1
|
Zhang D, Babayan L, Ho H, Heaney AP. Chromogranin A regulates neuroblastoma proliferation and phenotype. Biol Open 2019; 8:8/3/bio036566. [PMID: 30833285 PMCID: PMC6451332 DOI: 10.1242/bio.036566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Neuroblastoma is a commonly encountered solid tumor in early childhood with high neuroplasticity, and differentiation therapy is hypothesized to lead to tumor mass shrinkage and/or symptom relief. CgA is a tissue specific protein restricted to the diffuse neuroendocrine system, and widely expressed in neuroblastomas. Using knockdown and knockout approaches to deplete CgA levels, we demonstrated that CgA loss inhibits SH-SY5Y cell proliferation and leads to a morphological shift with increased expression of Schwann and extracellular matrix specific molecules, and suppression of chromaffin features. We further confirmed the effects of CgA in a series of neuroblastoma cells with [BE(2)-M17 and IMR-32] and without (SK-N-SH) N-Myc amplification. We demonstrated that CgA depletion reduced IGF-II and IGFBP-2 expression, increased IGFBP-3 levels, and suppresses IGF downstream signaling as evidenced by reduced AKT/ERK pathway activation. This was further supported by an increased anti-proliferative effect of the ERK inhibitor in the CgA depleted cells. In an in vivo xenograft neuroblastoma model, CgA knockdown led to increased S-phenotypic marker expression at both protein and mRNA levels. Together these results suggest that CgA maintains IGF secretion and intracellular signaling to regulate proliferation and differentiation in neuroblastomas.
Collapse
Affiliation(s)
- Dongyun Zhang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles 90095, USA
| | - Lilit Babayan
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles 90095, USA
| | - Hillary Ho
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles 90095, USA
| | - Anthony P Heaney
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles 90095, USA .,Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles 90095, USA
| |
Collapse
|
2
|
de Boisvilliers M, Perrin F, Hebache S, Balandre AC, Bensalma S, Garnier A, Vaudry D, Fournier A, Festy F, Muller JM, Chadéneau C. VIP and PACAP analogs regulate therapeutic targets in high-risk neuroblastoma cells. Peptides 2016; 78:30-41. [PMID: 26826611 DOI: 10.1016/j.peptides.2016.01.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 12/30/2015] [Accepted: 01/21/2016] [Indexed: 12/14/2022]
Abstract
Neuroblastoma (NB) is a pediatric cancer. New therapies for high-risk NB aim to induce cell differentiation and to inhibit MYCN and ALK signaling in NB. The vasoactive intestinal peptide (VIP) and the pituitary adenylate cyclase-activating polypeptide (PACAP) are 2 related neuropeptides sharing common receptors. The level of VIP increases with NB differentiation. Here, the effects of VIP and PACAP analogs developed for therapeutic use were studied in MYCN-amplified NB SK-N-DZ and IMR-32 cells and in Kelly cells that in addition present the F1174L ALK mutation. As previously reported by our group in IMR-32 cells, VIP induced neuritogenesis in SK-N-DZ and Kelly cells and reduced MYCN expression in Kelly but not in SK-N-DZ cells. VIP decreased AKT activity in the ALK-mutated Kelly cells. These effects were PKA-dependent. IMR-32, SK-NDZ and Kelly cells expressed the genes encoding the 3 subtypes of VIP and PACAP receptors, VPAC1, VPAC2 and PAC1. In parallel to its effect on MYCN expression, VIP inhibited invasion in IMR-32 and Kelly cells. Among the 3 PACAP analogs tested, [Hyp(2)]PACAP-27 showed higher efficiency than VIP in Kelly cells. These results indicate that VIP and PACAP analogs act on molecular and cellular processes that could reduce aggressiveness of high-risk NB.
Collapse
MESH Headings
- Anaplastic Lymphoma Kinase
- Cell Differentiation/drug effects
- Cell Line, Tumor
- Cell Movement/drug effects
- Cyclic AMP-Dependent Protein Kinases/genetics
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Mutation
- N-Myc Proto-Oncogene Protein/genetics
- N-Myc Proto-Oncogene Protein/metabolism
- Neurons/drug effects
- Neurons/metabolism
- Neurons/pathology
- Organ Specificity
- Pituitary Adenylate Cyclase-Activating Polypeptide/chemical synthesis
- Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Receptor Protein-Tyrosine Kinases/genetics
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I/genetics
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I/metabolism
- Receptors, Vasoactive Intestinal Peptide, Type II/genetics
- Receptors, Vasoactive Intestinal Peptide, Type II/metabolism
- Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics
- Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism
- Signal Transduction
- Structure-Activity Relationship
- Vasoactive Intestinal Peptide/chemical synthesis
- Vasoactive Intestinal Peptide/pharmacology
Collapse
Affiliation(s)
- Madryssa de Boisvilliers
- Université de Poitiers, Équipe Récepteurs, Régulations et Cellules Tumorales (2RCT), Pôle Biologie Santé, Bât. B36/B37, UFR Sciences Fondamentales et Appliquées, 1 rue Georges Bonnet TSA, 51106 86073 Poitiers Cedex 9, France
| | - Florian Perrin
- Université de Poitiers, Équipe Récepteurs, Régulations et Cellules Tumorales (2RCT), Pôle Biologie Santé, Bât. B36/B37, UFR Sciences Fondamentales et Appliquées, 1 rue Georges Bonnet TSA, 51106 86073 Poitiers Cedex 9, France
| | - Salima Hebache
- Université de Poitiers, Équipe Récepteurs, Régulations et Cellules Tumorales (2RCT), Pôle Biologie Santé, Bât. B36/B37, UFR Sciences Fondamentales et Appliquées, 1 rue Georges Bonnet TSA, 51106 86073 Poitiers Cedex 9, France
| | - Annie-Claire Balandre
- Université de Poitiers, Équipe Récepteurs, Régulations et Cellules Tumorales (2RCT), Pôle Biologie Santé, Bât. B36/B37, UFR Sciences Fondamentales et Appliquées, 1 rue Georges Bonnet TSA, 51106 86073 Poitiers Cedex 9, France
| | - Souheyla Bensalma
- Université de Poitiers, Équipe Récepteurs, Régulations et Cellules Tumorales (2RCT), Pôle Biologie Santé, Bât. B36/B37, UFR Sciences Fondamentales et Appliquées, 1 rue Georges Bonnet TSA, 51106 86073 Poitiers Cedex 9, France
| | - Agnès Garnier
- Université de Poitiers, Équipe Récepteurs, Régulations et Cellules Tumorales (2RCT), Pôle Biologie Santé, Bât. B36/B37, UFR Sciences Fondamentales et Appliquées, 1 rue Georges Bonnet TSA, 51106 86073 Poitiers Cedex 9, France
| | - David Vaudry
- Université de Rouen, INSERM U982, Equipe Neuropeptides, survie neuronale et plasticité cellulaire, IRIB, UFR Sciences et Techniques, Place E. Blondel, 76821 Mont-Saint-Aignan, France
| | - Alain Fournier
- INRS, Institut Armand-Frappier, 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| | - Franck Festy
- Université de la Réunion, Stemcis c/o CYROI, 2, rue Maxime Rivière, 97490 Sainte Clotilde, France
| | - Jean-Marc Muller
- Université de Poitiers, Équipe Récepteurs, Régulations et Cellules Tumorales (2RCT), Pôle Biologie Santé, Bât. B36/B37, UFR Sciences Fondamentales et Appliquées, 1 rue Georges Bonnet TSA, 51106 86073 Poitiers Cedex 9, France
| | - Corinne Chadéneau
- Université de Poitiers, Équipe Récepteurs, Régulations et Cellules Tumorales (2RCT), Pôle Biologie Santé, Bât. B36/B37, UFR Sciences Fondamentales et Appliquées, 1 rue Georges Bonnet TSA, 51106 86073 Poitiers Cedex 9, France.
| |
Collapse
|
3
|
Duskey JT, Rice KG. Nanoparticle ligand presentation for targeting solid tumors. AAPS PharmSciTech 2014; 15:1345-54. [PMID: 24927668 PMCID: PMC4179653 DOI: 10.1208/s12249-014-0143-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 05/07/2014] [Indexed: 01/10/2023] Open
Abstract
Among the many scientific advances to come from the study of nanoscience, the development of ligand-targeted nanoparticles to eliminate solid tumors is predicted to have a major impact on human health. There are many reports describing novel designs and testing of targeted nanoparticles to treat cancer. While the principles of the technology are well demonstrated in controlled lab experiments, there are still many hurdles to overcome for the science to mature into truly efficacious targeted nanoparticles that join the arsenal of agents currently used to treat cancer in humans. One of these hurdles is overcoming unwanted biodistribution to the liver while maximizing delivery to the tumor. This almost certainly requires advances in both nanoparticle stealth technology and targeting. Currently, it continues to be a challenge to control the loading of ligands onto polyethylene glycol (PEG) to achieve maximal targeting. Nanoparticle cellular uptake and subcellular targeting of genes and siRNA also remain a challenge. This review examines the types of ligands that have been most often used to target nanoparticles to solid tumors. As the science matures over the coming decade, careful control over ligand presentation on nanoparticles of precise size, shape, and charge will likely play a major role in achieving success.
Collapse
Affiliation(s)
- Jason T. Duskey
- Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City, Iowa 52242 USA
| | - Kevin G. Rice
- Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City, Iowa 52242 USA
| |
Collapse
|
4
|
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
|
5
|
Lee PH. Label-free optical biosensor: A tool for G protein-coupled receptors pharmacology profiling and inverse agonists identification. J Recept Signal Transduct Res 2009; 29:146-53. [DOI: 10.1080/10799890903064390] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
6
|
Lutz EM, Ronaldson E, Shaw P, Johnson MS, Holland PJ, Mitchell R. Characterization of novel splice variants of the PAC1 receptor in human neuroblastoma cells: consequences for signaling by VIP and PACAP. Mol Cell Neurosci 2005; 31:193-209. [PMID: 16226889 DOI: 10.1016/j.mcn.2005.09.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2004] [Revised: 08/24/2005] [Accepted: 09/12/2005] [Indexed: 10/25/2022] Open
Abstract
Expression of VPAC and PAC1 receptor isoforms was determined in six neuroblastoma cell lines as well as in human embryonic and adult brain using reverse transcriptase PCR and quantitative PCR. PAC1 receptor splice variants missing a 21 amino acid sequence in the amino terminal domain were found to be the major receptor variants in the neuroblastoma cell lines and also were highly expressed in embryonic brain compared to adult brain. In four of the neuroblastoma cell lines, VIP and PACAP stimulated cyclic AMP production with different potencies and levels of maximal stimulation. High potency and greatest maximal stimulation of cyclic AMP for each peptide were recorded in SH-SY5Y cells, indicating the presence of high affinity VIP and PACAP receptors. Further characterization of specific VPAC and PAC1 receptor isoforms was carried out in the SH-SY5Y cell line, where along with known PAC1 receptor splice variants and the VPAC2 receptor, a number of novel PAC1 receptor splice variants were identified. The comparatively low level expression of the VPAC2 receptor along with the poor responsiveness of SH-SY5Y cells to the VPAC2 receptor-specific agonist Ro 25-1553 indicated that this receptor did not contribute significantly to the observed VIP responses. When the individual PAC1 receptor isoforms were expressed in COS 7 cells, the ability of VIP to activate cyclic AMP production was increased more than 50-fold at the majority of the PAC1 receptor variants lacking the 21 amino acid amino terminal domain sequence compared to those with the complete domain. Smaller changes were seen in the potency of PACAP-38. Similar trends were seen with inositol phosphate responses, where in each case agonist potencies were lower than for cyclic AMP production. The results of this study show that the combination of different amino terminal and intracellular loop 3 splicing variants in the PAC1 receptor dictates the ability of agonists, particularly VIP, to activate signaling pathways. VIP has considerably greater potency at most PAC1 receptors with the short amino terminal domain, and these therefore may mediate physiological effects of both VIP and PACAP. Furthermore, there may be a phenotypic switch in the expression of different PAC1 receptor amino terminal splice variants between embryonic and mature nervous system, indicating that regulation of this event may have an important role in VIP/PACAP function, particularly in the developing nervous system.
