1
|
Tasma Z, Rees TA, Guo S, Tan S, O'Carroll SJ, Faull RLM, Curtis MA, Christensen SL, Hay DL, Walker CS. Pharmacology of PACAP and VIP receptors in the spinal cord highlights the importance of the PAC 1 receptor. Br J Pharmacol 2024; 181:2655-2675. [PMID: 38616050 DOI: 10.1111/bph.16376] [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] [Received: 12/06/2022] [Revised: 12/18/2023] [Accepted: 01/20/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND AND PURPOSE The spinal cord is a key structure involved in the transmission and modulation of pain. Pituitary adenylate cyclase-activating peptide (PACAP) and vasoactive intestinal peptide (VIP), are expressed in the spinal cord. These peptides activate G protein-coupled receptors (PAC1, VPAC1 and VPAC2) that could provide targets for the development of novel pain treatments. However, it is not clear which of these receptors are expressed within the spinal cord and how these receptors signal. EXPERIMENTAL APPROACH Dissociated rat spinal cord cultures were used to examine agonist and antagonist receptor pharmacology. Signalling profiles were determined for five signalling pathways. The expression of different PACAP and VIP receptors was then investigated in mouse, rat and human spinal cords using immunoblotting and immunofluorescence. KEY RESULTS PACAP, but not VIP, potently stimulated cAMP, IP1 accumulation and ERK and cAMP response element-binding protein (CREB) but not Akt phosphorylation in spinal cord cultures. Signalling was antagonised by M65 and PACAP6-38. PACAP-27 was more effectively antagonised than either PACAP-38 or VIP. The patterns of PAC1 and VPAC2 receptor-like immunoreactivity appeared to be distinct in the spinal cord. CONCLUSIONS AND IMPLICATIONS The pharmacological profile in the spinal cord suggested that a PAC1 receptor is the major functional receptor subtype present and thus likely mediates the nociceptive effects of the PACAP family of peptides in the spinal cord. However, the potential expression of both PAC1 and VPAC2 receptors in the spinal cord highlights that these receptors may play differential roles and are both possible therapeutic targets.
Collapse
MESH Headings
- Animals
- Spinal Cord/metabolism
- Spinal Cord/drug effects
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I/metabolism
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I/agonists
- Humans
- Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology
- Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism
- Vasoactive Intestinal Peptide/metabolism
- Vasoactive Intestinal Peptide/pharmacology
- Mice
- Rats
- Signal Transduction/drug effects
- Receptors, Vasoactive Intestinal Peptide/metabolism
- Receptors, Vasoactive Intestinal Peptide/antagonists & inhibitors
- Cells, Cultured
- Rats, Sprague-Dawley
- Male
- Mice, Inbred C57BL
- Cyclic AMP/metabolism
- Receptors, Vasoactive Intestinal Peptide, Type II/metabolism
- Receptors, Vasoactive Intestinal Peptide, Type II/agonists
Collapse
Affiliation(s)
- Zoe Tasma
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Tayla A Rees
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Song Guo
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Odontology, Panum Institute, Faculty of Health, University of Copenhagen, Copenhagen, Denmark
| | - Sheryl Tan
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, The University of Auckland, Auckland, New Zealand
| | - Simon J O'Carroll
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, The University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, The University of Auckland, Auckland, New Zealand
| | - Maurice A Curtis
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, The University of Auckland, Auckland, New Zealand
| | - Sarah L Christensen
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Debbie L Hay
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
- Department of Pharmacology and Toxicology, The University of Otago, Dunedin, New Zealand
| | - Christopher S Walker
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
2
|
Wnorowski A, Wójcik J, Maj M. Gene Expression Data Mining Reveals the Involvement of GPR55 and Its Endogenous Ligands in Immune Response, Cancer, and Differentiation. Int J Mol Sci 2021; 22:ijms222413328. [PMID: 34948125 PMCID: PMC8707311 DOI: 10.3390/ijms222413328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/05/2021] [Accepted: 12/08/2021] [Indexed: 12/04/2022] Open
Abstract
G protein-coupled receptor 55 (GPR55) is a recently deorphanized lipid- and peptide-sensing receptor. Its lipidic endogenous agonists belong to lysoglycerophospholipids, with lysophosphatidylinositol (LPI) being the most studied. Peptide agonists derive from fragmentation of pituitary adenylate cyclase-activating polypeptide (PACAP). Although GPR55 and its ligands were implicated in several physiological and pathological conditions, their biological function remains unclear. Thus, the aim of the study was to conduct a large-scale re-analysis of publicly available gene expression datasets to identify physiological and pathological conditions affecting the expression of GPR55 and the production of its ligands. The study revealed that regulation of GPR55 occurs predominantly in the context of immune activation pointing towards the role of the receptor in response to pathogens and in immune cell lineage determination. Additionally, it was revealed that there is almost no overlap between the experimental conditions affecting the expression of GPR55 and those modulating agonist production. The capacity to synthesize LPI was enhanced in various types of tumors, indicating that cancer cells can hijack the motility-related activity of GPR55 to increase aggressiveness. Conditions favoring accumulation of PACAP-derived peptides were different than those for LPI and were mainly related to differentiation. This indicates a different function of the two agonist classes and possibly the existence of a signaling bias.
Collapse
|
3
|
D’Amico AG, Maugeri G, Vanella L, Pittalà V, Reglodi D, D’Agata V. Multimodal Role of PACAP in Glioblastoma. Brain Sci 2021; 11:994. [PMID: 34439613 PMCID: PMC8391398 DOI: 10.3390/brainsci11080994] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the deadliest form of brain tumors. To date, the GBM therapeutical approach consists of surgery, radiation-therapy and chemotherapy combined with molecules improving cancer responsiveness to treatments. In this review, we will present a brief overview of the GBM classification and pathogenesis, as well as the therapeutic approach currently used. Then, we will focus on the modulatory role exerted by pituitary adenylate cyclase-activating peptide, known as PACAP, on GBM malignancy. Specifically, we will describe PACAP ability to interfere with GBM cell proliferation, as well as the tumoral microenvironment. Considering its anti-oncogenic role in GBM, synthesis of PACAP agonist molecules may open new perspectives for combined therapy to existing gold standard treatment.
Collapse
Affiliation(s)
- Agata Grazia D’Amico
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (A.G.D.); (L.V.); (V.P.)
| | - Grazia Maugeri
- Section of Anatomy, Histology and Movement Sciences, Department of Biomedical and Biotechnological Sciences, University of Catania, 95100 Catania, Italy;
| | - Luca Vanella
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (A.G.D.); (L.V.); (V.P.)
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (A.G.D.); (L.V.); (V.P.)
| | - Dora Reglodi
- MTA-PTE PACAP Research Group, Department of Anatomy, University of Pécs Medical School, 7624 Pécs, Hungary;
| | - Velia D’Agata
- Section of Anatomy, Histology and Movement Sciences, Department of Biomedical and Biotechnological Sciences, University of Catania, 95100 Catania, Italy;
| |
Collapse
|
4
|
Kambe Y, Yamauchi Y, Thanh Nguyen T, Thi Nguyen T, Ago Y, Shintani N, Hashimoto H, Yoshitake S, Yoshitake T, Kehr J, Kawamura N, Katsuura G, Kurihara T, Miyata A. The pivotal role of pituitary adenylate cyclase-activating polypeptide for lactate production and secretion in astrocytes during fear memory. Pharmacol Rep 2021; 73:1109-1121. [PMID: 33835466 DOI: 10.1007/s43440-021-00222-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/20/2021] [Accepted: 01/24/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Pituitary adenylate cyclase-activating polypeptide (PACAP) plays an essential role in the modulation of astrocyte functions. Although lactate secretion from astrocytes contributes to many forms of neuronal plasticity in the central nervous system, including fear learning and memory, the role of PACAP in lactate secretion from astrocytes is unclear. METHODS The amygdala and hippocampus of PACAP (+ / +) and PACAP (-/-) mice were acquired 1 h after memory acquisition and recall in the passive avoidance test. The concentration of glycogen and lactate in these regions was measured. The concentration of lactate in the hippocampus's extracellular fluid was also measured by microdialysis during memory acquisition or intracerebroventricular administration of PACAP. RESULTS We observed that memory acquisition caused a significant decrease in glycogen concentration and increased lactate concentration in the PACAP (+ / +) mice's hippocampus. However, memory acquisition did not increase in the lactate concentration in PACAP (-/-) mice's hippocampus. Further, memory retrieval evoked lactate production in the amygdala and the hippocampus of PACAP (+ / +) mice. Still, there was no significant increase in lactate concentration in the same regions of PACAP (-/-) mice. In vivo microdialysis in rats revealed that the hippocampus's extracellular lactate concentration increased after a single PACAP intracerebroventricular injection. Additionally, the hippocampus's extracellular lactate concentration increased with the memory acquisition in PACAP (+ / +) mice, but not in PACAP (-/-) mice. CONCLUSIONS PACAP may enhance lactate production and secretion in astrocytes during the acquisition and recall of fear memories.
