAP expression at a high level in the fetal pituitary. We propose that this perinatal decline in pituitary PACAP reduces pituitary follistatin which permits GnRH receptors and FSH-β to increase to facilitate activation of the neonatal gonad.
Collapse
Affiliation(s)
- Stephen J Winters
- Division of Endocrinology, Metabolism & Diabetes (S.J.W., D.T.G., J.B.H., A.R.W.O., J.M., J.P.M.), and Department of Anatomy and Neurobiology (R.Q.Y., J.P.M.), University of Louisville, Louisville, Kentucky 40202
| | | | | | | | | | | | | |
Collapse
|
15
|
Saito TH, Uda S, Tsuchiya T, Ozaki YI, Kuroda S. Temporal decoding of MAP kinase and CREB phosphorylation by selective immediate early gene expression. PLoS One 2013; 8:e57037. [PMID: 23469182 PMCID: PMC3587639 DOI: 10.1371/journal.pone.0057037] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 01/16/2013] [Indexed: 12/23/2022] Open
Abstract
A wide range of growth factors encode information into specific temporal patterns of MAP kinase (MAPK) and CREB phosphorylation, which are further decoded by expression of immediate early gene products (IEGs) to exert biological functions. However, the IEG decoding system remain unknown. We built a data-driven based on time courses of MAPK and CREB phosphorylation and IEG expression in response to various growth factors to identify how signal is processed. We found that IEG expression uses common decoding systems regardless of growth factors and expression of each IEG differs in upstream dependency, switch-like response, and linear temporal filters. Pulsatile ERK phosphorylation was selectively decoded by expression of EGR1 rather than c-FOS. Conjunctive NGF and PACAP stimulation was selectively decoded by synergistic JUNB expression through switch-like response to c-FOS. Thus, specific temporal patterns and combinations of MAPKs and CREB phosphorylation can be decoded by selective IEG expression via distinct temporal filters and switch-like responses. The data-driven modeling is versatile for analysis of signal processing and does not require detailed prior knowledge of pathways.
Collapse
Affiliation(s)
- Takeshi H. Saito
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shinsuke Uda
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takaho Tsuchiya
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yu-ichi Ozaki
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shinya Kuroda
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Bunkyo-ku, Tokyo, Japan
- CREST, Japan Science and Technology Corporation, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
16
|
Miura A, Odahara N, Tominaga A, Inoue K, Kambe Y, Kurihara T, Miyata A. Regulatory mechanism of PAC1 gene expression via Sp1 by nerve growth factor in PC12 cells. FEBS Lett 2012; 586:1731-5. [PMID: 22609358 DOI: 10.1016/j.febslet.2012.05.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 04/02/2012] [Accepted: 05/03/2012] [Indexed: 11/26/2022]
Abstract
In addition to VPAC1 and VPAC2, PAC1 is involved in the pleiotropic action of pituitary adenylate cyclase activating polypeptide (PACAP) in the CNS. A luciferase reporter assay for the human PAC1 gene (-2160/+268) revealed that NGF treatment significantly augments the promoter activity of the PAC1 gene. Moreover, the Sp1 site at -282/-273 was shown to be essential for the NGF-augmented promoter activity of the PAC1 gene. Treatment with U0126, an MEK inhibitor, or Mithramycin A, an Sp1 inhibitor, significantly attenuated promoter activity. These results indicate that activation of Sp1 by the Ras/MAPK pathway might participate in neuron specific expression of the PAC1 gene.
Collapse
Affiliation(s)
- Ayako Miura
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, University of Kagoshima, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | | | | | | | | | | | | |
Collapse
|
17
|
Raoult E, Roussel BD, Bénard M, Lefebvre T, Ravni A, Ali C, Vivien D, Komuro H, Fournier A, Vaudry H, Vaudry D, Galas L. Pituitary adenylate cyclase-activating polypeptide (PACAP) stimulates the expression and the release of tissue plasminogen activator (tPA) in neuronal cells: involvement of tPA in the neuroprotective effect of PACAP. J Neurochem 2011; 119:920-31. [DOI: 10.1111/j.1471-4159.2011.07486.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
18
|
Holighaus Y, Mustafa T, Eiden LE. PAC1hop, null and hip receptors mediate differential signaling through cyclic AMP and calcium leading to splice variant-specific gene induction in neural cells. Peptides 2011; 32:1647-55. [PMID: 21693142 PMCID: PMC3163081 DOI: 10.1016/j.peptides.2011.06.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 06/05/2011] [Accepted: 06/06/2011] [Indexed: 11/19/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP)-mediated activation of its G protein-coupled receptor PAC1 results in activation of the two G proteins Gs and Gq to alter second messenger generation and gene transcription in the nervous system, important for homeostatic responses to stress and injury. Heterologous expression of the three major splice variants of the rat PAC1 receptor, PAC1hop, null and hip, in neural NG108-15 cells conferred PACAP-mediated intracellular cAMP generation, while elevation of [Ca(2+)](i) occurred only in PAC1hop-, and to a lesser extent in PAC1null-expressing cells. Induction of vasoactive intestinal polypeptide (VIP) and stanniocalcin 1 (STC1), two genes potentially involved in PACAP's homeostatic responses, was examined as a function of the expressed PAC1 variant. VIP induction was greatest in PAC1hop-expressing cells, suggesting that a maximal transcriptional response requires combinatorial signaling through both cAMP and Ca(2+). STC1 induction was similar for all three receptor splice variants and was mimicked by the adenylate cyclase activator forskolin, indicating that cAMP elevation is sufficient to induce STC1. The degree of activation of two different second messenger pathways appears to determine the transcriptional response, suggesting that cellular responses to stressors are fine-tuned through differential receptor isoform expression. Signaling to the VIP gene proceeded through cAMP and protein kinase A (PKA) in these cells, independently of the MAP kinase ERK1/2. STC1 gene induction by PACAP was dependent on cAMP and ERK1/2, independently of PKA. Differential gene induction via different cAMP dependent signaling pathways potentially provides further targets for the design of treatments for stress-associated disorders.
Collapse
Affiliation(s)
- Yvonne Holighaus
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
19
|
Rat D, Schmitt U, Tippmann F, Dewachter I, Theunis C, Wieczerzak E, Postina R, van Leuven F, Fahrenholz F, Kojro E. Neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) slows down Alzheimer's disease-like pathology in amyloid precursor protein-transgenic mice. FASEB J 2011; 25:3208-18. [PMID: 21593432 PMCID: PMC3157688 DOI: 10.1096/fj.10-180133] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) has neuroprotective and neurotrophic properties and is a potent α-secretase activator. As PACAP peptides and their specific receptor PAC1 are localized in central nervous system areas affected by Alzheimer's disease (AD), this study aims to examine the role of the natural peptide PACAP as a valuable approach in AD therapy. We investigated the effect of PACAP in the brain of an AD transgenic mouse model. The long-term intranasal daily PACAP application stimulated the nonamyloidogenic processing of amyloid precursor protein (APP) and increased expression of the brain-derived neurotrophic factor and of the antiapoptotic Bcl-2 protein. In addition, it caused a strong reduction of the amyloid β-peptide (Aβ) transporter receptor for advanced glycation end products (RAGE) mRNA level. PACAP, by activation of the somatostatin-neprilysin cascade, also enhanced expression of the Aβ-degrading enzyme neprilysin in the mouse brain. Furthermore, daily PAC1-receptor activation via PACAP resulted in an increased mRNA level of both the PAC1 receptor and its ligand PACAP. Our behavioral studies showed that long-term PACAP treatment of APP[V717I]-transgenic mice improved cognitive function in animals. Thus, nasal application of PACAP was effective, and our results indicate that PACAP could be of therapeutic value in treating AD.—Rat, D., Schmitt, U., Tippmann, F., Dewachter, I., Theunis, C., Wieczerzak, E, Postina, R., van Leuven, F., Fahrenholz, F., Kojro, E. Neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) slows down Alzheimer's disease-like pathology in amyloid precursor protein-transgenic mice.
