Abstract
This paper reviews the potential role of honey as a therapeutic antioxidant to reduce oxidative stress and improve cognitive ageing. All articles indexed to PubMed Central (PMC) were searched using the following key words: honey, antioxidant, memory and ageing. Honey is a natural insect-derived product with therapeutic, medicinal and nutritional values. Antioxidant properties of honey quench biologically-circulating reactive oxygen species (ROS) and counter oxidative stress while restoring the cellular antioxidant defense system. Antioxidant properties of honey may complement its nootropic effects to reduce cognitive ageing.
Key Words: Ageing, Antioxidant, Cognition, Honey, Oxidative stress
Introduction
Ageing is associated with a slow deterioration of cognitive functions, especially learning and memory (1, 2). Increasing age is a significant risk factor for dementia, Alzheimer’s disease and other prevalent neurodegenerative disorders (3). The accumulation of oxidative damage and the reduction of the antioxidant defense system play key roles in an organism’s ageing and functional senescence. The central nervous system is particularly susceptible to oxidative stress due to its high oxygen consumption and its lower levels of scavenging enzymes for ROS as well as endogenous antioxidants compared with other organs (4). Based on previous studies, increased protein oxidation and lipid peroxidation in various regions of aged mammalian brains have been reported (5-7). These findings suggested that oxidative damage which is increased in age-related cognitive functions could not be prevented by natural antioxidant defense mechanisms and also stated that dietary intake of antioxidants might be beneficial for preserving brain function.
Honey is a therapeutic antioxidant which contains varying concentrations of flavonoids, phenolic acids, glucose oxidase, catalase, carotenoid derivatives, ascorbic acid, organic acids, Maillard reaction products, amino acids and proteins (8-16). It also contains choline and acetylcholine, which are essential for brain functions as they act as neurotransmitters (17). Hence, recent studies on the therapeutic properties of honey have revealed that honey consumption improved cognitive function in adult rats (18), middle-aged rats (19), aged rats (20, 21), ovariectomized rats (22) and postmenopausal women (23). It was hypothesized that honey’s cognitive-enhancing effects are mainly mediated by its antioxidant properties (20-22, 24). This review aims to assess the capability of honey as a therapeutic antioxidant to protect against oxidative damage in ageing brain and improve cognition.
Cognitive changes in the ageing brain
The brain undergoes pronounced age-related structural and physiological changes that might influence the functional changes. Age-related structural changes can be detected at the macroscopic level (e.g., prefrontal volume loss, grey matter atrophy, changes in white matter frontostriatal systems, decline in white matter integrity, parietal cortical thinning and hippocampal damage) (25-31), the cellular level (e.g., mitochondrial changes, synaptic pruning, axonal loss and alterations to glial cell numbers) (32-34) and the molecular level (e.g., disrupted calcium signaling, altered gene expression and epigenetic changes) (32, 35, 36).
Firstly, age-related prefrontal volume loss has been shown to result in the deterioration of executive functions (37). Similarly, frontal areas and the striatum undergo significant structural changes with age (30). Buckner (38) suggested that the relatively mild memory problems often seen in physiological ageing are caused by grey matter atrophy and by changes in the white matter frontostriatal systems, in addition to the changes in the corresponding neurotransmitter systems. Other studies, on the other hand, have reported that the integrity of white matter declines with age (26, 28, 29, 31, 39) and that this deterioration is accompanied by a cognitive decline (26, 39, 40). Besides, the thinning of the parietal cortex in the elderly was also associated with episodic memory-retrieval problems (27, 38, 41).
The hippocampus, an extension of the temporal part of the cerebral cortex, plays an important role in the memory of both humans and rodents (42-44). Damage to the rat hippocampus leads to non-spatial task impairments (25), including object recognition memory (45-46), transitive odor associations (47), temporal order memory (48, 49) and social transmission of food preferences (50, 51). In young animals, the hippocampus receives highly processed multi-modal information from widespread cortical association areas (52). The hippocampus receives sensory information largely through the perforant path, which projects from the entorhinal cortex (EC) to the dentate gyrus (DG), through the CA3 and CA1 areas, to the subiculum (53). Moreover, Scheff et al. (54) reported a reduced number of synapses receiving EC input in the outer molecular layer of the DG in elderly subjects with mild cognitive impairment compared to cognitively-intact elderly subjects. Where the extent of synapse reduction correlated with memory ability. Similarly, aged memory-impaired humans exhibited marked atrophy and synapse loss in the perforant path (55).
On the other hand, the hippocampal neurons also undergo age-related changes, such as decreased synaptic plasticity, decreased neuronal excitability, slower neurogenesis and faster potentiation decay (2, 56-59). Adult neurogenesis is present in the aged brain, but it is dramatically reduced in early adulthood in both the subventricular zone (60, 61) and the subgranular layer of the DG (62-66). This age-related decline in hippocampal neurogenesis, together with other cellular changes, contributes to a physiological decline in cognitive functions (67, 68).
Oxidative stress, antioxidants and the ageing brain
Oxidative Stress
The brain is particularly susceptible to oxidative damage due to its rich abundance of polyunsaturated fatty acids and other lipids, its high-energy demands and high oxygen consumption rate, and its limited antioxidant capacity compared to other organs (69). Oxidative damage has been found to be the highest in the brain regions that are responsible for the cognitive dysfunctions found in Alzheimer’s disease (70-72). Oxidative stress might cause neuron dysfunction through a cascade of molecular events: (i) oxidative damage in mitochondria leads to additional free radical production, (ii) increased oxidation leads to increased levels of β-amyloid (Aβ), which generates additional amyloid precursor protein (APP), (iii) Aβ and the accumulating oxidative damage activate caspases, which cleave APP and generate additional Aβ and oxidative damage, and (iv) progressive oxidative damage and increasing Aβ concentrations cause a decrease in BCL-2 levels, leaving neurons more vulnerable to oxidative damages and insults (73).
Oxidative damage may also affect replication and transcription of mitochondrial DNA (mtDNA) which leads to a decline in mitochondrial function, consecutively leading to enhanced ROS production and further damage to mtDNA (74). Evidently, age-dependent increases in DNA oxidative damage have also been demonstrated in the brain of Alzheimer’s disease patients (69, 75-77). Apart from humans, the accumulation of DNA oxidative damage in different tissues including the brain during ageing has also been observed in other mammals like rodents (78-83).
