Treatment of a multiple sclerosis animal model by a novel nanodrop formulation of a natural antioxidant

Orli Binyamin, Liraz Larush, Kati Frid, Guy Keller, Yael Friedman-Levi, Haim Ovadia, Oded Abramsky, Shlomo Magdassi, Ruth Gabizon, Orli Binyamin, Liraz Larush, Kati Frid, Guy Keller, Yael Friedman-Levi, Haim Ovadia, Oded Abramsky, Shlomo Magdassi, Ruth Gabizon

Abstract

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system and is associated with demyelination, neurodegeneration, and sensitivity to oxidative stress. In this work, we administered a nanodroplet formulation of pomegranate seed oil (PSO), denominated Nano-PSO, to mice induced for experimental autoimmune encephalomyelitis (EAE), an established model of MS. PSO comprises high levels of punicic acid, a unique polyunsaturated fatty acid considered as one of the strongest natural antioxidants. We show here that while EAE-induced mice treated with natural PSO presented some reduction in disease burden, this beneficial effect increased significantly when EAE mice were treated with Nano-PSO of specific size nanodroplets at much lower concentrations of the oil. Pathological examinations revealed that Nano-PSO administration dramatically reduced demyelination and oxidation of lipids in the brains of the affected animals, which are hallmarks of this severe neurological disease. We propose that novel formulations of natural antioxidants such as Nano-PSO may be considered for the treatment of patients suffering from demyelinating diseases. On the mechanistic side, our results demonstrate that lipid oxidation may be a seminal feature in both demyelination and neurodegeneration.

Keywords: EAE; PSO; nanodrops; neurodegeneration; oxidative stress.

Figures

Figure 1
Figure 1
Droplet size analysis. Notes: (A) DLS results for Nano-PSO and (B) cryo-TEM image of Nano-PSO. Abbreviations: DLS, dynamic light scattering; PSO, pomegranate seed oil; cryo-TEM, cryogenic transmission electron microscope.
Figure 2
Figure 2
Nano-PSO as an α-EAE agent. Notes: Mice were induced for EAE and treated from day 1 of the induction either with PSO or with Nano-PSO. (A) Designated EAE-induced groups were fed either with normal mouse chow (untreated group; n=10) or with chow enriched with PSO at the concentration in which 3 g (daily intake) comprises the levels designated in the figure insert: 100 (n=10) or 300 μL (n=8) PSO. P<0.05 for all PSO-treated groups versus the untreated group. (B) Designated EAE-induced groups were either left untreated or treated (by gavage) with 150 μL solution comprising 0.2, 0.8 (n=6), or 10 μL (n=7) PSO in the form of Nano-PSO. P<0.05 for 0.8 and 10 μL PSO-treated group versus the untreated group. Abbreviations: PSO, pomegranate seed oil; EAE, experimental autoimmune encephalomyelitis.
Figure 3
Figure 3
Individual α-EAE activity of Nano-PSO ingredients. Notes: Mice were induced for EAE and treated from day 1 of induction (by gavage) with the reagents described in the insert of the figure (n=7 for each of the groups). Nano-PSO was administrated at a dose of 2 μL PSO per 150 μL solution. Mice were scored daily for EAE signs for 2 additional weeks. P<0.05 for the results in the Nano-PSO group versus all others. Abbreviations: PSO, pomegranate seed oil; EAE, experimental autoimmune encephalomyelitis.
Figure 4
Figure 4
Nano-PSO in the prevention and treatment of EAE. Notes: Mice induced for EAE were administered Nano-PSO in two different start points. As shown in the insert, while one group of induced mice was left untreated (n=8), a second group was treated with Nano-PSO from day 1 of the induction (n=6) and a third group from day 7 of the induction (n=7). Mice were scored daily for EAE signs for 2 additional weeks. P<0.05 for both Nano-PSO treatments. Abbreviations: PSO, pomegranate seed oil; EAE, experimental autoimmune encephalomyelitis.
Figure 5
Figure 5
Small Nano-PSO particles are inactive against EAE. Notes: Mice induced for EAE were treated with Nano-PSO in different droplet sizes. As shown in the insert, while one group of induced mice was left untreated (n=8), a second group was treated with 180 nm droplets of Nano-PSO (n=7), and a third group with 30 nm Nano-PSO droplets (n=7). P<0.05 for the untreated group versus the group treated with 180 nm droplets of Nano-PSO. Abbreviations: PSO, pomegranate seed oil; EAE, experimental autoimmune encephalomyelitis.
Figure 6
Figure 6
Pathological markers of EAE in Nano-PSO treated and untreated mice. Notes: Nano-PSO treated and untreated mice were sacrificed 3 weeks after induction of EAE, and their formalin-fixed, paraffin-embedded brain sections as well as those of age-matched naïve mice (C and J) were stained by mAb EO6 (AD and F), H&E (E and G), and LFB/PAS (brains and spinal cords) (HJ). (D) and (F) represent an enlargement of the squares in (A) and (B); (E) and (G) are serial sections of (D) and (F), respectively. Arrows in (DG) indicate immune infiltrates. Arrows in (H) represent demyelinated areas. Abbreviations: EAE, experimental autoimmune encephalomyelitis; PSO, pomegranate seed oil; mAb, monoclonal antibody; H&E, hematoxylin and eosin; LFB, Luxol fast blue; PAS, periodic acid Schiff.
Figure 7
Figure 7
TBARS levels in EAE brains. Notes: Samples from naïve, as well as treated and untreated EAE brains were subjected to the TBARS test. Abbreviations: TBARS, thiobarbituric acid reactive substances; EAE, experimental autoimmune encephalomyelitis; MDA, malonaldehyde.