Collapse
Affiliation(s)
- Dorothea Rat
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
The role of PACAP in central cardiorespiratory regulation. Respir Physiol Neurobiol 2010; 174:65-75. [PMID: 20470908 DOI: 10.1016/j.resp.2010.05.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 05/03/2010] [Accepted: 05/03/2010] [Indexed: 11/22/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) plays a role in almost every biological process from reproduction to hippocampal function. One area where a role for PACAP is not clearly delineated is central cardiorespiratory regulation. PACAP and its receptors (PAC1, VPAC1 and VPAC2) are present in cardiovascular areas of the ventral medulla and spinal cord and in the periphery. Central administration of PACAP generally increases arterial pressure. Knowledge about the role of PACAP in central cardiovascular regulation is growing, but even less is known about PACAP in central respiratory regulation. No specific data is currently available regarding the presence of PACAP or receptors in key respiratory centers, although it is known that neonatal PACAP knock-out mice die suddenly in a manner similar to sudden infant death syndrome (SIDS). Future studies in mature preparations investigating the role of PACAP in the physiology and integration of central cardiorespiratory reflexes are clearly essential for a full understanding of this important neuropeptide in breathing.
Collapse
|
21
|
Increased ethanol preference and serotonin 1A receptor-dependent attenuation of ethanol-induced hypothermia in PACAP-deficient mice. Biochem Biophys Res Commun 2010; 391:773-7. [DOI: 10.1016/j.bbrc.2009.11.136] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Accepted: 11/24/2009] [Indexed: 01/02/2023]
|
22
|
Vaudry D, Falluel-Morel A, Bourgault S, Basille M, Burel D, Wurtz O, Fournier A, Chow BKC, Hashimoto H, Galas L, Vaudry H. Pituitary adenylate cyclase-activating polypeptide and its receptors: 20 years after the discovery. Pharmacol Rev 2009; 61:283-357. [PMID: 19805477 DOI: 10.1124/pr.109.001370] [Citation(s) in RCA: 860] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a 38-amino acid C-terminally alpha-amidated peptide that was first isolated 20 years ago from an ovine hypothalamic extract on the basis of its ability to stimulate cAMP formation in anterior pituitary cells (Miyata et al., 1989. PACAP belongs to the vasoactive intestinal polypeptide (VIP)-secretin-growth hormone-releasing hormone-glucagon superfamily. The sequence of PACAP has been remarkably well conserved during evolution from protochordates to mammals, suggesting that PACAP is involved in the regulation of important biological functions. PACAP is widely distributed in the brain and peripheral organs, notably in the endocrine pancreas, gonads, respiratory and urogenital tracts. Characterization of the PACAP precursor has revealed the existence of a PACAP-related peptide, the activity of which remains unknown. Two types of PACAP binding sites have been characterized: type I binding sites exhibit a high affinity for PACAP and a much lower affinity for VIP, whereas type II binding sites have similar affinity for PACAP and VIP. Molecular cloning of PACAP receptors has shown the existence of three distinct receptor subtypes: the PACAP-specific PAC1-R, which is coupled to several transduction systems, and the PACAP/VIP-indifferent VPAC1-R and VPAC2-R, which are primarily coupled to adenylyl cyclase. PAC1-Rs are particularly abundant in the brain, the pituitary and the adrenal gland, whereas VPAC receptors are expressed mainly in lung, liver, and testis. The development of transgenic animal models and specific PACAP receptor ligands has strongly contributed to deciphering the various actions of PACAP. Consistent with the wide distribution of PACAP and its receptors, the peptide has now been shown to exert a large array of pharmacological effects and biological functions. The present report reviews the current knowledge concerning the pleiotropic actions of PACAP and discusses its possible use for future therapeutic applications.
Collapse
Affiliation(s)
- David Vaudry
- Institut National de la Santé et de la Recherche Médicale U413, European Institute for Peptide Research (Institut Fédératif de Recherches Multidisciplinaires sur les Peptides 23), Mont-Saint-Aignan, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Fila T, Trazzi S, Crochemore C, Bartesaghi R, Ciani E. Lot1 is a key element of the pituitary adenylate cyclase-activating polypeptide (PACAP)/cyclic AMP pathway that negatively regulates neuronal precursor proliferation. J Biol Chem 2009; 284:15325-38. [PMID: 19346254 DOI: 10.1074/jbc.m109.002329] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The tumor suppressor gene Lot1 is highly expressed during brain development. During cerebellar development, Lot1 is expressed by proliferating granule cells with a time course matching the expression of the pituitary adenylate cyclase-activating polypeptide (PACAP) receptor, a neuropeptide receptor that plays an important role in the regulation of granule cell proliferation/survival. Although it has become clear that Lot1 is a negative regulator of cell division in tumor cells, its role in neuronal proliferation is not understood. We previously demonstrated that in cerebellar granule cells Lot1 expression is regulated by the PACAP/cAMP system. The aim of this study was to investigate the role played by Lot1 in neuron proliferation/survival and to identify the molecular mechanisms underlying its actions. Using a Lot1-inducible expression system, we found that in PC12 cells Lot1 negatively regulates proliferation and favors differentiation by up-regulating the expression of the PACAP receptor. In cerebellar granule cells in culture, an increase in Lot1 expression was paralleled by inhibition of proliferation and up-regulation of the PACAP receptor, which in turn positively regulated Lot1 expression. Silencing of Lot1 leads to an increase in granule cell proliferation and a reduction in survival. Confirming the in vitro results, in vivo experiments showed that PACAP induced an increase in Lot1 expression that was paralleled by inhibition of cerebellar granule cell proliferation. These data show that Lot1 is a key element of the PACAP/cAMP pathway that negatively regulates neuronal precursor proliferation. The existence of a PACAP receptor/Lot1-positive feedback loop may powerfully regulate neural proliferation during critical phases of cerebellar development.
Collapse
Affiliation(s)
- Tatiana Fila
- Department of Human and General Physiology, University of Bologna, Piazza di Porta San Donato 2, Bologna, Italy
| | | | | | | | | |
Collapse
|
24
|
Grafer CM, Thomas R, Lambrakos L, Montoya I, White S, Halvorson LM. GnRH stimulates expression of PACAP in the pituitary gonadotropes via both the PKA and PKC signaling systems. Mol Endocrinol 2009; 23:1022-32. [PMID: 19342443 DOI: 10.1210/me.2008-0477] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Recent studies have demonstrated a clear role for pituitary adenylate cyclase-activating polypeptide (PACAP) in the regulation of gonadotropin biosynthesis and secretion, both alone and in conjunction with GnRH. First defined as a hypothalamic releasing factor, PACAP subsequently has been identified in the gonadotrope subpopulation of the anterior pituitary gland, suggesting that PACAP may act as an autocrine-paracrine factor in this tissue. In initial studies, we determined that GnRH markedly stimulated endogenous PACAP mRNA levels and promoter-reporter activity in the mature gonadotrope cell line, LbetaT2. GnRH-stimulated rat PACAP promoter activity was blunted with deletion from position -915 to -402 and eliminated with further truncation to position -77 relative to the transcriptional start site. Site-directed mutagenesis demonstrated a functional requirement for a cAMP response element (CRE)-like site at position -205 and an activating protein-1 (AP-1)-like site at position -275, both of which bound CRE binding protein and AP-1 family members on EMSA. Treatment with pharmacological activators or inhibitors of second messenger signaling pathways implicated the protein kinase A, protein kinase C, and MAPK pathways in the GnRH response. In support of these in vitro data, we demonstrate that JunB binds to the rat PACAP gene promoter by chromatin immunoprecipitation assay and that small interfering RNA knockdown of JunB, cFos, and CRE binding protein factors blunts PACAP expression. In summary, these results further elucidate the complex functional interactions between PACAP and GnRH in the anterior pituitary. Specifically, these studies demonstrate that GnRH-stimulated PACAP gene expression is mediated via multiple signaling pathways acting on CRE/AP-1 sites in the proximal gene promoter. Because both PACAP and GnRH regulate gonadotropin biosynthesis and secretion, these results provide important insight into the critical fine tuning of gonadotrope function and, thereby, the maintenance of normal reproductive function.
