Generic placeholder image

Current Enzyme Inhibition

Editor-in-Chief

ISSN (Print): 1573-4080
ISSN (Online): 1875-6662

Research Article

Lactobacillus Rhamnosus UBLR-58 and Diclofenac Potentiate the Anti- Alzheimer Activity of Curcumin in Mice

Author(s): Sonal Pande, Chirag Patel*, Dipta Sarkar and Sanjeev Acharya

Volume 17, Issue 1, 2021

Published on: 17 August, 2020

Page: [49 - 56] Pages: 8

DOI: 10.2174/1573408016999200817170821

Price: $65

Abstract

Background: Curcumin, a providential spice, has its role in protecting the brain from neurodegeneration. Despite its ubiquitous role, it is not exploited alone due to its hampered bioavailability. By restraining the intestinal and liver enzymatic metabolism, one can boost the bioavailability of curcumin and promotes reabsorption of the curcumin. Diclofenac inhibits uridine 5'-diphosphoglucuronosyltransferase enzymes specifically responsible for the metabolism and elimination of curcumin. Lactobacillus rhamnosus is able to synthesize and release the β -d-glucuronidase enzyme, which reverts curcumin into the active form.

Objective: In this research, we aim to combine curcumin with Lactobacillus rhamnosus and diclofenac as an adjuvant with curcumin to potentiate anti-Alzheimer effect in mice impaired with memory by scopolamine.

Methods: To induce amnesia, scopolamine was used in mice model (1mg/kg, daily for 10 days i.p.). After execution of behavioural tests (Morris Water Maze test), brain and liver were isolated for further neurochemical and histopathology examination.

Results: Our finding showed a marked rise in the level of antioxidant enzymes in curcumin with L. rhamnosus and diclofenac compared to curcumin alone. Additionally, the behavioural study revealed that cognition in mice with curcumin adjuvant with L. rhamnosus and diclofenac showed a marked improvement. The histology study proves that curcumin alone possesses less and a non-significant neuroprotective effect as compared to curcumin with L. rhamnosus and diclofenac.

Conclusion: This entire outcome ratifies that curcumin with L. rhamnosus and diclofenac has higher activity as compared to curcumin alone, which reversed the cognition in the Alzheimer disease model.

Keywords: Alzheimer's disease, curcumin, dementia, diclofenac, Lactobacillus rhamnosus, probiotics.