The long-term oxidative stress in brain tissues possibly contributes to declining cognitive processes (84). Based on a previous study, aged hippocampal neurons which appear to be under oxidative stress is more severe in the learning-impaired subjects, suggesting a possible basis for age-induced cognitive decline (85). Indeed, increased oxidative damage is related to reductions in learning (7, 86-88), memory (87, 89, 90), and increased anxiety (91, 92). All these findings suggested that oxidative stress contributes to learning and memory deficits due to brain oxidative damages during ageing. Hence, protection from oxidative damage offers a basic approach to the controlled strategy of age-related cognitive impairment.
Antioxidants
ROS are highly reactive molecules that consist of a number of diverse chemical species including superoxide anion (O2•−), hydroxyl radical (•OH), and hydrogen peroxide (H2O2). ROS readily attack DNA and generate a variety of DNA lesions, such as oxidized DNA bases, abasic sites and DNA strand breaks, which ultimately lead to genomic instability. Due to their potentiality to causing oxidative damage, ROS have been implicated as one of the mechanisms underlying cognitive ageing (94).
In order to protect cells from oxidative damage, aerobic metabolism generally depends on stringent control of ROS by antioxidants. Antioxidants are substances that, when present at low concentrations than that of the oxidizable substrates, significantly inhibits or delays oxidative processes of that substrate by oxidizing itself (95). In tissues functioning normally, a balance is maintained between ROS generation and antioxidant protection, which is mediated through antioxidant enzymes and antioxidant molecules that are obtained naturally through nutrition (84). Oxidative stress leading to lipids, proteins, ribonucleic acid (RNA) and DNA oxidative damage occurs when the balance between free radical generation and antioxidant capacities shift toward free radical generation (69).
There are two types of antioxidants in the human body, enzymatic and non-enzymatic (96). Enzymatic antioxidants, which are also known as natural antioxidants, neutralize excessive ROS and prevent them from damaging the cellular structure. Enzymatic antioxidants include superoxide dismutase (SOD), glutathione peroxidase (GPx), ascorbate peroxidase, mono- and dehydroascorbate reductases, glutathione reductase (GR), and catalase (CAT). The levels of enzymatic antioxidants tend to decrease with age, leaving the biological system vulnerable to oxidative stress, thus, contributing to the senescence processes. Tiana et al. (97) reported that SOD, GPx, and CAT activities in most tissues including brain, liver, heart, and kidney significantly decreased during ageing. The continuous decline of these enzymatic enzymes is associated with an increase in brain oxidative stress and age-related decline in cognitive performance (98). In addition to enzymatic antioxidants, non-enzymatic antioxidants (synthetic antioxidants) also serve as an essential biological defense against environmental pro-oxidant conditions. The complex antioxidant system of the body is influenced by the dietary intake of antioxidant vitamins and minerals, such as vitamin C, vitamin E, glutathione, melatonin, selenium, zinc, taurine, beta carotene, and carotene (99).
Additionally, several studies have found possible inverse associations between high plasma levels of antioxidants and cognitive decline (100-103) as well as dementia (104-106). Hence, the consumption of antioxidant compounds was found to improve cognitive performance in aged animals (73, 107, 108) as well as in humans (109, 110). Research by Fahnestock et al. (111) revealed an increase in brain-derived neurotrophic factor (BDNF) levels, which is one of the factors underlying improvements in learning and memory in aged dogs receiving antioxidant-fortified diet. Overall, these results suggested that an increase in the intake of natural antioxidants may help maintain an acceptable antioxidant status, perhaps normal physiological functioning including cognitive function (112).
Honey, its constituents and medicinal effects
Honey is a naturally sweet substance. It is produced by honey bees from the nectar of plants or from secretion of living parts of plants or excretion of plant-sucking insects on the living parts of plants, which the bees collect, transform by combining with specific substances of their own, deposit, dehydrate, store, and leave in honeycombs to ripen and mature (113). The color of the honey usually ranging from water white to dark amber, depends on the floral source and its mineral content collected by the bees. Moreover, the flavor of the honey depends upon the color, normally the darker the honey the stronger the flavor and quality. Honey is often named according to the geographical location where it was produced, the floral source of the honey or the trees on which the hives are found (114).
In general, honey can be grouped into either monofloral or multi-floral types (115). Monofloral honey is made primarily from the nectar of one type of flower, whereas multi-floral honey, also known as polyfloral or wildflower honey, is derived from the nectar of many types of flowers (115, 116). Examples of monofloral honey are manuka honey, sourwood honey, tupelo honey, orange blossom honey, acacia honey and palmetto honey, whereas examples of polyfloral honey are tualang honey, Georgia wildflower honey, savannah honey and Charleston honey.
A number of factors such as geographical origin, botanical sources of nectar, environmental, and climatic conditions as well as the processing techniques can influence the composition of honey (117, 118). Approximately 79.6% of honey is of sugars and the major sugars are comprised of levulose and dextrose which constitutes 38.2% and 31.3% correspondingly, while the remaining 1.3% is sucrose and 7.3% is maltose (119). The minor constituents of honey include 0.57% acids, 0.27% protein, 0.04% nitrogen, 0.1% amino acids, 0.17% minerals, and other compounds such as pigments, flavor and aroma substances, phenolic compounds, colloids, sugar alcohols, and vitamins which all together account for the 2.1% of whole honey composition (119). Vitamins such as vitamin A, C, and E are also found in honey (119-121). Besides, honey also contains other bioactive constituents such as flavonoids, carotenoid-derived compounds, nitric oxide metabolites, and Maillard reaction products (118, 123-125). The primary flavonoid compounds found in honey are triacetin, quercetin, and luteolin (126). Honey also contains a number of enzymes such as glucose oxidase, diastase, invertase, phosphatase, catalase, and peroxidase (124, 127).
The use of honey in folk medicine dates back to 2100-2000 BC (128). Initially, most of the health benefits ascribed to honey were based on mere observations or generalizations without any scientific evidence. In the last few years, however, there has been a renewed interest in research related to potential health benefits of honey in the treatment of various diseases resulting in findings attributable to several medicinal effects of honey. These include cardioprotective (129), hepatoprotective (131, 132), hypoglycemia (133, 134), reproductive (139, 140), and antihypertensive effects (121, 142). Other effects such as antibacterial (142-144), antifungal (145, 146), antiviral (147), anti-inflammatory (148-150), and antitumor (119, 151-155) have also been documented and attributed to honey.