Collapse
Affiliation(s)
- Constance M Grafer
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9032, USA
| | | | | | | | | | | |
Collapse
|
25
|
Sanchez A, Chiriva-Internati M, Grammas P. Transduction of PACAP38 protects primary cortical neurons from neurotoxic injury. Neurosci Lett 2008; 448:52-5. [PMID: 18938212 DOI: 10.1016/j.neulet.2008.10.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2008] [Revised: 09/25/2008] [Accepted: 10/01/2008] [Indexed: 10/21/2022]
Abstract
Neurotrophic factors such as pituitary adenylate cyclase activating polypeptide (PACAP38) are promising therapeutics for neurodegenerative diseases. However, delivery of trophic factors into brain neurons remains a challenge. The objective of this study is to determine whether adeno-associated virus (AAV) can mediate PACAP38 gene delivery into neurons in vitro and if transduction of AAV/PACAP38 into cortical neurons protects cells against neurotoxic insult. Primary cortical neuronal cultures are transduced with rAAV/PACAP38/GFP and cell survival against the nitric oxide releasing neurotoxin sodium nitroprusside (SNP) determined. GFP expression, a surrogate marker for successful transduction, is detected using fluorescent microscopy. The results show expression of GFP transgene and AAV capsid proteins in neurons. PACAP38 transduction significantly increases cell survival of neurons exposed to SNP. These results support the feasibility of using AAV-mediated delivery of PACAP38 to enhance neuronal survival and suggest that AAV-delivered PACAP38 maybe a therapeutic strategy for neurodegenerative diseases.
Collapse
Affiliation(s)
- Alma Sanchez
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | | |
Collapse
|
26
|
Implication of Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) for Neuroprotection of Nicotinic Acetylcholine Receptor Signaling in PC12 Cells. J Mol Neurosci 2008; 36:73-8. [DOI: 10.1007/s12031-008-9127-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Accepted: 06/12/2008] [Indexed: 10/21/2022]
|
27
|
Baba A. [Molecular pharmacologic approaches to functional analysis of new biological target molecules for drug discovery]. YAKUGAKU ZASSHI 2007; 127:1643-54. [PMID: 17917422 DOI: 10.1248/yakushi.127.1643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This review focuses on two pharmacologic approaches to the functional evaluation of new target molecules for drug discovery. One is the development of a novel specific antagonist of the Na(+)-Ca(++) exchanger (NCX) SEA0400. The other is a comprehensive analysis of the functions of pituitary adenylate cyclase-activating polypeptide (PACAP), a neuropeptide ligand for G protein-coupled receptors. NCX is the one of the last target molecules regulating the cellular Ca(++) concentration. There was no efficient way to address the pathophysiologic roles of NCX until a specific antagonist, 2-[4-[(2,5-difluorophenyl)methoxy]phenoxy]-5-ethoxyaniline (SEA0400), was developed. Our recent studies using SEA0400 clearly showed the possible roles of NCX in several pathologic states of cardiovascular and nervous tissues. In our second approach including gene-targeting methods, we found new, unexpected roles of PACAP in higher brain functions, such as psychomotor, cognition, photoentrainment, and nociception. Based on these experimental findings, a genetic association study in schizophrenia patients revealed that the single-nucleotide polymorphisms of the PACAP gene are significantly associated with the hypofunction of the hippocampus. Regarding the peripheral roles of PACAP, we found that PACAP is involved not only in the regulation of insulin secretion in pancreatic islets, but also in the regulation of islet turnover. In subsequent phenotypic analysis of PACAP transgenic mice, we identified novel candidate genes that probably have promising functional roles.
Collapse
Affiliation(s)
- Akemichi Baba
- Molecular Pharmacological Laboratory, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita City 565-0871, Japan.
| |
Collapse
|
28
|
Braas KM, Schutz KC, Bond JP, Vizzard MA, Girard BM, May V. Microarray analyses of pituitary adenylate cyclase activating polypeptide (PACAP)-regulated gene targets in sympathetic neurons. Peptides 2007; 28:1856-70. [PMID: 17512639 PMCID: PMC2744890 DOI: 10.1016/j.peptides.2007.04.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Revised: 03/29/2007] [Accepted: 04/10/2007] [Indexed: 11/23/2022]
Abstract
The high and preferential expression of the PAC(1)(short)HOP1 receptor in postganglionic sympathetic neurons facilitates microarray studies for mechanisms underlying PACAP-mediate neurotrophic signaling in a physiological context. Replicate primary sympathetic neuronal cultures were treated with 100 nM PACAP27 either acutely (9 h) or chronically (96 h) before RNA extraction and preparation for Affymetrix microarray analysis. Compared to untreated control cultures, acute PACAP treatment modulated significantly the expression of 147 transcripts of diverse functional groups, including peptides, growth factors/cytokines, transcriptional factors, receptors/signaling effectors and cell cycle regulators, that collectively appeared to facilitate neuronal plasticity, differentiation and/or regeneration processes. Some regulated transcripts, for example, were related to BDNF/TrkB, IL-6/Jak2/Socs2 and TGF/follistatin signaling; many transcripts affected bioactive peptide and polyamine biosynthesis. Although chronic PACAP treatments altered the expression of 109 sympathetic transcripts, only 43 transcripts were shared between the acute and chronic treatment data sets. The PACAP-mediated changes in transcript expression were corroborated independently by quantitative PCR measurement. The PACAP-regulated transcripts in sympathetic neurons did not bear strong resemblance to those in PACAP-treated pheochromocytoma cells. However, many PACAP-targeted sympathetic transcripts, especially those related to peptide plasticity and nerve regeneration processes, coincided significantly with genes altered after peripheral nerve injury. The ability for sympathetic PAC(1)(short)HOP1 receptors to engage multiple downstream signaling cascades appeared to be reflected in the number and diversity of genes targeted in a multifaceted strategy for comprehensive neurotrophic responses.
Collapse
Affiliation(s)
- Karen M. Braas
- Department of Anatomy and Neurobiology, The University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Kristin C. Schutz
- Department of Anatomy and Neurobiology, The University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Jeffrey P. Bond
- Department of Bioinformatics Core Facility, The University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Margaret A. Vizzard
- Department of Anatomy and Neurobiology, The University of Vermont College of Medicine, Burlington, Vermont 05405
- Department of Neurology, The University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Beatrice M. Girard
- Department of Anatomy and Neurobiology, The University of Vermont College of Medicine, Burlington, Vermont 05405
- Department of Neurology, The University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Victor May
- Department of Anatomy and Neurobiology, The University of Vermont College of Medicine, Burlington, Vermont 05405
- Corresponding Author: Victor May, Ph.D., Departments of Anatomy & Neurobiology, and Pharmacology, University of Vermont College of Medicine, 149 Beaumont Avenue, Health Science Research Facility, Room 428, Burlington, Vermont 05405, (802) 656-4579 (voice), (802) 656-8704 (facsimile),
| |
Collapse
|
29
|
Girard BM, Young BA, Buttolph TR, White SL, Parsons RL. Regulation of neuronal pituitary adenylate cyclase-activating polypeptide expression during culture of guinea-pig cardiac ganglia. Neuroscience 2007; 146:584-93. [PMID: 17367946 PMCID: PMC2048657 DOI: 10.1016/j.neuroscience.2007.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2006] [Revised: 01/17/2007] [Accepted: 02/01/2007] [Indexed: 10/23/2022]
Abstract
The trophic neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) increases in many different neuron types following injury; a response postulated to support cell survival and regeneration. In acutely isolated cardiac ganglia, approximately 1% of the cardiac neurons exhibited PACAP immunoreactivity whereas after 72 h in culture, approximately 25% of the neurons were PACAP immunoreactive. In contrast, there was no increase in vasoactive intestinal polypeptide (VIP)-immunoreactive (IR) cells. Using a combination of immunocytochemical and molecular techniques, we have quantified PACAP expression, during explant culture of guinea-pig cardiac ganglia. Using real time polymerase chain reaction, PACAP transcript levels increased progressively up to 48 h in culture with no further increase after 72 h. PACAP transcript levels were reduced by neurturin at 48 h in culture but not after 24 or 72 h in culture. In addition, neurturin partially suppressed the percentage of PACAP-IR neurons after 72 h in culture, an effect mediated by activation of the phosphatidylinositol 3-kinase and mitogen-activated protein kinase signaling pathways. The addition of different known regulatory molecules, including ciliary neurotrophic factor (CNTF), interleukin-1 beta (Il-1beta), tumor necrosis factor-alpha (TNFalpha), fibroblast growth factor basic (bFGF), transforming growth factor-beta (TGF-beta) and nerve growth factor (NGF) did not increase the percentage of PACAP-IR neurons after 24 h in culture; a result indicating that the generation and secretion of these factors did not stimulate PACAP expression. The presence of 20 nM PACAP or 10 muM forskolin increased the percentage of PACAP-IR cardiac neurons in 24 h cultures, but not in 72 h cultures. Neither treatment enhanced the number of VIP-IR neurons. The addition of the PACAP selective receptor (PAC(1)) receptor antagonist, M65 (100 nM) suppressed the 20 nM PACAP-induced increase in percentage of PACAP-IR cells in 24 h cultures indicating the effect of PACAP was mediated through the PAC(1) receptor. However, 100 nM M65 had no effect on the percentage of PACAP-IR cells in either 24 or 48 h cultures not treated with exogenous PACAP, suggesting that endogenous release of PACAP likely did not contribute to the enhanced peptide expression. We postulate that the enhanced PACAP expression, which occurs in response to injury is facilitated in the explant cultured cardiac ganglia by the loss of a target-derived inhibitory factor, very likely neurturin. In intact tissues the presence of neurturin would normally suppress PACAP expression. Lastly, our results indicate that many common trophic factors do not enhance PACAP expression in the cultured cardiac neurons. However, the stimulatory role of an, as yet, unidentified factor cannot be excluded.