Collapse
Affiliation(s)
- E M Lutz
- Molecular Signalling Group, Department of Bioscience, University of Strathclyde, Royal College, 204 George St., Glasgow G1 1XW, UK.
| | | | | | | | | | | |
Collapse
|
7
|
Abstract
As a neuroendocrine tumor, neuroblastoma expresses various gastrointestinal (GI) hormones, such as vasoactive intestinal peptide, gastrin-releasing peptide (GRP), neurotensin, and somatostatin, which exert diverse cellular functions in neuroblastoma. In particular, we have recently found that GRP and its cell surface receptor, GRP-R, are abundantly expressed in neuroblastomas. Moreover, more advanced-stage neuroblastomas demonstrated an increased level of GRP-R, suggesting an important role of GRP in aggressive tumor behavior. This review describes the role of several GI hormones commonly expressed in neuroblastoma and discusses in depth the mitogenic actions of GRP in neuroblastoma. In addition, the molecular mechanisms involved in the GRP-induced stimulation of neuroblastoma cell growth are discussed. Our study results demonstrate a role of GRP as an autocrine/paracrine growth factor and elucidate involvement of specific intracellular signaling, the phosphatidylinositol 3-kinase pathway, in the growth regulation of neuroblastoma.
Collapse
Affiliation(s)
- W Clay Gustafson
- Department of Surgery, The University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas, 77555, USA
| | | | | | | |
Collapse
|
8
|
Alleaume C, Eychène A, Harnois T, Bourmeyster N, Constantin B, Caigneaux E, Muller JM, Philippe M. Vasoactive intestinal peptide-induced neurite remodeling in human neuroblastoma SH-SY5Y cells implicates the Cdc42 GTPase and is independent of Ras-ERK pathway. Exp Cell Res 2004; 299:511-24. [PMID: 15350548 DOI: 10.1016/j.yexcr.2004.06.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2004] [Revised: 06/15/2004] [Indexed: 11/28/2022]
Abstract
Vasoactive intestinal peptide (VIP) is known to regulate proliferation or differentiation in normal and tumoral cells. SH-SY5Y is a differentiated cell subclone derived from the SK-N-SH human neuroblastoma cell line and possess all the components for an autocrine action of VIP. In the present study, we investigated the morphological changes and intracellular signaling pathways occurring upon VIP treatment of SH-SY5Y cells. VIP induced an early remodeling of cell projections: a branched neurite network spread out and prominent varicosities developed along neurites. Although activated by VIP, the Ras/ERK pathway was not required for the remodeling process. In contrast, pull-down experiments revealed a strong Cdc42 activation by VIP while expression of a dominant-negative Cdc42 prevented the VIP-induced neurite changes, suggesting an important role for this small GTPase in the process. These data provide the first evidence for a regulation of the activity of Rho family GTPases by VIP and bring new insights in the signaling pathways implicated in neurite remodeling process induced by VIP in neuroblastoma cells.
Collapse
Affiliation(s)
- Céline Alleaume
- Equipe Neuropeptides, Institut de Physiologie et Biologie Cellulaires, CNRS UMR 6187, Université de Poitiers-Pôle Biologie Santé, 86022 Poitiers Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Bourgois B, Boman F, Nelken B, Bonnevalle M, Turck D. [Intractable diarrhoea revealing a neuroblastoma hypersecreting the vasoactive intestinal peptide]. Arch Pediatr 2004; 11:340-3. [PMID: 15051093 DOI: 10.1016/j.arcped.2004.01.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2003] [Accepted: 01/19/2004] [Indexed: 10/26/2022]
Abstract
In children, the watery diarrhoea-hypokalemia-achlorhydria (WDHA) syndrome is uncommon and usually due to a neuroblastic tumour hypersecreting the vasoactive intestinal peptide (VIP). We report a case of WDHA syndrome secondary to hypersecretion of VIP that revealed a neuroblastoma in a 13-month-old girl. A secretory diarrhoea, characterised by the persistence of diarrhoea despite the cessation of oral feeding, led to the search of a neuroblastic tumour in the patient. The serum concentration of VIP decreased to normal values soon after the surgical excision of the tumour.
Collapse
Affiliation(s)
- B Bourgois
- Clinique de pédiatrie, hôpital Jeanne-de-Flandre, 2, avenue Oscar-Lambret, 59037 Lille, France
| | | | | | | | | |
Collapse
|
10
|
Héraud C, Hilairet S, Muller JM, Leterrier JF, Chadéneau C. Neuritogenesis induced by vasoactive intestinal peptide, pituitary adenylate cyclase-activating polypeptide, and peptide histidine methionine in SH-SY5y cells is associated with regulated expression of cytoskeleton mRNAs and proteins. J Neurosci Res 2004; 75:320-9. [PMID: 14743445 DOI: 10.1002/jnr.10866] [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: 11/06/2022]
Abstract
Vasoactive intestinal peptide (VIP) and the related peptides pituitary adenylate cyclase-activating polypeptide (PACAP) and peptide histidine methionine (PHM) are known to regulate proliferation and/or differentiation in normal and tumoral cells. In this study, neuritogenesis in human neuroblastoma SH-SY5Y cells cultured in serum-free medium was induced by VIP, PACAP, and PHM. The establishment of this process was followed by the quantification of neurite length and branching and the expression of neurofilament mRNAs, neurofilament proteins, and other cytoskeletal protein markers of neuronal differentiation: neuron-specific MAPs and beta-tubulin III. Neurite length and branching and the expression of most markers tested were increased by VIP and PACAP in a similar, although slightly different, fashion. In contrast, neuritic elongation induced by PHM was correlated with neither an increase in branching or neurofilament mRNAs nor a clear change in the expression of cytoskeleton proteins, with the exception of the stimulation by PHM of doublecortin, a microtubule-associated marker of migrating neuroblasts. These findings are the first evidence from a human neuron-like cell line for 1) a direct regulation of the metabolism of neurofilaments by VIP and PACAP and 2) the induction by PHM of neuritic processes of an apparent immature character.
Collapse
Affiliation(s)
- Céline Héraud
- Laboratoire des Biomembranes et Signalisation Cellulaire, Poitiers, France
| | | | | | | | | |
Collapse
|
11
|
Dufes C, Alleaume C, Montoni A, Olivier JC, Muller JM. Effects of the vasoactive intestinal peptide (VIP) and related peptides on glioblastoma cell growth in vitro. J Mol Neurosci 2004; 21:91-102. [PMID: 14593209 DOI: 10.1385/jmn:21:2:91] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2003] [Accepted: 03/29/2003] [Indexed: 11/11/2022]
Abstract
The growth rate of numerous cancer cell lines is regulated in part by actions of neuropeptides of the vasoactive intestinal peptide (VIP) family, which also includes pituitary adenylate cyclase-activating peptide (PACAP), glucagon, and peptide histidine/isoleucine (PHI). The aim of this work was to investigate the effect of these peptides on the growth of the rat glioblastoma cell line C6 in vitro. We also sought to determine which binding sites were correlated with the effects observed. Proliferation studies performed by means of a CyQuant trade mark assay showed that VIP and PACAP strongly stimulated C6 cell proliferation at most of the concentrations tested, whereas PHI increased cell proliferation only when associated with VIP. Two growth hormone-releasing factor (GRF) derivatives and the VIP antagonist hybrid peptide neurotensin-VIP were able to inhibit VIP-induced cell growth stimulation, even at very low concentrations. Binding experiments carried out on intact cultured C6 cells, using 125I-labeled VIP and PACAP as tracers, revealed that the effects of the peptides on cell growth were correlated with the expression on C6 cells of polyvalent high-affinity VIP-PACAP binding sites and of a second subtype corresponding to very high-affinity VIP-selective binding species. The latter subtype, which interacted poorly with PACAP with a 10,000-fold lower affinity than VIP, might mediate the antagonist effects of neurotensin- VIP and of both GRF derivatives on VIP-induced cell growth stimulation.
Collapse
Affiliation(s)
- Christine Dufes
- Laboratoire de Biologie des Interactions Cellulaires, CNRS UMR 6558, Faculté de Sciences, Université de Poitiers, 86022 Poitiers, France
| | | | | | | | | |
Collapse
|
12
|
Kenakin T. Efficacy as a vector: the relative prevalence and paucity of inverse agonism. Mol Pharmacol 2004; 65:2-11. [PMID: 14722230 DOI: 10.1124/mol.65.1.2] [Citation(s) in RCA: 179] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
This article describes the expected phenotypic behavior of all types of ligands in constitutively active receptor systems and, in particular, the molecular mechanisms of inverse agonism. The possible physiological relevance of inverse agonism also is discussed. Competitive antagonists with the molecular property of negative efficacy demonstrate inverse agonism in constitutively active receptor systems. This is a phenotypic behavior that can only be observed in the appropriate assay; a lack of observed inverse agonism is evidence that the ligand does not possess negative efficacy only if it can be shown that constitutive receptor activity is present. In the absence of constitutive activity, inverse agonists behave as simple competitive antagonists. A survey of 105 articles on the activity of 380 antagonists on 73 biological G-protein-coupled receptor targets indicates that, in this sample dataset, 322 are inverse agonists and 58 (15%) are neutral antagonists. The predominance of inverse agonism agrees with theoretical predictions which indicate that neutral antagonists are the minority species in pharmacological space.
