Brain targeting of 9c,11t-Conjugated Linoleic Acid, a natural calpain inhibitor, preserves memory and reduces Aβ and P25 accumulation in 5XFAD mice

Orli Binyamin, Keren Nitzan, Kati Frid, Yael Ungar, Hanna Rosenmann, Ruth Gabizon, Orli Binyamin, Keren Nitzan, Kati Frid, Yael Ungar, Hanna Rosenmann, Ruth Gabizon

Abstract

Deregulation of Cyclin-dependent kinase 5 (CDK5) by binding to the activated calpain product p25, is associated with the onset of neurodegenerative diseases, such as Alzheimer's disease (AD). Conjugated Linoleic Acid (CLA), a calpain inhibitor, is a metabolite of Punicic Acid (PA), the main component of Pomegranate seed oil (PSO). We have shown recently that long-term administration of Nano-PSO, a nanodroplet formulation of PSO, delays mitochondrial damage and disease advance in a mouse model of genetic Creutzfeldt Jacob disease (CJD). In this project, we first demonstrated that treatment of mice with Nano-PSO, but not with natural PSO, results in the accumulation of CLA in their brains. Next, we tested the cognitive, biochemical and pathological effects of long-term administration of Nano-PSO to 5XFAD mice, modeling for Alzheimer's disease. We show that Nano-PSO treatment prevented age-related cognitive deterioration and mitochondrial oxidative damage in 5XFAD mice. Also, brains of the Nano-PSO treated mice presented reduced accumulation of Aβ and of p25, a calpain product, and increased expression of COX IV-1, a key mitochondrial enzyme. We conclude that administration of Nano-PSO results in the brain targeting of CLA, and suggest that this treatment may prevent/delay the onset of neurodegenerative diseases, such as AD and CJD.

Conflict of interest statement

R.G. is one of the founders of Granalix Biotechnologies LTD, a company producing Granagard, a commercial version of Nano-PSO. O.B., K.N., K.F., Y.U. declare no potential conflict of interest.