Collapse
Affiliation(s)
- B M Girard
- Department of Anatomy and Neurobiology, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | | | | | | | | |
Collapse
|
30
|
Pavelock KA, Girard BM, Schutz KC, Braas KM, May V. Bone morphogenetic protein down-regulation of neuronal pituitary adenylate cyclase-activating polypeptide and reciprocal effects on vasoactive intestinal peptide expression. J Neurochem 2006; 100:603-16. [PMID: 17181550 DOI: 10.1111/j.1471-4159.2006.04293.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Among bone morphogenetic proteins (BMPs), the decapentaplegic (Dpp; BMP2, BMP4) and glass bottom boat (Gbb/60A; BMP5, BMP6, BMP7) subgroups have well-described functions guiding autonomic and sensory neuronal development, fiber formation and neurophenotypic identities. Evaluation of rat superior cervical ganglia (SCG) post-ganglionic sympathetic neuron developmental regulators identified that selected BMPs of the transforming growth factor beta superfamily have reciprocal effects on neuronal pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) expression. Dpp and Gbb/60A BMPs rapidly down-regulated PACAP expression, while up-regulating other sympathetic neuropeptides, including PACAP-related VIP. The suppressive effects of BMP on PACAP mRNA and peptide expression were potent, efficacious and phosphorylated mothers against decapentaplegic homolog (Smad) signaling-dependent. Axotomy of SCG dramatically increases PACAP expression, and the possibility that abrogation of inhibitory retrograde target tissue BMP signaling may contribute to this up-regulation of sympathetic neuron PACAP was investigated. Replacement of BMP6 to SCG explant preparations significantly blunted the injury-induced elevated PACAP expression, with a concomitant decrease in sympathetic PACAP-immunoreactive neuron numbers. These studies suggested that BMPs modulate neuropeptide identity and diversity by stimulating or restricting the expression of specific peptidergic systems. Furthermore, the liberation of SCG neurons from target-derived BMP inhibition following axotomy may be one participating mechanism associated with injury-induced neuropeptidergic plasticity.
Collapse
Affiliation(s)
- Kristen A Pavelock
- Department of Anatomy and Neurobiology, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | | | | | | | | |
Collapse
|
31
|
Tai TC, Wong-Faull DC, Claycomb R, Wong DL. Nerve growth factor regulates adrenergic expression. Mol Pharmacol 2006; 70:1792-801. [PMID: 16926281 DOI: 10.1124/mol.106.026237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The mechanism by which nerve growth factor (NGF) regulates adrenergic expression was examined in PC-12 cells transfected with a rat phenylethanolamine N-methyl-transferase (PNMT) promoter-luciferase reporter gene construct pGL3RP893. NGF treatment increased PNMT promoter-driven luciferase activity in a dose- and time-dependent manner. Induction was attenuated by inhibition of the extracellular signal-regulated kinase mitogen-activated protein kinase (MAPK) pathway ( approximately 60%) but not by inhibition of the protein kinase A (PKA), protein kinase C, phosphoinositol kinase, or p38 MAPK pathways. Deletion PNMT promoter-luciferase reporter gene constructs showed that the NGF-responsive sequences lay within the proximal -392 base pairs (bp) of PNMT promoter, wherein binding elements for Egr-1 (-165 bp) and Sp1 (-48 bp) reside. Western analysis further showed that NGF increased nuclear levels of Egr-1, but not Sp1 or the catalytic subunit of PKA. Gel mobility shift assays showed increased potential for Egr-1, but not Sp1, protein-DNA binding complex formation. Mutation of either the Egr-1 or Sp1 binding sites in the PNMT promoter attenuated NGF activation. NGF, combined with pituitary adenylyl cyclase-activating protein (PACAP), another PNMT transcriptional activator, cooperatively stimulated PNMT promoter driven-luciferase activity beyond levels observed with either neurotrophin alone. Finally, post-transcriptional control seems to be another important mechanism by which neurotrophins regulate the adrenergic phenotype. NGF, PACAP, and a combination of the two stimulated both intron-retaining and intronless PNMT mRNA and PNMT protein, but to different extents.
Collapse
Affiliation(s)
- T C Tai
- Laboratory of Molecular and Developmental Neurobiology, Department of Psychiatry, McLean Hospital, Harvard Medical School, 115 Mill St., MRC 116, Belmont, MA 02478, USA
| | | | | | | |
Collapse
|
32
|
Ravni A, Bourgault S, Lebon A, Chan P, Galas L, Fournier A, Vaudry H, Gonzalez B, Eiden LE, Vaudry D. The neurotrophic effects of PACAP in PC12 cells: control by multiple transduction pathways. J Neurochem 2006; 98:321-9. [PMID: 16805827 DOI: 10.1111/j.1471-4159.2006.03884.x] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal polypeptide (VIP) are closely related members of the secretin superfamily of neuropeptides expressed in both the brain and peripheral nervous system, and they exhibit neurotrophic and neurodevelopmental effects in vivo. Like the index member of the Trk receptor ligand family, nerve growth factor (NGF), PACAP promotes the differentiation of PC12 cells, a well-established cell culture model, to investigate neuronal differentiation, survival and function. Stimulation of catecholamine secretion and enhanced neuropeptide biosynthesis are effects exerted by PACAP at the adrenomedullary synapse in vivo and on PC12 cells in vitro through stimulation of the specific PAC1 receptor. Induction of neuritogenesis, growth arrest, and promotion of cell survival are effects of PACAP that occur in developing cerebellar, hippocampal and cortical neurons, as well as in the more tractable PC12 cell model. Study of the mechanisms through which PACAP exerts its various effects on cell growth, morphology, gene expression and survival, i.e. its actions as a neurotrophin, in PC12 cells is the subject of this review. The study of neurotrophic signalling by PACAP in PC12 cells reveals that multiple independent pathways are coordinated in the PACAP response, some activated by classical and some by novel or combinatorial signalling mechanisms.
Collapse
Affiliation(s)
- Aurélia Ravni
- Laboratory of Cellular and Molecular Neuroendocrinology, European Institute for Peptide Research, University of Rouen, Mont-Saint-Aignan, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Greene LA, Angelastro JM. You can't go home again: transcriptionally driven alteration of cell signaling by NGF. Neurochem Res 2006; 30:1347-52. [PMID: 16341597 DOI: 10.1007/s11064-005-8807-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2005] [Indexed: 11/30/2022]
Abstract
Here we review findings indicating that neurotrophins such as NGF promote changes in gene transcription that in turn influence the ways that cells subsequently respond to trophic factors. As a result, initial responses of "naïve" cells to NGF and other trophic agents differ from those of cells with prior NGF exposure. We discuss specific examples based on reports in the literature as well as on data derived from a serial analysis of gene expression (SAGE) study of NGF-promoted transcriptional changes in PC12 pheochromocytoma cells.
Collapse
Affiliation(s)
- Lloyd A Greene
- Department of Pathology, Center for Neurobiology and Behavior, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| | | |
Collapse
|
34
|
Kohama K, Tsukamoto Y, Furuya M, Okamura K, Tanaka H, Miki N, Taira E. Molecular cloning and analysis of the mouse gicerin gene. Neurochem Int 2005; 46:465-70. [PMID: 15769548 DOI: 10.1016/j.neuint.2004.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2004] [Revised: 12/20/2004] [Accepted: 12/21/2004] [Indexed: 11/15/2022]
Abstract
Gicerin is a cell adhesion molecule, which has five immunoglobulin-like loop structures in an extracellular domain followed by a single transmembrane domain and a short cytoplasmic tail. We have reported that gicerin participates in neurite extension and structural organization of the nervous system, and its expression in the nervous system is high during the development and dramatically decreased after birth. To elucidate the mechanism how the expression of gicerin is regulated, we performed a genomic cloning of a mouse gicerin. A fragment of 16 kbp genomic clone contained 8 kbp gicerin gene composed of 16 exons with 6 kbp upstream region. Genomic cloning revealed that two isoforms of gicerin were generated by an alternative splicing of exon 15 results in cytoplasmic domains composed of either 63 or 21 amino acids. As for an expressional regulation of gicerin, we found that the mRNA content of gicerin in PC12 cells was regulated by cAMP. Quantitative-PCR analysis revealed that forskolin induced four-fold increase of gicerin mRNA. To characterize the involvement of its promoter region, we examined the promoter activity in PC12 cells by a luciferase-reporter assay. We found that a CRE site located at 60 bp upstream of gicerin gene was responsible for the increase of its mRNA induced by forskolin.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, CD/biosynthesis
- Antigens, CD/chemistry
- Antigens, CD/genetics
- Base Sequence
- CD146 Antigen
- Cloning, Molecular
- Colforsin/pharmacology
- Cyclic AMP/metabolism
- Exons/genetics
- Gene Expression Regulation, Developmental/genetics
- Genes/genetics
- Genes, Regulator/drug effects
- Genes, Regulator/genetics
- Genes, Reporter
- Integrases/genetics
- Mice
- Molecular Sequence Data
- Neural Cell Adhesion Molecules/biosynthesis
- Neural Cell Adhesion Molecules/chemistry
- Neural Cell Adhesion Molecules/genetics
- PC12 Cells
- Promoter Regions, Genetic
- Protein Structure, Tertiary/physiology
- RNA, Messenger/metabolism
- Rats
- Up-Regulation/drug effects
- Up-Regulation/genetics
Collapse
Affiliation(s)
- Keiko Kohama
- Department of Pharmacology, Osaka University Medical School, Room# A6, 2-2 Yamada-oka Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | |
Collapse
|
35
|
Hirose M, Hashimoto H, Shintani N, Nakanishi M, Arakawa N, Iga J, Niwa H, Miyazaki JI, Baba A. Differential expression of mRNAs for PACAP and its receptors during neural differentiation of embryonic stem cells. ACTA ACUST UNITED AC 2005; 126:109-13. [PMID: 15620423 DOI: 10.1016/j.regpep.2004.08.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The expressions of mRNAs for pituitary adenylate cyclase-activating polypeptide (PACAP), vasoactive intestinal peptide (VIP), and their receptors (PAC1, VPAC1 and VPAC2) were examined in the five steps of the in vitro neuronal culture model of embryonic stem (ES) cell differentiation. mRNAs for PACAP, VIP, PAC1 receptor, and VPAC2 receptor were moderately expressed in neural stem cell-enriched cultures, while VPAC1 receptor mRNA was most prominently expressed in embryoid bodies (EBs). The expression of PAC1 receptor mRNA was further upregulated after terminal differentiation into neurons. In contrast, the expressions of PAC1 receptor and PACAP mRNAs were markedly decreased after glial differentiation. These results suggest that this in vitro neuronal culture system will be a useful model for future studies on the functional role of the PACAPergic system during different stages of neuronal development.
Collapse
MESH Headings
- Animals
- Cell Differentiation/physiology
- Cells, Cultured
- Mice
- Multipotent Stem Cells/physiology
- Nerve Growth Factors/biosynthesis
- Nerve Growth Factors/genetics
- Nerve Tissue/embryology
- Neurons/physiology
- Neuropeptides/biosynthesis
- Neuropeptides/genetics
- Neurotransmitter Agents/biosynthesis
- Neurotransmitter Agents/genetics
- Pituitary Adenylate Cyclase-Activating Polypeptide
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Receptors, Cell Surface/biosynthesis
- Receptors, Cell Surface/genetics
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I
- Receptors, Vasoactive Intestinal Peptide, Type II
- Receptors, Vasoactive Intestinal Polypeptide, Type I
Collapse
Affiliation(s)
- Megumi Hirose
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Shintani N, Suetake S, Hashimoto H, Koga K, Kasai A, Kawaguchi C, Morita Y, Hirose M, Sakai Y, Tomimoto S, Matsuda T, Baba A. Neuroprotective action of endogenous PACAP in cultured rat cortical neurons. ACTA ACUST UNITED AC 2005; 126:123-8. [PMID: 15620425 DOI: 10.1016/j.regpep.2004.08.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) exerts neurotrophic effects both in vitro and in vivo. Here we demonstrate the upregulation of PACAP mRNA expression in cultured rat cortical neurons after excitotoxic glutamate exposure, and the exacerbating effect of the PACAP receptor antagonist, PACAP(6-38), on neuronal viability. PACAP mRNA levels were increased up to 3.5-fold 8 h after glutamate exposure. PACAP(6-38) decreased the viability of cortical neurons, irrespective of whether the cells were exposed to glutamate or not. PACAP(6-38) also inhibited glutamate-induced expression of PACAP mRNA, suggesting that PACAP acts via an autocrine or paracrine mechanism to enhance PACAP expression itself. Glutamate exposure is known to increase brain-derived neurotrophic factor (BDNF) mRNA expression. This increased expression was markedly suppressed by PACAP(6-38). Our previous study has shown that PACAP stimulates the PACAP gene transcription in PC12 cells. Taken together, these data may suggest that endogenous PACAP regulates the expression of PACAP itself and BDNF. Although it may also be possible that PACAP(6-38)-induced death of PACAP and BDNF mRNA-expressing cells, per se, results in reduced levels of these mRNAs, the present results support the idea that endogenous PACAP has a neuroprotective action.
Collapse
Affiliation(s)
- Norihito Shintani
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kasai A, Shintani N, Oda M, Kakuda M, Hashimoto H, Matsuda T, Hinuma S, Baba A. Apelin is a novel angiogenic factor in retinal endothelial cells. Biochem Biophys Res Commun 2005; 325:395-400. [PMID: 15530405 DOI: 10.1016/j.bbrc.2004.10.042] [Citation(s) in RCA: 216] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2004] [Indexed: 01/11/2023]
Abstract
There has been much focus recently on the possible functions of apelin, an endogenous ligand for the orphan G-protein-coupled receptor APJ, in cardiovascular and central nervous systems. We report a new function of apelin as a novel angiogenic factor in retinal endothelial cells. The retinal endothelial cell line RF/6A highly expressed both apelin and APJ transcripts, while human umbilical venous endothelial cells (HUVECs) only expressed apelin mRNA. In accordance with these observations, apelin at concentrations of 1 pM-1 microM significantly enhanced migration, proliferation, and capillary-like tube formation of RF/6A cells, but not those of HUVECs, whereas VEGF stimulates those parameters of both cell types. In vivo Matrigel plug assay for angiogenesis, the inclusion of 1 nM apelin in the Matrigel resulted in clear capillary-like formations with an increase of hemoglobin content in the plug. This is the first report showing that apelin is an angiogenic factor in retinal endothelial cells.