References

    1. Butterfield DA, Kanski J. Brain protein oxidation in age-related neurodegenerative disorders that are associated with aggregated proteins. Mech Ageing Dev. 2001;122(9):945–962.
    1. Aguzzi A, O’Connor T. Protein aggregation diseases: pathogenicity and therapeutic perspectives. Nat Rev Drug Discov. 2010;9(3):237–248.
    1. Morales R, Green KM, Soto C. Cross currents in protein misfolding disorders: interactions and therapy. CNS Neurol Disord Drug Targets. 2009;8(5):363–371.
    1. Canello T, Frid K, Gabizon R, et al. Oxidation of Helix-3 methionines precedes the formation of PK resistant PrP. PLoS Pathog. 2010;6(7):e1000977.
    1. Uttara B, Singh AV, Zamboni P, Mahajan RT. Oxidative stress and neurodegenerative diseases: a review of upstream and downstream antioxidant therapeutic options. Curr Neuropharmacol. 2009;7(1):65–74.
    1. Haider L, Fischer MT, Frischer JM, et al. Oxidative damage in multiple sclerosis lesions. Brain. 2011;134(pt 7):1914–1924.
    1. Perluigi M, Coccia R, Butterfield DA. 4-Hydroxy-2-nonenal, a reactive product of lipid peroxidation, and neurodegenerative diseases: a toxic combination illuminated by redox proteomics studies. Antioxid Redox Signal. 2012;17(11):1590–1609.
    1. Adibhatla RM, Hatcher JF. Lipid oxidation and peroxidation in CNS health and disease: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal. 2010;12(1):125–169.
    1. Reed TT. Lipid peroxidation and neurodegenerative disease. Free Radic Biol Med. 2011;51(7):1302–1319.
    1. Singh M, Dang TN, Arseneault M, Ramassamy C. Role of by-products of lipid oxidation in Alzheimer’s disease brain: a focus on acrolein. J Alzheimers Dis. 2010;21(3):741–756.
    1. Lassmann H. Mechanisms of inflammation induced tissue injury in multiple sclerosis. J Neurol Sci. 2008;274(1–2):45–47.
    1. Ellwardt E, Zipp F. Molecular mechanisms linking neuroinflammation and neurodegeneration in MS. Exp Neurol. 2014;262(pt A):8–17.
    1. Herz J, Zipp F, Siffrin V. Neurodegeneration in autoimmune CNS inflammation. Exp Neurol. 2010;225(1):9–17.
    1. Witte ME, Bol JG, Gerritsen WH, et al. Parkinson’s disease-associated parkin colocalizes with Alzheimer’s disease and multiple sclerosis brain lesions. Neurobiol Dis. 2009;36(3):445–452.
    1. Castegna A, Palmieri L, Spera I, et al. Oxidative stress and reduced glutamine synthetase activity in the absence of inflammation in the cortex of mice with experimental allergic encephalomyelitis. Neuroscience. 2011;185:97–105.
    1. Palinski W, Hörkkö S, Miller E, et al. Cloning of monoclonal autoantibodies to epitopes of oxidized lipoproteins from apolipoprotein E-deficient mice. Demonstration of epitopes of oxidized low density lipoprotein in human plasma. J Clin Invest. 1996;98(3):800–814.
    1. Mizrahi M, Friedman-Levi Y, Larush L, et al. Pomegranate seed oil nanoemulsions for the prevention and treatment of neurodegenerative diseases: the case of genetic CJD. Nanomedicine. 2014;10(6):1353–1363.
    1. Schubert SY, Lansky EP, Neeman I. Antioxidant and eicosanoid enzyme inhibition properties of pomegranate seed oil and fermented juice flavonoids. J Ethnopharmacol. 1999;66(1):11–17.