Collapse
Affiliation(s)
- Terry Kenakin
- Department of Assay Development and Compound Profiling, GlaxoSmithKline Research and Development, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
13
|
Karacay B, O'Dorisio MS, Summers M, Robinson M, Bonthius DJ. VIP receptor 1 (VPAC1) promoter targets the expression of a reporter gene to cerebellum and adrenal medulla in transgenic mice. ACTA ACUST UNITED AC 2003; 116:1-12. [PMID: 14599709 DOI: 10.1016/s0167-0115(03)00170-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Vasoactive intestinal peptide (VIP) is a neurotransmitter with neurotropic effects. VIP functions through two distinct G-protein-coupled receptor subtypes (VPAC1 and VPAC2). We have demonstrated expression of VPAC1 in pediatric nervous system tumors, including medulloblastoma arising in the cerebellum and neuroblastoma arising in the adrenal medulla. More recently, we have reported the differentiation of neuroblastoma cells by upregulation of VIP type 1 receptor suggesting a role for VPAC1 in neuronal development. To understand the molecular mechanisms regulating VPAC1 expression in both cerebellum and adrenal medulla, we have cloned the human VPAC1 gene and sequenced 2.6-kb of the 5'-flanking sequence. Expression of the luciferase reporter gene under the control of this 2.6-kb human VPAC1 promoter was induced 35-fold in a human medulloblastoma cell line (DAOY) and 36-fold in a human neuroblastoma cell line (SKNSH). Analysis of 5'-unidirectional deletion derivatives of the 2.6-kb fragment demonstrated that a 241-bp sequence immediately upstream of the VPAC1 coding region retains high activity, suggesting that it contains the core promoter region. Quantitative RT-PCR analysis demonstrated that VPAC1 is expressed in mouse cerebellar and adrenal tissues. The VPAC1 promoter also directed expression of a reporter gene in cerebellum and adrenal medulla in transgenic mice. Along with our previous findings, these results suggest that VPAC1 may play a functional role in development of both cerebellum and adrenal medulla.
Collapse
MESH Headings
- Adrenal Medulla/metabolism
- Animals
- Base Sequence
- Cerebellum/metabolism
- Cloning, Molecular
- Female
- Gene Expression Regulation
- Genes, Reporter/genetics
- Humans
- Male
- Mice
- Mice, Transgenic
- Molecular Sequence Data
- Myocardium/metabolism
- Organ Specificity
- Promoter Regions, Genetic/genetics
- Rats
- Receptors, Vasoactive Intestinal Peptide/isolation & purification
- Receptors, Vasoactive Intestinal Peptide/metabolism
- Receptors, Vasoactive Intestinal Polypeptide, Type I
- Response Elements/genetics
- Sp1 Transcription Factor/metabolism
- Transcription, Genetic/genetics
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Bahri Karacay
- Division of Hematology/Oncology, Department of Pediatrics, University of Iowa Hospitals and Clinics, 200 Hawkins Drive-2526 JCP, Iowa City, IA 52242, USA
| | | | | | | | | |
Collapse
|
14
|
Abstract
During the past decade, proof of the principle that peptide receptors can be used successfully for in vivo targeting of human cancers has been provided. The molecular basis for targeting rests on the in vitro observation that peptide receptors can be expressed in large quantities in certain tumors. The clinical impact is at the diagnostic level: in vivo receptor scintigraphy uses radiolabeled peptides for the localization of tumors and their metastases. It is also at the therapeutic level: peptide receptor radiotherapy of tumors emerges as a serious treatment option. Peptides linked to cytotoxic agents are also considered for therapeutic applications. The use of nonradiolabeled, noncytotoxic peptide analogs for long-term antiproliferative treatment of tumors appears promising for only a few tumor types, whereas the symptomatic treatment of neuroendocrine tumors by somatostatin analogs is clearly successful. The present review summarizes and critically evaluates the in vitro data on peptide and peptide receptor expression in human cancers. These data are considered to be the molecular basis for peptide receptor targeting of tumors. The paradigmatic peptide somatostatin and its receptors are extensively reviewed in the light of in vivo targeting of neuroendocrine tumors. The role of the more recently described targeting peptides vasoactive intestinal peptide, gastrin-releasing peptide, and cholecystokinin/gastrin is discussed. Other emerging and promising peptides and their respective receptors, including neurotensin, substance P, and neuropeptide Y, are introduced. This information relates to established and potential clinical applications in oncology.
Collapse
Affiliation(s)
- Jean Claude Reubi
- Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, CH-3010 Berne, Switzerland
| |
Collapse
|
15
|
Abstract
The effects of vasoactive intestinal peptide (VIP) on the proliferation of central nervous system (CNS) and cancer cells were investigated. VIP has important actions during CNS development. During neurogenesis, VIP stimulates the proliferation and differentiation of brain neurons. Addition of VIP to embryonic mouse spinal cord cultures increases neuronal survival and activity dependent neurotrophic factor (ADNF) secretion from astroglial cells. VIP is an integrative regulator of brain growth and development during neurogenesis and embryogenesis. Also, VIP causes increased proliferation of human breast and lung cancer cells in vitro. VIP binds with high affinity to cancer cells, elevates the cAMP and increases gene expression of c-fos, c-jun, c-myc and vascular endothelial cell growth factor. The effects of VIP on cancer cells are reversed by VIPhybrid, a synthetic VPAC(1) receptor antagonist. VIPhyb inhibits the basal growth of lung cancer cells in vitro and tumors in vivo and potentiates the ability of chemotherapeutic drugs to kill cancer cells. Due to the high density of VPAC(1) receptors in cancer cells, VIP has been radiolabeled with 123I, 18F and 99mTc to image tumors. It remains to be determined if radiolabeled VIP analogs will be useful agents for early detection of cancer in patients.
Collapse
Affiliation(s)
- Terry W Moody
- NCI Office of the Director, Center for Cancer Research, National Cancer Institute, Bldg 31, Rm 3A34, 31 Center Dr, Bethesda, MD, USA.
| | | | | |
Collapse
|
16
|
The Biological Significance of PACAP and PACAP Receptors in Human Tumors: From Cell Lines to Cancers. PITUITARY ADENYLATE CYCLASE-ACTIVATING POLYPEPTIDE 2003. [DOI: 10.1007/978-1-4615-0243-2_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
17
|
O'Dorisio MS, Hauger M, O'Dorisio TM. Age-dependent levels of plasma neuropeptides in normal children. REGULATORY PEPTIDES 2002; 109:189-92. [PMID: 12409232 DOI: 10.1016/s0167-0115(02)00203-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Several neuropeptides are secreted in high amounts in pediatric tumors such as neuroblastoma and have been used as markers of residual or recurrent disease. Plasma levels of neuropeptides might be expected to change during development, but have not been determined in normal children. We have obtained fresh plasma from cord blood of six full-term infants and from peripheral blood in 41 healthy children, ages 1 month to 21 years. Levels of six neuropeptides, vasoactive intestinal peptide (VIP), somatostatin, gastrin releasing peptide (GRP), substance P, pancreastatin and neuropeptide Y (NPY) were measured by radioimmunoassay along with insulin-like growth factor-1 (IGF-1) whose plasma levels are known to vary during development. A child with neuroblastoma was treated with the somatostatin analogue, octreotide, and the effect on plasma neuropeptides quantified. Octreotide doses of 2-3 microg/kg daily resulted in a 40-60% decrease in plasma levels of IGF-1, pancreastatin and GRP. These results are the first publication of plasma neuropeptide levels in normal children.
Collapse
Affiliation(s)
- M Sue O'Dorisio
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA.
| | | | | |
Collapse
|
18
|
Balster DA, O'Dorisio MS, Albers AR, Park SK, Qualman SJ. Suppression of tumorigenicity in neuroblastoma cells by upregulation of human vasoactive intestinal peptide receptor type 1. REGULATORY PEPTIDES 2002; 109:155-65. [PMID: 12409228 DOI: 10.1016/s0167-0115(02)00199-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We hypothesize that vasoactive intestinal peptide (VIP) promotes neural crest differentiation through VIP receptor type I (VPAC1). In order to test this hypothesis, SKNSH neuroblastoma cells were stably transfected with VPAC1 and receptor expression was verified by real-time RT-PCR. Overexpression of VPAC1 in SKNSH cells resulted in upregulation of endogenous retinoic acid receptor expression for both RARalpha and RXRalpha with no change in expression of RARbeta. Transfected cells demonstrated high affinity binding of VIP (K(D)=0.2 nM) and VIP-mediated stimulation of adenylate cyclase and a shift in cell cycle kinetics to a near triploid DNA index in G1. SKNSH/VPAC1 cells treated with VIP were observed to express a more differentiated phenotype compared to wild type cells as characterized by an increase in tissue transglutaminase II and a decrease in bcl-2 immunostaining. VIP-induced differentiation effects were potentiated by retinoic acid. This differentiation resulted in decreased proliferative potential in a xenograft model. Whereas, wild type SKNSH cells induced tumor growth in 100% of nude mice within 13 days post-injection, SKNSH transfected with VPAC1 demonstrated no tumor formation in xenografts followed for 6 months. Taken together, these data support the hypothesis that VIP modulation of neural crest differentiation is mediated via VPAC1 and that high expression of VPAC1 induces differentiation in and decreases tumorigenicity of neuroblastoma cells.
Collapse
Affiliation(s)
- Douglas A Balster
- Department of Pathology, The Ohio State University, Columbus, OH 43205, USA
| | | | | | | | | |
Collapse
|
19
|
Karacay B, O'Dorisio MS, Kasow K, Hollenback C, Krahe R. Expression and fine mapping of murine vasoactive intestinal peptide receptor 1. J Mol Neurosci 2002. [PMID: 11859927 DOI: 10.1385/jmn: 17: 3: 311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Vasoactive intestinal peptide (VIP) plays multiple roles in the nervous, endocrine, and immune systems as a neurotransmitter, a hormone, and a cytokine. VIP is widely distributed in neurons of the central and peripheral nervous systems (CNS/PNS), and recently has been found to be an important neuroprotective agent. VIP actions are mediated through specific G protein-coupled receptors. We have cloned the cDNA of VIP receptor subtype 1 (VIPR1 or VPAC1) and have demonstrated the quantitative expression profile in mice. Fluorometric real-time reverse transcription-polymerase chain reaction (RT-PCR) analysis demonstrated that VPAC1 is expressed in all tissues examined. Expression was highest in the small intestine and colon followed by the liver and brain. The high level of VPAC1 expression in forebrain and cerebellum suggests that VPAC1 may mediate the neuroprotective effect of VIP. We have refined the chromosomal localization of the mouse, rat, and human VPAC1 genes. This fine mapping of the VPAC1 gene extends the respective regions of synteny between the distal region of mouse chromosome 9, rat chromosome 8q32, and human chromosome 3p21.33-p21.31. Thus, VPAC, constitutes a functional-positional candidate for the tumor-suppressor function mapped to human 3p22-p21 where loss-of-heterozygosity is observed in small-cell lung carcinoma (SCLC) cell lines and primary tumors. Availability of the cDNA sequences for mouse VPAC1 will facilitate the generation of VPAC1 null mutant animals. Such studies will ultimately enhance our understanding of the role of VIP in the nervous system.
Collapse
Affiliation(s)
- B Karacay
- Department of Pediatrics, University of Iowa, Iowa City 52242, USA
| | | | | | | | | |
Collapse
|
20
|
Moody TW, Jensen RT, Fridkin M, Gozes I. (N-stearyl, norleucine17)VIPhybrid is a broad spectrum vasoactive intestinal peptide receptor antagonist. J Mol Neurosci 2002; 18:29-35. [PMID: 11931347 DOI: 10.1385/jmn:18:1-2:29] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2001] [Accepted: 08/26/2001] [Indexed: 11/11/2022]
Abstract
The effects of a (N-stearyl, Norleucine17) vasoactive intestinal peptide hybrid ((SN)VIPhybrid) on cells stably transfected with VPAC,, VPAC2, or PAC1 receptors were investigated. (SN)VIPhybrid inhibited specific 125I-VIP binding to membranes derived from CHO cells transfected with VPAC, or VPAC2 receptors with high affinity (IC50 = 30 and 50 nM). (SN)VIPhyb inhibited specific 125I-PACAP-27 binding to membranes derived from NIH/3T3 cells transfected with PAC1 receptors with high affinity (IC50 = 65 nM). PACAP-27 caused cAMP elevation in NIH/3T3 cells transfected with PAC1 receptors and the increase cAMP caused by pituitary adenylated cyclase (PACAP) was inhibited by (SN)VIPhyb. Also, the increase in cAMP caused by VIP using CHO cells transfected with VPAC1 or VPAC2 receptors was antagonized by (SN)VIPhyb. These results indicate that (SN)VIPhyb is an antagonist for VPAC1, VPAC2, and PAC1 receptors.
Collapse
MESH Headings
- 3T3 Cells
- Animals
- Binding, Competitive/drug effects
- Binding, Competitive/physiology
- CHO Cells
- Cricetinae
- Cyclic AMP/metabolism
- Dose-Response Relationship, Drug
- Iodine Radioisotopes
- Mice
- Peptide Fragments/pharmacology
- Radioligand Assay
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I
- Receptors, Pituitary Hormone/drug effects
- Receptors, Pituitary Hormone/genetics
- Receptors, Pituitary Hormone/metabolism
- Receptors, Vasoactive Intestinal Peptide/drug effects
- Receptors, Vasoactive Intestinal Peptide/genetics
- Receptors, Vasoactive Intestinal Peptide/metabolism
- Receptors, Vasoactive Intestinal Peptide, Type II
- Receptors, Vasoactive Intestinal Polypeptide, Type I
- Recombinant Fusion Proteins/pharmacology
- Up-Regulation/drug effects
- Up-Regulation/physiology
- Vasoactive Intestinal Peptide/metabolism
- Vasoactive Intestinal Peptide/pharmacology
Collapse
Affiliation(s)
- Terry W Moody
- National Cancer Institute, Medicine Branch, Rockville, MD 20850, USA.
| | | | | | | |
Collapse
|
21
|
Sharma A, Walters J, Gozes Y, Fridkin M, Brenneman D, Gozes I, Moody TW. A vasoactive intestinal peptide antagonist inhibits the growth of glioblastoma cells. J Mol Neurosci 2001; 17:331-9. [PMID: 11859929 PMCID: PMC8767806 DOI: 10.1385/jmn:17:3:331] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The effects of a vasoactive intestinal peptide (VIP) receptor antagonist (VIPhyb) on human glioblastoma cells were characterized. Pituitary adenylate cyclase activating polypeptide (125I-PACAP-27) bound with high affinity to U87, U118, and U373 cells. Specific 125I-PACAP-27 binding to U87 cells was inhibited, with high affinity, by PACAP but not VIP or VIPhyb (IC50 = 10, 1500, and 500 nM, respectively). By reverse transcriptase-polymerase chain reaction (RT-PCR), a major 305 bp band was observed indicative of PAC1 receptors. PACAP-27 caused cAMP elevation and the increase in cAMP caused by PACAP-27, was inhibited by the VIPhyb. Also, PACAP-27 caused cytosolic Ca2+ elevation in Fura-2AM loaded U87 cells and the VIPhyb inhibited this increase. Using the MTT growth assay, the VIPhyb was shown to inhibit glioblastoma growth in a concentration-dependent manner. Using a clonogenic assay in vitro, 10 microM VIPhyb significantly inhibited proliferation of U87, U118, and U373 cells. In vivo, 0.4 microg/kg VIPhyb inhibited U87 xenograft proliferation in nude mice. These results suggest that the VIPhyb antagonizes PAC1 receptors on glioblastoma cells and inhibits their proliferation.
Collapse
Affiliation(s)
- Anita Sharma
- National Cancer Institute, Medicine Branch, Rockville, MD 20850
| | - James Walters
- National Cancer Institute, Medicine Branch, Rockville, MD 20850
| | - Yehoshua Gozes
- Israel Institute for Biological Research, Ness-Ziona 74100, Israel
| | - Mati Fridkin
- Department of Organic Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Douglas Brenneman
- Section on Developmental and Molecular Pharmacology, Laboratory of Developmental Neurobiology, National Institute for Child Health and Human Development, Bethesda, MD 20892
| | - Illana Gozes
- Department of Clinical Biochemistry, Sackler School of Medicine, Tel Aviv Univ. Tel Aviv 69978, Israel
| | - Terry W. Moody
- National Cancer Institute, Medicine Branch, Rockville, MD 20850
| |
Collapse
|
22
|
Karacay B, O'Dorisio MS, Kasow K, Hollenback C, Krahe R. Expression and fine mapping of murine vasoactive intestinal peptide receptor 1. J Mol Neurosci 2001; 17:311-24. [PMID: 11859927 DOI: 10.1385/jmn:17:3:311] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Vasoactive intestinal peptide (VIP) plays multiple roles in the nervous, endocrine, and immune systems as a neurotransmitter, a hormone, and a cytokine. VIP is widely distributed in neurons of the central and peripheral nervous systems (CNS/PNS), and recently has been found to be an important neuroprotective agent. VIP actions are mediated through specific G protein-coupled receptors. We have cloned the cDNA of VIP receptor subtype 1 (VIPR1 or VPAC1) and have demonstrated the quantitative expression profile in mice. Fluorometric real-time reverse transcription-polymerase chain reaction (RT-PCR) analysis demonstrated that VPAC1 is expressed in all tissues examined. Expression was highest in the small intestine and colon followed by the liver and brain. The high level of VPAC1 expression in forebrain and cerebellum suggests that VPAC1 may mediate the neuroprotective effect of VIP. We have refined the chromosomal localization of the mouse, rat, and human VPAC1 genes. This fine mapping of the VPAC1 gene extends the respective regions of synteny between the distal region of mouse chromosome 9, rat chromosome 8q32, and human chromosome 3p21.33-p21.31. Thus, VPAC, constitutes a functional-positional candidate for the tumor-suppressor function mapped to human 3p22-p21 where loss-of-heterozygosity is observed in small-cell lung carcinoma (SCLC) cell lines and primary tumors. Availability of the cDNA sequences for mouse VPAC1 will facilitate the generation of VPAC1 null mutant animals. Such studies will ultimately enhance our understanding of the role of VIP in the nervous system.
Collapse
Affiliation(s)
- B Karacay
- Department of Pediatrics, University of Iowa, Iowa City 52242, USA
| | | | | | | | | |
Collapse
|
23
|
Lelièvre V, Pineau N, Hu Z, Ioffe Y, Byun JY, Muller JM, Waschek JA. Proliferative actions of natriuretic peptides on neuroblastoma cells. Involvement of guanylyl cyclase and non-guanylyl cyclase pathways. J Biol Chem 2001; 276:43668-76. [PMID: 11553633 DOI: 10.1074/jbc.m107341200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To identify neural tumor cell lines that could be used as models to study growth-related natriuretic peptide actions, we determined the effects of these peptides on the proliferation of human and rodent neuroblastoma cell lines. Subnanomolar concentrations of atrial natriuretic peptide (ANP) and type C natriuretic peptide (CNP) stimulated proliferation in all four cell lines. These actions were associated with cGMP elevation and were blocked by a protein kinase G inhibitor. These data imply the involvement of guanylyl cyclase (GC)-coupled natriuretic receptors. However, higher concentrations of ANP and CNP, and low concentrations of des-[Gln(18),Ser(19),Gly(20),Leu(21),Gly(22)]-ANP(4-23)-NH(2) (desANP(4-23)) (analog for NPR-C receptor) exerted antiproliferative actions in three of the cell lines. These effects were insensitive to a protein kinase G inhibitor and to HS-142-1, suggesting that growth-inhibitory actions involved a non-GC receptor. They did not appear to involve cAMP, protein kinase A, protein kinase C, or calcium mobilization but were abolished when constitutive mitogen-activated protein kinase activity was inhibited. Radioligand binding experiments revealed the presence of a uniform class of binding sites in NG108 cells and multiple binding sites in Neuro2a cells. Northern and reverse transcriptase-polymerase chain reaction analyses revealed differential gene expression for NPR-A/B/C in NG108 and Neuro2a cells. The results indicate that natriuretic peptides stimulate neuroblastoma cell proliferation through type NPR-A/B (GC) receptors. Higher concentrations of ANP and CNP exerted a mitogen-activated protein kinase-dependent antiproliferative action mediated by a non-GC receptor that interacts with desANP(4-23) with relatively high affinity.
Collapse
Affiliation(s)
- V Lelièvre
- Department of Psychiatry and Mental Retardation Research Center, UCLA, Neuropsychiatric Institute, Los Angeles, California 90024, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Jensen JA, Carroll RE, Benya RV. The case for gastrin-releasing peptide acting as a morphogen when it and its receptor are aberrantly expressed in cancer. Peptides 2001; 22:689-99. [PMID: 11311741 DOI: 10.1016/s0196-9781(01)00380-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Gastrin-releasing peptide (GRP) and its receptor (GRP-R) are frequently expressed by cancers of the gastrointestinal tract, breast, lung, and prostate. Most studies have found that GRP and its amphibian homologue bombesin act to increase tumor cell proliferation, leading to the hypothesis that this peptide hormone is a mitogen important for the growth of various cancers. Yet GRP/GRP-R co-expression in cancer promotes the development of a well-differentiated phenotype; while multiple studies suggest that the presence of these 2 proteins confer a survival advantage. Along with recent reports showing that GRP and its receptor critically regulate aspects of colon and lung organogenesis, we argue that these proteins do not function primarily as mitogens when aberrantly expressed in cancer. Rather, we postulate that GRP/GRP-R are onco-fetal antigens that function as morphogens, with their effect on tumor cell proliferation being a component property of their ability to regulate differentiation. Thus aberrant GRP/GRP-R expression in cancer recapitulates, albeit in a dysfunctional manner, their normal role in development.
Collapse
Affiliation(s)
- J A Jensen
- Department of Medicine, University of Illinois at Chicago and Chicago Veterans Administration Medical Center (West Side Division), Chicago, Illinois 60612, USA
| | | | | |
Collapse
|
25
|
Abstract
Concepts regarding the mechanisms by which drugs activate receptors to produce physiological response have progressed beyond considering the receptor as a simple on-off switch. Current evidence suggests that the idea that agonists produce only varying degrees of receptor activation is obsolete and must be reconciled with data to show that agonist efficacy has texture as well as magnitude. Thus, agonists can block system constitutive response (inverse agonists), behave as positive and inverse agonists on the same receptor (protean agonists), and differ in the stimulus pattern they produce in physiological systems (ligand-selective agonists). The molecular mechanism for this seemingly diverse array of activities is the same, namely, the selective microaffinity of ligands for different conformational states of the receptor. This paper reviews evidence for the existence of the various types of agonism and the potential therapeutic utility of different agonist types.-Kenakin, T. Inverse, protean, and ligand-selective agonism: matters of receptor conformation.
Collapse
Affiliation(s)
- T Kenakin
- Department of Receptor Biochemistry, Glaxo SmithKline Research and Development, Research Triangle Park, North Carolina 27709, USA.
| |
Collapse
|
26
|
Karacay B, O'Dorisio MS, Summers M, Bruce J. Regulation of vasoactive intestinal peptide receptor expression in developing nervous systems. Ann N Y Acad Sci 2001; 921:165-74. [PMID: 11193820 DOI: 10.1111/j.1749-6632.2000.tb06963.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Vasoactive intestinal peptide (VIP) is a 28-amino acid peptide that has several functions, including the regulation of water and electrolyte secretion, hormone and cytokine release, bronchodilitation, and neurogenesis. VIP effects are mediated by specific G-protein coupled receptors. Three distinct receptor subtypes, with differing affinity for VIP, have been cloned and characterized as receptors 1 and 2 (VPAC1 and VPAC2) and pituitary adenylate cyclase activating polypeptide receptor (PAC1). Our laboratory has demonstrated that upregulation of VPAC1 in SK-N-SH neuroblastoma cells results in marked shift in cell type to the glial lineage with a corresponding loss of neuronal lineage and suppression of xenograft tumor growth. To understand the molecular mechanisms responsible for regulation of the VPAC1 gene in neuronal lineage, we have cloned and sequenced 2.6-kb of the 5'-flanking sequences of the human VPAC1 gene. Sequence analysis demonstrated that the human VPAC1 promoter sequence contains putative binding sites for several known transcription factors, including Sp1, NFkB, and cETS-1. To study the temporal and spatial expression pattern of human VPAC1 promoter sequences, we have generated transgenic mice expressing the bacterial beta-galactosidase gene under the control of the 2.6-kb 5'-flanking and promoter sequence of the human VPAC1 gene. Transgene expression was detected in brain, spinal cord, and lung in 14-day-old animals. Taken together, these results demonstrate that VPAC1 may play an important role in the nervous system, and suggest a role for VIP in neuronal differentiation.
Collapse
Affiliation(s)
- B Karacay
- Department of Pediatrics and Comprehensive Cancer Center, Ohio State University, College of Medicine and Public Health, Columbus, Ohio, USA
| | | | | | | |
Collapse
|
27
|
Jakowlew SB, Zakowicz H, Moody TW. Retinoic acid down-regulates VPAC(1) receptors and TGF-beta 3 but up-regulates TGF-beta 2 in lung cancer cells. Peptides 2000; 21:1831-7. [PMID: 11150643 DOI: 10.1016/s0196-9781(00)00344-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The effects of retinoic acid (RA) on lung cancer cells were investigated. Both all-trans (t-RA) and 13-cis RA (c-RA) decreased specific (125)I-VIP binding to NCI-H1299 cells in a time- and concentration-dependent manner. After 20 hr, 30 microM t-RA decreased specific (125)I-VIP binding by 60%. By Scatchard analysis, the density of VIP binding sites but not the affinity was reduced by 42%. NCI-H1299 VPAC(1) receptor mRNA was reduced by 48%. VIP caused a 3-fold elevation in the NCI-H1299 cAMP, and the increase in cAMP caused by VIP was reduced by 38% if the NCI-H1299 cells were treated with t-RA. Using the MTT assay, 3 microM t-RA and 3 microM c-RA inhibited NCI-H1299 proliferation by 60 and 23% respectively. Also, transforming growth factor (TGF)-beta2 increased after treatment of NCI-H1299 cells with t-RA whereas TGF-beta 1 mRNA was unaffected and TGF-beta 3 mRNA was decreased. These results suggest that RA may inhibit lung cancer growth by down-regulating VPAC(1) receptor and TGF-beta 3 mRNA but up-regulating TGF-beta 2 mRNA.
Collapse
Affiliation(s)
- S B Jakowlew
- Cell & Cancer Biology Dept., Medicine Branch, National Cancer Institute, Rockville, MD 20850, USA
| | | | | |
Collapse
|
28
|
Frühwald MC, O'Dorisio MS, Pietsch T, Reubi JC. High expression of somatostatin receptor subtype 2 (sst2) in medulloblastoma: implications for diagnosis and therapy. Pediatr Res 1999; 45:697-708. [PMID: 10231868 DOI: 10.1203/00006450-199905010-00016] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Medulloblastoma is a pediatric malignancy, which arises in cerebellum. The neuropeptide somatostatin (SS-14) is a neuromodulator and growth regulator in the developing cerebellum. SS-14 has previously been demonstrated in medulloblastomas with immunohistochemical techniques, but somatostatin receptor (sst) expression is less well understood. We analyzed somatostatin and sst subtype expression (sst1-5) in central primitive neuroectodermal tumors (cPNET), including 23 medulloblastomas, 6 supratentorial PNET, and 10 cPNET cell lines. The expression of SS-14 and sst genes in cPNET was compared with expression of these genes in 17 tumors of the Ewing's sarcoma family of tumors using reverse transcriptase-PCR, Southern hybridization, quantitative in vitro receptor autoradiography, and competitive membrane binding assays. The sst1 subtype was expressed in similar frequency in cPNET (83%) and Ewing's sarcoma family of tumors (71%). Nine of the 10 cell lines and 76% of the cPNET expressed mRNA for sst2 compared with 35% of the Ewing's sarcoma family of tumors. High-affinity binding of SS-14 was demonstrated in cPNET by quantitative autoradiography as well as by competitive binding assays. The cPNET cell line D283 Med bound SS-14 and octreotide with high affinity; SS-14 inhibited proliferation of D283 Med cells as measured by a decrease in [3H]thymidine uptake. We conclude that both sst1 and sst2 are highly expressed in cPNET and suggest that somatostatin may regulate proliferation and differentiation in these developmental tumors.
Collapse
MESH Headings
- Adolescent
- Adult
- Cerebellar Neoplasms/diagnosis
- Cerebellar Neoplasms/genetics
- Cerebellar Neoplasms/pathology
- Cerebellar Neoplasms/therapy
- Child
- Child, Preschool
- DNA Primers
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Infant
- Infant, Newborn
- Male
- Medulloblastoma/diagnosis
- Medulloblastoma/genetics
- Medulloblastoma/pathology
- Medulloblastoma/therapy
- Neuroectodermal Tumors, Primitive, Peripheral/diagnosis
- Neuroectodermal Tumors, Primitive, Peripheral/genetics
- Neuroectodermal Tumors, Primitive, Peripheral/therapy
- Receptors, Somatostatin/analysis
- Receptors, Somatostatin/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- M C Frühwald
- Department of Pediatrics, Ohio State University, Columbus 43205, USA
| | | | | | | |
Collapse
|
29
|
Frühwald MC, O'Dorisio MS, Fleitz J, Pietsch T, Reubi JC. Vasoactive intestinal peptide (VIP) and VIP receptors: gene expression and growth modulation in medulloblastoma and other central primitive neuroectodermal tumors of childhood. Int J Cancer 1999; 81:165-73. [PMID: 10188714 DOI: 10.1002/(sici)1097-0215(19990412)81:2<165::aid-ijc1>3.0.co;2-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Vasoactive intestinal peptide (VIP) is a neuromodulator and growth regulator in the developing nervous system. We analyzed 10 primitive neuroectodermal tumor (PNET) cell lines, 29 central PNET (cPNET) and 17 tumors of the Ewing's sarcoma/peripheral PNET family (ESFT) using reverse transcriptase-polymerase chain reaction (RT-PCR) and Southern hybridization. Each of the 10 cell lines and 86.2% of cPNET expressed mRNA for VIP receptor 1 (VIPR1) compared to 52.9% of ESFT. VIPR2 was expressed in 75.8% of cPNET, in 28.6% of ESFT and in all 10 cell lines. cPNET demonstrated high-affinity binding of 125I-VIP on quantitative autoradiography and in competitive binding assays. VIP inhibited tumor cell proliferation in a dose-dependent manner in 5 of 7 PNET cell lines. We conclude that VIPR1 and VIPR2 are highly expressed in cPNET and demonstrate that VIP is a growth modulator in these tumors.
Collapse
Affiliation(s)
- M C Frühwald
- Department of Pediatrics, Ohio State University, Columbus, USA
| | | | | | | | | |
Collapse
|
30
|
Carroll RE, Matkowskyj KA, Chakrabarti S, McDonald TJ, Benya RV. Aberrant expression of gastrin-releasing peptide and its receptor by well-differentiated colon cancers in humans. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 276:G655-65. [PMID: 10070042 DOI: 10.1152/ajpgi.1999.276.3.g655] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Epithelial cells lining the adult human colon do not normally express gastrin-releasing peptide (GRP) or its receptor (GRPR). In contrast, approximately one-third of human colon cancers and cancer cell lines have been shown to express GRP-binding sites. Because GRPR activation causes the proliferation of many cancer cell lines, GRP has been presumed to act as a clinically significant growth factor. Yet GRP has not been shown to be expressed by colon cancers in humans nor has the effect of GRP and/or GRPR coexpression on tumor behavior been investigated. We therefore determined GRP and GRPR expression by immunohistochemistry in 50 randomly selected colon cancers resected between 1980 and 1997, all 37 associated lymph node and liver metastases, and 20 polyps. Tumor sections studied were those that contained the margin and adjacent nonmalignant epithelium. Overall, 84% of cancers aberrantly expressed GRP or GRPR, with 62% expressing both ligand and receptor, whereas expression was not observed in adjacent normal epithelium. Consistent with the previously established mitogenic capabilities of GRP, tissues coexpressing GRP and GRPR were more likely to express proliferating cell nuclear antigen than tissues not expressing both ligand and receptor. Yet GRP/GRPR coexpression was seen with equal frequency in stage A as in stage D cancers and was only detected in 1 in 37 metastases. Furthermore, Kaplan-Meier analysis did not reveal any difference in patient survival between those whose tumors did or did not express GRP/GRPR. In contrast, GRP/GRPR coexpression was found in all well-differentiated tumor regions, whereas poorly differentiated tissues never coexpressed GRP/GRPR. Overall, these data indicate that, although GRP is a mitogen, it is not a clinically significant growth factor in human colon cancers. Rather, the strong association of GRP/GRPR coexpression with tumor differentiation raises the possibility that these proteins primarily act in vivo as morphogens.
Collapse
Affiliation(s)
- R E Carroll
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | | | | | | | | |
Collapse
|
31
|
Woltering EA, O'Dorisio MS, Murphy WA, Chen F, Drouant GJ, Espenan GD, Fisher DR, Sharma C, Diaco DS, Maloney TM, Fuselier JA, Nelson JA, O'Dorisio TM, Coy DH. Synthesis and characterization of multiply-tyrosinated, multiply-iodinated somatostatin analogs. THE JOURNAL OF PEPTIDE RESEARCH : OFFICIAL JOURNAL OF THE AMERICAN PEPTIDE SOCIETY 1999; 53:201-13. [PMID: 10195457 DOI: 10.1111/j.1397-002x.1999.00019.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Radio-labeled somatostatin analogs have recently gained popularity as agents useful in intraoperative tumor localization, external scintigraphy and in situ radiotherapy. We have synthesized and characterized a series of novel N-terminally extended multiply-tyrosinated somatostatin analogs that possess high binding affinity for somatostatin receptors, exhibit biological activity comparable to the native peptide and retain these characteristics after iodination. These analogs can be radio-iodinated to high specific activities. Following radioiodination, these analogs exhibit minimal radiolysis and may be clinically useful for tumor localization, scanning and therapy.
Collapse
Affiliation(s)
- E A Woltering
- Louisiana State University University Medical Center, Department of Surgery, Stanley S. Scott Cancer Center, New Orleans 70012-2822, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Carroll RE, Carroll R, Benya RV. Characterization of gastrin-releasing peptide receptors aberrantly expressed by non-antral gastric adenocarcinomas. Peptides 1999; 20:229-37. [PMID: 10422879 DOI: 10.1016/s0196-9781(98)00164-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Epithelial cells lining the GI tract except in the gastric antrum do not normally express gastrin-releasing peptide receptors (GRP-R). Because GRP-R activation causes the proliferation of many GI cancer cell lines, aberrant expression has been presumed to negatively influence patient survival. We therefore determined the incidence and quality of GRP-R aberrantly expressed by non-antral gastric adenocarcinomas, and evaluated the impact of receptor expression on patient survival. We studied RNA isolated from 20 consecutive non-antral gastric adenocarcinomas, and determined that 8 (40%) aberrantly expressed GRP-R. Of these, 6 (75%) were found to be mutated. Pharmacologically, the effect of these mutations ranged from rendering the GRP-R non-functional to constitutively active. Contrary to expectations, however, survival of patients whose tumor expressed functional GRP-R (18.5 +/- 9.8 months) was not statistically different from those that did not (8.3 +/- 1.8 months; p = 0.24). Thus our data indicate that mutated isoforms of GRP-R are commonly expressed by non-antral gastric adenocarcinomas. However, expression of functional GRP-R does not alter patient survival, suggesting that this receptor may not be clinically important to the growth of gastric cancers.
Collapse
Affiliation(s)
- R E Carroll
- Department of Medicine, University of Illinois at Chicago, and Chicago Veterans Administration Medical Center, 60612, USA
| | | | | |
Collapse
|
33
|
Frühwald MC, O'Dorisio MS, Cottingham SL, Qualman SJ, O'Dorisio TM. Neuropeptides in developmental tumors of the central and peripheral nervous system. Ann N Y Acad Sci 1998; 865:420-6. [PMID: 9928042 DOI: 10.1111/j.1749-6632.1998.tb11208.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- M C Frühwald
- Department of Pediatrics, Hematology and Oncology, Ohio State University, Columbus 43205, USA
| | | | | | | | | |
Collapse
|
34
|
Lelièvre V, Pineau N, Du J, Wen CH, Nguyen T, Janet T, Muller JM, Waschek JA. Differential effects of peptide histidine isoleucine (PHI) and related peptides on stimulation and suppression of neuroblastoma cell proliferation. A novel VIP-independent action of PHI via MAP kinase. J Biol Chem 1998; 273:19685-90. [PMID: 9677397 DOI: 10.1074/jbc.273.31.19685] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The growth rate of rodent embryonic neuroblasts and human neuroblastoma cell lines is regulated in part by autocrine or paracrine actions of neuropeptides of the family that includes vasoactive intestinal peptide (VIP), peptide histidine isoleucine (PHI), and pituitary adenylate cyclase-activating peptide (PACAP). These peptides act via seven transmembrane G-protein-linked receptors coupled to cAMP elevation, phospholipase C activation, intracellular Ca2+ release, and/or of mitogen-activated protein (MAP) kinase activation. Here we investigated the action of these peptides on the mouse neuroblastoma cell line Neuro2a. PHI and VIP inhibited proliferation at concentrations as low as 10(-13) M and 10(-10) M, respectively. In contrast, PACAP action was biphasic, with stimulation occurring at subnanomolar doses and inhibition at higher doses. Peptide actions were studied further by measuring cAMP and ERK1/2 MAP kinase activity and by assessing 3H-thymidine incorporation in conjunction with a panel of signal transduction pathways inhibitors. The data obtained indicated that the PHI-inhibitory and PACAP-stimulatory activities were mediated by corresponding changes in activity of the MAP kinase pathway and independent of protein kinase A (PKA) or protein kinase C (PKC). In contrast, the inhibitory actions of VIP and PACAP were specifically blocked by antagonists of PKA. Northern blot analysis revealed gene expression for only the PACAP-preferring (PAC1) receptor. However, binding experiments using 125I-labeled PACAP27, PHI, and VIP, demonstrated the presence of PACAP-preferring sites, bivalent VIP/PACAP sites, and PHI-binding sites that did not interact with VIP. The studies demonstrate potent regulatory actions of PACAP, PHI, and VIP on neuroblastoma cell proliferation which appear to be mediated by multiple subsets of receptors which differentially couple to MAP kinase and PKA signaling pathways.
Collapse
Affiliation(s)
- V Lelièvre
- Department of Psychiatry, Mental Retardation Research Center, UCLA, Neuropsychiatric Institute, Los Angeles, CA 90024, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Neurons in the adult rat superior cervical sympathetic ganglion (SCG) dramatically increase their content of vasoactive intestinal peptide (VIP) and its mRNA after axotomy in vivo and after explantation. Because the VIP gene contains a functional cAMP response element, the effects of cAMP-elevating agents on VIP expression were examined. VIP, forskolin, or isoproterenol increased cAMP accumulation in explanted ganglia. Secretin, a peptide chemically related to VIP, or forskolin increased VIP levels above those seen in ganglia cultured in control medium, whereas treatment with VIP or secretin increased the level of peptide histidine isoleucine (PHI), a peptide coded for by the same mRNA that encodes VIP. VIP or forskolin also increased VIP-PHI mRNA. In contrast, isoproterenol did not alter levels of VIP, PHI, or VIP-PHI mRNA. Although VIP or forskolin increased cAMP levels in both dissociated neurons and in non-neuronal cells, isoproterenol significantly stimulated cAMP accumulation only in the latter. VIP6-28 was an effective antagonist of the actions of exogenous VIP on cAMP and VIP-PHI mRNA in neuron-enriched cultures. When adult SCG explants were cultured in defined medium, endogenous VIP immunoreactivity was released. When VIP6-28 was added to such cultures, it significantly inhibited the increase in VIP-PHI mRNA that normally occurs. These data indicate that VIP, or a closely related molecule, produced by adult neurons after injury can enhance the expression of VIP. Such a mechanism may prolong the period during which VIP is elevated after axonal damage. The possibility is also discussed that, because VIP is present in preganglionic neurons in normal animals, its release during periods of increased sympathetic nerve activity could alter VIP expression in the SCG.
Collapse
|
36
|
Turman MA, Apple CA. Human proximal tubular epithelial cells express somatostatin: regulation by growth factors and cAMP. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 274:F1095-101. [PMID: 9841501 DOI: 10.1152/ajprenal.1998.274.6.f1095] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Somatostatin modulates several renal tubular cell functions, including gluconeogenesis and proliferation. In this study, we demonstrate that cultured human proximal tubular epithelial cells (PTEC) express somatostatin. We also demonstrate positive and negative regulation of PTEC somatostatin production. We found that PTEC derived from 14 different human donors consistently expressed somatostatin mRNA and/or peptide as detected by RT-PCR and enzyme-linked immunoassay. Furthermore, Northern blot analysis revealed that PTEC express the same size mRNA transcript (750 nucleotides) as human thyroid carcinoma (TT) cells. The PTEC mitogens, epidermal growth factor(EGF) and hydrocortisone, inhibit PTEC somatostatin secretion, whereas forskolin (a direct stimulator of adenylate cyclase) and fetal bovine serum stimulate secretion. These findings raise the possibility that renal-derived somatostatin modulates tubular cell function in an autocrine/paracrine manner. Manipulation of this pathway may lead to novel methods with which to alter tubular cell proliferation and function in vivo.
Collapse
Affiliation(s)
- M A Turman
- Department of Pediatrics, Ohio State University, Columbus, Ohio 43205, USA
| | | |
Collapse
|
37
|
Lelièvre V, Meunier AC, Caigneaux E, Falcon J, Muller JM. Differential expression and function of PACAP and VIP receptors in four human colonic adenocarcinoma cell lines. Cell Signal 1998; 10:13-26. [PMID: 9502113 DOI: 10.1016/s0898-6568(97)00067-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Human colonic adenocarcinoma cell lines have conserved several features of the native tissue. Among these is the expression of cell surface receptors for hormones and neurotransmitters that may be involved in the regulation of proliferation and differentiation processes in these cancer cells. Here, we confirm that high-affinity binding sites for the Vasoactive Intestinal Polypeptide (VIP) and for the VIP analogue Pituitary Adenylate-Cyclase Activating Polypeptide (PACAP), were expressed in 4 human colonic adenocarcinoma cell lines, HT29, SW403, DLD-1 and Caco-2, that spontaneously displayed variable phenotypic properties in culture. We demonstrated that after long-term treatments, VIP and PACAP significantly reduced cell proliferation in the 4 cell lines and modulated intracellular cAMP and cGMP levels. Furthermore, conspicuous differences were observed from one cell type to another concerning expression of the receptor subsets or the effects of the neuropeptides on cell growth and on cyclic nucleotides production.
Collapse
Affiliation(s)
- V Lelièvre
- Laboratoire de Biologie des Interactions Cellulaires, CNRS UMR 6558, Université de Poitiers, Faculté des Sciences, France
| | | | | | | | | |
Collapse
|
38
|
Vernallis AB, Hudson KR, Heath JK. An antagonist for the leukemia inhibitory factor receptor inhibits leukemia inhibitory factor, cardiotrophin-1, ciliary neurotrophic factor, and oncostatin M. J Biol Chem 1997; 272:26947-52. [PMID: 9341130 DOI: 10.1074/jbc.272.43.26947] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The leukemia inhibitory factor receptor (LIF-R) is activated not only by LIF, but also by cardiotrophin-1, ciliary neurotrophic factor with its receptor, and oncostatin M (OSM). Each of these cytokines induces the hetero-oligomerization of LIF-R with gp130, a signal-transducing subunit shared with interleukin-6 and interleukin-11. The introduction of mutations into human LIF that reduced the affinity for gp130 while retaining affinity for LIF-R has generated antagonists for LIF. In the current study, a LIF antagonist that was free of detectable agonistic activity was tested for antagonism against the family of LIF-R ligands. On cells that express LIF-R and gp130, all LIF-R ligands were antagonized. On cells that also express OSM receptor, OSM was not antagonized, demonstrating that the antagonist is specific for LIF-R. Ligand-triggered tyrosine phosphorylation of both LIF-R and gp130 was blocked by the antagonist. The antagonist is therefore likely to work by preventing receptor oligomerization.
Collapse
Affiliation(s)
- A B Vernallis
- CRC Growth Factor Group, School of Biochemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | | | | |
Collapse
|
39
|
Kogner P, Borgström P, Bjellerup P, Schilling FH, Refai E, Jonsson C, Dominici C, Wassberg E, Bihl H, Jacobsson H, Theodorsson E, Hassan M. Somatostatin in neuroblastoma and ganglioneuroma. Eur J Cancer 1997; 33:2084-9. [PMID: 9516858 DOI: 10.1016/s0959-8049(97)00212-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Neuroblastoma, a childhood tumour of the sympathetic nervous system, may in some cases differentiate to a benign ganglioneuroma or regress due to apoptosis. Somatostatin may inhibit neuroblastoma growth and induce apoptosis in vitro and was therefore investigated. Using a radioimmunoassay, we found that all ganglioneuromas contained high somatostatin concentrations (> 16 pmol/g), significantly higher than neuroblastomas (n = 117, median 2.8 pmol/g), healthy adrenals, Wilms' tumours, phaeochromocytomas and other neuroendocrine tumours (P < 0.001). Neuroblastomas contained more somatostatin than control tumours (P < 0.001-0.05). Neuroblastomas amplified for the MYCN oncogene contained less somatostatin than non-amplified tumours (1.2 pmol/g versus 4.0 pmol/g, respectively; P = 0.026). In a clinically unfavourable neuroblastoma subset (age > 12 months, stage 3 or 4) 16 children with high concentrations of somatostatin in primary tumours had a better prognosis than 23 with low somatostatin (46.7% versus 0% survival at 5 years, P < 0.005). Scintigraphy using 111In-pentetreotide identified tumours expressing high-affinity somatostatin receptors in vivo. However, no significant correlation was found between somatostatin receptor expression and peptide content in 15 tumours. Similarly, human SH-SY5Y neuroblastoma xenografts grown in nude rats showed low somatostatin concentrations, but were positive for somatostatin receptor scintigraphy. Treatment of these rats with the somatostatin analogue octreotide seemed to upregulate in vivo receptor expression of somatostatin and vasoactive intestinal peptide more effectively than 13-cis retinoic acid. In conclusion, somatostatin in neuroblastoma is associated with differentiation to benign ganglioneuromas in vivo and favourable outcome in advanced tumours. Furthermore, somatostatin receptor scintigraphy may identify tumours with high-affinity receptors in children that might benefit from targeted therapy using synthetic somatostatin analogues.
Collapse
Affiliation(s)
- P Kogner
- Dept. of Woman and Child Health, Karolinska Institute, Karolinska Hospital, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Vertongen P, De Clerck P, Fournet JC, Martelli H, Hélardot P, Devalck C, Peeters T, Sariban E, Robberecht P. Comparison between vasoactive intestinal polypeptide and pituitary adenylate cyclase activating polypeptide levels in neuroblastoma tumour tissues. Neuropeptides 1997; 31:409-13. [PMID: 9413016 DOI: 10.1016/s0143-4179(97)90033-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Vasoactive intestinal polypeptide (VIP) is reported to exert an autocrine control on neuroblastoma cell tumours: VIP is produced by the tumour and stimulates cell differentiation. This study tested the hypothesis that the parent peptide; the pituitary adenylate cyclase activating polypeptide (PACAP) may have a similar role. It was found that PACAP mRNA and PACAP were expressed in 12/12 tumours; it was also observed that PACAP receptor mRNA and functional PACAP receptors were expressed in 12/12 and 5/9 tumours, respectively. VIP mRNA and VIP were detected in 9/12 tumours. VIP receptor mRNA was expressed in 5/12 tumours and functional VIP receptors were never demonstrated. The tumours having the highest VIP levels also had the highest PACAP contents and were associated with a watery diarrhoea syndrome due to activation of intestinal VIP receptors. As PACAP recognizes the PACAP receptors and the VIP receptors with the same high affinity it may contribute to the syndrome and is a likely candidate for an autocrine control of neuroblastoma cell growth and differentiation.
Collapse
Affiliation(s)
- P Vertongen
- Laboratoire de Chimie Biologique et de la Nutrition, Faculté de Médecine, Université Libre de Bruxelles, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Tanaka J, Koshimura K, Murakami Y, Sohmiya M, Yanaihara N, Kato Y. Neuronal protection from apoptosis by pituitary adenylate cyclase-activating polypeptide. REGULATORY PEPTIDES 1997; 72:1-8. [PMID: 9404727 DOI: 10.1016/s0167-0115(97)01038-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is known to have trophic effects on neurons. Apoptosis of PC12 cells was induced by depletion of serum and nerve growth factor (NGF) from culture medium. Not only high potassium-induced Ca2+ channel activation but PACAP-38 at physiological concentrations (10[-10] to 10[-8] M) protected PC12 cells from apoptosis. PACAP-38 increased Ca2+ uptake and intracellular Ca2+ concentrations in PC12 cells. The effects of PACAP-38 on cell survival and Ca2+ channels were eliminated by inhibitors for Ca2+ channels and protein kinase A, and mimicked by 8-bromo-cAMP. Mitogen-activated protein (MAP) kinase activity was stimulated by PACAP-38. These findings implicate that PACAP protects PC12 cells from apoptosis by activating Ca2+ channels via the cAMP-protein kinase A pathway to stimulate MAP kinase cascade.
Collapse
Affiliation(s)
- J Tanaka
- Department of Medicine, Shimane Medical University, Izumo, Japan
| | | | | | | | | | | |
Collapse
|
42
|
Zigmond RE, Sun Y. Regulation of neuropeptide expression in sympathetic neurons. Paracrine and retrograde influences. Ann N Y Acad Sci 1997; 814:181-97. [PMID: 9160971 DOI: 10.1111/j.1749-6632.1997.tb46157.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Sympathetic neurons and other peripheral neurons exhibit a great deal of plasticity in their neuropeptide phenotype in adulthood. In this review, two phenotypes have been described in detail: that of normal sympathetic neurons and that of axotomized neurons. Two factors produced by nonneuronal cells, LIF and NGF, determine which of these phenotypes is expressed. Under normal conditions, the neurons receive NGF primarily, if not exclusively, from the target tissues they innervate. Prior to surgery, the nonneuronal cells within the ganglion and nerve tract express little, if any, LIF. This milieu favors the expression of NPY and suppresses the expression of VIP, galanin, and substance P (Fig. 6). After axotomy, however, this situation is reversed. The neuronal cell bodies are deprived of target-derived NGF and are exposed to LIF both within the ganglion and at the site of the injury (Fig 6). Both the removal of NGF and the exposure to LIF inhibit NPY expression, while promoting the expression of VIP and galanin. Expression of substance P after axotomy occurs primarily, if not entirely, because of the effects of LIF, with the removal of NGF playing no obvious role in the regulation of this peptide.
Collapse
Affiliation(s)
- R E Zigmond
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106-4975, USA.
| | | |
Collapse
|
43
|
Turman MA, O'Dorisio MS, O'Dorisio TM, Apple CA, Albers AR. Somatostatin expression in human renal cortex and mesangial cells. REGULATORY PEPTIDES 1997; 68:15-21. [PMID: 9094750 DOI: 10.1016/s0167-0115(96)00136-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Somatostatin modulates important physiologic functions of the kidney, including mesangial cell contraction, glomerular prostaglandin synthesis, and phosphate, water and sodium excretion. In diabetic nephropathy, somatostatin inhibits renal hypertrophy. High affinity somatostatin receptors are expressed in the kidney. Circulating somatostatin concentrations, however, are generally well below the affinity constants of known somatostatin receptors. Thus, we hypothesized that somatostatin is produced in the kidney and released locally to act in an autocrine/paracrine manner. Using reverse transcriptase and polymerase chain reaction (RT-PCR) analysis, we found that fresh human renal cortex and cultured human mesangial cells express somatostatin mRNA. Restriction enzyme and Southern blot analysis confirmed that RT-PCR cDNA products were derived from somatostatin mRNA. Radioimmunoassay of mesangial cell culture supernatants demonstrated SS-immunoreactive peptide (87 +/- 30 pg/ml compared to 19 +/- 9 pg/ml in medium not exposed to cells; P < 0.05). In contrast, renal cells did not transcribe detectable levels of vasoactive intestinal peptide (VIP) or neuropeptide Y (NPY) mRNA, nor did they synthesize measurable peptide. Our results demonstrate that renal cells produce somatostatin and suggest that kidney-derived somatostatin may regulate renal function in an autocrine/paracrine manner. Characterization of this pathway may lead to novel methods to alter the course of diabetic nephropathy and other renal diseases.
Collapse
Affiliation(s)
- M A Turman
- Department of Pediatrics, Ohio State University, Wexner Institute for Pediatric Research, Children's Hospital, Columbus 43205, USA
| | | | | | | | | |
Collapse
|
44
|
Waschek JA, Lelievre V, Bravo DT, Nguyen T, Muller JM. Retinoic acid regulation of the VIP and PACAP autocrine ligand and receptor system in human neuroblastoma cell lines. Peptides 1997; 18:835-41. [PMID: 9285932 DOI: 10.1016/s0196-9781(97)00015-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Neuroendocrine tumors, neuroblastoma in particular, commonly express the neuropeptides vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase activating peptide (PACAP) and their receptors. Retinoic acid (RA) has been shown to induce differentiation of neuroblastoma cell lines, possibly by augmenting or interfering with neuropeptide autocrine loops. We sought to determine which receptor gene subtypes are expressed in selected human neuroblastoma cell lines (SH-SY5Y, IMR-32, and LA-N-5), and the effect of RA on the VIP/PACAP ligand/receptor system. Expression of both PACAP1 and VIP1/PACAP2 receptor genes was detected by Northern analysis, which characteristically encode Type I (PACAP-preferring), and Type II (bivalent VIP/PACAP) receptors, respectively. Binding experiments carried out on IMR-32 cells, using 125I VIP and 125I PACAP-27 as tracers, corroborated that both receptor subtypes were expressed. In contrast to RA upregulation of VIP binding (confirmed here in IMR-32 cells), levels of both receptor mRNAs were reduced after RA treatment. VIP mRNA in each cell line was increased by RA, whereas PACAP mRNA, detected in IMR-32 cells only, was reduced. The studies indicate that several components of the VIP/PACAP autocrine system are regulated in neuroblastoma cell lines during RA differentiation.