Collapse
Affiliation(s)
- Atsushi Kasai
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Japan
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Reglodi D, Tamás A, Lubics A, Szalontay L, Lengvári I. Morphological and functional effects of PACAP in 6-hydroxydopamine-induced lesion of the substantia nigra in rats. ACTA ACUST UNITED AC 2004; 123:85-94. [PMID: 15518897 DOI: 10.1016/j.regpep.2004.05.016] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) has several different actions in the nervous system, including neuroprotective effects. In the present study, we investigated the effects of different doses of PACAP on the functional and morphological outcome in a rat model of Parkinson's disease. Rats were given unilateral injections of 6-hydroxydopamine (6-OHDA) into the substantia nigra. PACAP-treated animals received 1, 0.1 or 0.01 microg PACAP as a pretreatment. Control animals without PACAP treatment displayed severe hypokinesia at 1 and 10 days post-lesion when compared to normal animals or those receiving saline only. PACAP treatment resulted in less severe acute hypokinesia, and complete recovery by 10 days. Asymmetrical signs were observed in all lesioned animals 1 day post-lesion. PACAP-treated animals, however, showed better recovery as they ceased to display asymmetrical signs 10 days later and showed markedly less apomorphine-induced rotations. Best behavioral outcome was observed in animals treated with 0.1 microg PACAP. Tyrosine-hydroxylase (TH) immunohistochemistry revealed increased number of dopaminergic neurons in the substantia nigra pars compacta and in the ventral tegmental area in all PACAP-treated rats in contrast to the severe cell loss in control animals. These results indicate that PACAP may be a promising therapeutic agent in Parkinson's disease.
Collapse
Affiliation(s)
- Dóra Reglodi
- Department of Anatomy, Pécs University Medical Faculty and Neurohumoral Regulations Research Group of the Hungarian Academy of Sciences, Szigeti u 12, 7624 Pécs, Hungary.
| | | | | | | | | |
Collapse
|
39
|
Fukuchi M, Tabuchi A, Tsuda M. Activity-dependent transcriptional activation and mRNA stabilization for cumulative expression of pituitary adenylate cyclase-activating polypeptide mRNA controlled by calcium and cAMP signals in neurons. J Biol Chem 2004; 279:47856-65. [PMID: 15355970 DOI: 10.1074/jbc.m409090200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although it has been established that an activity-dependent gene transcription is induced by the calcium (Ca(2+)) signals in neurons, it is unclear how the specific mRNA moieties are transiently accumulated in response to synaptic transmission which evokes multiple intracellular signals including Ca(2+) and cAMP ones. The expression of pituitary adenylate cyclase activating polypeptide (PACAP), a neuropeptide, is controlled by Ca(2+) signals evoked via membrane depolarization in neurons, and, in cultured rat cortical neuronal cells, we found that the Ca(2+) signal-mediated activation of the PACAP gene promoter was critically controlled by a single cAMP-response element (CRE) located at around -200, to which the CRE-binding protein predominantly bound. The Ca(2+) signal-induced expression of PACAP mRNA was enhanced by forskolin, which evokes cAMP signals. In support, the PACAP gene promoter was synergistically enhanced by Ca(2+) and cAMP signals through the CRE, accompanying a prolonged activation of extracellular signal-related protein kinase 1/2 and CRE-binding protein. On the other hand, sole administration of forskolin markedly reduced the cellular content of PACAP mRNA, which was restored by the addition of Ca(2+) signals. We found that the stability of PACAP mRNA was increased in response to Ca(2+) signals but not that of activity-regulated cytoskeleton-associated protein (Arc) mRNA, indicating an activity-dependent stabilization of specific mRNA species in neurons, which can antagonize the regulation mediated by cAMP signals. Thus, the transcriptional activation and mRNA stabilization are coordinately regulated by Ca(2+) and cAMP signals for the cumulative expression of PACAP mRNA in neurons.
Collapse
Affiliation(s)
- Mamoru Fukuchi
- Department of Biological Chemistry, Faculty of Pharmaceutical Sciences, Toyama Medical and Pharmaceutical University, Sugitani 2630, Toyama 931-0194, Japan
| | | | | |
Collapse
|
40
|
Sakai Y, Hashimoto H, Shintani N, Katoh H, Negishi M, Kawaguchi C, Kasai A, Baba A. PACAP activates Rac1 and synergizes with NGF to activate ERK1/2, thereby inducing neurite outgrowth in PC12 cells. ACTA ACUST UNITED AC 2004; 123:18-26. [PMID: 15046862 DOI: 10.1016/j.molbrainres.2003.12.013] [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] [Accepted: 12/23/2003] [Indexed: 11/29/2022]
Abstract
The mechanisms linked to the neuritogenic effect of PACAP acting in synergy with NGF were analyzed in PC12 cells. Recently, we have shown that PACAP synergizes with NGF to stimulate PACAP gene transcription and neurite outgrowth, differentially dependent on both the ERK1/2 and p38 MAP kinase pathways in PC12 cells. This suggests that PACAP modulates mitogen signaling pathways governing cell differentiation, in part through MAP kinase activation and an autocrine mechanism. Here, we studied the mechanism of the underlying neuritogenic actions of PACAP. PACAP induced transient activation of Rac1, a small GTPase involved in neurite outgrowth, in a PI3-kinase-independent manner, and stimulated accumulation of active Rac1 at filamentous actin-rich protrusions on the cell surface to induce subsequent neurite formation. PACAP had no additional effect on the activity of Rac1 beyond the effect of NGF and failed to activate Ras or Cdc42. By contrast, simultaneous treatment with PACAP and NGF acts in synergy to induce prolonged activation of ERK1/2. These results indicate for the first time that PACAP induces activation of Rac1 associated with neurite outgrowth and suggest that the synergistic effect of PACAP and NGF on neurite extension is due to enhanced activation of ERK1/2.
Collapse
Affiliation(s)
- Yoshiyuki Sakai
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Miyata A, Sugawara H, Iwata SI, Shimizu T, Kangawa K. [The regulatory mechanism for neuron specific expression of PACAP gene]. Nihon Yakurigaku Zasshi 2004; 123:235-42. [PMID: 15056938 DOI: 10.1254/fpj.123.235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP), a pleiotropic neuropeptide, is present abundantly in the central nervous system. In the 5'-flanking region of the PACAP gene, we found and characterized two negative regulatory elements, which are homologous to the neural-restrictive silencer element (NRSE). Their sequence and position were significantly conserved among mouse, human, and rat PACAP genes. NRSE is a crucial negative-acting DNA regulatory element for neuron-specific gene expression. NRSE acts through the transcription factor known as neural-restrictive silencer factor (NRSF). In non-neuronal cells, NRSF suppresses the expression of neuron-specific genes. On the other hand, in neuronal cells, NRnV, a NRSF truncated form, repress their expressions in a dominant negative manner. The electrophoretic mobility shift assay with 3T3 cells extract demonstrated the identical complexes among NRSLE-1, NRSLE2, and the NRSE of rat type II sodium channel gene. In the luciferase reporter assay, NRSLEs suppressed SV40 promoter activity in 3T3 cells, but not in PC12 cells. RT-PCR analysis revealed that PACAP and NRnV mRNAs are expressed in neuronal cells (differentiated PC12), but not in non-neuronal cells (3T3 or C6). These results suggested that the NRSE-NRSF system might be involved in the regulatory mechanism of neuron-specific expression of the PACAP gene.
Collapse
Affiliation(s)
- Atsuro Miyata
- Department of Pharmacology, Postgraduate School of Medical and Dental Sciences, Kagoshima University, Japan.
| | | | | | | | | |
Collapse
|
42
|
Tomimoto S, Hashimoto H, Shintani N, Yamamoto K, Kawabata Y, Hamagami KI, Yamagata K, Miyagawa JI, Baba A. Overexpression of pituitary adenylate cyclase-activating polypeptide in islets inhibits hyperinsulinemia and islet hyperplasia in agouti yellow mice. J Pharmacol Exp Ther 2004; 309:796-803. [PMID: 14742740 DOI: 10.1124/jpet.103.062919] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is an intraislet neuropeptide and shares insulinotropic and insulin-sensitizing properties with glucagon-like peptide-1 (GLP-1); however, the pathophysiological significance of PACAP in diabetes remains largely unknown. To assess this, we crossed our recently developed transgenic mice overexpressing PACAP in pancreatic beta-cells (Tg/+), with lethal yellow agouti (KKA(y)) mice (A(y)/+), a genetic model for obesity-diabetes, and examined the metabolic and morphological phenotypes of F(1) animals. Tg/+ mice with the A(y) allele (Tg/+:A(y)/+) developed maturity-onset obesity and diabetes associated with hyperglycemia, hyperlipidemia, and hyperphagia, similar to those of A(y)/+ mice, but hyperinsulinemia was significantly ameliorated in Tg/+:A(y)/+ mice. Although A(y)/+ mice exhibited a marked increase in islet mass resulting from hyperplasia and hypertrophy, this increase was significantly attenuated in Tg/+:A(y)/+ mice. Size frequency distribution analysis revealed that the very large islets comprising one-fourth of islets of A(y)/+ mice were selectively reduced in Tg/+:A(y)/+ mice. Because functional defects have been demonstrated in the large islets of obese animal models, together these findings suggest that PACAP regulates hyperinsulinemia and the abnormal increase in islet mass that occurs during the diabetic process.