Malaysian tualang honey is produced by the rock bee (Apis dorsata). The rock bees build hives on tualang trees (Koompassia excelsa) found mainly in the north-western region of Peninsular Malaysia. Tualang honey has a dark brown appearance with a pH of 3.55–4.00 and a specific gravity of 1.335 (156). Hydrocarbons constitute more than half (58.5%) of its composition. These include alcohols, ketones, aldehydes, furans, terpenes, flavonoids, and phenols (157). A total of six phenolic acids (gallic, syringic, benzoic, trans-cinnamic, p-coumaric, and caffeic acids) and five flavonoids (catechin, kaempferol, naringenin, luteolin, and apigenin) were found in tualang honey (157-159). It was reported that tualang honey contains more phenolic acids and flavonoids than manuka honey and other local Malaysian honey (159). It was also reported that tualang honey possessed the highest antioxidant activity compared to gelam honey, Indian forest honey, and pineapple honey (159). In addition, Moniruzzaman et al. (160) reported that among four different types of local honey (tualang, acacia, pineapple, and Borneo), tualang honey possessed the highest phenolic compounds, flavonoid contents, ferric reducing power values along with the greatest color intensity, indicating its antioxidant potential. Tualang honey has been reported to possess medicinal properties such as anticancer (153), antiproliferative (161), anti-inflammatory, and antibacterial properties (142, 162). It has also been reported to improve male (122, 162-164) and female reproductive systems (139, 165, 167).
Antioxidant and cognitive enhancing properties of honey
The therapeutic role of honey in the treatment of various diseases has been attributed to its antioxidant (168, 169), anti-inflammatory and anti-acetylcholinesterase (AChE) properties (Table 1). Honey contains both enzymatic (catalase, glucose oxidase, peroxidase) and non-enzymatic antioxidants (ascorbic acid, α-tocopherol, carotenoids, amino acids, proteins, flavonoids, and phenolic acids) (179-181). The amount and type of these antioxidants depend largely upon the types of honey. Some reports have established a correlation between floral origins and phenolic compounds and flavonoids (179, 182-184). Correspondingly, a correlation between total phenolic content in honey and its antioxidant activity has been demonstrated (169,180). Other studies have also demonstrated that honey is able to increase plasma antioxidants and ameliorate oxidative stress in tissues (121, 133, 134, 137, 138, 149, 185-188).
Table 1.
Properties | Actions | Reference(s) |
---|---|---|
Antioxidant | Tualang honey supplementation (20 mg/day) for 16 weeks was able to reduce blood oxidative stress levels/activities and improve memory function of postmenopausal women compared to those who received estrogen-progestin therapy | (170) |
Daily consumption of tualang honey (200 mg/kg/day) for 18 days in female ovariectomized rats is able to reduce blood oxidative stress levels/activities in the brain homogenate and improve memory performance of tualang honey-treated group as compared with the untreated stressed ovariectomized rats | (171) | |
Both short and long term (short term - 3 weeks and long term - 12 weeks) honey supplementations at a dose of 250 mg/kg body weight increased the brain protein and CAT activities of brain cells | (172) | |
Tualang honey supplementation was able to improve memory performance and decrease oxidative stress levels in rats exposed to noise stress | (20, 21) | |
Treatment with tualang honey and its methanolic fraction markedly reduced oxidative damage in rats’ hippocampus as evidence by restoration in the activities of antioxidant enzymes (CAT, GPx and GPr) and decreased in MDA level comparable to LPS rats treated with memantine (a controlled drug) | (173) | |
Tualang honey has therapeutic potential against KA-induced oxidative stress and neurodegeneration through its antioxidant effect | (174) | |
Anti-inflammatory | Tualang honey and its methanolic fraction significantly reduced the concentration of COX-2 and TNF-α expression, as well as amyloid deposition in the hippocampus of LPS rats comparable to memantine group | (175) |
Tualang honey reduced neuroinflammation and caspase-3 activity after kainic acid-induced status epilepticus | (176) | |
Anti AChE | Reduced memory performance, low concentrations of ACh and high AChE in the brain homogenates of stressed ovariectomized rats compared with non-stressed sham-operated controls and the effects were reversed after treatment with Tualang honey | (177) |
Tualang honey supplementation was able to improve memory performance and decrease acetylcholinesterase activity in noise-induced male rats | (20) | |
BDNF | BDNF concentration is significantly decreased in stressed ovariectomized rats compared to other experimental groups where the concentration was restored to normal following tualang honey treatment | (178) |
Tualang honey supplementation was able to improve memory performance, increase BDNF concentration in noise-induced male rats | (20) |
These antioxidant properties of honey may help in reducing oxidative damage and improving brain cell structure and integrity, hence enhancing cognitive performance. Al-Himyari (24) reported that one tablespoon of honey daily to mild cognitive impaired subjects aged 65 and older was able to prevent cognitive decline and development of dementia. In addition, supplementation of a combination of astragalus honey, sedge, and saffron in major neurocognitive disorder patients was able to improve their cognitive scores, as assessed by Addenbrooke’s Cognitive Scale (189). However, another study which was conducted in rats reported that reduction in anxiety (190) and improvement in spatial memory among honey-fed middle-aged rats compared to those sucrose-fed or on a sugar-free diet (19).
Tualang honey, in particular, has been shown to improve immediate memory of postmenopausal women, a result comparable with the memory improvement seen in women receiving estrogen plus progestin therapy (23). Additionally, tualang honey also exhibited neuroprotective potential in neurodegenerative disorders as it protects the viability of hippocampal cells in chronic cerebral hypoperfusion induced by permanent bilateral common carotid arteries ligation in rats (191). Similarly, tualang honey improved short- and long-term memory of ovariectomized rats via enhancing neuronal proliferation in hippocampal CA2, CA3, and DG regions (22). Tualang honey has also been shown to improve memory performance in aged male rats, while its antioxidant properties are thought to be one of the possible mechanism (20, 21). These reports suggest the potential use of honey as an alternative therapy to prevent memory decline due to oxidative stress exposure and/or ageing in humans.