Collapse
Affiliation(s)
- Yuki Kambe
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544, Japan
| | - Yu Yamauchi
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544, Japan
| | - Trung Thanh Nguyen
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544, Japan
| | - Thu Thi Nguyen
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544, Japan
| | - Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Norihito Shintani
- Laboratories of Molecular Neuropharmacology and Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University Medical School, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Hitoshi Hashimoto
- Laboratories of Molecular Neuropharmacology and Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University Medical School, Osaka University, Suita, Osaka, 565-0871, 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, Osaka, 565-0871, Japan.,Division of Bioscience, Institute for Datability Science, Osaka University, Suita, Osaka, 565-0871, Japan.,Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Shimako Yoshitake
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Takashi Yoshitake
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544, Japan.,Department of Physiology and Pharmacology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Jan Kehr
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Namiko Kawamura
- Department of Drug Discovery of Next-Generation GcMAF, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544, Japan
| | - Goro Katsuura
- Department of Drug Discovery of Next-Generation GcMAF, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544, Japan
| | - Takashi Kurihara
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544, Japan
| | - Atsuro Miyata
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544, Japan.
| |
Collapse
|
5
|
Moody TW, Jensen RT. Pituitary adenylate cyclase-activating polypeptide/vasoactive intestinal peptide [Part 1]: biology, pharmacology, and new insights into their cellular basis of action/signaling which are providing new therapeutic targets. Curr Opin Endocrinol Diabetes Obes 2021; 28:198-205. [PMID: 33449573 PMCID: PMC7957349 DOI: 10.1097/med.0000000000000617] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW To discuss recent advances of vasoactive intestinal peptide (VIP)/pituitary adenylate cyclase-activating polypeptide (PACAP) receptors in pharmacology, cell biology, and intracellular signaling in cancer. RECENT FINDINGS Recent studies provide new insights into the pharmacology, cell biology of the VIP/PACAP system and show they play important roles in a number of human cancers, as well as in tumor growth/differentiation and are providing an increased understanding of their signaling cascade that is suggesting new treatment targets/approaches. SUMMARY Recent insights from studies of VIP/PACAP and their receptors in both central nervous system disorders and inflammatory disorders suggest possible new treatment approaches. Elucidation of the exact roles of VIP/PACAP in these disorders and development of new therapeutic approaches involving these peptides have been limited by lack of specific pharmacological tools, and exact signaling mechanisms involved, mediating their effects. Reviewed here are recent insights from the elucidation of structural basis for VIP/PACAP receptor activation as well as the signaling cascades mediating their cellular effects (using results primarily from the study of their effects in cancer) that will likely lead to novel targets and treatment approaches in these diseases.
Collapse
Affiliation(s)
- Terry W Moody
- Department of Health and Human Services, National Cancer Institute, Center for Cancer Training
| | - Robert T Jensen
- National Institutes of Health, National Institute of Diabetes, Digestive and Kidney Diseases, Digestive Diseases Branch, Bethesda, Maryland 20892, USA
| |
Collapse
|
6
|
Moody TW, Ramos-Alvarez I, Jensen RT. Bombesin, endothelin, neurotensin and pituitary adenylate cyclase activating polypeptide cause tyrosine phosphorylation of receptor tyrosine kinases. Peptides 2021; 137:170480. [PMID: 33385499 DOI: 10.1016/j.peptides.2020.170480] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 12/12/2022]
Abstract
Numerous peptides including bombesin (BB), endothelin (ET), neurotensin (NTS) and pituitary adenylate cyclase-activating polypeptide (PACAP) are growth factors for lung cancer cells. The peptides bind to G protein-coupled receptors (GPCRs) resulting in elevated cAMP and/or phosphatidylinositol (PI) turnover. In contrast, growth factors such as epidermal growth factor (EGF) or neuregulin (NRG)-1 bind to receptor tyrosine kinases (RTKs) such as the EGFR or HER3, increasing tyrosine kinase activity, resulting in the phosphorylation of protein substrates such as PI3K or phospholipase (PL)C. Peptide GPCRs can transactivate numerous RTKs, especially members of the EGFR/HER family resulting in increased phosphorylation of ERK, leading to cellular proliferation or increased phosphorylation of AKT, leading to cellular survival. GRCR antagonists and tyrosine kinase inhibitors are useful agents to prevent RTK transactivation and inhibit proliferation of cancer cells.
Collapse
Affiliation(s)
- Terry W Moody
- Department of Health and Human Services, National Institutes of Health, National Cancer Institute, Center for Cancer Training, Bethesda, MD, 20892, USA.
| | - Irene Ramos-Alvarez
- National Institute of Diabetes, Digestive and Kidney Disease, Digestive Diseases Branch, 9000 Rockville Pike, Bethesda, MD, 20892 USA
| | - Robert T Jensen
- National Institute of Diabetes, Digestive and Kidney Disease, Digestive Diseases Branch, 9000 Rockville Pike, Bethesda, MD, 20892 USA
| |
Collapse
|
7
|
Effects of Pacap on Schwann Cells: Focus on Nerve Injury. Int J Mol Sci 2020; 21:ijms21218233. [PMID: 33153152 PMCID: PMC7663204 DOI: 10.3390/ijms21218233] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/23/2020] [Accepted: 11/02/2020] [Indexed: 12/27/2022] Open
Abstract
Schwann cells, the most abundant glial cells of the peripheral nervous system, represent the key players able to supply extracellular microenvironment for axonal regrowth and restoration of myelin sheaths on regenerating axons. Following nerve injury, Schwann cells respond adaptively to damage by acquiring a new phenotype. In particular, some of them localize in the distal stump to form the Bungner band, a regeneration track in the distal site of the injured nerve, whereas others produce cytokines involved in recruitment of macrophages infiltrating into the nerve damaged area for axonal and myelin debris clearance. Several neurotrophic factors, including pituitary adenylyl cyclase-activating peptide (PACAP), promote survival and axonal elongation of injured neurons. The present review summarizes the evidence existing in the literature demonstrating the autocrine and/or paracrine action exerted by PACAP to promote remyelination and ameliorate the peripheral nerve inflammatory response following nerve injury.