    1. Sawant RR, Torchilin VP. Multifunctionality of lipid-core micelles for drug delivery and tumour targeting. Mol Membr Biol. 2010;27(7):232–246.
    1. Merian J, Boisgard R, Decleves X, Theze B, Texier I, Tavitian B. Synthetic lipid nanoparticles targeting steroid organs. J Nucl Med. 2013;54(11):1996–2003.
    1. Margulis-Goshen K, Magdassi S. Formation of simvastatin nanoparticles from microemulsion. Nanomedicine. 2009;5(3):274–281.
    1. Kim D, Park JH, Kweon DJ, Han GD. Bioavailability of nanoemulsified conjugated linoleic acid for an antiobesity effect. Int J Nanomedicine. 2013;8:451–459.
    1. Dhopeshwarkar GA, Mead JF. Uptake and transport of fatty acids into the brain and the role of the blood–brain barrier system. Adv Lipid Res. 1973;11(0):109–142.
    1. Spector R. Fatty acid transport through the blood–brain barrier. J Neurochem. 1988;50(2):639–643.
    1. Avellini L, Terracina L, Gaiti A. Linoleic acid passage through the blood–brain barrier and a possible effect of age. Neurochem Res. 1994;19(2):129–133.
    1. Yuan G, Sinclair AJ, Xu C, Li D. Incorporation and metabolism of punicic acid in healthy young humans. Mol Nutr Food Res. 2009;53(10):1336–1342.
    1. Saha SS, Ghosh M. Antioxidant effect of vegetable oils containing conjugated linolenic acid isomers against induced tissue lipid peroxidation and inflammation in rat model. Chem Biol Interact. 2011;190(2–3):109–120.
    1. Shi C, Wu F, Zhu XC, Xu J. Incorporation of beta-sitosterol into the membrane increases resistance to oxidative stress and lipid peroxidation via estrogen receptor-mediated PI3K/GSK3beta signaling. Biochim Biophys Acta. 2013;1830(3):2538–2544.
    1. Kaufman M, Wiesman Z. Pomegranate oil analysis with emphasis on MALDI-TOF/MS triacylglycerol fingerprinting. J Agric Food Chem. 2007;55(25):10405–10413.
    1. Ben-Nun A, Mendel I, Bakimer R, et al. The autoimmune reactivity to myelin oligodendrocyte glycoprotein (MOG) in multiple sclerosis is potentially pathogenic: effect of copolymer 1 on MOG-induced disease. J Neurol. 1996;243(4 suppl 1):S14–S22.
    1. Friedman-Levi Y, Ovadia H, Hoftberger R, et al. Fatal neurological disease in scrapie-infected mice induced for experimental autoimmune encephalomyelitis. J Virol. 2007;81(18):9942–9949.
    1. Steinman L, Zamvil SS. Virtues and pitfalls of EAE for the development of therapies for multiple sclerosis. Trends Immunol. 2005;26(11):565–571.
    1. Gold R, Linington C, Lassmann H. Understanding pathogenesis and therapy of multiple sclerosis via animal models: 70 years of merits and culprits in experimental autoimmune encephalomyelitis research. Brain. 2006;129(pt 8):1953–1971.
    1. Brown DA, Sawchenko PE. Time course and distribution of inflammatory and neurodegenerative events suggest structural bases for the pathogenesis of experimental autoimmune encephalomyelitis. J Comp Neurol. 2007;502(2):236–260.
    1. Jose S, Anju SS, Cinu TA, Aleykutty NA, Thomas S, Souto EB. In vivo pharmacokinetics and biodistribution of resveratrol-loaded solid lipid nanoparticles for brain delivery. Int J Pharm. 2014;474(1–2):6–13.
    1. Blasi P, Schoubben A, Traina G, et al. Lipid nanoparticles for brain targeting III. Long-term stability and in vivo toxicity. Int J Pharm. 2013;454(1):316–323.
    1. Chen S, Tam YY, Lin PJ, Leung AK, Tam YK, Cullis PR. Development of lipid nanoparticle formulations of siRNA for hepatocyte gene silencing following subcutaneous administration. J Control Release. 2014;196:106–112.
    1. Wang P, Zhang L, Peng H, Li Y, Xiong J, Xu Z. The formulation and delivery of curcumin with solid lipid nanoparticles for the treatment of on non-small cell lung cancer both in vitro and in vivo. Mater Sci Eng C Mater Biol Appl. 2013;33(8):4802–4808.
    1. Gabizon R, Ovadia H, Abramsky O, Magdassi S, Larush L, inventors. Hadasit Medical Ressearch Services And Development Ltd., Yissum Research Development Company Of Tthe Hebrew University Of Jerusalem Ltd., assignees Pomegranate Oil For Preventing And Treating Neurodegenerative Diseases. 20150044314 A1. United States patent US. 2015 Feb 12;
    1. Guillen-Sans R, Guzman-Chozas M. The thiobarbituric acid (TBA) reaction in foods: a review. Crit Rev Food Sci Nutr. 1998;38(4):315–330.
    1. Karlik M, Valkovic P, Hancinova V, Krizova L, Tothova L, Celec P. Markers of oxidative stress in plasma and saliva in patients with multiple sclerosis. Clin Biochem. 2015;48(1–2):24–28.
    1. Kong W, Yen JH, Ganea D. Docosahexaenoic acid prevents dendritic cell maturation, inhibits antigen-specific Th1/Th17 differentiation and suppresses experimental autoimmune encephalomyelitis. Brain Behav Immun. 2013;25(5):872–882.
    1. Zhang Z, Gao F, Jiang S, et al. Bile salts enhance the intestinal absorption of lipophilic drug loaded lipid nanocarriers: mechanism and effect in rats. Int J Pharm. 2012;452(1–2):374–381.
    1. Schubert R, Schmidt KH. Structural changes in vesicle membranes and mixed micelles of various lipid compositions after binding of different bile salts. Biochemistry. 1988;27(24):8787–8794.
    1. Ljubisavljevic S. Oxidative stress and neurobiology of demyelination. Mol Neurobiol. 2014 Dec 11; Epub.
    1. Ferretti G, Bacchetti T. Peroxidation of lipoproteins in multiple sclerosis. J Neurol Sci. 2011;311(1–2):92–97.
    1. Leitinger N. The role of phospholipid oxidation products in inflammatory and autoimmune diseases: evidence from animal models and in humans. Subcell Biochem. 2008;49:325–350.
    1. Sellebjerg F, Sorensen PS. Therapeutic interference with leukocyte recirculation in multiple sclerosis. Eur J Neurol. 2015;22(3):434–442.
    1. Kizelsztein P, Ovadia H, Garbuzenko O, Sigal A, Barenholz Y. Pegylated nanoliposomes remote-loaded with the antioxidant tempamine ameliorate experimental autoimmune encephalomyelitis. J Neuroimmunol. 2009;213(1–2):20–25.
    1. Han JM, Lee YJ, Lee SY, et al. Protective effect of sulforaphane against dopaminergic cell death. J Pharmacol Exp Ther. 2007;321(1):249–256.
    1. Hatcher H, Planalp R, Cho J, Torti FM, Torti SV. Curcumin: from ancient medicine to current clinical trials. Cell Mol Life Sci. 2008;65(11):1631–1652.
    1. Choi YT, Jung CH, Lee SR, et al. The green tea polyphenol (−)- epigallocatechin gallate attenuates beta-amyloid-induced neurotoxicity in cultured hippocampal neurons. Life Sci. 2001;70(5):603–614.

Source: PubMed

3
Iratkozz fel