Apart from its cognitive enhancing properties in aged subjects (rats), honey has also shown similar effects in young or adult subjects. Azman et al. (18) reported that tualang honey could improve working memory performance in rats exposed to loud noise stress, as assessed by the novel object recognition test. Tualang honey has also been shown to improve spatial memory performance in adult male rats as assessed by the radial arm maze, via enhancing numbers of CA1 and CA3 hippocampal pyramidal cells (192). Additionally, honey exhibited neuroprotective effects against lead-induced cognitive deficit via enhancing antioxidant activities as evidenced by increased brain SOD, GST, and GSH activities (193). Another study demonstrated that honey effectively inhibited apoptosis or neuronal cell death in the hippocampus of streptozotocin-induced diabetic rats (194).
Also, at old age, the antioxidant defense systems may adopt less efficiently to defend against ROS generation compared to young age (195, 196). Azman et al. (21) reported that the neuroprotective effects of tualang honey were more prominent in young than old rats. While tualang honey was able to improve memory performance in both young and aged rats, it was noted the young rats exhibited lower brain MDA level and higher antioxidant enzymes activities compared to the aged rats (21). This finding is probably due to the optimal antioxidant protection system in younger groups which efficiently reduces oxidative stress. Therefore, for old subjects, longer duration or a higher dose of honey is probably needed to produce the same results as in young subjects.
Based on the results from these previous studies, positive correlations between neuronal cell counts, antioxidant activities, and memory performance were unravelled (20-22, 192). These studies confirmed the notion that exposure to oxidative stress in the course of ageing leads to damages of neuronal cells and neuronal death which eventually results in cognitive decline. Hence, antioxidant properties of honey may help combat against damages to the neuronal cells by neutralizing the free radicals, by maintaining healthy neuronal cells to preserve cognitive functions.
Conclusion
Oxidative stress plays an important role in ageing and neurodegenerative disorders. Based on the in vitro and in vivo studies discussed in this review, honey has the potential to be a therapeutic agent against oxidative damage and cognitive decline associated with ageing. Honey supplementation is likely to enhance the antioxidant defense system via attenuating ROS attacks on cell structures, particularly brain cells, hence preserving brain functions and cognitive ability.
Conflicts of Interest
The authors declare that there is no conflict of interest regarding the publication of this paper.
Acknowledgment
The authors wish to acknowledge funding from USM short-term research grant (304/PPSP/6315093).
References
- 1.Grady CL, Craik FI. Changes in memory processing with age. Curr Opin Neurobiol. 2000;10:224–231. doi: 10.1016/s0959-4388(00)00073-8. [DOI] [PubMed] [Google Scholar]
- 2.Rosenzweig ES, Barnes CA. Impact of ageing on hippocampal function: plasticity, network dynamics, and cognition. Prog Neurobiol. 2003;69:143–179. doi: 10.1016/s0301-0082(02)00126-0. [DOI] [PubMed] [Google Scholar]
- 3.Yankner B, Lu T, Loerch P. The ageing brain. Annu Rev Pathol. 2008;3:41–66. doi: 10.1146/annurev.pathmechdis.2.010506.092044. [DOI] [PubMed] [Google Scholar]
- 4.Schmitt-Schillig S, Schaffer S, Weber C, Eckert G, Muller W. Flavonoids and the ageing brain. J Physiol Pharmacol. 2005;56:23–26. [PubMed] [Google Scholar]
- 5.Leutner S, Eckert A, Muller W. ROS generation, lipid peroxidation and antioxidant enzyme activities in the ageing brain. J Neural Transm. 2001;108:955–967. doi: 10.1007/s007020170015. [DOI] [PubMed] [Google Scholar]
- 6.Liu J, Head E, Gharib AM, Yuan W, Ingersoll RT, Hagen TM, et al. Memory loss in old rats is associated with brain mitochondrial decay and RNA/DNA oxidation: partial reversal by feeding acetyl-L-carnitine and/or R-alpha-lipoic acid. Proc Natl Acad Sci U.S.A. 2002;99:2356–2361. doi: 10.1073/pnas.261709299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Liu R, Liu IY, Bi X, Thompson RF, Doctrow SR, Malfroy B, et al. Reversal of age-related learning deficits and brain oxidative stress in mice with superoxide dismutase/catalase mimetics. Proc Natl Acad Sci U.S.A. 2003;100:8526–8531. doi: 10.1073/pnas.1332809100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Beretta G, Granata P, Ferrero M, Orioli M, Facino RM. Standardization of antioxidant properties of honey by a combination of spectrophotometric/fluorimetric assays and chemometrics. Anal Chim Acta. 2005;533:185–191. [Google Scholar]
- 9.D’Arcy BR. Antioxidants in Australian floral honeys – Identification of health enhancing nutrient components. RIRDC Publication. 2005;05/040:1. [Google Scholar]
- 10.Frankel S, Robinson GE, Berenbaum MR. Antioxidant capacity and correlated characteristics of 14 unifloral honeys. J Apic Res. 1998;37:27–31. [Google Scholar]
- 11.Aljadi AM, Kamaruddin MY. Evaluation of the phenolic contents and antioxidant capacities of two Malaysian floral honeys. Food Chem. 2004;85:513–518. [Google Scholar]
- 12.Inoue K, Murayarna S, Seshimo F, Takeba K, Yoshimura Y, Nakazawa H. Identification of phenolic compound in manuka honey as specific superoxide anion radical scavenger using electron spin resonance (ESR) and liquid chromatography with coulometric array detection. J Sci Food Agric. 2005;85:872–878. [Google Scholar]
- 13.Fahey JW, Stephenson KK. Pinostrobin from honey and Thai ginger (Boesenbergia pandurata): A potent flavonoid inducer of mammalian phase 2 chemoprotective and antioxidant enzymes. J Agric Food Chem. 2002;50:7472–7476. doi: 10.1021/jf025692k. [DOI] [PubMed] [Google Scholar]
- 14.Blasa M, Candiracci M, Accorsi A, Piacentini M, Albertini M, Piatti E. Raw Millefiori honey is packed full of antioxidants. Food Chem. 2006;97:217–222. [Google Scholar]
- 15.Nagai T, Inoue R, Kanamori N, Suzuki N, Nagashima T. Characterization of honey from different floral sources Its functional properties and effects of honey species on storage of meat. Food Chem. 2006;97:256–262. [Google Scholar]
- 16.Perez RA, Iglesias MT, Pueyo E, Gonzalez M, de Lorenzo C. Amino acid composition and antioxidant capacity of Spanish honeys. J Agric Food Chem. 2007;55:360–365. doi: 10.1021/jf062055b. [DOI] [PubMed] [Google Scholar]
- 17.White J. Composition of honey. In: Crane E, editor. Honey: A comprehensive survey. London: Heinemann; 1975. [Google Scholar]
- 18.Azman KF, Zakaria R, Abd Aziz CB, Othman Z, Al-Rahbi B. Tualang honey improves memory performance and decreases depressive-like behavior in rats exposed to loud noise stress. Noise and Health. 2015;17:83–89. doi: 10.4103/1463-1741.153388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Chepulis LM, Starkey NJ. Waas JR, Molan PC. The effects of long-term honey, sucrose or sugar-free diets on memory and anxiety in rats. Physiol Behav. 2009;97:359–368. doi: 10.1016/j.physbeh.2009.03.001. [DOI] [PubMed] [Google Scholar]
- 20.Azman KF, Zakaria R, Abd Aziz CB, Othman Z. Tualang honey attenuates noise stress-induced memory deficits in aged rats. Oxid Med Cell Longev. 2016;Article ID 1549158:11 pages. doi: 10.1155/2016/1549158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Azman KF, Zakaria R, Othman Z, Abd Aziz CB. Neuroprotective effects of Tualang honey against oxidative stress and memory decline in young and aged rats exposed to noise stress. J Taibah Univ Sci. 2018;12:273–284. [Google Scholar]
- 22.Al-Rahbi B, Zakaria R, Othman Z, Hassan A, Mohd Ismail ZI, Muthuraju S. Tualang honey supplement improves memory performance and hippocampal morphology in stressed overiectomized rats. Acta Histochem. 2014;116:79–88. doi: 10.1016/j.acthis.2013.05.004. [DOI] [PubMed] [Google Scholar]
- 23.Othman Z, Shafin N, Zakaria R, Hussain NHN, Mohammad WMZW. Improvement in immediate memory after 16 weeks of Tualang honey (Agro Mas) supplement in healthy postmenopausal women. Menopause. 2011;18:1219–1224. doi: 10.1097/gme.0b013e31821e2044. [DOI] [PubMed] [Google Scholar]
- 24.Al-Himyari FA. The use of honey as a natural preventive therapy of cognitive decline and dementia in the Middle East. Alzheimers Dement. 2009;5:P247. [Google Scholar]
- 25.Eichenbaum H, Dudchenko P, Wood E, Shapiro M, Tanila H. The hippocampus, memory, and place cells: Is it spatial memory or memory space? Neuron. 1999;23:209–226. doi: 10.1016/s0896-6273(00)80773-4. [DOI] [PubMed] [Google Scholar]
- 26.Moseley M. Diffusion tensor imaging and ageing - A review. NMR Biomed. 2002;15:553–560. doi: 10.1002/nbm.785. [DOI] [PubMed] [Google Scholar]
- 27.Salat D, Buckner R, Snyder A, Greve D, Desikan R, Busa E, et al. Thinning of the cerebral cortex in ageing. Cereb Cortex. 2004;14:721–730. doi: 10.1093/cercor/bhh032. [DOI] [PubMed] [Google Scholar]
- 28.Pfefferbaum A, Adalsteinsson E, Sullivan EV. Frontal circuitry degradation marks healthy adult ageing: evidence from diffusion tensor imaging. Neuroimage. 2005;26:891–899. doi: 10.1016/j.neuroimage.2005.02.034. [DOI] [PubMed] [Google Scholar]
- 29.Salat D, Tuch D, Greve D, Van Der Kouwe A, Hevelone N, Zaleta A, et al. Age-related alterations in white matter microstructure measured by diffusion tensor imaging. Neurobiol Aging. 2005;26:1215–1227. doi: 10.1016/j.neurobiolaging.2004.09.017. [DOI] [PubMed] [Google Scholar]
- 30.Walhovd KB, Fjell AM, Reinvang I, Lundervold A, Dale AM, Eilertsen DE, et al. Effects of age on volumes of cortex, white matter and subcortical structures. Neurobiol Aging. 2005;26:1261–1270. doi: 10.1016/j.neurobiolaging.2005.05.020. [DOI] [PubMed] [Google Scholar]
- 31.Sullivan EV, Adalsteinsson E, Pfefferbaum A. Selective age-related degradation of anterior callosal fiber bundles quantified in vivo with fiber tracking. Cereb Cortex. 2006;16:1030–1039. doi: 10.1093/cercor/bhj045. [DOI] [PubMed] [Google Scholar]
- 32.Marner L, Nyengaard JR, Tang Y, Pakkenberg B. Marked loss of myelinated nerve fibers in the human brain with age. J Comp Neurol. 2003;462:144–152. doi: 10.1002/cne.10714. [DOI] [PubMed] [Google Scholar]
- 33.Toescu EC. Normal brain ageing: Models and mechanisms. Philos Trans R Soc B Biol Sci. 2005;360:2347–2354. doi: 10.1098/rstb.2005.1771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Pelvig DP, Pakkenberg H, Stark AK, Pakkenberg B. Neocortical glial cell numbers in human brains. Neurobiol Aging. 2008;29:1754–1762. doi: 10.1016/j.neurobiolaging.2007.04.013. [DOI] [PubMed] [Google Scholar]
- 35.Soreq L, UK Brain Expression Consortium; North American Brain Expression Consortium, Rose J, Soreq E, Hardy J, Trabzuni D, et al. Major shifts in glial regional identity are a transcriptional hallmark of human brain ageing. Cell Rep. 2017;18:557–570. doi: 10.1016/j.celrep.2016.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Horvath S. DNA methylation age of human tissues and cell types. Genome Biol. 2013;14:R115–133. doi: 10.1186/gb-2013-14-10-r115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Head D, Snyder A, Girton L, Morris J, Buckner R. Frontal-hippocampal double dissociation between normal ageing and Alzheimer’s disease. Cereb Cortex. 2005;15:732–739. doi: 10.1093/cercor/bhh174. [DOI] [PubMed] [Google Scholar]
- 38.Buckner RL. Memory and executive function in ageing and AD: multiple factors that cause decline and reserve factors that compensate. Neuron. 2004;44:195–208. doi: 10.1016/j.neuron.2004.09.006. [DOI] [PubMed] [Google Scholar]
- 39.Charlton R, Barrick T, McIntyre D, Shen Y, O’Sullivan M, Howe FA, et al. White matter damage on diffusion tensor imaging correlates with age-related cognitive decline. Neurology. 2006;66:217–222. doi: 10.1212/01.wnl.0000194256.15247.83. [DOI] [PubMed] [Google Scholar]
- 40.Grieve S, Williams L, Paul R, Clark C, Gordon E. Cognitive ageing, executive function, and fractional anisotropy: a diffusion tensor MR imaging study. Am J Neuroradiol. 2007;28:226–235. [PMC free article] [PubMed] [Google Scholar]
- 41.Cavanna AE, Trimble MR. The precuneus: a review of its functional anatomy and behavioural correlates. Brain. 2006;129:564–583. doi: 10.1093/brain/awl004. [DOI] [PubMed] [Google Scholar]
- 42.Burgess N, Maguire EA, O’Keefe J. The human hippocampus and spatial and episodic memory. Neuron. 2002;35:625–641. doi: 10.1016/s0896-6273(02)00830-9. [DOI] [PubMed] [Google Scholar]
- 43.O’Keefe J. Do hippocampal pyramidal cells signal non-spatial as well as spatial information? Hippocampus. 1999;9:352–364. doi: 10.1002/(SICI)1098-1063(1999)9:4<352::AID-HIPO3>3.0.CO;2-1. [DOI] [PubMed] [Google Scholar]
- 44.Hall J. Guyton and Hall textbook of medical physiology. Philadelphia, USA: Saunders Elsevier; 2010. [Google Scholar]
- 45.Clark R, Zola S, Squire L. Impaired recognition memory in rats after damage to the hippocampus. J Neurosci. 2000;20:8853–8860. doi: 10.1523/JNEUROSCI.20-23-08853.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Fortin N, Wright S, Eichenbaum H. Recollection-like memory retrieval in rats is dependent on the hippocampus. Nature. 2004;431:188–191. doi: 10.1038/nature02853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Bunsey M, Eichenbaum H. Conservation of hippocampal memory function in rats and humans. Nature. 1996;379:255–257. doi: 10.1038/379255a0. [DOI] [PubMed] [Google Scholar]
- 48.Fortin N, Agster K, Eichenbaum H. Critical role of the hippocampus in memory for sequences of events. Nat Neurosci. 2002;5:458–462. doi: 10.1038/nn834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kesner R, Gilbert P, Barua L. The role of the hippocampus in memory for the temporal order of a sequence of odors. Behavioral Neurosci. 2002;116:286–290. doi: 10.1037//0735-7044.116.2.286. [DOI] [PubMed] [Google Scholar]
- 50.Alvarez P, Lipton PA, Melrose R, Eichenbaum H. Differential effects of damage within the hippocampal region on memory for a natural, nonspatial odor-odor association. Learn Mem. 2001;8:79–86. doi: 10.1101/lm.38201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Clark R, Broadbent N, Zola S, Squire L. Anterograde amnesia and temporally graded retrograde amnesia for a nonspatial memory task after lesions of hippocampus and subiculum. J Neurosci. 2002;22:4663–4669. doi: 10.1523/JNEUROSCI.22-11-04663.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Amaral D, Witter M. Hippocampal formation. In: Paxinos G, editor. The rat nervous system. (2nd edition) Academic Press; 1995. [Google Scholar]
- 53.Wilson IA, Gallagher M, Eichenbaum H, Tanila H. Neurocognitive ageing: prior memories hinder new hippocampal encoding. Trends Neurosci. 2006;29:662–670. doi: 10.1016/j.tins.2006.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Scheff SW, Price DA, Schmitt FA, Mufson EJ. Hippocampal synaptic loss in early Alzheimer’s disease and mild cognitive impairment. Neurobiol Aging. 2006;27:1372–1384. doi: 10.1016/j.neurobiolaging.2005.09.012. [DOI] [PubMed] [Google Scholar]
- 55.Kalus P, Slotboom J, Gallinat J, Mahlberg R, Cattapan-Ludewig K, Wiest R, et al. Examining the gateway to the limbic system with diffusion tensor imaging: the perforant pathway in dementia. Neuroimage. 2006;30:713–720. doi: 10.1016/j.neuroimage.2005.10.035. [DOI] [PubMed] [Google Scholar]
- 56.Barnes CA, Rao G, Houston FP. LTP induction threshold change in old rats at the perforant path-granule cell synapse. Neurobiol Aging. 2000;21:613–620. doi: 10.1016/s0197-4580(00)00163-9. [DOI] [PubMed] [Google Scholar]
- 57.Dieguez DJ, Barea-Rodriguez E. Ageing impairs the late phase of long-term potentiation at the medial perforant path-CA3 synapse in awake rats. Synapse. 2004;52:53–61. doi: 10.1002/syn.20004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Burke SN, Barnes CA. Neural plasticity in the ageing brain. Nat Rev Neurosci. 2006;7:30–40. doi: 10.1038/nrn1809. [DOI] [PubMed] [Google Scholar]
- 59.Leuner B, Kozorovitskiy Y, Gross C, Gould E. Diminished adult neurogenesis in the marmoset brain precedes old age. Proc Natl Acad Sci USA. 2007;104:17169–17173. doi: 10.1073/pnas.0708228104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Mirich JM, Williams NC, Berlau DJ, Brunjes PC. Comparative study of ageing in the mouse olfactory bulb. J Comp Neurol. 2002;454:361–372. doi: 10.1002/cne.10426. [DOI] [PubMed] [Google Scholar]
- 61.Shook BA, Manz DH, Peters JJ, Kang S, Conover JC. Spatiotemporal changes to the subventricular zone stem cell pool through ageing. J Neurosci. 2012;32:6947–6956. doi: 10.1523/JNEUROSCI.5987-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Bondolfi L, Ermini F, Long JM, Ingram DK, Jucker M. Impact of age and caloric restriction on neurogenesis in the dentate gyrus of C57BL/6 mice. Neurobiol Aging. 2004;25:333–340. doi: 10.1016/S0197-4580(03)00083-6. [DOI] [PubMed] [Google Scholar]
- 63.Kronenberg G, Bick-Sander A, Bunk E, Wolf C, Ehninger D, Kempermann G. Physical exercise prevents age-related decline in precursor cell activity in the mouse dentate gyrus. Neurobiol Aging. 2006;27:1505–1513. doi: 10.1016/j.neurobiolaging.2005.09.016. [DOI] [PubMed] [Google Scholar]
- 64.Ben Abdallah NM-B, Slomianka L, Vyssotski AL, Lipp H-P. Early age-related changes in adult hippocampal neurogenesis in C57 mice. Neurobiol Aging. 2010;31:151–161. doi: 10.1016/j.neurobiolaging.2008.03.002. [DOI] [PubMed] [Google Scholar]
- 65.Encinas JM, Michurina TV, Peunova N, Park J-H, Tordo J, Peterson DA, et al. Division-coupled astrocytic differentiation and age-related depletion of neural stem cells in the adult hippocampus. Cell Stem Cell. 2011;8:566–579. doi: 10.1016/j.stem.2011.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Miranda CJ, Braun L, Jiang Y, Hester ME, Zhang L, Riolo M, et al. Ageing brain microenvironment decreases hippocampal neurogenesis through Wnt-mediated survivin signaling. Aging Cell. 2012;11:542–552. doi: 10.1111/j.1474-9726.2012.00816.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Bizon J, Gallagher M. Production of new cells in the rat dentate gyrus over the lifespan: relation to cognitive decline. Eur J Neurosci. 2003;18:215–219. doi: 10.1046/j.1460-9568.2003.02733.x. [DOI] [PubMed] [Google Scholar]
- 68.Drapeau E, Mayo W, Aurousseau C, Le M, Piazza P, Abrous D. Spatial memory performances of aged rats in the water maze predict levels of hippocampal neurogenesis. Proc Natl Acad Sci U.S.A. 2003;100:14385–14390. doi: 10.1073/pnas.2334169100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Lovell MA, Markesbery WR. Oxidative DNA damage in mild cognitive impairment and late-stage Alzheimer’s disease. Nucleic Acids Res. 2007;35:7497–7504. doi: 10.1093/nar/gkm821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Nunomura A, Perry G, Aliev G, Hirai K, Takeda A, Balraj E, et al. Oxidative damage is the earliest event in Alzheimer disease. J Neuropathol Exp Neurol. 2001;60:759–767. doi: 10.1093/jnen/60.8.759. [DOI] [PubMed] [Google Scholar]
- 71.Giasson B, Ischiropoulos H, Lee V, Trojanowski J. The relationship between oxidative/nitrative stress and pathological inclusions in Alzheimer’s and Parkinson’s diseases. Free Radic Biol Med. 2002;32:1264–1275. doi: 10.1016/s0891-5849(02)00804-3. [DOI] [PubMed] [Google Scholar]
- 72.Zhu X, Raina A, Lee H, Casadesus G, Smith M, Perry G. Oxidative stress signalling in Alzheimer’s disease. Brain Res. 2004;1000:32–39. doi: 10.1016/j.brainres.2004.01.012. [DOI] [PubMed] [Google Scholar]
- 74.Cui H, Kong Y, Zhang H. Oxidative stress, mitochondrial dysfunction, and ageing. J Signal Transduct. 2012;2012:646354. doi: 10.1155/2012/646354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Lyras L, Cairns N, Jenner A, Jenner P, Halliwell B. An assessment of oxidative damage to proteins, lipids and DNA in brain from patients with Alzheimer’s disease. J Neurochem. 1997;68:2061–2069. doi: 10.1046/j.1471-4159.1997.68052061.x. [DOI] [PubMed] [Google Scholar]
- 76.Gabbita S, Lovell M, Markesbery W. Increased nuclear DNA oxidation in the brain in Alzheimer’s disease. J Neurochem. 1998;71:2034–2040. doi: 10.1046/j.1471-4159.1998.71052034.x. [DOI] [PubMed] [Google Scholar]
- 77.Mecocci P, Polidori M, Cherubini A, Ingegni T, Mattioli P, Catani M, et al. Lymphocyte oxidative DNA damage and plasma antioxidants in Alzheimer disease. Arch Neurol. 2002;59:794–798. doi: 10.1001/archneur.59.5.794. [DOI] [PubMed] [Google Scholar]
- 78.Su J, Deng G, Cotman C. Neuronal DNA damage precedes tangle formation and is associated with up-regulation of nitrotyrosine in Alzheimer’s disease brain. Brain Res Reviews. 1997;774:193–199. doi: 10.1016/s0006-8993(97)81703-9. [DOI] [PubMed] [Google Scholar]
- 79.Hamilton M, Guo Z, Fuller C, Remmen H, Ward W, Austad S, et al. A reliable assessment of 8-oxo-2-deoxyguanosine levels in nuclear and mitochondrial DNA using the sodium iodide method to isolate DNA. Nucleic Acids Res. 2001;29:2117–2126. doi: 10.1093/nar/29.10.2117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Dorszewska J, Adamczewska-Goncerzewicz Z. Oxidative damage to DNA, p53 gene expression and p53 protein level in the process of ageing in rat brain. Respir Physiol Neurobiol. 2004;139:227–236. doi: 10.1016/j.resp.2003.10.005. [DOI] [PubMed] [Google Scholar]
- 81.Sedelnikova O, Horikawa I, Zimonjic D, Popescu N, Bonner W, Barrett J. Senescing human cells and ageing mice accumulate DNA lesions with unrepairable double-strand breaks. Nat Cell Biol. 2004;6:168–170. doi: 10.1038/ncb1095. [DOI] [PubMed] [Google Scholar]
- 82.Von Figura G, Hartmann D, Song Z, Rudolph K. Role of telomere dysfunction in ageing and its detection by biomarkers. J Mole Med. 2009;87:1165–1171. doi: 10.1007/s00109-009-0509-5. [DOI] [PubMed] [Google Scholar]