Collapse
MESH Headings
- Autocrine Communication
- Gene Expression
- Humans
- Ligands
- Neuroblastoma/metabolism
- Neuropeptides/genetics
- Neuropeptides/metabolism
- Pituitary Adenylate Cyclase-Activating Polypeptide
- RNA, Messenger/metabolism
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I
- Receptors, Pituitary Hormone/genetics
- Receptors, Pituitary Hormone/metabolism
- Receptors, Vasoactive Intestinal Peptide/genetics
- Receptors, Vasoactive Intestinal Peptide/metabolism
- Receptors, Vasoactive Intestinal Polypeptide, Type I
- Tretinoin/pharmacology
- Tumor Cells, Cultured
- Up-Regulation/drug effects
- Vasoactive Intestinal Peptide/genetics
- Vasoactive Intestinal Peptide/metabolism
Collapse
Affiliation(s)
- J A Waschek
- Department of Psychiatry, University of California at Los Angeles 90024, USA.
| | | | | | | | | |
Collapse
|
45
|
Waschek JA. VIP and PACAP receptor-mediated actions on cell proliferation and survival. Ann N Y Acad Sci 1996; 805:290-300; discussion 300-1. [PMID: 8993411 DOI: 10.1111/j.1749-6632.1996.tb17491.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- J A Waschek
- Department of Psychiatry, University of California at Los Angeles 90024-1759, USA.
| |
Collapse
|
46
|
Park SK, O'Dorisio MS, O'Dorisio TM. Vasoactive intestinal polypeptide-secreting tumours: biology and therapy. BAILLIERE'S CLINICAL GASTROENTEROLOGY 1996; 10:673-96. [PMID: 9113317 DOI: 10.1016/s0950-3528(96)90018-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
VIP-secreting tumours are rare, but they produce a dramatic clinical picture, the most prominent feature being profuse, watery diarrhoea and hypokalaemia. VIPomas are malignant and require sophisticated diagnostic and localization techniques in order to identify their presence. Delays in diagnosis are the rule rather than the exception. Improvements in the diagnosis of VIPomas appear to result in an increase in resectability rates. A definitive diagnosis is aided by the determination of plasma VIP concentrations through the use of sensitive radioimmunoassays. With heightened awareness of this syndrome, increasing numbers of patients can be identified and more effective treatments developed for the refractory and recurrent tumours.
Collapse
Affiliation(s)
- S K Park
- Department of Pediatrics, Ohio State University College of Medicine, Columbus 43210, USA
| | | | | |
Collapse
|
47
|
Lelièvre V, Becq-Giraudon L, Meunier AC, Muller JM. Switches in the expression and function of PACAP and VIP receptors during phenotypic interconversion in human neuroblastoma cells. Neuropeptides 1996; 30:313-22. [PMID: 8914856 DOI: 10.1016/s0143-4179(96)90019-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Clonal human neuroblastoma cells SH-IN undergo a very conspicuous phenotypic change in culture. Large substrate-adherent cells with a slow growth rate give rise to small cells emerging in focal aggregates and growing to high cell densities. This is accompanied by a dramatic switch in the expression of receptors for the structurally related neuropeptides VIP (vasoactive intestinal polypeptide) and PACAP (pituitary adenylate cyclase activating polypeptide). Large cells expressed mainly PACAP-specific receptors that triggered stimulation of intracellular cGMP production. On the other hand, polyvalent VIP/PACAP receptors positively coupled to adenylate cyclase were mostly observed in the small cells. Both neuropeptides stimulated cell proliferation in large and small cells. These data, together with the previous demonstration of autocrine/paracrine actions of VIP and PACAP in human neuroblastomas, support the idea that these neuropeptides may participate in the establishment of the apparent phenotype in these cancer cells.
Collapse
Affiliation(s)
- V Lelièvre
- Laboratoire de Biologie des Interactions Cellulaires, CNRS, Université de Poitiers, France
| | | | | | | |
Collapse
|
48
|
Vertongen P, Devalck C, Sariban E, De Laet MH, Martelli H, Paraf F, Hélardot P, Robberecht P. Pituitary adenylate cyclase activating peptide and its receptors are expressed in human neuroblastomas. J Cell Physiol 1996; 167:36-46. [PMID: 8698838 DOI: 10.1002/(sici)1097-4652(199604)167:1<36::aid-jcp4>3.0.co;2-d] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Vasoactive intestinal peptide (VIP) has been considered as an autocrine growth factor in neuroblastomas. Pituitary adenylate cyclase activating polypeptides (PACAPs) are newly recognized members of the VIP family of neurohormones. As compared to VIP, PACAP has been reported to be biologically more potent and more efficient in tissues expressing selective PACAP receptors rather than common VIP/PACAP receptors. PACAPs and VIP interact with the same affinity and stimulate adenylate cyclase activity with the same efficacy and potency on the VIP receptors, but PACAPs act also on a more selective PACAP receptor that also recognizes VIP but with a 100- to 1,000-fold lower affinity. Thus, depending on the type of receptors expressed at a cell surface, PACAP may be more potent and efficient than VIP. The capacity of 22 surgical specimens of neuroblastomas and of 5 established cell lines to synthesize PACAP and VIP and to synthesize and express PACAP receptors and VIP receptors was studied. Using the reverse transcriptase-polymerase chain (RT-PCR) method with specific primers, we detected the mRNAs coding for PACAP and VIP in 19 and 3 out of 22 samples, respectively. PACAP mRNA was expressed in 3 of the 5 cell lines studied and VIP mRNA in 4. Using the same techniques, PACAP and VIP receptors mRNA were detected in 21, and 13 of the 22 tumor samples and in 5 and 1 of the cell lines studied, respectively. The expression of the PACAP receptor was demonstrated by direct binding studies and/or by the relative potency of PACAPs and VIP to stimulate adenylate cyclase activity in 16 of the 22 tumors and in all the cell lines. In addition, there was no correlation between tumor stage and the expression of mRNA coding for the peptides and the receptors. The present results demonstrated that PACAP could also be a candidate as an autocrine regulator of neuroblastoma which a higher activity than VIP.
Collapse
Affiliation(s)
- P Vertongen
- Department of Biochemistry and Nutrition, Medical School, Université Libre de Bruxelles, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Martinez DA, O'Dorisio MS, O'Dorisio TM, Qualman SJ, Caniano DA, Teich S, Besner GE, King DR. Intraoperative detection and resection of occult neuroblastoma: a technique exploiting somatostatin receptor expression. J Pediatr Surg 1995; 30:1580-9. [PMID: 8583329 DOI: 10.1016/0022-3468(95)90161-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Tumor cell expression of specific high-affinity somatostatin receptors has been associated with a favorable prognosis in children with neuroblastoma. The purpose of this study was (1) to document intraoperatively the in vivo binding of the somatostatin analogue 125I-tyr3-octreotide to high-affinity somatostatin receptors expressed on human neuroblastoma, using a hand-held gamma detector; (2) to determine whether gamma-probe detection of radioligand binding to tumor receptors could identify occult malignancy; and (3) to determine the safety and biodistribution of 125I-tyr3-octreotide in children. Six children with stage III or IV neuroblastoma received an intravenous injection of 125I-tyr3-octreotide and underwent operative exploration using gamma-probe detection of radioligand binding to tumor somatostatin receptors. Tissue that demonstrated in vivo binding of 125I-tyr3-octreotide, or that was suspicious for tumor, was extirpated and analyzed by histopathology, immunohistochemistry, and microautoradiography. The biodistribution of 125I-tyr3-octreotide was recorded intraoperatively over time. Tumor tissue from each child also was assayed in vitro for somatostatin receptor expression by competitive binding studies using 125I-tyr3-octreotide. In vivo binding of 125I-tyr3-octreotide to malignant tissue was documented in the five children with a known tumor burden. Seventeen sites of radioreceptor binding were amenable to resection. Histopathological analysis confirmed neuroblastoma in 15 of these specimens. Four of the 15 proven tumor foci were occult malignancies. Every site of histologically proven neuroblastoma demonstrated in vivo binding of 125I-tyr3-octreotide. Five of seven sites histologically negative for neuroblastoma also were negative for in vivo radioreceptor binding. Microautoradiography confirmed in vivo binding of 125I-tyr3-octreotide to tumor cells. Uptake of 125I-tyr3-octreotide in abdominal organs occurred within 15 minutes of injection, was highest in the liver and gallbladder, and decreased over 24 hours. The conclusions were as follows. (1) 125I-tyr3-octreotide binds, in vivo, to somatostatin receptors on neuroblastoma, with 100% sensitivity and 71% specificity. (2) Occult neuroblastoma is found through gamma-probe detection of radioligand binding to receptors. (2) The biodistribution of 125I-tyr3-octreotide reflects the hepatobiliary clearance of this radionuclide. (4) Radioreceptor-guided surgery may safely provide more complete operative staging and cytoreduction of neuroblastoma.
Collapse
MESH Headings
- Biomarkers, Tumor/metabolism
- Child, Preschool
- Female
- Gamma Rays
- Humans
- Intraoperative Care
- Iodine Radioisotopes
- Male
- Neoplasm Recurrence, Local
- Neoplasm, Residual/diagnostic imaging
- Neoplasm, Residual/metabolism
- Neoplasm, Residual/surgery
- Neoplasms, Unknown Primary/diagnostic imaging
- Neoplasms, Unknown Primary/metabolism
- Neoplasms, Unknown Primary/surgery
- Neuroblastoma/diagnostic imaging
- Neuroblastoma/metabolism
- Neuroblastoma/surgery
- Octreotide
- Radioimmunoassay
- Radionuclide Imaging
- Receptors, Somatostatin/metabolism
- Sensitivity and Specificity
Collapse
Affiliation(s)
- D A Martinez
- Department of Pediatrics, Ohio State University College of Medicine, Columbus 43205-2696, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Waschek JA, Richards ML, Bravo DT. Differential expression of VIP/PACAP receptor genes in breast, intestinal, and pancreatic cell lines. Cancer Lett 1995; 92:143-9. [PMID: 7600524 DOI: 10.1016/0304-3835(95)03768-r] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating peptide (PACAP) are structurally-related neuropeptides that function as trophic factors in addition to their more classical roles as neurotransmitters. Binding and molecular cloning studies have shown that their actions are mediated by receptors encoded by at least three different genes. VIP binding has been demonstrated on many tumor types, and radiolabeled VIP has recently been used as a novel method to localize intestinal tumors in humans and their sites of metastasis. To determine the receptor subtype and level of gene expression, we screened breast, intestinal, and pancreatic, cell lines by Northern blot analysis. Breast lines expressed VIP/PACAP1 receptor mRNA levels comparable to intestinal lines, in agreement with the studies showing particularly high VIP binding in these tumors and their derived cell lines. Pancreatic cell lines expressed mRNA for several receptor types. This extends the potential utility of VIP and PACAP in the localization of tumors, and because VIP and PACAP may regulate the growth rate of some tumors by autocrine or other mechanisms, the identification of receptor subtypes on these lines sets the stage for studies in which the activity of these individual receptors in growth and other processes can be investigated.
Collapse
Affiliation(s)
- J A Waschek
- Department of Psychiatry, University of California at Los Angeles 90024-1759, USA
| | | | | |
Collapse
|