Graphical Abstract
[1]
Chesser AS, Pritchard SM, Johnson GV. Tau clearance mechanisms and their possible role in the pathogenesis of Alzheimer disease. Front Neurol 2013; 4: 122.
[http://dx.doi.org/10.3389/fneur.2013.00122] [PMID: 24027553]
[2]
Kametani F, Hasegawa M. Reconsideration of amyloid hypothesis and tau hypothesis in Alzheimer’s disease. Front Neurosci 2018; 12: 25.
[http://dx.doi.org/10.3389/fnins.2018.00025] [PMID: 29440986]
[3]
Weller J, Budson A. Current understanding of Alzheimer’s disease diagnosis and treatment. F1000. Res Rev 2018; 1161: 7.
[4]
Sathianathan R, Kantipudi SJ. The dementia epidemic: Impact, prevention, and challenges for India. Indian J Psychiatry 2018; 60(2): 165-7.
[http://dx.doi.org/10.4103/psychiatry.IndianJPsychiatry_261_18] [PMID: 30166671]
[5]
Li HL, Liu C, de Couto G, et al. Curcumin prevents and reverses murine cardiac hypertrophy. J Clin Invest 2008; 118(3): 879-93.
[http://dx.doi.org/10.1172/JCI32865] [PMID: 18292803]
[6]
Tang M, Taghibiglou C. The mechanisms of action of curcumin in Alzheimer’s disease. J Alzheimers Dis 2017; 58(4): 1003-16.
[http://dx.doi.org/10.3233/JAD-170188] [PMID: 28527218]
[7]
Gupta SC, Kismali G, Aggarwal BB. Curcumin, a component of turmeric: from farm to pharmacy. Biofactors 2013; 39(1): 2-13.
[http://dx.doi.org/10.1002/biof.1079] [PMID: 23339055]
[8]
Anand P, Kunnumakkara AB, Newman RA, Aggarwal BB, Ajaikumar B. Bioavailability of curcumin: problems and promises. Mol Pharm 2007; 4(6): 807-18.
[http://dx.doi.org/10.1021/mp700113r] [PMID: 17999464]
[9]
Prasad S, Tyagi AK, Aggarwal BB. Recent developments in delivery, bioavailability, absorption and metabolism of curcumin: the golden pigment from golden spice. Cancer Res Treat 2014; 46(1): 2-18.
[http://dx.doi.org/10.4143/crt.2014.46.1.2] [PMID: 24520218]
[10]
Chhouk K, Kanda H, Kawasaki SI, Goto M. Micronization of curcumin with the biodegradable polymer by supercritical anti-solvent using micro swirl mixer. Front ChemSciEng 2018; 12(1): 184-93.
[http://dx.doi.org/10.1007/s11705-017-1678-3]
[11]
Xue J, Wang T, Hu Q, Zhou M, Luo Y. Insight into natural biopolymer-emulsified solid lipid nanoparticles for encapsulation of curcumin: Effect of loading methods. Food Hydrocoll 2018; 79: 110-6.
[http://dx.doi.org/10.1016/j.foodhyd.2017.12.018]
[12]
Abdel-Hafez SM, Hathout RM, Sammour OA. Curcumin-loaded ultradeformable nanovesicles as a potential delivery system for breast cancer therapy. Colloids Surf B Biointerfaces 2018; 167: 63-72.
[http://dx.doi.org/10.1016/j.colsurfb.2018.03.051] [PMID: 29626721]
[13]
Patel C, Pande S, Acharya S. Potentiation of anti-Alzheimer activity of curcumin by probiotic Lactobacillus rhamnosus UBLR-58 against scopolamine-induced memory impairment in mice. Naunyn Schmiedebergs Arch Pharmacol 2020; 25: 1-8.
[PMID: 32448977]
[14]
Rowland A, Miners JO, Mackenzie PI. The UDP-glucuronosyl transferases: their role in drug metabolism and detoxification. Int J Biochem Cell Biol 2013; 45(6): 1121-32.
[http://dx.doi.org/10.1016/j.biocel.2013.02.019] [PMID: 23500526]
[15]
Uchaipichat V, Mackenzie PI, Guo XH, et al. Human udp-glucuronosyltransferases: isoform selectivity and kinetics of 4-methylumbelliferone and 1-naphthol glucuronidation, effects of organic solvents, and inhibition by diclofenac and probenecid. Drug Metab Dispos 2004; 32(4): 413-23.
[http://dx.doi.org/10.1124/dmd.32.4.413] [PMID: 15039294]
[16]
Kuehl GE, Lampe JW, Potter JD, Bigler J. Glucuronidation of nonsteroidal anti-inflammatory drugs: identifying the enzymes responsible in human liver microsomes. Drug Metab Dispos 2005; 33(7): 1027-35.
[http://dx.doi.org/10.1124/dmd.104.002527] [PMID: 15843492]
[17]
Joo J, Kim YW, Wu Z, et al. Screening of non-steroidal anti-inflammatory drugs for inhibitory effects on the activities of six UDP-glucuronosyltransferases (UGT1A1, 1A3, 1A4, 1A6, 1A9 and 2B7) using LC-MS/MS. Biopharm Drug Dispos 2015; 36(4): 258-64.
[http://dx.doi.org/10.1002/bdd.1933] [PMID: 25522350]
[18]
Ozawa H, Imaizumi A, Sumi Y, et al. Curcumin β-D-glucuronide plays an important role to keep high levels of free-form curcumin in the blood. Biol Pharm Bull 2017; 40(9): 1515-24.
[http://dx.doi.org/10.1248/bpb.b17-00339] [PMID: 28867734]
[19]
Davidson LE, Fiorino AM, Snydman DR, Hibberd PL. Lactobacillus GG as an immune adjuvant for live-attenuated influenza vaccine in healthy adults: a randomized double-blind placebo-controlled trial. Eur J Clin Nutr 2011; 65(4): 501-7.
[http://dx.doi.org/10.1038/ejcn.2010.289] [PMID: 21285968]
[20]
Pham PL, Dupont I, Roy D, Lapointe G, Cerning J. Production of exopolysaccharide by Lactobacillus rhamnosus R and analysis of its enzymatic degradation during prolonged fermentation. Appl Environ Microbiol 2000; 66(6): 2302-10.