Collapse
|
8
|
Moody TW, Lee L, Jensen RT. The G Protein–Coupled Receptor PAC1 Regulates Transactivation of the Receptor Tyrosine Kinase HER3. J Mol Neurosci 2020; 71:1589-1597. [DOI: 10.1007/s12031-020-01711-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/14/2020] [Indexed: 11/30/2022]
|
9
|
Biran J, Gliksberg M, Shirat I, Swaminathan A, Levitas-Djerbi T, Appelbaum L, Levkowitz G. Splice-specific deficiency of the PTSD-associated gene PAC1 leads to a paradoxical age-dependent stress behavior. Sci Rep 2020; 10:9559. [PMID: 32533011 PMCID: PMC7292827 DOI: 10.1038/s41598-020-66447-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023] Open
Abstract
The pituitary adenylate cyclase-activating polypeptide receptor (PAC1, also known as ADCYAP1R1) is associated with post-traumatic stress disorder and modulation of stress response in general. Alternative splicing of PAC1 results in multiple gene products, which differ in their mode of signalling and tissue distribution. However, the roles of distinct splice variants in the regulation of stress behavior is poorly understood. Alternative splicing of a short exon, which is known as the "hop cassette", occurs during brain development and in response to stressful challenges. To examine the function of this variant, we generated a splice-specific zebrafish mutant lacking the hop cassette, which we designated 'hopless'. We show that hopless mutant larvae display increased anxiety-like behavior, including reduced dark exploration and impaired habituation to dark exposure. Conversely, adult hopless mutants displayed superior ability to rebound from an acute stressor, as they exhibited reduced anxiety-like responses to an ensuing novelty stress. We propose that the developmental loss of a specific PAC1 splice variant mimics prolonged mild stress exposure, which in the long term, predisposes the organism's stress response towards a resilient phenotype. Our study presents a unique genetic model demonstrating how early-life state of anxiety paradoxically correlates with reduced stress susceptibility in adulthood.
Collapse
Affiliation(s)
- Jakob Biran
- Department of Poultry and Aquaculture, Agricultural Research Organization, Rishon, Letziyon, 7528809, Israel.
| | - Michael Gliksberg
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot, 7610001, Israel
| | - Ido Shirat
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot, 7610001, Israel
| | - Amrutha Swaminathan
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot, 7610001, Israel
| | - Talia Levitas-Djerbi
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Lior Appelbaum
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Gil Levkowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot, 7610001, Israel.
| |
Collapse
|
10
|
Biran J, Gliksberg M, Shirat I, Swaminathan A, Levitas-Djerbi T, Appelbaum L, Levkowitz G. Splice-specific deficiency of the PTSD-associated gene PAC1 leads to a paradoxical age-dependent stress behavior. Sci Rep 2020. [PMID: 32533011 DOI: 10.1038/s41598-020-66447-2.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The pituitary adenylate cyclase-activating polypeptide receptor (PAC1, also known as ADCYAP1R1) is associated with post-traumatic stress disorder and modulation of stress response in general. Alternative splicing of PAC1 results in multiple gene products, which differ in their mode of signalling and tissue distribution. However, the roles of distinct splice variants in the regulation of stress behavior is poorly understood. Alternative splicing of a short exon, which is known as the "hop cassette", occurs during brain development and in response to stressful challenges. To examine the function of this variant, we generated a splice-specific zebrafish mutant lacking the hop cassette, which we designated 'hopless'. We show that hopless mutant larvae display increased anxiety-like behavior, including reduced dark exploration and impaired habituation to dark exposure. Conversely, adult hopless mutants displayed superior ability to rebound from an acute stressor, as they exhibited reduced anxiety-like responses to an ensuing novelty stress. We propose that the developmental loss of a specific PAC1 splice variant mimics prolonged mild stress exposure, which in the long term, predisposes the organism's stress response towards a resilient phenotype. Our study presents a unique genetic model demonstrating how early-life state of anxiety paradoxically correlates with reduced stress susceptibility in adulthood.
Collapse
Affiliation(s)
- Jakob Biran
- Department of Poultry and Aquaculture, Agricultural Research Organization, Rishon, Letziyon, 7528809, Israel.
| | - Michael Gliksberg
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot, 7610001, Israel
| | - Ido Shirat
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot, 7610001, Israel
| | - Amrutha Swaminathan
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot, 7610001, Israel
| | - Talia Levitas-Djerbi
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Lior Appelbaum
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Gil Levkowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot, 7610001, Israel.
| |
Collapse
|
11
|
Pleiotropic pituitary adenylate cyclase-activating polypeptide (PACAP): Novel insights into the role of PACAP in eating and drug intake. Brain Res 2019; 1729:146626. [PMID: 31883848 PMCID: PMC6953419 DOI: 10.1016/j.brainres.2019.146626] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/20/2019] [Accepted: 12/24/2019] [Indexed: 01/30/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) was discovered thirty years ago, but its role in eating and drug use disorders has only recently begun to be investigated. The present review develops the hypothesis that, although PACAP normally functions to tightly regulate intake, inhibiting it through negative feedback, this relationship can become dysregulated with the development of dependence, such that PACAP instead acts through positive feedback to promote excessive intake. We propose that repeated exposure to palatable food and drugs of abuse can alter the downstream responses of specific populations of neurons to stimulation by PACAP, leading to the perpetuation of the addiction cycle. Thus, this review will first describe published literature on homeostatic food intake, which shows that PACAP suppresses food intake, while its levels are themselves increased by overfeeding. Next, it will present literature on palatable food, cocaine, alcohol, and nicotine, which overall demonstrates that PACAP in specific limbic brain regions can promote their seeking and intake and itself is stimulated by their intake. Then, it will present literature on affective behavior, which shows that chronic stress increases levels of PACAP, which then promotes anxiety and depression, factors that can trigger substance seeking. Finally, the review will address mechanisms through which chronic substance exposure may dysregulate the PACAP system, proposing that it alters expression of PACAP receptor splice variants. While many questions remain to be addressed, the current evidence suggests that PACAP could be a viable medication target for the treatment of binge eating and drug and alcohol use disorders.
Collapse
|
12
|
Moody TW, Lee L, Iordanskaia T, Ramos-Alvarez I, Moreno P, Boudreau HE, Leto TL, Jensen RT. PAC1 regulates receptor tyrosine kinase transactivation in a reactive oxygen species-dependent manner. Peptides 2019; 120:170017. [PMID: 30273693 PMCID: PMC6438776 DOI: 10.1016/j.peptides.2018.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/10/2018] [Accepted: 09/19/2018] [Indexed: 11/22/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a growth factor for lung cancer cells. PACAP-27 or PACAP-38 binds with high affinity to non-small cell lung cancer (NSCLC) cells, causing elevated cytosolic Ca2+, increased proliferation and increased phosphorylation of extracellular regulated kinase (ERK) and the epidermal growth factor receptor (EGFR). The role of reactive oxygen species (ROS) was investigated in these processes. Addition of PACAP-38 to NCI-H838 or A549 cells increased the tyrosine phosphorylation of the EGFR, HER2 and ERK significantly by 4-, 3-, and 2-fold, respectively. The transactivation of the EGFR and HER2 was inhibited by gefitinib or lapatinib (tyrosine kinase inhibitors), PACAP (6-38) (PAC1 antagonist), N-acetylcysteine (NAC is an anti-oxidant) or dipheyleneiodonium (DPI is an inhibitor of Nox and Duox enzymes). PACAP-38 addition to NSCLC cells increased ROS which was inhibited by PACAP (6-38), NAC or DPI. Nox1, Nox2, Nox3, Nox4, Nox5, Duox1 and Duox2 mRNA was present in many NSCLC cell lines. PACAP-38 stimulated the growth of NSCLC cells whereas PACAP (6-38), gefitinib or DPI inhibited proliferation. The results show that ROS are essential for PAC1 to regulate EGFR and HER2 transactivation as well as proliferation of NSCLC cells.
Collapse
Affiliation(s)
- Terry W Moody
- Department of Health and Human Services, National Institutes of Health, National Cancer Institute, Center for Cancer Research, 9609 Medical Center Drive, Room 2W-340, Bethesda, MD, 20892, USA.
| | - Lingaku Lee
- National Institute of Diabetes, Digestive and Kidney Disease, Digestive Diseases Branch, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Tatiana Iordanskaia
- National Institute of Diabetes, Digestive and Kidney Disease, Digestive Diseases Branch, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Irene Ramos-Alvarez
- National Institute of Diabetes, Digestive and Kidney Disease, Digestive Diseases Branch, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Paola Moreno
- National Institute of Diabetes, Digestive and Kidney Disease, Digestive Diseases Branch, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Howard E Boudreau
- National Institute of Allergy and Infectious Diseases, Lab. Host Defenses, 12441 Parklawn Dr., Rockville, MD, 20852, USA
| | - Thomas L Leto
- National Institute of Allergy and Infectious Diseases, Lab. Host Defenses, 12441 Parklawn Dr., Rockville, MD, 20852, USA
| | - Robert T Jensen
- National Institute of Diabetes, Digestive and Kidney Disease, Digestive Diseases Branch, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| |
Collapse
|
13
|
Moody TW. Peptide receptors as cancer drug targets. Ann N Y Acad Sci 2019; 1455:141-148. [PMID: 31074514 DOI: 10.1111/nyas.14100] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 03/08/2019] [Accepted: 03/26/2019] [Indexed: 12/18/2022]
Abstract
Neuropeptides function as neuromodulators in the brain, whereby they are released in a paracrine manner and activate G protein-coupled receptors (GPCRs) in adjacent cells. Because neuropeptides are made in, and secreted from, cancer cells, then bind to cell surface receptors, they function in an autocrine manner. Bombesin (BB)-like peptides synthesized by neuroendocrine tumor small cell lung cancer (SCLC) bind to BB receptors (BBRs), causing phosphatidylinositol turnover and phosphorylation of extracellular signal-regulated kinase (ERK). Phosphorylated ERK enters the nucleus and alters gene expression of SCLC cells, stimulating growth. Vasoactive intestinal peptide (VIP) addition to SCLC cells increases their release rate of BB-like peptides via activation of VIP receptors (VIPR), leading to activation of adenylyl cyclase and subsequent elevation of cAMP. Protein kinase A is then stimulated, leading to phosphorylation of cyclic AMP response element binding protein (CREB), which alters gene expression and stimulates proliferation. The growth of SCLC is inhibited by BBR and VIPR antagonists. This review will focus on how GPCRs for VIP and BB are molecular targets for early detection and treatment of cancer.
Collapse
Affiliation(s)
- Terry W Moody
- Department of Health and Human Services, Center for Cancer Training, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
14
|
Moody TW, Ramos-Alvarez I, Jensen RT. Neuropeptide G Protein-Coupled Receptors as Oncotargets. Front Endocrinol (Lausanne) 2018; 9:345. [PMID: 30008698 PMCID: PMC6033971 DOI: 10.3389/fendo.2018.00345] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 06/11/2018] [Indexed: 12/15/2022] Open
Abstract
Neuropeptide G protein-coupled receptors (GPCRs) are overexpressed on numerous cancer cells. In a number of tumors, such as small cell lung cancer (SCLC), bombesin (BB) like peptides and neurotensin (NTS) function as autocrine growth factors whereby they are secreted from tumor cells, bind to cell surface receptors and stimulate growth. BB-drug conjugates and BB receptor antagonists inhibit the growth of a number of cancers. Vasoactive intestinal peptide (VIP) increases the secretion rate of BB-like peptide and NTS from SCLC leading to increased proliferation. In contrast, somatostatin (SST) inhibits the secretion of autocrine growth factors from neuroendocrine tumors (NETs) and decreases proliferation. SST analogs such as radiolabeled octreotide can be used to localize tumors, is therapeutic for certain cancer patients and has been approved for four different indications in the diagnosis/treatment of NETs. The review will focus on how BB, NTS, VIP, and SST receptors can facilitate the early detection and treatment of cancer.
Collapse
Affiliation(s)
- Terry W. Moody
- Department of Health and Human Services, National Cancer Institute, Center for Cancer Research, National Institute of Diabetes, Digestive, and Kidney Disease (NIDDK), Bethesda, MD, United States
| | - Irene Ramos-Alvarez
- Digestive Diseases Branch, National Institute of Diabetes, Digestive, and Kidney Disease (NIDDK), Bethesda, MD, United States
| | - Robert T. Jensen
- Digestive Diseases Branch, National Institute of Diabetes, Digestive, and Kidney Disease (NIDDK), Bethesda, MD, United States
| |
Collapse
|
15
|
Gupte RP, Kadunganattil S, Shepherd AJ, Merrill R, Planer W, Bruchas MR, Strack S, Mohapatra DP. Convergent phosphomodulation of the major neuronal dendritic potassium channel Kv4.2 by pituitary adenylate cyclase-activating polypeptide. Neuropharmacology 2015; 101:291-308. [PMID: 26456351 DOI: 10.1016/j.neuropharm.2015.10.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 09/29/2015] [Accepted: 10/03/2015] [Indexed: 12/30/2022]
Abstract
The endogenous neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) is secreted by both neuronal and non-neuronal cells in the brain and spinal cord, in response to pathological conditions such as stroke, seizures, chronic inflammatory and neuropathic pain. PACAP has been shown to exert various neuromodulatory and neuroprotective effects. However, direct influence of PACAP on the function of intrinsically excitable ion channels that are critical to both hyperexcitation as well as cell death, remain largely unexplored. The major dendritic K(+) channel Kv4.2 is a critical regulator of neuronal excitability, back-propagating action potentials in the dendrites, and modulation of synaptic inputs. We identified, cloned and characterized the downstream signaling originating from the activation of three PACAP receptor (PAC1) isoforms that are expressed in rodent hippocampal neurons that also exhibit abundant expression of Kv4.2 protein. Activation of PAC1 by PACAP leads to phosphorylation of Kv4.2 and downregulation of channel currents, which can be attenuated by inhibition of either PKA or ERK1/2 activity. Mechanistically, this dynamic downregulation of Kv4.2 function is a consequence of reduction in the density of surface channels, without any influence on the voltage-dependence of channel activation. Interestingly, PKA-induced effects on Kv4.2 were mediated by ERK1/2 phosphorylation of the channel at two critical residues, but not by direct channel phosphorylation by PKA, suggesting a convergent phosphomodulatory signaling cascade. Altogether, our findings suggest a novel GPCR-channel signaling crosstalk between PACAP/PAC1 and Kv4.2 channel in a manner that could lead to neuronal hyperexcitability.
Collapse
Affiliation(s)
- Raeesa P Gupte
- Department of Pharmacology, The University of Iowa Roy J. and Lucile A. Carver College of Medicine, Iowa City, IA 52242, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Suraj Kadunganattil
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew J Shepherd
- Department of Pharmacology, The University of Iowa Roy J. and Lucile A. Carver College of Medicine, Iowa City, IA 52242, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ronald Merrill
- Department of Pharmacology, The University of Iowa Roy J. and Lucile A. Carver College of Medicine, Iowa City, IA 52242, USA
| | - William Planer
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael R Bruchas
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Stefan Strack
- Department of Pharmacology, The University of Iowa Roy J. and Lucile A. Carver College of Medicine, Iowa City, IA 52242, USA
| | - Durga P Mohapatra
- Department of Pharmacology, The University of Iowa Roy J. and Lucile A. Carver College of Medicine, Iowa City, IA 52242, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
16
|
Pituitary adenylate cyclase activating polypeptide (PACAP) signalling exerts chondrogenesis promoting and protecting effects: implication of calcineurin as a downstream target. PLoS One 2014; 9:e91541. [PMID: 24643018 PMCID: PMC3958376 DOI: 10.1371/journal.pone.0091541] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 02/13/2014] [Indexed: 01/20/2023] Open
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is an important neurotrophic factor influencing differentiation of neuronal elements and exerting protecting role during traumatic injuries or inflammatory processes of the central nervous system. Although increasing evidence is available on its presence and protecting function in various peripheral tissues, little is known about the role of PACAP in formation of skeletal components. To this end, we aimed to map elements of PACAP signalling in developing cartilage under physiological conditions and during oxidative stress. mRNAs of PACAP and its receptors (PAC1,VPAC1, VPAC2) were detectable during differentiation of chicken limb bud-derived chondrogenic cells in micromass cell cultures. Expression of PAC1 protein showed a peak on days of final commitment of chondrogenic cells. Administration of either the PAC1 receptor agonist PACAP 1-38, or PACAP 6-38 that is generally used as a PAC1 antagonist, augmented cartilage formation, stimulated cell proliferation and enhanced PAC1 and Sox9 protein expression. Both variants of PACAP elevated the protein expression and activity of the Ca-calmodulin dependent Ser/Thr protein phosphatase calcineurin. Application of PACAPs failed to rescue cartilage formation when the activity of calcineurin was pharmacologically inhibited with cyclosporine A. Moreover, exogenous PACAPs prevented diminishing of cartilage formation and decrease of calcineurin activity during oxidative stress. As an unexpected phenomenon, PACAP 6-38 elicited similar effects to those of PACAP 1-38, although to a different extent. On the basis of the above results, we propose calcineurin as a downstream target of PACAP signalling in differentiating chondrocytes either in normal or pathophysiological conditions. Our observations imply the therapeutical perspective that PACAP can be applied as a natural agent that may have protecting effect during joint inflammation and/or may promote cartilage regeneration during degenerative diseases of articular cartilage.