Collapse
Affiliation(s)
- Shuhei Tomimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hashimoto H, Kunugi A, Arakawa N, Shintani N, Fujita T, Kasai A, Kawaguchi C, Morita Y, Hirose M, Sakai Y, Baba A. Possible involvement of a cyclic AMP-dependent mechanism in PACAP-induced proliferation and ERK activation in astrocytes. Biochem Biophys Res Commun 2004; 311:337-43. [PMID: 14592419 DOI: 10.1016/j.bbrc.2003.10.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
In cultured astrocytes, PACAP activates extracellular signal-regulated kinase (ERK) and induces cell proliferation at picomolar concentrations. Here, we examined the role of cyclic AMP signaling underlying the effects of PACAP. PACAP38 induced accumulation of cyclic AMP in astrocytes at concentrations as low as 10(-12)M. PACAP38 (10(-12)-10(-9)M)-stimulated cell proliferation was completely abolished by the cyclic AMP antagonist Rp-cAMP, whereas the protein kinase A (PKA) inhibitor H89 had no effect. This PACAP38-mediated effect was also abolished by the ERK kinase inhibitor PD98059, suggesting the involvement of ERK in PACAP-induced proliferation. PACAP38 (10(-12)M)-stimulated phosphorylation of ERK lasted for at least 60 min. This effect was completely abolished by Rp-cAMP but not by H89. Dibutyryl cyclic AMP maximally stimulated the incorporation of thymidine and activation of ERK at 10(-10)M. These results suggest that PACAP-mediated stimulation of ERK activity and proliferation of astrocytes may involve a cyclic AMP-dependent, but PKA-independent, pathway.
Collapse
Affiliation(s)
- Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Jongsma Wallin H, Pettersson LME, Verge VMK, Danielsen N. Effect of anti-nerve growth factor treatment on pituitary adenylate cyclase activating polypeptide expression in adult sensory neurons exposed to adjuvant induced inflammation. Neuroscience 2003; 120:325-31. [PMID: 12890505 DOI: 10.1016/s0306-4522(03)00118-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Expression of pituitary adenylate cyclase activating polypeptide (PACAP) is increased in sensory neurons exposed to adjuvant induced peripheral inflammation. Local elevation in expression of the neurotrophin nerve growth factor (NGF) is a main factor contributing to the neuronal response to inflammation. This study examines the role of endogenous NGF in inflammation-associated increases in PACAP expression using the adjuvant-induced peripheral inflammation model with or without systemic administration of antibodies against NGF. Quantitative in situ hybridization was used to detect changes in neuronal PACAP mRNA expression and to correlate this expression with neuronal mRNA expression of the NGF receptor tyrosine kinase (trk) A. The results from this study show that inflammation triggered increases in PACAP expression occurs in small- to medium-sized dorsal root ganglion (DRG) neurons that also express trkA, and that this elevation in PACAP expression is prevented by systemic injection of anti-NGF. This supports a role for NGF as a positive regulator of PACAP expression during inflammation.
Collapse
Affiliation(s)
- H Jongsma Wallin
- Department of Physiological Sciences, BMC F10, Lund University, SE-221 84 Lund, Sweden.
| | | | | | | |
Collapse
|
45
|
Kimura H, Kawatani M, Ito E, Ishikawa K. Effects of pituitary adenylate cyclase-activating polypeptide on facial nerve recovery in the Guinea pig. Laryngoscope 2003; 113:1000-6. [PMID: 12782812 DOI: 10.1097/00005537-200306000-00016] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES/HYPOTHESIS Pituitary adenylate cyclase-activating polypeptide (PACAP) has neurotrophic effects of neural regeneration and gives protection to the nervous system. We investigated whether PACAP had a neurotrophic effect on peripheral motoneurons and whether PACAP could facilitate glial cell line-derived neurotrophic factor (GDNF), a neurotrophin, in nerve regeneration. The presence and distribution of PACAP receptors following facial nerve transection were also investigated. STUDY DESIGN Animal experiment. METHODS Unilateral transection of the facial nerve was performed in male Hartley guinea pigs, and PACAP was injected at the site. Saline was substituted as a control. Compound muscle action potentials were recorded to measure the changes of latency. Glial cell line-derived neurotrophic factor (GDNF) content in facial target muscle was measured using enzyme-linked immunosorbent assay. The regenerating site was taken for histological studies. RESULTS Pituitary adenylate cyclase-activating polypeptide hastened the appearance of compound muscle action potentials and shortened the latency. Pituitary adenylate cyclase-activating polypeptide increased and prolonged the nerve transection-induced GDNF increase in the facial muscles. The number of myelinated fibers at 1 to 4 weeks after the transection was increased. PAC1 receptor or VPAC1 receptor or both were identified in the injury area at 2 to 4 weeks. CONCLUSIONS Pituitary adenylate cyclase-activating polypeptide facilitated the recovery of latency of compound muscle action potentials or the number of myelinated axons, or both. Pituitary adenylate cyclase-activating polypeptide prolonged the GDNF levels in target organs. These data indicated that PACAP promoted regeneration of the facial nerve.
Collapse
Affiliation(s)
- Hiromoto Kimura
- Department of Otolaryngology, Akita University School of Medicine, Japan.
| | | | | | | |
Collapse
|
46
|
Yamamoto K, Hashimoto H, Tomimoto S, Shintani N, Miyazaki JI, Tashiro F, Aihara H, Nammo T, Li M, Yamagata K, Miyagawa JI, Matsuzawa Y, Kawabata Y, Fukuyama Y, Koga K, Mori W, Tanaka K, Matsuda T, Baba A. Overexpression of PACAP in transgenic mouse pancreatic beta-cells enhances insulin secretion and ameliorates streptozotocin-induced diabetes. Diabetes 2003; 52:1155-62. [PMID: 12716746 DOI: 10.2337/diabetes.52.5.1155] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP), a member of the vasoactive intestinal peptide/secretin/glucagon family, stimulates insulin secretion from islets in a glucose-dependent manner at femtomolar concentrations. To assess PACAP's pancreatic function in vivo, we generated transgenic mice overexpressing PACAP in the pancreas under the control of human insulin promoter. Northern blot and immunohistochemical analyses showed that PACAP is overexpressed in pancreatic islets, specifically in transgenic mice. Plasma glucose and glucagon levels during a glucose tolerance test were not different between PACAP transgenic mice and nontransgenic littermates. However, plasma insulin levels in transgenic mice were higher after glucose loading. Also, increases of streptozotocin-induced plasma glucose were attenuated in transgenic compared with nontransgenic mice. Notably, an increase in 5-bromo-2-deoxyuridine-positive beta-cells in the streptozotocin-treated transgenic mice was observed but without differences in the staining patterns by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling. Morphometric analysis revealed that total islet mass tends to increase in 12-month-old transgenic mice but showed no difference between 12-week-old transgenic and nontransgenic littermates. This is the first time that PACAP has been observed to play an important role in the proliferation of beta-cells.