[http://dx.doi.org/10.1128/AEM.66.6.2302-2310.2000 PMID: 10831403]
[21]
Biernat KA, Pellock SJ, Bhatt AP, et al. Structure, function, and inhibition of drug reactivating human gut microbial β-glucuronidases. Sci Rep 2019; 9(1): 825.
[http://dx.doi.org/10.1038/s41598-018-36069-w] [PMID: 30696850]
[22]
Athari Nik Azm S, Djazayeri A, Safa M, et al. Lactobacilli and bifidobacteria ameliorate memory and learning deficits and oxidative stress in β-amyloid (1-42) injected rats. Appl Physiol Nutr Metab 2018; 43(7): 718-26.
[http://dx.doi.org/10.1139/apnm-2017-0648] [PMID: 29462572]
[23]
Vorhees CV, Williams MT. Morris water maze: procedures for assessing spatial and related forms of learning and memory. Nat Protoc 2006; 1(2): 848-58.
[http://dx.doi.org/10.1038/nprot.2006.116] [PMID: 17406317]
[24]
Gacar N, Mutlu O, Utkan T, Komsuoglu Celikyurt I, Gocmez SS, Ulak G. Beneficial effects of resveratrol on scopolamine but not mecamylamine induced memory impairment in the passive avoidance and Morris water maze tests in rats. Pharmacol Biochem Behav 2011; 99(3): 316-23.
[http://dx.doi.org/10.1016/j.pbb.2011.05.017] [PMID: 21624386]
[25]
Ellman GL, Courtney KD, Andres V Jr, Feather-Stone RM. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem Pharmacol 1961; 7(2): 88-95.
[http://dx.doi.org/10.1016/0006-2952(61)90145-9] [PMID: 13726518]
[26]
Ohkawa H, Ohishi N, Yagi K. Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem 1979; 95(2): 351-8.
[http://dx.doi.org/10.1016/0003-2697(79)90738-3] [PMID: 36810]
[27]
Rinwa P, Kumar A. Piperine potentiates the protective effects of curcumin against chronic unpredictable stress-induced cognitive impairment and oxidative damage in mice. Brain Res 2012; 1488: 38-50.
[http://dx.doi.org/10.1016/j.brainres.2012.10.002] [PMID: 23099054]
[28]
Weydert CJ, Cullen JJ. Measurement of superoxide dismutase, catalase and glutathione peroxidase in cultured cells and tissue. Nat Protoc 2010; 5(1): 51-66.
[http://dx.doi.org/10.1038/nprot.2009.197] [PMID: 20057381]
[29]
Luck H, Bergmeyer HU. Catalase Methods of Enzymatic Analysis. 1st ed. NewYork, USA: Academic Press 1971; p. 1088.
[30]
Goverdhan P, Sravanthi A, Mamatha T. Neuroprotective effects of meloxicam and selegiline in scopolamine-induced cognitive impairment and oxidative stress. Int J Alzheimers Dis 2012.
[http://dx.doi.org/10.1155/2012/974013]
[31]
Konar A, Gupta R, Shukla RK, et al. M1 muscarinic receptor is a key target of neuroprotection, neuroregeneration and memory recovery by i-Extract from Withania somnifera. Sci Rep 2019; 9(1): 13990.
[http://dx.doi.org/10.1038/s41598-019-48238-6] [PMID: 31570736]
[32]
Rami A, Krieglstein J. Muscarinic-receptor antagonist scopolamine rescues hippocampal neurons from death induced by glutamate. Brain Res 1998; 788(1-2): 323-6.
[http://dx.doi.org/10.1016/S0006-8993(98)00041-9] [PMID: 9555079]
[33]
Tang KS. The cellular and molecular processes associated with scopolamine-induced memory deficit: A model of Alzheimer’s biomarkers. Life Sci 2019.233116695
[http://dx.doi.org/10.1016/j.lfs.2019.116695] [PMID: 31351082]
[34]
Suthprasertporn N, Mingchinda N, Fukunaga K, Thangnipon W. Neuroprotection of SAK3 on scopolamine-induced cholinergic dysfunction in human neuroblastoma SH-SY5Y cells. Cytotechnology 2020; 72(1): 155-64.
[http://dx.doi.org/10.1007/s10616-019-00366-7] [PMID: 31933104]
[35]
Ponne S, Kumar CR, Boopathy R. Verapamil attenuates scopolamine induced cognitive deficits by averting oxidative stress and mitochondrial injury - A potential therapeutic agent for Alzheimer’s Disease. Metab Brain Dis 2020; 35(3): 503-15.
[http://dx.doi.org/10.1007/s11011-019-00498-x] [PMID: 31691145]
[36]
Maurer SV, Williams CL. The cholinergic system modulates memory and hippocampal plasticity via its interactions with non-neuronal cells. Front Immunol 2017; 8: 1489.
[http://dx.doi.org/10.3389/fimmu.2017.01489] [PMID: 29167670]
[37]
Decker AL, Duncan K. Acetylcholine and the complex interdependence of memory and attention. Curr Opin Behav 2020; 32: 21-8.
[http://dx.doi.org/10.1016/j.cobeha.2020.01.013]
[38]
Basaure P, Guardia-Escote L, Cabré M, et al. Learning, memory and the expression of cholinergic components in mice are modulated by the pesticide chlorpyrifos depending upon age at exposure and apolipoprotein E (APOE) genotype. Arch Toxicol 2019; 93(3): 693-707.
[http://dx.doi.org/10.1007/s00204-019-02387-9] [PMID: 30656380]
[39]
Telles-Longui M, Mourelle D, Schöwe NM, et al. α7 nicotinic ACh receptors are necessary for memory recovery and neuroprotection promoted by attention training in amyloid-β-infused mice. Br J Pharmacol 2019; 176(17): 3193-205.
[http://dx.doi.org/10.1111/bph.14744] [PMID: 31144293]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy