We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Chemical synthesis and characterization of silver-protected vasoactive intestinal peptide nanoparticles

    Rafael Fernandez-Montesinos*

    CABIMER-Andalusian Center for Molecular Biology and Regenerative Medicine, Seville, Spain.

    ,
    Paula M Castillo*

    CABIMER-Andalusian Center for Molecular Biology and Regenerative Medicine, Seville, Spain.

    University Pablo de Olavide, Seville, Spain

    ,
    Rebecca Klippstein

    CABIMER-Andalusian Center for Molecular Biology and Regenerative Medicine, Seville, Spain.

    ,
    Elena Gonzalez-Rey

    CABIMER-Andalusian Center for Molecular Biology and Regenerative Medicine, Seville, Spain.

    University of Seville, Seville, Spain

    ,
    Jose A Mejias

    University Pablo de Olavide, Seville, Spain

    ,
    Ana P Zaderenko

    University Pablo de Olavide, Seville, Spain

    &
    David Pozo

    † Author for correspondence

    CABIMER-Andalusian Center for Molecular Biology and Regenerative Medicine, Seville, Spain.

    Published Online:https://doi.org/10.2217/nnm.09.79

    We characterized a method to conjugate functional silver nanoparticles with vasoactive intestinal peptide (VIP), which could be used as a working model for further tailor-made applications based on VIP surface functionality. Despite sustained interest in the therapeutic applications of VIP, and the fact that its drugability could be largely improved by the attachament to functionalized metal nanoparticles, no methods have been described so far to obtain them. Materials & methods: VIP was conjugated to tiopronin-capped silver nanoparticles of a narrow size distribution, by means of proper linkers, to obtain VIP functionalized silver nanoparticles with two different VIP orientations (Ag–tiopronin–PEG–succinic–[His]VIP and Ag–tiopronin–PEG–VIP[His]). VIP intermediate nanoparticles were characterized by transmission-electron microscopy and Fourier transform infrared spectroscopy. VIP functionalized silver nanoparticles cytotoxicity was determined by lactate dehydrogenase release from mixed glial cultures prepared from cerebral cortices of 1–3 days-old C57/Bl mice. Cells were used for lipopolysaccharide stimulation at day 18–22 of culture. Results: Two different types of VIP-functionalized silver nanoparticles were obtained; both expose the C-terminal part of the neuropeptide, but in the first type VIP is attached to silver nanoparticle through its free amine terminus (Ag–tiopronin–PEG–succinic–[His]VIP), while in the second type, VIP N-terminus remains free (Ag–tiopronin–PEG–VIP[His]). VIP-functionalized silver nanoparticles did not compromise cellular viability and inhibited microglia-induced stimulation under inflammatory conditions. Conclusion: The chemical synthesis procedure developed to obtain VIP-functionalized silver nanoparticles rendered functional products, in terms of biological activity. The two alternative orientations designed, reduced the constraints for chemical synthesis that depends on the nanosurface to be functionalized. Our study provides, for the first time, a proof of principle to enhance the therapeutic potential of VIP with the valuable properties of metal nanoparticles for imaging, targeting and drug delivery.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    Bibliography

    • Wilson BB: Brain targeting PBCA nanoparticles and the blood–brain barrier. Nanomedicine4,499–502 (2009).
    • Blow N: Nanotechnology in biology: big collaborations for a small world. Nat. Methods5,569–574 (2008).
    • Jain KK: Role of nanobiotechnology in the development of personalized medicine. Nanomedicine4,249–252 (2009).
    • Dalby MJ: Nanostructured surfaces: cell engineering and cell biology. Nanomedicine4,247–248 (2009).
    • Bally M, Voros J: Nanoscale labels: nanoparticles and liposomes in the development of high-performance biosensors. Nanomedicine4,447–467 (2009).
    • Lu Y, Liu Gl, Lee LP: High-density silver nanoparticle film with temperature-controllable interparticle spacing for a tunable surface enhanced raman scattering substrate. Nano Lett.5,5–9 (2005).
    • Zheng J, Nicovich PR, Dickson RM: Highly fluorescent noble-metal quantum dots. Annu. Rev. Phys. Chem.58,409–431 (2007).
    • Souza GR, Christianson DR, Staquicini FI et al.: Networks of gold nanoparticles and bacteriophage as biological sensors and cell-targeting agents. Proc. Natl Acad. Sci. USA103,1215–1220 (2006).
    • Gong Jl, Liang Y, Huang Y et al.: Ag/SiO2 core-shell nanoparticle-based surface-enhanced RAMAN probes for immunoassay of cancer marker using silica-coated magnetic nanoparticles as separation tools. Biosens. Bioelectron.22,1501–1507 (2007).
    • 10  Tkachenko A, Xie H, Franzen S, Feldheim DL: Assembly and characterization of biomolecule-gold nanoparticle conjugates and their use in intracellular imaging. Methods Mol. Biol.303,85–99 (2005).
    • 11  Murphy CJ, Sau TK, Gole AM et al.: Anisotropic metal nanoparticles: synthesis, assembly, and optical applications. J. Phys. Chem. B109,13857–13870 (2005).
    • 12  Nowacek A, Gendelman HE: NanoART, neuroAIDS and CNS drug delivery. Nanomedicine4,557–574 (2009).
    • 13  Tsoli M, Kuhn H, Brandau W, Esche H, Schmid G: Cellular uptake and toxicity of Au55 clusters. Small1,841–844 (2005).
    • 14  Bhattacharya R, Mukherjee P, Xiong Z, Atala A, Soker S, Mukhopadhyay D: Gold nanoparticles inhibit VEGF165-induced proliferation of HUVEC cells. Nano Lett.4,2479–2481 (2004).
    • 15  Mukherjee P, Bhattacharya R, Wang P et al.: Antiangiogenic properties of gold nanoparticles. Clin. Cancer Res.11,3530–3534 (2005).
    • 16  Tsai CY, Shiau Al, Chen SY et al.: Amelioration of collagen-induced arthritis in rats by nanogold. Arthritis Rheum.56,544–554 (2007).
    • 17  Castillo PM, Herrera JL, Fernández-Montesinos R et al.: Tiopronin monolayer-protected silver nanoparticles modulates interleukin-6 secretion mediated by toll-like receptor ligands. Nanomedicine3,627–635 (2008).
    • 18  Connor EE, Mwamuka J, Gole A, Murphy CJ, Wyatt MD: Gold nanoparticles are taken up by human cells but do not cause acute cytotoxicity. Small1,325–327 (2005).
    • 19  Thrall L: A nano trojan horse. Environ. Sci. Technol.41,3791–3792 (2007).
    • 20  Stern ST, McNeil SE: Nanotechnology safety concerns revisited. Toxicol. Sci.101,4–21 (2008).
    • 21  Linkov I, Satterstrom FK, Corey LM: Nanotoxicology and Nanomedicine: making hard decisions. Nanomedicine4,167–171 (2008).
    • 22  Resnik DB, Tinkle SS: Ethics in nanomedicine. Nanomedicine2,345–350 (2007).
    • 23  Huang T, Murray RW: Visible luminescence of water-soluble monolayer-protected gold clusters. J. Phys. Chem. B105,12498–12502 (2001).
    • 24  Kogan MJ, Olmedo I, Hosta L, Guerrero AR, Cruz LJ, Albericio F: Peptides and metallic nanoparticles for biomedical applications. Nanomedicine2,287–306 (2007).
    • 25  Kobayashi H, Ogawa M, Kosaka N, Choyke Pl, Urano Y: Multicolor imaging of lymphatic function with two nanomaterials: quantum dot-labeled cancer cells and dendrimer-based optical agents. Nanomedicine4,411–419 (2009).
    • 26  Said S, Mutt V: Polypeptide with broad biological activity: isolation from small intestine. Science169,1217–1218 (1970).▪ First report on the isolation of vasoactive intestinal peptide (VIP).
    • 27  Gozes I: VIP, from gene to behavior and back: summarizing my 25 years of research. J. Mol. Neurosci.36,115–124 (2008).▪▪ Personal account of the main findings on the VIP biological roles by one of the leading experts in the field.
    • 28  Harmar A, Arimura A, Gozes I et al.: International union of pharmacology. XVIII. Nomenclature of receptors for vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide. Pharmacol. Rev.50,265–270 (1998).
    • 29  Gonzalez-Rey E, Chorny A, Delgado M: Regulation of immune tolerance by anti-inflammatory neuropeptides. Nat. Rev. Immunol.7,52–63 (2007).▪ Extensive reviews on the role of VIP in immune responses.
    • 30  Pozo D, Delgado M: The many faces of VIP in neuroimmunology: a cytokine rather a neuropeptide? FASEB J.18,1325–1334 (2004).▪ Extensive reviews on the role of VIP in immune responses.
    • 31  Gonzalez-Rey E, Fernandez-Martin A, Chorny A et al.: Therapeutic effect of vasoactive intestinal peptide on experimental autoimmune encephalomyelitis: down-regulation of inflammatory and autoimmune responses. Am. J. Pathol.168,1179–1188 (2006).
    • 32  Zhang K, Aruva MR, Shanthly N et al.: PET imaging of VPAC1 expression in experimental and spontaneous prostate cancer. J. Nucl. Med.49,112–121 (2008).
    • 33  Reubi JC, Maecke HR: Peptide-based probes for cancer imaging. J. Nucl. Med.49,1735–1738 (2008).
    • 34  Reubi JC, Horisberger U, Kappeler A, Laissue JA: Localization of receptors for vasoactive intestinal peptide, somatostatin, and substance P in distinct compartments of human lymphoid organs. Blood92,191–197 (1998).
    • 35  El Battari A, Martin JM, Luis J et al.: Solubilization of the active vasoactive intestinal peptide receptor from human colonic adenocarcinoma cells. J. Biol. Chem.263,17685–17689 (1988).
    • 36  Harmar AJ: Family-B G-protein-coupled receptors. Genome Biol.2,1–10 (2001).
    • 37  Nicole P, Lins L, Rouyer-Fessard C et al.: Identification of key residues for interaction of vasoactive intestinal peptide with human VPAC1 and VPAC2 receptors and development of a highly selective VPAC1 receptor agonist. Alanine scanning and molecular modeling of the peptide. J. Biol. Chem.275,24003–24012 (2000).
    • 38  Ceraudo E, Murail S, Tan YV et al.: The vasoactive intestinal peptide (VIP) α-helix up to c terminus interacts with the N-terminal ectodomain of the human VIP/pituitary adenylate cyclase-activating peptide 1 receptor: photoaffinity, molecular modeling, and dynamics. Mol. Endocrinol.22,147–155 (2008).▪ Structure–function analysis of the interaction between the C-terminal VIP and VIP receptors.
    • 39  Tan YV, Couvineau A, Murail S et al.: Peptide agonist docking in the N-terminal ectodomain of a class II G protein-coupled receptor, the VPAC1 receptor. Photoaffinity, NMR, and molecular modeling. J. Biol. Chem.281,12792–12798 (2006).▪ Structure–function analysis of the interaction between the C-terminal VIP and VIP receptors.
    • 40  Tan YV, Couvineau A, Van Rampelbergh J, Laburthe M: Photoaffinity labeling demonstrates physical contact between vasoactive intestinal peptide and the N-terminal ectodomain of the human VPAC1 receptor. J. Biol. Chem.278,36531–36536 (2003).▪ Structure–function analysis of the interaction between the C-terminal VIP and VIP receptors.
    • 41  Tan YV, Couvineau A, Laburthe M: Diffuse pharmacophoric domains of vasoactive intestinal peptide (VIP) and further insights into the interaction of VIP with the N-terminal ectodomain of human VPAC1 receptor by photoaffinity labeling with [BPA6]-VIP. J. Biol. Chem.279,38889–38894 (2004).▪ Structure–function analysis of the interaction between the C-terminal VIP and VIP receptors.
    • 42  Ceraudo E, Tan YV, Nicole P, Couvineau A, Laburthe M: The N-terminal parts of VIP and antagonist PG97–269 physically interact with different regions of the human VPAC1 receptor. J. Mol. Neurosci.36,245–248. (2008).
    • 43  Gao X, Wu B, Zhang Q et al.: Brain delivery of vasoactive intestinal peptide enhanced with the nanoparticles conjugated with wheat germ agglutinin following intranasal administration. J. Control. Release121,156–167 (2007).
    • 44  Lim S, Rubinstein I, Onyuksel H: Freeze drying of peptide drugs self-associated with long-circulating, biocompatible and biodegradable sterically stabilized phospholipid nanomicelles. Int. J. Pharm.356,345–350 (2008).
    • 45  de la Fuente JM, Riehle MO, Curtis AS: Nanoparticle targeting at cells. Langmuir22,3286–3293 (2006).▪ Pioneering work for peptide functionalization of metal nanoparticles.
    • 46  Han S, Kim Y: Recent development of peptide coupling reagents in organic synthesis. Tetrahedron60,2447–2467 (2004).
    • 47  Montalbetti C, Falque V: Amide bond formation and peptide coupling Tetrahedron61,10827–10852 (2005).
    • 48  Saura J, Tusell JM, Serratosa J: High-yield isolation of murine microglia by mild trypsinization. Glia44,183–189 (2003).
    • 49  Delgado M, Ganea D: Vasoactive intestinal peptide prevents activated microglia-induced neurodegeneration under inflammatory conditions: potential therapeutic role in brain trauma. FASEB J.17,1922–1924 (2003).
    • 50  Detar DF, Silverstein SR: The mechanisms of the reactions of acetic acid with dicyclohexylcarbodiimide. J. Am. Chem. Soc.88,1013–1019 (1996).
    • 51  Grabarek Z, Gerjely J: Zero-length crosslinking procedure with the use of active esters. Anal. Biochem.185,131–135 (1990).
    • 52  Staros JV, Wright RW, Swingle DM: Enhancement by N-hydroxysulfosuccinimide of water-soluble carbodiimide-mediated coupling reactions. Anal. Biochem.156,220–222 (1986).
    • 53  Owens III, Peppas, N: Opsonization, biodistribution, and pharmacokinetics of polymeric nanoparticles. Int. J. Pharm.307,93–102 (2006).
    • 54  Dangoor D, Rubinraut S, Fridkin M, Gozes I: Novel extended and branched N-terminal analogs of VIP. Regul. Pept.137,42–49 (2006).▪ Extensive report on the interaction between NH2 terminal VIP and VIP receptors.
    • 55  Delgado M, Ganea D: Neuroprotective effect of vasoactive intestinal peptide (VIP) in a mouse model of Parkinson’s Disease by blocking microglial activation. FASEB J.17,944–946 (2003).
    • 56  Pozo D, Gonzalez-Rey E, Chorny A, Anderson P, Varela N, Delgado M: Tuning immune tolerance with vasoactive intestinal peptide: a new therapeutic approach for immune disorders. Peptides28,1833–1846 (2007).▪ Extensive reviews on the role of VIP in immune responses.
    • 57  Gonzalez-Rey E, Delgado M: Vasoactive intestinal peptide inhibits cyclooxygenase-2 expression in activated macrophages, microglia, and dendritic cells. Brain Behav. Immun.22,35–41 (2008).
    • 58  Stewart AJ, Fox A, Morimoto BH, Gozes I: Looking for novel ways to treat the hallmarks of Alzheimer’s disease. Expert Opin. Investig. Drugs16,1183–1196 (2007).
    • 59  Rolls A, Shechter R, Schwartz M: The bright side of the glial scar in CNS repair. Nat. Rev. Neurosci.10,235–241 (2009).
    • 60  Franklin RJ, Ffrench-Constant C: Remyelination in the CNS: from biology to therapy. Nat. Rev. Neurosci.9,839–855 (2008).
    • 61  Yu DG: Formation of colloidal silver nanoparticles stabilized by Na+-poly(g-glutamic acid)-silver nitrate complex via chemical reduction process. Colloids Surf. B Biointerfaces59,171–178 (2007).
    • 62  Lucarelli M, Gatti AM, Savarino G et al.: Innate defence functions of macrophages can be biased by nano-sized ceramic and metallic particles. Eur. Cytokine Netw.15,339–346 (2004).
    • 63  Wagner AJ, Bleckmann CA, Murdock RC, Schrand AM, Schlager JJ, Hussain SM: Cellular interaction of different forms of aluminum nanoparticles in rat alveolar macrophages. J. Phys. Chem. B111,7353–7359 (2007).
    • 64  Delgado M, Leceta J, Ganea D: Vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide inhibit the production of inflammatory mediators by activated microglia. J. Leukoc. Biol.73,155–164 (2003).
    • 65  Fonseca JE, Santos MJ, Canhao H, Choy E: Interleukin-6 as a key player in systemic inflammation and joint destruction. Autoimmun. Rev.8,538–542 (2009).
    • 66  Bromberg J, Wang TC: Inflammation and cancer: IL-6 and STAT3 complete the link. Cancer Cell15,79–80 (2009).
    • 67  Tweedie D, Luo W, Short RG et al.: A cellular model of inflammation for identifying TNF-α synthesis inhibitors. J. Neurosci. Methods183(2),182–187 (2009).
    • 68  Pozo D: Immune-based disorders: the challenges for translational immunology. J. Cell. Mol. Med.12,1085–1086 (2008).
    • 69  Anderson P, Delgado M: Endogenous anti-inflammatory neuropeptides and proresolving lipid mediators: a new therapeutic approach for immune disorders. J. Cell. Mol. Med.12,1830–1847 (2008).
    • 70  Delgado M, Muñoz-Elias E, Gomariz R, Ganea D: Vasoactive intestinal peptide and pituitary adenylate cyclase activating polypeptide enhance IL-10 production by murine macrophages: in vitro and in vivo studies. J. Immunol.162,1707–1716 (1999).
    • 71  Kim WK, Ganea D, Jonakait GM: Inhibition of microglial CD40 expression by pituitary adenylate cyclase-activating polypeptide is mediated by interleukin-10. J. Neuroimmunol.126,16–24 (2002).
    • 72  Campos-Salinas J, Gonzalez-Rey E: Autophagy and neuropeptides at the crossroad for parasites: to survive or to die? Autophagy5,551–554 (2009).
    • 73  Delgado M, Anderson P, Garcia-Salcedo JA, Caro M, Gonzalez-Rey E: Neuropeptides kill african trypanosomes by targeting intracellular compartments and inducing autophagic-like cell death. Cell Death Differ.16,406–416 (2009).