Collapse
Affiliation(s)
- Kyohei Yamamoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Yuhara A, Ishii K, Nishio C, Abiru Y, Yamada M, Nawa H, Hatanaka H, Takei N. PACAP and NGF cooperatively enhance choline acetyltransferase activity in postnatal basal forebrain neurons by complementary induction of its different mRNA species. Biochem Biophys Res Commun 2003; 301:344-9. [PMID: 12565866 DOI: 10.1016/s0006-291x(02)03037-1] [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: 01/07/2023]
Abstract
Both nerve growth factor (NGF) and pituitary adenylate cyclase activating polypeptide (PACAP) have neurotrophic effects on basal forebrain cholinergic neurons. They promote differentiation, maturation, and survival of these cholinergic neurons in vivo and in vitro. Here we report on the cooperative effects of NGF and PACAP on postnatal, but not embryonic, cholinergic neurons cultured from rat basal forebrain. Combined treatment with NGF, brain-derived neurotrophic factor (BDNF), neurotrophin-4 (NT-4), and PACAP induced an additive increase in choline acetyltransferase (ChAT) activity. There were no cooperative effects on the number of cholinergic neurons, suggesting that ChAT mRNA expression had been induced in each cholinergic neuron. Further analysis revealed that NGF and PACAP led to complementary induction of different ChAT mRNA species, thus enhancing total ChAT mRNA expression. These results explain the cooperative neurotrophic action of NGF and PACAP on postnatal cholinergic neurons.
Collapse
Affiliation(s)
- Aki Yuhara
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, 565-0871, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Vaudry D, Chen Y, Ravni A, Hamelink C, Elkahloun AG, Eiden LE. Analysis of the PC12 cell transcriptome after differentiation with pituitary adenylate cyclase-activating polypeptide (PACAP). J Neurochem 2002; 83:1272-84. [PMID: 12472882 PMCID: PMC4186721 DOI: 10.1046/j.1471-4159.2002.01242.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) promotes neurite outgrowth and inhibits proliferation of rat pheochromocytoma (PC12) cells. Characterizing the PACAP-differentiated PC12 cell transcriptome should provide genetic insight into how these processes occur in these cells, and in neuronal precursors in vivo. For this purpose, RNA samples were collected from PC12 cells before or after a 6-h treatment with PACAP, from which a labeled cDNA was hybridized to a high-density cDNA array containing 15 365 genes. The genomic response to PACAP involves at least 73 genes. Among the genes differentially expressed in the presence of PACAP, 71% were up regulated, and 29% down regulated, 2-fold or more. Sixty-six percent of the messages affected by PACAP code for functionally categorized proteins, most not previously known to be regulated during PC12 cell differentiation. PACAP has been shown to induce PC12 cell neurite outgrowth through the mitogen-activated protein kinase kinase (MEK) pathway independently of protein kinase A (PKA). Therefore treatments were conducted in the absence or presence of the PKA inhibitor H89, or the MEK inhibitor U0126 in order to identify subsets of genes involved in specific aspects of PC12 cell differentiation. Co-treatment of PC12 cells with PACAP plus H89 revealed a cluster of five genes specifically regulated through the PKA pathway and co-treatment of the cells with PACAP and U0126 revealed a cluster of 13 messages specifically activated through the MEK pathway. Many of the known genes regulated by PACAP have been associated with neuritogenesis (i.e. villin 2 or annexin A2) or cell growth (i.e. growth arrest specific 1 or cyclin B2). Thus, some of the expressed sequence tags (ESTs) that exhibit the same regulation pattern (i.e. AU016391 or AW552690) may also be involved in the neuritogenic and anti-mitogenic effects of PACAP in PC12 cells. Among the 73 PACAP regulated genes, 10 are disqualified on pharmacological grounds as actors in PACAP-mediated neurite outgrowth or growth arrest, leaving 63 new PACAP-regulated genes implicated in neuronal differentiation. Thirteen of these are candidates for mediating ERK-dependent neurite outgrowth, and 47 are possibly involved in the ERK-independent growth arrest induced by PACAP.
Collapse
Affiliation(s)
- David Vaudry
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health
| | - Yun Chen
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health
| | - Aurélia Ravni
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health
| | - Carol Hamelink
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health
| | - Abdel G. Elkahloun
- Cancer Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, USA
| | - Lee E. Eiden
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health
| |
Collapse
|
49
|
Hashimoto H. [Physiological significance of pituitary adenylate cyclase-activating polypeptide (PACAP) in the nervous system]. YAKUGAKU ZASSHI 2002; 122:1109-21. [PMID: 12510388 DOI: 10.1248/yakushi.122.1109] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) has been conserved remarkably during evolution and is widely expressed in the nervous system across phyla. PACAP has an amino acid sequence homology of 68% with that of vasoactive intestinal polypeptide (VIP) and of 37% with that of secretin, indicating that PACAP is a member of the VIP/glucagon/secretin superfamily. PACAP exerts its actions via three heptahelical G-protein-linked receptors: one PACAP-specific (PAC1) receptor and two receptors (VPAC1 and VPAC2) shared with VIP. PACAP stimulates several different signaling cascades in neurons, leading to the activation of adenylate cyclase, phospholipase C, and mitogen-activated protein kinase and mobilization of calcium. Although PACAP and VIP have no apparent homology with calcitonin and parathyroid hormone (PTH), PAC1, VPAC, secretin, glucagon, glucagon-like peptide 1, growth hormone-releasing hormone, calcitonin, and PTH/PTH-related peptide receptors are related to each other and constitute a subfamily of the G-protein-coupled receptors. Distribution analysis of PACAP and its receptors and pharmacological studies have elucidated its pleiotropic effects in the central and peripheral nervous systems. However, the relevance of the pharmacological PACAP effects to the actual physiological activities of endogenous PACAP has not been addressed, because potent and selective low-molecular-weight PACAP antagonists have not yet been developed. To assess the function of PACAP in vivo, we have recently generated PAC1 receptor- and PACAP-targeted mice, and provided evidence that PACAP plays a previously uncharacterized role in the regulation of psychomotor behaviors. In this review, we focus on the physiological and or pathophysiological roles mediated by PACAP in the nervous system.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Cell Differentiation
- Gene Expression Regulation
- Ligands
- Mice
- Molecular Sequence Data
- Neurons/cytology
- Neuropeptides/chemistry
- Neuropeptides/genetics
- Neuropeptides/physiology
- Pituitary Adenylate Cyclase-Activating Polypeptide
- Psychomotor Performance
- RNA, Messenger/metabolism
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I
- Receptors, Pituitary Hormone/chemistry
- Receptors, Pituitary Hormone/genetics
- Receptors, Pituitary Hormone/physiology
- Receptors, Vasoactive Intestinal Peptide, Type II
- Receptors, Vasoactive Intestinal Polypeptide, Type I
- Signal Transduction
Collapse
Affiliation(s)
- Hitoshi Hashimoto
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
50
|
Sakai Y, Hashimoto H, Shintani N, Ichibori A, Tomimoto S, Tanaka K, Hirose M, Baba A. Involvement of intracellular Ca2+ elevation but not cyclic AMP in PACAP-induced p38 MAP kinase activation in PC12 cells. REGULATORY PEPTIDES 2002; 109:149-53. [PMID: 12409227 DOI: 10.1016/s0167-0115(02)00198-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
We have recently shown that in PC12 cells, pituitary adenylate cyclase-activating polypeptide (PACAP) and NGF synergistically stimulate PACAP mRNA expression primarily via a mechanism involving a p38 mitogen-activated protein kinase (MAPK)-dependent pathway. Here we have analyzed p38 MAPK activation by PACAP and the mechanism underlying this action of PACAP in PC12 cells. PACAP increased phosphorylation of p38 MAPK with a bell-shaped dose-response relationship and a maximal effect was obtained at 10(-8) M. PACAP (10(-8) M)-induced p38 MAPK phosphorylation was already evident at 2.5 min, maximal at 5 min, and rapidly declined thereafter. PACAP-induced p38 MAPK phosphorylation was potently inhibited by depletion of Ca(2+) stores with thapsigargin and partially inhibited by the phospholipase C inhibitor U-73122, L-type voltage-dependent calcium channel inhibitors nifedipine and nimodipine, and the Ca(2+) chelator EGTA, whereas the protein kinase C inhibitor calphostin C, the protein kinase A inhibitor H-89, the cAMP antagonist Rp-cAMP, and the nonselective cation channel blocker SKF96365 had no effect. These results indicate that PACAP activates p38 MAPK in PC12 cells through activation of a phospholipase C, mobilization of intracellular Ca(2+) stores, and Ca(2+) influx through voltage-dependent Ca(2+) channels, but not cyclic AMP-dependent mechanisms.
Collapse
Affiliation(s)
- Yoshiyuki Sakai
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | | | | | | | | | | | | | | |
Collapse
|