Collapse
|
17
|
Blechman J, Levkowitz G. Alternative Splicing of the Pituitary Adenylate Cyclase-Activating Polypeptide Receptor PAC1: Mechanisms of Fine Tuning of Brain Activity. Front Endocrinol (Lausanne) 2013; 4:55. [PMID: 23734144 PMCID: PMC3659299 DOI: 10.3389/fendo.2013.00055] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Accepted: 04/24/2013] [Indexed: 12/11/2022] Open
Abstract
Alternative splicing of the precursor mRNA encoding for the neuropeptide receptor PAC1/ADCYAP1R1 generates multiple protein products that exhibit pleiotropic activities. Recent studies in mammals and zebrafish have implicated some of these splice isoforms in control of both cellular and body homeostasis. Here, we review the regulation of PAC1 splice variants and their underlying signal transduction and physiological processes in the nervous system.
Collapse
Affiliation(s)
- Janna Blechman
- Department of Molecular Cell Biology, Weizmann Institute of ScienceRehovot, Israel
| | - Gil Levkowitz
- Department of Molecular Cell Biology, Weizmann Institute of ScienceRehovot, Israel
- *Correspondence: Gil Levkowitz, Department of Molecular Cell Biology, Weizmann Institute of Science, P. O. Box 26, Rehovot 76100, Israel. e-mail:
| |
Collapse
|
18
|
Furness SGB, Wootten D, Christopoulos A, Sexton PM. Consequences of splice variation on Secretin family G protein-coupled receptor function. Br J Pharmacol 2012; 166:98-109. [PMID: 21718310 DOI: 10.1111/j.1476-5381.2011.01571.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The Secretin family of GPCRs are endocrine peptide hormone receptors that share a common genomic organization and are the subject of a wide variety of alternative splicing. All GPCRs contain a central seven transmembrane domain responsible for transducing signals from the outside of the cell as well as extracellular amino and intracellular carboxyl termini. Members of the Secretin receptor family have a relatively large N-terminus and a variety of lines of evidence support a common mode of ligand binding and a common ligand binding fold. These receptors are best characterized as coupling to intracellular signalling pathways via G(αs) and G(αq) but are also reported to couple to a multitude of other signalling pathways. The intracellular loops are implicated in regulating the interaction between the receptor and heterotrimeric G protein complexes. Alternative splicing of exons encoding both the extracellular N-terminal domain as well as the extracellular loops of some family members has been reported and as expected these splice variants display altered ligand affinity as well as differential activation by endogenous ligands. Various forms of alternative splicing have also been reported to alter intracellular loops 1 and 3 as well as the C-terminus and as one might expect these display differences in signalling bias towards downstream effectors. These diverse pharmacologies require that the physiological role of these splice variants be addressed but should provide unique opportunities for drug design and development.
Collapse
|
19
|
Baun M, Pedersen MHF, Olesen J, Jansen-Olesen I. Dural mast cell degranulation is a putative mechanism for headache induced by PACAP-38. Cephalalgia 2012; 32:337-45. [PMID: 22421901 DOI: 10.1177/0333102412439354] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Pituitary adenylate cyclase activating peptide-38 (PACAP-38) has been shown to induce migraine in migraineurs, whereas the related peptide vasoactive intestinal peptide (VIP) does not. In the present study we examine the hypothesis that PACAP-38 and its truncated version PACAP-27 but not VIP cause degranulation of mast cells in peritoneum and in dura mater. METHODS The degranulatory effects of PACAP-38, PACAP-27 and VIP were investigated by measuring the amount of N-acetyl-β-hexosaminidase released from isolated peritoneal mast cells and from dura mater attached to the skull of the rat in vitro. In peritoneal mast cells N-truncated fragments of PACAP-38 (PACAP(6-38), PACAP(16-38) and PACAP(28-38)) were also studied. To investigate transduction pathways involved in mast cell degranulation induced by PACAP-38, PACAP-27 and VIP, the phospholipase C inhibitor U-73122 and the adenylate cyclase inhibitor SQ 22536 were used. RESULTS The peptides induced degranulation of isolated peritoneal mast cells of the rat with the following order of potency: PACAP-38 = PACAP(6-38) = PACAP(16-38) » PACAP-27 = VIP = PACAP(28-38). In the dura mater we found that 10(-5) M PACAP-38 was significantly more potent in inducing mast cell degranulation than the same concentration of PACAP-27 or VIP. Inhibition of intracellular mechanisms demonstrated that PACAP-38-induced degranulation is mediated by the phospholipase C pathway. Selective blockade of the PAC(1) receptor did not attenuate degranulation. CONCLUSION These findings correlate with clinical studies and support the hypothesis that mast cell degranulation is involved in PACAP-induced migraine. PACAP-38 has a much stronger degranulatory effect on rat peritoneal and dural mast cells than VIP and PACAP-27. The difference in potency between PACAP-38- and PACAP-27/VIP-induced peritoneal mast cell degranulation is probably not related to the PAC(1) receptor but is caused by a difference in efficacy on phospholipase C.
Collapse
Affiliation(s)
- Michael Baun
- Department of Neurology, Glostrup Hospital, University of Copenhagen, Glostrup, Denmark
| | | | | | | |
Collapse
|
20
|
Moody TW, Osefo N, Nuche-Berenguer B, Ridnour L, Wink D, Jensen RT. Pituitary adenylate cyclase-activating polypeptide causes tyrosine phosphorylation of the epidermal growth factor receptor in lung cancer cells. J Pharmacol Exp Ther 2012; 341:873-881. [PMID: 22389426 PMCID: PMC3362890 DOI: 10.1124/jpet.111.190033] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 02/28/2012] [Indexed: 11/22/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is an autocrine growth factor for some lung cancer cells. The activated PACAP receptor (PAC1) causes phosphatidylinositol turnover, elevates cAMP, and increases the proliferation of lung cancer cells. PAC1 and epidermal growth factor receptor (EGFR) are present in non-small-cell lung cancer (NSCLC) cells, and the growth of NSCLC cells is inhibited by the PAC1 antagonist PACAP(6-38) and the EGFR tyrosine kinase inhibitor gefitinib. Here, the ability of PACAP to transactivate the EGFR was investigated. Western blot analysis indicated that the addition of PACAP but not the structurally related vasoactive intestinal peptide increased EGFR tyrosine phosphorylation in NCI-H838 or H345 cells. PACAP-27, in a concentration-dependent manner, increased EGFR transactivation 4-fold 2 min after addition to NCI-H838 cells. The ability of 100 nM PACAP-27 to increase EGFR or extracellular signal-regulated kinase tyrosine phosphorylation in NCI-H838 cells was inhibited by PACAP(6-38), gefitinib, 4-amino-5-(4-chlorophenyl)-7-(dimethylethyl)pyrazolo[3,4-d]pyrimidine (PP2; Src inhibitor), (R)-N4-hydroxy-N1-[(S)-2-(1H-indol-3-yl)-1-methylcarbamoyl-ethyl]-2-isobutyl-succinamide (GM6001; matrix metalloprotease inhibitor), or antibody to transforming growth factor α (TGFα). By enzyme-linked immunosorbent assay, PACAP addition to NCI-H838 cells increased TGFα secretion into conditioned media. EGFR transactivation caused by the addition of PACAP to NCI-H838 cells was inhibited by N-acetyl-cysteine (antioxidant), tiron (superoxide scavenger), diphenylene iodonium (NADPH oxidase inhibitor), or 1-[6-[[(17β)-3-methoxyestra-1,3,5(10)-trien-17-yl]amino]hexyl]-1H-pyrrole-2,5-dione (U73122; phospholipase C inhibitor), but not N-[2-[[3-(4-bromophenyl)-2-propenyl]amino]ethyl]-5-isoquinolinesulfonamide (H89; protein kinase A inhibitor). PACAP addition to NCI-H838 cells significantly increased reactive oxygen species, and the increase was inhibited by tiron. The results indicate that PACAP causes transactivation of the EGFR in NSCLC cells in an oxygen-dependent manner that involves phospholipase C but not protein kinase A.
Collapse
Affiliation(s)
- Terry W Moody
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 31, Room 4A48, 31 Center Drive, Bethesda, MD 20892, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Resch JM, Boisvert JP, Hourigan AE, Mueller CR, Yi SS, Choi S. Stimulation of the hypothalamic ventromedial nuclei by pituitary adenylate cyclase-activating polypeptide induces hypophagia and thermogenesis. Am J Physiol Regul Integr Comp Physiol 2011; 301:R1625-34. [PMID: 21957159 DOI: 10.1152/ajpregu.00334.2011] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Numerous studies have demonstrated that the hypothalamic ventromedial nuclei (VMN) regulate energy homeostasis by integrating and utilizing behavioral and metabolic mechanisms. The VMN heavily express pituitary adenylate cyclase-activating polypeptide (PACAP) type I receptors (PAC1R). Despite the receptor distribution, most PACAP experiments investigating affects on feeding have focused on intracerebroventricular administration or global knockout mice. To identify the specific contribution of PACAP signaling in the VMN, we injected PACAP directly into the VMN and measured feeding behavior and indices of energy expenditure. Following an acute injection of PACAP, nocturnal food intake was significantly reduced for 6 h after injections without evidence of malaise. In addition, PACAP-induced suppression of feeding also occurred following an overnight fast and could be blocked by a specific PAC1R antagonist. Metabolically, VMN-specific injections of PACAP significantly increased both core body temperature and spontaneous locomotor activity with a concurrent increase in brown adipose uncoupling protein 1 mRNA expression. To determine which signaling pathways were responsive to PACAP administration into the VMN, we measured mRNA expression of well-characterized hypothalamic neuropeptide regulators of feeding. One hour after PACAP administration, expression of pro-opiomelanocortin mRNA was significantly increased in the arcuate nuclei (ARC), with no changes in neuropeptide Y and agouti-related polypeptide mRNA levels. This suggests that PAC1R expressing VMN neurons projecting to pro-opiomelanocortin neurons contribute to hypophagia by involving melanocortin signaling. While the VMN also abundantly express PACAP protein, the present study demonstrates that PACAP input to the VMN can influence the control of energy homeostasis.
Collapse
Affiliation(s)
- Jon M Resch
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | | | | | | | | | | |
Collapse
|
22
|
Gasperini L, Piubelli C, Carboni L. Proteomics of rat hypothalamus, hippocampus and pre-frontal/frontal cortex after central administration of the neuropeptide PACAP. Mol Biol Rep 2011; 39:2921-35. [PMID: 21687973 DOI: 10.1007/s11033-011-1054-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2010] [Accepted: 06/08/2011] [Indexed: 11/25/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide that exerts pleiotropic functions, acting as a hypophysiotropic factor, a neurotrophic and a neuroprotective agent. The molecular pathways activated by PACAP to exert its physiological roles in brain are incompletely understood. In this study, adrenocorticotropic hormone (ACTH), prolactin, luteinising hormone (LH), follicle-stimulating hormone (FSH), thyroid-stimulating hormone (TSH), brain-derived neurotrophic factor and corticosterone blood levels were determined before and 20, 40, 60, and 120 min after PACAP intracerebroventricular administration. PACAP treatment increased ACTH, corticosterone, LH and FSH blood concentrations, while it decreased TSH levels. A proteomics investigation was carried out in hypothalamus, hippocampus and pre-frontal/frontal cortex (P/FC) using 2-dimensional gel electrophoresis at 120 min, the end-point suggested by studies on PACAP hypophysiotropic activities. Spots showing statistically significant alterations after PACAP treatment were identified by Matrix-assisted laser desorption/ionization-Time of flight mass spectrometry. Identified proteins were consistent with PACAP involvement in different molecular processes in brain. Altered expression levels were observed for proteins involved in cytoskeleton modulation and synaptic plasticity: actin in the hypothalamus; stathmin, dynamin, profilin and cofilin in hippocampus; synapsin in P/FC. Proteins involved in cellular differentiation were also modulated: glutathione-S-transferase α and peroxiredoxin in hippocampus; nucleoside diphosphate kinase in P/FC. Alterations were detected in proteins involved in neuroprotection, neurodegeneration and apoptosis: ubiquitin carboxyl-terminal hydrolase isozyme L1 and heat shock protein 90-β in hypothalamus; α-synuclein in hippocampus; glyceraldehyde-3-phosphate dehydrogenase and prohibitin in P/FC. This proteomics study identified new proteins involved in molecular mechanisms mediating PACAP functions in the central nervous system.
Collapse
Affiliation(s)
- Lisa Gasperini
- Neurosciences CEDD, GlaxoSmithKline Medicines Research Centre, Via A Fleming 4, 37135 Verona, Italy
| | | | | |
Collapse
|
23
|
Moody TW, Ito T, Osefo N, Jensen RT. VIP and PACAP: recent insights into their functions/roles in physiology and disease from molecular and genetic studies. Curr Opin Endocrinol Diabetes Obes 2011; 18:61-67. [PMID: 21157320 PMCID: PMC3075877 DOI: 10.1097/med.0b013e328342568a] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) as well as the three classes of G-protein-coupled receptors mediating their effects, are widely distributed in the central nervous system (CNS) and peripheral tissues. These peptides are reported to have many effects in different tissues, which are physiological or pharmacological, and which receptor mediates which effect, has been difficult to determine, primarily due to lack of potent, stable, selective agonists/antagonists. Recently the use of animals with targeted knockout of the peptide or a specific receptor has provided important insights into their role in normal physiology and disease states. RECENT FINDINGS During the review period, considerable progress and insights has occurred in the understanding of the role of VIP/PACAP as well as their receptors in a number of different disorders/areas. Particularly, insights into their roles in energy metabolism, glucose regulation, various gastrointestinal processes including gastrointestinal inflammatory conditions and motility and their role in the CNS as well as CNS diseases has greatly expanded. SUMMARY PACAP/VIP as well as their three classes of receptors are important in many physiological/pathophysiological processes, some of which are identified in these studies using knockout animals. These studies may lead to new novel treatment approaches. Particularly important are their roles in glucose metabolism and on islets leading to possible novel approaches in diabetes; their novel anti-inflammatory, cytoprotective effects, their CNS neuroprotective effects, and their possible roles in diseases such as schizophrenia and chronic depression.
Collapse
Affiliation(s)
- Terry W. Moody
- Department of Health and Human Services, National Cancer Institute Office of the Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Tetsuhide Ito
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Nuramy Osefo
- Department of Health and Human Services, National Cancer Institute Office of the Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Robert T. Jensen
- Digestive Diseases Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
24
|
Hu E, Demmou L, Cauli B, Gallopin T, Geoffroy H, Harris-Warrick RM, Paupardin-Tritsch D, Lambolez B, Vincent P, Hepp R. VIP, CRF, and PACAP act at distinct receptors to elicit different cAMP/PKA dynamics in the neocortex. ACTA ACUST UNITED AC 2010; 21:708-18. [PMID: 20699230 DOI: 10.1093/cercor/bhq143] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The functional significance of diverse neuropeptide coexpression and convergence onto common second messenger pathways remains unclear. To address this question, we characterized responses to corticotropin-releasing factor (CRF), pituitary adenylate cyclase-activating peptide (PACAP), and vasoactive intestinal peptide (VIP) in rat neocortical slices using optical recordings of cyclic adenosine monophosphate (cAMP) and protein kinase A (PKA) sensors, patch-clamp, and single-cell reverse transcription-polymerase chain reaction. Responses of pyramidal neurons to the 3 neuropeptides markedly differed in time-course and amplitude. Effects of these neuropeptides on the PKA-sensitive slow afterhyperpolarization current were consistent with those observed with cAMP/PKA sensors. CRF-1 receptors, primarily expressed in pyramidal cells, reportedly mediate the neocortical effects of CRF. PACAP and VIP activated distinct PAC1 and VPAC1 receptors, respectively. Indeed, a selective VPAC1 antagonist prevented VIP responses but had a minor effect on PACAP responses, which were mimicked by a specific PAC1 agonist. While PAC1 and VPAC1 were coexpressed in pyramidal cells, PAC1 expression was also frequently detected in interneurons, suggesting that PACAP has widespread effects on the neuronal network. Our results suggest that VIP and CRF, originating from interneurons, and PACAP, expressed mainly by pyramidal cells, finely tune the excitability and gene expression in the neocortical network via distinct cAMP/PKA-mediated effects.
Collapse
Affiliation(s)
- Emilie Hu
- Neurobiologie des Processus Adaptatifs, CNRS UMR7102, Université Pierre et Marie Curie-P6, Paris, F75005-France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Yu R, Li J, Wang J, Liu X, Huang L, Ding Y, Chen J. The functional recombinant first extracellular (EC1) domain of PACAP receptor PAC1 normal form (PAC1-EC1(N)) recognizes selective ligands and stimulates the proliferation of PAC1-CHO cells. Neurosci Lett 2010; 480:73-7. [DOI: 10.1016/j.neulet.2010.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Revised: 05/26/2010] [Accepted: 06/02/2010] [Indexed: 11/16/2022]
|
26
|
PACAP protects against TNFα-induced cell death in olfactory epithelium and olfactory placodal cell lines. Mol Cell Neurosci 2010; 45:345-54. [PMID: 20654718 DOI: 10.1016/j.mcn.2010.07.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 07/09/2010] [Accepted: 07/13/2010] [Indexed: 11/20/2022] Open
Abstract
In mouse olfactory epithelium (OE), pituitary adenylate cyclase-activating peptide (PACAP) protects against axotomy-induced apoptosis. We used mouse OE to determine whether PACAP protects neurons during exposure to the inflammatory cytokine TNFα. Live slices of neonatal mouse OE were treated with 40 ng/ml TNFα ± 40nM PACAP for 6h and dying cells were live-labeled with 0.5% propidium iodide. TNFα significantly increased the percentage of dying cells while co-incubation with PACAP prevented cell death. PACAP also prevented TNFα-mediated cell death in the olfactory placodal (OP) cell lines, OP6 and OP27. Although OP cell lines express all three PACAP receptors (PAC1, VPAC1,VPAC2), PACAP's protection of these cells from TNFα was mimicked by the specific PAC1 receptor agonist maxadilan and abolished by the PAC1 antagonist PACAP6-38. Treatment of OP cell lines with blockers or activators of the PLC and AC/MAPKK pathways revealed that PACAP-mediated protection from TNFα involved both pathways. PACAP may therefore function through PAC1 receptors to protect neurons from cell death during inflammatory cytokine release in vivo as would occur upon viral infection or allergic rhinitis-associated injury.
Collapse
|
27
|
Yu R, Wang J, Li J, Wang Y, Zhang H, Chen J, Huang L, Liu X. A novel cyclopeptide from the cyclization of PACAP(1-5) with potent activity towards PAC1 attenuates STZ-induced diabetes. Peptides 2010; 31:1062-7. [PMID: 20307608 DOI: 10.1016/j.peptides.2010.03.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2010] [Revised: 03/06/2010] [Accepted: 03/08/2010] [Indexed: 11/23/2022]
Abstract
The N-terminal deletion of pituitary adenylate cyclase-activating polypeptide (PACAP)(1-5) generates its own antagonist. The cyclopeptide C*HSDGIC*, which results from the cyclization of PACAP(1-5) with disulfide, was designed and synthesized. CHO cells expressing a PAC1 N/R splice variant (PAC1-CHO) were used to detect the potent activation of PAC1 by C*HSDGIC*. In vitro cell assays showed that C*HSDGIC* stimulated cAMP production and increased the viability of PAC1-CHO cells at micromolar concentrations, about 1000 fold that of PACAP. PACAP(6-38) blocked the effects of PACAP on the proliferation of PAC1-CHO cells but did not interfere with the effects of C*HSDGIC*, suggesting that the activation of PAC1 by C*HSDGIC* was independent of the binding of PAC1 to the C-terminus of PACAP. In experiments in vivo, 10mumol/kg C*HSDGIC* decreased the plasma glucose level, increased the plasma insulin level and improved glucose tolerance significantly (P<0.01) when co-injected with STZ for 5 days. The results of these in vitro and in vivo studies of the biological characteristics of C*HSDGIC* reveal that it is a potent activator of PAC1.
Collapse
Affiliation(s)
- Rongjie Yu
- Engineering Institute of Jinan University, Jinan University, Guangzhou 510630, PR China. rongjie
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Ushiyama M, Ikeda R, Yoshida M, Mori K, Kangawa K, Sugawara H, Inoue K, Yamada K, Miyata A. Alternative splicing of the pituitary adenylate cyclase-activating polypetide (PACAP) receptor contributes to function of PACAP-27. J Mol Neurosci 2010; 42:341-8. [PMID: 20473586 DOI: 10.1007/s12031-010-9385-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Accepted: 04/23/2010] [Indexed: 11/25/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP)-27 and PACAP-38 are neuropeptides performing a variety of physiological functions. The PACAP-specific receptor PAC1 has several variants that result mainly from alternative splicing in the mRNA region encoding the first extracellular (EC1) domain and the third intracellular cytoplasmic (IC3) loop. To characterize the molecular forms of alternative splicing variants of PAC1, we examined the binding affinity and activation of two major second messenger pathways (cAMP production and changes in [Ca(2+)]( i )) by PACAP-27. Activation of cAMP was influenced by the variant in both of the EC1 domain and IC3 loops. In the N form in the EC1 domain, the suppressive effect of the HOP1 form in the IC3 loop was enhanced. Regarding the intracellular calcium mobilization assay, the rank order of the potency of PACAP-27 for the different PAC1 isoforms was S/HOP1>>N/R~S/R>>N/HOP1. In particular, PACAP-27 exhibited remarkable potency of calcium mobilization in the S/HOP1-expressing cells at sub-picomolar concentrations even though the affinities of PACAP-27 to the four PAC1 isoforms were not significantly different. This suggests the specific functions of PACAP-27 due to PACAP-27 preferring PAC1 activation, and leads in clarification of the pleiotoropic function of PACAP.
Collapse
Affiliation(s)
- Mina Ushiyama
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Sakuragaoka, Kagoshima, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Giunta S, Castorina A, Adorno A, Mazzone V, Carnazza ML, D'Agata V. PACAP and VIP affect NF1 expression in rat malignant peripheral nerve sheath tumor (MPNST) cells. Neuropeptides 2010; 44:45-51. [PMID: 19919880 DOI: 10.1016/j.npep.2009.10.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Revised: 09/14/2009] [Accepted: 10/15/2009] [Indexed: 10/20/2022]
Abstract
In our previous study we have identified PACAP, VIP and their receptors in rat malignant peripheral nerve sheath tumor (MPNST) cells, thus showing anti-apoptotic roles. Recently it has been shown that the tumor suppressor neurofibromin, encoded by the Neurofibromatosis type I (NF1) gene, promotes MPNST cells sensitivity to apoptosis after serum withdrawal. In the present study we investigated whether PACAP or VIP negatively regulate NF1 expression under normal or serum-dependent pro-apoptotic culture conditions. Results indicated that serum itself significantly influenced gene and protein levels. In fact, the low NF1 levels of cells cultured in normal serum-containing medium were remarkably increased in cells switched to low- or no-serum after 24h and 48 h. Treatment with 100 nM PACAP or VIP did not affect NF1 expression when using normal amounts of serum, whereas it significantly inhibited transcript and protein levels both in low- or no-serum cultured cells. In particular, PACAP reduced NF1 levels already after 24h in low-serum cultured cells, while VIP showed a similar effect only after serum deprivation. However, both PACAP and VIP downregulated gene and protein levels within 48 h either in low-dose and serum-starved cells. Results were confirmed by fluorescence microscopy, showing that 100 nM PACAP or VIP attenuated neurofibromin cytoplasmic localization only in low- or no-serum cultured cells. The present study provides a comprehensive analysis of both neuropeptides effect on NF1 expression in normal, low- or serum-starved MPNST cells, ameliorating the hypothesis that resistance to apoptosis in serum-deprived cells might be correlated to PACAP-/VIP-induced NF1 inhibition.
Collapse
Affiliation(s)
- Salvatore Giunta
- Department of Anatomy, Diagnostic Pathology, Legal Medicine, Hygiene and Public Health, University of Catania, Catania, Italy
| | | | | | | | | | | |
Collapse
|
30
|
Hoover DB, Tompkins JD, Parsons RL. Differential activation of guinea pig intrinsic cardiac neurons by the PAC1 agonists maxadilan and pituitary adenylate cyclase-activating polypeptide 27 (PACAP27). J Pharmacol Exp Ther 2009; 331:197-203. [PMID: 19602551 PMCID: PMC2766232 DOI: 10.1124/jpet.109.155747] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Accepted: 07/10/2009] [Indexed: 11/22/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) evokes tachycardia followed by a larger cholinergic bradycardia in isolated guinea pig hearts. We used the selective PAC1 receptor agonist maxadilan and vasoactive intestinal polypeptide (VIP) to test the hypothesis that PACAP27-evoked tachycardia and bradycardia are mediated by VPAC and PAC1 receptors, respectively. Chronotropic actions of these peptides were evaluated in isolated perfused hearts. Direct neuronal actions were determined by intracellular voltage recordings from cholinergic neurons in atrial ganglion whole mounts. Administration of 1 nmol of PACAP27 to isolated hearts evoked typical biphasic rate responses, whereas 1 nmol of maxadilan caused only a minor rate decrease. Desensitization with VIP eliminated the positive chronotropic effect of PACAP27 selectively. Local application of PACAP27 to cardiac neurons frequently evoked slow depolarization and caused prolonged increase of neuronal excitability. Maxadilan rarely affected membrane potential but consistently increased excitability. VIP had no effect on excitability and evoked depolarization in only a few neurons. Because maxadilan increased neuronal excitability but did not trigger action potentials as PACAP often does, we evaluated the interaction of maxadilan with substance P (SP) in isolated hearts. SP depolarizes cardiac neurons more consistently than PACAP, often triggers neuronal action potentials, and causes bradycardia but does not increase neuronal excitability. Maxadilan had a persistent effect to augment negative chronotropic responses to SP. These findings support our hypothesis that PACAP evokes tachycardia and bradycardia through VPAC and PAC1 receptors, respectively. They also suggest that maxadilan and PACAP27 differ in activating PAC1 receptors on cardiac neurons and/or stimulating downstream signaling mechanisms.
Collapse
Affiliation(s)
- Donald B Hoover
- Department of Pharmacology, East Tennessee State University, Johnson City, 37614, USA.
| | | | | |
Collapse
|
31
|
Gericke MT, Kosacka J, Koch D, Nowicki M, Schröder T, Ricken AM, Nieber K, Spanel-Borowski K. Receptors for NPY and PACAP differ in expression and activity during adipogenesis in the murine 3T3-L1 fibroblast cell line. Br J Pharmacol 2009; 157:620-32. [PMID: 19422400 DOI: 10.1111/j.1476-5381.2009.00164.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Neuropeptides are involved in the regulation of food intake in the central nervous system, but they might also act on peripheral fat tissue via neuropeptide receptors. EXPERIMENTAL APPROACH We investigated the receptor expression and activity of pituitary adenylate cyclase-activating polypeptide (PACAP) and of neuropeptide Y at the mRNA and protein levels in the 3T3-L1 fibroblast line during differentiation into adipocytes. Intracellular calcium concentration was measured by calcium imaging. KEY RESULTS The PACAP receptors PAC(1) and VPAC(2) as well as the neuropeptide Y(1) receptor were expressed at the mRNA level in fibroblasts, pre-adipocytes and adipocytes. The mRNA profile of the PAC(1) receptor isoforms showed the HOP sequence, whereas the HIP-isoform was present in subconfluent 3T3-L1 fibroblasts only. At the protein level, the mature 3T3-L1 adipocytes produced the PAC(1) and Y(1) receptors; only the PAC(1) receptor showed carbohydrate residues. Both neuropeptides induced an increase of intracellular calcium in mature adipocytes, which was absent in the precursor cells. These changes in calcium were mediated by Y(1) and PAC(1) receptors as demonstrated by the effects of specific receptor agonists and antagonists. CONCLUSIONS AND IMPLICATIONS As the PAC(1)-HOP receptor variant seems to be responsible for PACAP-mediated calcium influx in many cell types, the HOP sequence might also mediate the increase in intracellular calcium in adipocytes. Because a high intracellular calcium level is associated with lipogenesis, peptidergic innervation of adipose tissue might be involved in stress-induced obesity.
Collapse
|
32
|
Wheat WH, Pauken KE, Morris RV, Titus RG. Lutzomyia longipalpis salivary peptide maxadilan alters murine dendritic cell expression of CD80/86, CCR7, and cytokine secretion and reprograms dendritic cell-mediated cytokine release from cultures containing allogeneic T cells. THE JOURNAL OF IMMUNOLOGY 2008; 180:8286-98. [PMID: 18523295 DOI: 10.4049/jimmunol.180.12.8286] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Leishmania protozoan parasites, the etiologic agent of leishmaniasis, are transmitted exclusively by phlebotomine sand flies of the genera Phlebotomus and Lutzomyia. In addition to parasites, the infectious bite inoculum contains arthropod salivary components. One well-characterized salivary component from Lutzomyia longipalpis is maxadilan (MAX), a vasodilator acting via the type I receptor for the pituitary cyclic AMP activating peptide. MAX has been shown to elicit immunomodulatory effects potentially dictating immune responses to Leishmania parasites. When exposed to MAX, both resting and LPS-stimulated dendritic cells (DCs) show reduced CD80 and CD86 expression on most DCs in vitro. However, CD86 expression is increased significantly on a subpopulation of DCs. Furthermore, MAX treatment promoted secretion of type 2 cytokines (IL-6 and IL-10) while reducing production of type 1 cytokines (IL-12p40, TNF-alpha, and IFN-gamma) by LPS-stimulated DCs. A similar trend was observed in cultures of MAX-treated DCs containing naive allogeneic CD4(+) T cells: type 2 cytokines (IL-6 and IL-13) increased while type 1 cytokines (TNF-alpha and IFN-gamma) decreased. Additionally, the proinflammatory cytokine IL-1beta was increased in cultures containing MAX-treated mature DCs. MAX treatment of LPS-stimulated DCs also prevented optimal surface expression of CCR7 in vitro. These MAX-dependent effects were evident in DCs from both Leishmania major-susceptible (BALB/c) and -resistant (C3H/HeN) murine strains. These data suggest that modification of DC phenotype and function by MAX likely affects crucial cellular components that determine the pathological response to infection with Leishmania.
Collapse
Affiliation(s)
- William H Wheat
- Department of Microbiology Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA.
| | | | | | | |
Collapse
|