Figures

Figure 1
Figure 1
Brain targeting of CLA following administration of Nano-PSO to WT mice. C57B mice were treated either with PSO or with Nano-PSO as described in the methods and subsequently lipids were extracted from brains and livers for detection of PA or CLA. (a) PA and CLA after a single dose of PSO or Nano-PSO administration (b) PA and CLA after continuous administration of PSO and Nano-PSO. Unpaired t-test: *p < 0.05; ***p < 0.001.
Figure 2
Figure 2
Nano-PSO administration to both 5XFAD and WT mice improved their performance on the T-maze test. 5XFAD and WT mice treated with or without Nano-PSO were subjected to the T-Maze test at 7 months of age as described in the methods. (a) cartoon of the experiment (b) Number of entries to the new arm on day 2. Statistical analysis were performed by planned contrast test of numbers of entries to unfamiliar arm reveal significant difference between the groups [f(3,38) = 14.009, p < 0.001)].
Figure 3
Figure 3
Effect of Nano-PSO treatment on long-term non-associative spatial memory in the open field habituation task. 5XFAD and WT mice treated with or without Nano-PSO were subjected to the open field habituation test at 7 months of age as described in the methods. (a) Cartoon of the experiment (b) Difference in movement duration between day 1 and day 2. One-way Anova (Tukey post hoc analysis) present significant difference between Tg untreated group (n = 12) and Tg treated group (***p < 0.001; n = 18).
Figure 4
Figure 4
Recognition of a novel object is improved by Nano-PSO treatment of 5XFAD mice. Nano-PSO treated and untreated 5XFAD and WT mice were subjected to the novel object recognition task at 10 months of age as described in the methods. (a) Cartoon of the experiment. (b) Percentage of time spent in boundary of new object in day 2. Statistical analysis by simple contrast test was performed and showed a trend toward a difference between the groups ([f (3,39) = 0.49 p = 0.077].
Figure 5
Figure 5
Nano-PSO treatment restored mitochondrial COX IV-1 expression in brains of 5XFAD mice. (a) Paraffin embedded brain sections of untreated 5XFAD mice (at 3, 5 and 10 months of age), treated Tg (10 months) as well as untreated and treated WT (10 months) mice were stained with α COX IV-1 mAb (red) and counterstained with dapi (blue). The magnification of cortex area presented in this figure is (x20). (b) Ten months old 5XFAD mice, treated (I) and untreated (II) with Nano-PSO, were double-stained with the 6E10 antibody (green) and α COX IV-1 mAb(red) and presented at a magnification of x60. Magnification of single cells (x100) presented for treated (III) and untreated (IV) 5XFAD brains. Cortical neurons from 10 mounts WT brain also stained with 6E10 and COX IV-1, presented in picture V (x100).
Figure 6
Figure 6
Amyloid plaque burden is decreased in brains of Nano-PSO treated 5XFAD mice. (a) Sagittal serial brains sections of - 5XFAD untreated mice in ages of 3, 5, 7 and 10 months and 10 months old Nano-PSO treated mice were immunostained with α-Aβ antibody (green; 6E10 mAb) (b) quantitative assessment of Aβ plaques burden per hemisphere of 10 months 5XFAD mice, treated (green) or untreated (red) with Nano-PSO, (four section of each brain; n = 5 per group: unpaired t-test; **p = 0.01; ***p < 0.001). (c) Enlargement of Aβ plaques from 10 m old untreated 5XFAD brain -and Nano-PSO treated 5XFAD brain. (d) Quantitative assessment of Thioflavin S dense-core plaques per hemisphere of treated and untreated 10 months 5Xfad mice (four section of each brain; n = 5 per group; unpaired t-test; ***p < 0.001). (e) Sagittal section of 10 months 5Xfad mice, treated and untreated, were stain with Thioflavin S, to label dense-core plaques in cortex (x20).
Figure 7
Figure 7
Decreased levels of Aβ in brains of 5XFAD mice treated with Nano-PSO. Brain homogenates of WT, untreated and treated 5XFAD transgenic mice were immunoblotted with the anti Aβ 6E10mAb. Figure (a) shows levels of APP and Aβ oligomers. Figure (b) shows Aβ monomers in brains. Beta-actin served as loading control (lower panel of b). In some cases, blots have been cropped and increased in exposure; full length original blots are presented in Supplementary Fig. 1. Figure (c) represent quantitative analysis of immunoreactive bands. The bars represent the relative levels of Aβ compared with beta actin and are expressed as percentage of the 3m WT value. Unpaired t-test (***p = 0.006).
Figure 8
Figure 8
Nano-PSO treatment reduced p25 levels in neurodegenerative brains. (a) Brain homogenates of 5XFAD and TgMHu2ME199K mice treated and untreated with Nano-PSO as well as brain homogenates from WT mice of different ages were immunoblotted with a p35/ p25 antibody. Blots have been cropped for conciseness; full length original blots are presented in Supplementary Fig. 2. (b) Quantitative analysis of immunoreactive bands of p25. The bars are expressed as percentage of the 3 m WT value. Unpaired t-test revealed significant difference between the treated and untreated 5XFAD mice (**p < 0.05) and treated and untreated TgMHu2ME199K mice (p < 0.02).

References

    1. Kovacs GG, Budka H. Prion diseases: from protein to cell pathology. Am J Pathol. 2008;172:555–565. doi: 10.2353/ajpath.2008.070442.
    1. Di Carlo M, Giacomazza D, Picone P, Nuzzo D, San Biagio PL. Are oxidative stress and mitochondrial dysfunction the key players in the neurodegenerative diseases? Free Radic Res. 2012;46:1327–1338. doi: 10.3109/10715762.2012.714466.
    1. Faris, R., Moore, R. A., Ward, A., Sturdevant, D. E. & Priola, S. A. Mitochondrial Respiration Is Impaired during Late-Stage Hamster Prion Infection. J Virol91, 10.1128/JVI.00524-17 (2017).
    1. Redmann M, Darley-Usmar V, Zhang J. The Role of Autophagy, Mitophagy and Lysosomal Functions in Modulating Bioenergetics and Survival in the Context of Redox and Proteotoxic Damage: Implications for Neurodegenerative Diseases. Aging Dis. 2016;7:150–162. doi: 10.14336/AD.2015.0820.
    1. Brown K, Mastrianni JA. The prion diseases. J Geriatr Psychiatry Neurol. 2010;23:277–298. doi: 10.1177/0891988710383576.
    1. Scheckel Claudia, Aguzzi Adriano. Prions, prionoids and protein misfolding disorders. Nature Reviews Genetics. 2018;19(7):405–418. doi: 10.1038/s41576-018-0011-4.
    1. Bhounsule AS, Bhatt LK, Prabhavalkar KS, Oza M. Cyclin dependent kinase 5: A novel avenue for Alzheimer’s disease. Brain Res Bull. 2017;132:28–38. doi: 10.1016/j.brainresbull.2017.05.006.
    1. Lee MS, et al. Neurotoxicity induces cleavage of p35 to p25 by calpain. Nature. 2000;405:360–364. doi: 10.1038/35012636.
    1. Shah K, Lahiri DK. Cdk5 activity in the brain - multiple paths of regulation. J Cell Sci. 2014;127:2391–2400. doi: 10.1242/jcs.147553.
    1. Camins A, Verdaguer E, Folch J, Pallas M. Involvement of calpain activation in neurodegenerative processes. CNS Drug Rev. 2006;12:135–148. doi: 10.1111/j.1527-3458.2006.00135.x.
    1. Liang B, Duan BY, Zhou XP, Gong JX, Luo ZG. Calpain activation promotes BACE1 expression, amyloid precursor protein processing, and amyloid plaque formation in a transgenic mouse model of Alzheimer disease. J Biol Chem. 2010;285:27737–27744. doi: 10.1074/jbc.M110.117960.
    1. Trinchese F, et al. Inhibition of calpains improves memory and synaptic transmission in a mouse model of Alzheimer disease. J Clin Invest. 2008;118:2796–2807. doi: 10.1172/JCI34254.
    1. Friedman-Levi Y, et al. Fatal prion disease in a mouse model of genetic E200K Creutzfeldt-Jakob disease. PLoS Pathog. 2011;7:e1002350. doi: 10.1371/journal.ppat.1002350.
    1. Keller G, Binyamin O, Frid K, Saada A, Gabizon R. Mitochondrial dysfunction in preclinical genetic prion disease: A target for preventive treatment? Neurobiol Dis. 2019;124:57–66. doi: 10.1016/j.nbd.2018.11.003.
    1. Mizrahi M, et al. Pomegranate seed oil nanoemulsions for the prevention and treatment of neurodegenerative diseases: the case of genetic CJD. Nanomedicine. 2014;10:1353–1363. doi: 10.1016/j.nano.2014.03.015.
    1. Yuan G, Sinclair AJ, Xu C, Li D. Incorporation and metabolism of punicic acid in healthy young humans. Mol Nutr Food Res. 2009;53:1336–1342. doi: 10.1002/mnfr.200800520.
    1. Tsuzuki T, et al. Conjugated linolenic acid is slowly absorbed in rat intestine, but quickly converted to conjugated linoleic acid. J Nutr. 2006;136:2153–2159. doi: 10.1093/jn/136.8.2153.
    1. Yuan GF, Yuan JQ, Li D. Punicic acid from Trichosanthes kirilowii seed oil is rapidly metabolized to conjugated linoleic acid in rats. J Med Food. 2009;12:416–422. doi: 10.1089/jmf.2007.0541.
    1. Pereira de Melo Illana Louise, de Oliveira e Silva Ana Mara, Yoshime Luciana Tedesco, Gasparotto Sattler José Augusto, Teixeira de Carvalho Eliane Bonifácio, Mancini-Filho Jorge. Punicic acid was metabolised and incorporated in the form of conjugated linoleic acid in different rat tissues. International Journal of Food Sciences and Nutrition. 2018;70(4):421–431. doi: 10.1080/09637486.2018.1519528.
    1. Lee E, et al. Effect of conjugated linoleic acid, mu-calpain inhibitor, on pathogenesis of Alzheimer’s disease. Biochim Biophys Acta. 2013;1831:709–718. doi: 10.1016/j.bbalip.2012.12.003.
    1. Oakley H, et al. Intraneuronal beta-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: potential factors in amyloid plaque formation. J Neurosci. 2006;26:10129–10140. doi: 10.1523/JNEUROSCI.1202-06.2006.
    1. Bilkei-Gorzo A. Genetic mouse models of brain ageing and Alzheimer’s disease. Pharmacol Ther. 2014;142:244–257. doi: 10.1016/j.pharmthera.2013.12.009.
    1. Webster SJ, Bachstetter AD, Nelson PT, Schmitt FA, Van Eldik LJ. Using mice to model Alzheimer’s dementia: an overview of the clinical disease and the preclinical behavioral changes in 10 mouse models. Front Genet. 2014;5:88. doi: 10.3389/fgene.2014.00088.
    1. Patrick GN, et al. Conversion of p35 to p25 deregulates Cdk5 activity and promotes neurodegeneration. Nature. 1999;402:615–622. doi: 10.1038/45159.
    1. Kimura R, Ohno M. Impairments in remote memory stabilization precede hippocampal synaptic and cognitive failures in 5XFAD Alzheimer mouse model. Neurobiol Dis. 2009;33:229–235. doi: 10.1016/j.nbd.2008.10.006.
    1. Girard SD, et al. Evidence for early cognitive impairment related to frontal cortex in the 5XFAD mouse model of Alzheimer’s disease. J Alzheimers Dis. 2013;33:781–796. doi: 10.3233/JAD-2012-120982.
    1. Higuchi M, Iwata N, Saido TC. Understanding molecular mechanisms of proteolysis in Alzheimer’s disease: progress toward therapeutic interventions. Biochim Biophys Acta. 2005;1751:60–67. doi: 10.1016/j.bbapap.2005.02.013.
    1. Binyamin O, et al. Continues administration of Nano-PSO significantly increased survival of genetic CJD mice. Neurobiol Dis. 2017;108:140–147. doi: 10.1016/j.nbd.2017.08.012.
    1. Wenk Gary L. Assessment of Spatial Memory Using the T Maze. Current Protocols in Neuroscience. 1998;4(1):8.5B.1-8.5A.7. doi: 10.1002/0471142301.ns0805bs04.
    1. Vianna MR, et al. Role of hippocampal signaling pathways in long-term memory formation of a nonassociative learning task in the rat. Learn Mem. 2000;7:333–340. doi: 10.1101/lm.34600.
    1. Hammond RS, Tull LE, Stackman RW. On the delay-dependent involvement of the hippocampus in object recognition memory. Neurobiol Learn Mem. 2004;82:26–34. doi: 10.1016/j.nlm.2004.03.005.
    1. Caspersen C, et al. Mitochondrial Abeta: a potential focal point for neuronal metabolic dysfunction in Alzheimer’s disease. FASEB J. 2005;19:2040–2041. doi: 10.1096/fj.05-3735fje.
    1. Manczak M, et al. Mitochondria are a direct site of A beta accumulation in Alzheimer’s disease neurons: implications for free radical generation and oxidative damage in disease progression. Hum Mol Genet. 2006;15:1437–1449. doi: 10.1093/hmg/ddl066.
    1. Twig G, Shirihai OS. The interplay between mitochondrial dynamics and mitophagy. Antioxid Redox Signal. 2011;14:1939–1951. doi: 10.1089/ars.2010.3779.
    1. Arnold S. Cytochrome c oxidase and its role in neurodegeneration and neuroprotection. Adv Exp Med Biol. 2012;748:305–339. doi: 10.1007/978-1-4614-3573-0_13.
    1. Shi XZ, Wei X, Sha LZ, Xu Q. Comparison of beta-Amyloid Plaque Labeling Methods: Antibody Staining, Gallyas Silver Staining, and Thioflavin-S Staining. Chin Med Sci J. 2018;33:167–173. doi: 10.24920/03476.
    1. Liu P, et al. Quantitative Comparison of Dense-Core Amyloid Plaque Accumulation in Amyloid-beta Protein Precursor Transgenic Mice. J Alzheimers Dis. 2017;56:743–761. doi: 10.3233/JAD-161027.
    1. Katsouri L, Georgopoulos S. Lack of LDL receptor enhances amyloid deposition and decreases glial response in an Alzheimer’s disease mouse model. PLoS One. 2011;6:e21880. doi: 10.1371/journal.pone.0021880.
    1. Vassar R. beta-Secretase, APP and Abeta in Alzheimer’s disease. Subcell Biochem. 2005;38:79–103. doi: 10.1007/0-387-23226-5_4.
    1. Binyamin O, et al. Treatment of a multiple sclerosis animal model by a novel nanodrop formulation of a natural antioxidant. Int J Nanomedicine. 2015;10:7165–7174. doi: 10.2147/IJN.S92704.
    1. Shelton VJ, Shelton AG, Azain MJ, Hargrave-Barnes KM. Incorporation of conjugated linoleic acid into brain lipids is not necessary for conjugated linoleic acid-induced reductions in feed intake or body fat in mice. Nutr Res. 2012;32:827–836. doi: 10.1016/j.nutres.2012.10.003.
    1. Bevers MB, et al. Knockdown of m-calpain increases survival of primary hippocampal neurons following NMDA excitotoxicity. J Neurochem. 2009;108:1237–1250. doi: 10.1111/j.1471-4159.2008.05860.x.
    1. Hung KS, et al. Calpain inhibitor inhibits p35-p25-Cdk5 activation, decreases tau hyperphosphorylation, and improves neurological function after spinal cord hemisection in rats. J Neuropathol Exp Neurol. 2005;64:15–26. doi: 10.1093/jnen/64.1.15.
    1. Wang KK, Yuen PW. Development and therapeutic potential of calpain inhibitors. Adv Pharmacol. 1997;37:117–152. doi: 10.1016/S1054-3589(08)60949-7.
    1. Liu P, et al. Characterization of a Novel Mouse Model of Alzheimer’s Disease–Amyloid Pathology and Unique beta-Amyloid Oligomer Profile. PLoS One. 2015;10:e0126317. doi: 10.1371/journal.pone.0126317.
    1. Kanungo J, Zheng YL, Amin ND, Pant HC. Targeting Cdk5 activity in neuronal degeneration and regeneration. Cell Mol Neurobiol. 2009;29:1073–1080. doi: 10.1007/s10571-009-9410-6.
    1. Lopes JP, Oliveira CR, Agostinho P. Role of cyclin-dependent kinase 5 in the neurodegenerative process triggered by amyloid-Beta and prion peptides: implications for Alzheimer’s disease and prion-related encephalopathies. Cell Mol Neurobiol. 2007;27:943–957. doi: 10.1007/s10571-007-9224-3.
    1. Robin G, et al. Calcium dysregulation and Cdk5-ATM pathway involved in a mouse model of fragile X-associated tremor/ataxia syndrome. Hum Mol Genet. 2017;26:2649–2666. doi: 10.1093/hmg/ddx148.
    1. Watts Joel C., Prusiner Stanley B. Experimental Models of Inherited PrP Prion Diseases. Cold Spring Harbor Perspectives in Medicine. 2017;7(11):a027151. doi: 10.1101/cshperspect.a027151.
    1. Hassen GW, et al. Effects of Novel Calpain Inhibitors in Transgenic Animal Model of Parkinson’s disease/dementia with Lewy bodies. Sci Rep. 2018;8:18083. doi: 10.1038/s41598-018-35729-1.
    1. Scialò Carlo, De Cecco Elena, Manganotti Paolo, Legname Giuseppe. Prion and Prion-Like Protein Strains: Deciphering the Molecular Basis of Heterogeneity in Neurodegeneration. Viruses. 2019;11(3):261. doi: 10.3390/v11030261.
    1. Fainstein N, Dan-Goor N, Ben-Hur T. Resident brain neural precursor cells develop age-dependent loss of therapeutic functions in Alzheimer’s mice. Neurobiol Aging. 2018;72:40–52. doi: 10.1016/j.neurobiolaging.2018.07.020.
    1. Creighton SD, et al. Dissociable cognitive impairments in two strains of transgenic Alzheimer’s disease mice revealed by a battery of object-based tests. Sci Rep. 2019;9:57. doi: 10.1038/s41598-018-37312-0.
    1. Benhamron S, Rozenstein-Tsalkovich L, Nitzan K, Abramsky O, Rosenmann H. Phos-tau peptide immunization of amyloid-tg-mice reduced non-mutant phos-tau pathology, improved cognition and reduced amyloid plaques. Exp Neurol. 2018;303:48–58. doi: 10.1016/j.expneurol.2018.02.004.
    1. Lotan A, et al. Differential effects of chronic stress in young-adult and old female mice: cognitive-behavioral manifestations and neurobiological correlates. Mol Psychiatry. 2018;23:1432–1445. doi: 10.1038/mp.2017.237.
    1. Picada JN, Schroder N, Izquierdo I, Henriques JA, Roesler R. Differential neurobehavioral deficits induced by apomorphine and its oxidation product, 8-oxo-apomorphine-semiquinone, in rats. Eur J Pharmacol. 2002;443:105–111. doi: 10.1016/s0014-2999(02)01553-4.
    1. Deacon RM, Koros E, Bornemann KD, Rawlins JN. Aged Tg2576 mice are impaired on social memory and open field habituation tests. Behav Brain Res. 2009;197:466–468. doi: 10.1016/j.bbr.2008.09.042.

Source: PubMed

3
Iratkozz fel