- 83.Gan W, Nie B, Shi F, Xu X, Qian J, Takagi Y, et al. Age-dependent increases in the oxidative damage of DNA, RNA, and their metabolites in normal and senescence-accelerated mice analyzed by LC-MS/MS: Urinary 8-oxoguanosine as a novel biomarker of ageing. Free Radic Biol Med. 2012;52:1700–1707. doi: 10.1016/j.freeradbiomed.2012.02.016. [DOI] [PubMed] [Google Scholar]
- 84.Meydani M. Antioxidants and cognitive function. Nutr Rev. 2001;59:S75–S82. doi: 10.1111/j.1753-4887.2001.tb05505.x. [DOI] [PubMed] [Google Scholar]
- 85.Nicolle M, Gonzales J, Sugaya K, Baskerville K, Bryan D, Lund K, et al. Signatures of hippocampal oxidative stress in aged spatial learning-impaired rodents. Neuroscience. 2001;107:415–431. doi: 10.1016/s0306-4522(01)00374-8. [DOI] [PubMed] [Google Scholar]
- 86.Vishnevskaya G, Gern E, Adzhimolaev T. Influence of the inhibition of free radical oxidation processes in early postnatal ontogenesis on learning in adult rats. Neurosci Behav Physiol. 1991;21:568–571. doi: 10.1007/BF01185951. [DOI] [PubMed] [Google Scholar]
- 87.Jhoo J, Kim H, Nabeshima T, Yamada K, Shin E, Jhoo W, et al. β-amyloid (1-42)-induced learning and memory deficits in mice: involvement of oxidative burdens in the hippocampus and cerebral cortex. Behav Brain Res. 2004;155:185–196. doi: 10.1016/j.bbr.2004.04.012. [DOI] [PubMed] [Google Scholar]
- 88.Wu A, Ying Z, Gomez-Pinilla F. Dietary omega-3 fatty acids normalize BDNF levels, reduce oxidative damage, and counteract learning disability after traumatic brain injury in rats. J Neurotrauma. 2004;21:1457–1467. doi: 10.1089/neu.2004.21.1457. [DOI] [PubMed] [Google Scholar]
- 89.Fukui K, Onodera K, Shinkai T, Suzuki S, Urano S. Impairment of learning and memory in rats caused by oxidative stress and ageing, and changes in antioxidative defense systems. Ann N Y Acad Sci. 2001;928:168–175. doi: 10.1111/j.1749-6632.2001.tb05646.x. [DOI] [PubMed] [Google Scholar]
- 90.Pietá Dias C, Martins De Lima MN, Presti-Torres J, Dornelles A, Garcia VA, Siciliani Scalco F, et al. Memantine reduces oxidative damage and enhances long-term recognition memory in aged rats. Neuroscience. 2007;146:1719–1725. doi: 10.1016/j.neuroscience.2007.03.018. [DOI] [PubMed] [Google Scholar]
- 91.Arranz L, Guayerbas N, De La Fuente M. Impairment of several immune functions in anxious women. J Psychosom Res. 2007;62:1–8. doi: 10.1016/j.jpsychores.2006.07.030. [DOI] [PubMed] [Google Scholar]
- 92.Bouayed J, Rammal H, Dicko A, Younos C, Soulimani R. Chlorogenic acid, a polyphenol from Prunus domestica (Mirabelle), with coupled anxiolytic and antioxidant effects. J Neurol Sci. 2007;262:77–84. doi: 10.1016/j.jns.2007.06.028. [DOI] [PubMed] [Google Scholar]
- 93.Krokan H, Standal R, Slupphaug G. DNA glycosylases in the base excision repair of DNA. Biochem J. 1997;325:1–16. doi: 10.1042/bj3250001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Harman D. Ageing: a theory based on free radical and radiation chemistry. J Gerontol. 1956;11:298–300. doi: 10.1093/geronj/11.3.298. [DOI] [PubMed] [Google Scholar]
- 95.Halliwell B, Gutteridge J. Free radicals in biology and medicine. Oxford University Press. 1999 [Google Scholar]
- 96.Pierce JD, Cackler AB, Arnett MG. Why should you care about free radicals? RN Journal. 2004;67:38–42. [PubMed] [Google Scholar]
- 97.Tiana L, Caib Q, Wei H. Alterations of antioxidant enzymes and oxidative damage to macromolecules in different organs of rats during ageing. Free Radic Biol Med. 1998;24:1477–1484. doi: 10.1016/s0891-5849(98)00025-2. [DOI] [PubMed] [Google Scholar]
- 98.Clausen A, Doctrow S, Baudry M. Prevention of cognitive deficits and brain oxidative stress with superoxide dismutase/catalase mimetics in aged mice. Neurobiol Aging. 2010;31:425–433. doi: 10.1016/j.neurobiolaging.2008.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Agarwal A, Saleh RA, Bedaiwy MA. Role of reactive oxygen species in the pathophysiology of human reproduction. Fertil Steril. 2003;79:829–843. doi: 10.1016/s0015-0282(02)04948-8. [DOI] [PubMed] [Google Scholar]
- 100.La Rue A, Koehler K, Wayne S, Chiulli S, Haaland K, Garry P. Nutritional status and cognitive functioning in a normally ageing sample: a 6-y reassessment. Am J Clin Nutr. 1997;65:20–29. doi: 10.1093/ajcn/65.1.20. [DOI] [PubMed] [Google Scholar]
- 101.Perrig WJ, Perrig P, Stahelin HB. The relation between antioxidants and memory performance in the old and very old. J Am Geriatr Soc. 1997;45:718–724. doi: 10.1111/j.1532-5415.1997.tb01476.x. [DOI] [PubMed] [Google Scholar]
- 102.Schmidt R, Hayn M, Reinhart B, Roob G, Schmidt H, Schumacher M, et al. Plasma antioxidants and cognitive performance in middle-aged and older adults: results of the Austrian Stroke Prevention Study. J Am Geriatr Soc. 1998;46:1407–1410. doi: 10.1111/j.1532-5415.1998.tb06008.x. [DOI] [PubMed] [Google Scholar]
- 103.Berr C, Balansard B, Arnaud J, Roussel A, Alperovitch A. Cognitive decline is associated with systemic oxidative stress: the EVA study. J Am Geriatr Soc. 2000;48:1285. doi: 10.1111/j.1532-5415.2000.tb02603.x. [DOI] [PubMed] [Google Scholar]
- 104.Bourdel-Marchasson I, Delmas-Beauvieux M, Peuchant E, Richard-Harston S, Decamps A, Regnier B, et al. Antioxidant defences and oxidative stress markers in erythorcytes and plasma from normally nourished elderly Alzheimer patients. Age Ageing. 2001;30:235–241. doi: 10.1093/ageing/30.3.235. [DOI] [PubMed] [Google Scholar]
- 105.Cherubini A, Martin A, Andres-Lacueva C, Di Iorio A, Lamponi M, Mecocci P, et al. Vitamin E levels, cognitive impairment and dementia in older persons: the In CHIANTI study. Neurobiol Aging. 2005;26:987–994. doi: 10.1016/j.neurobiolaging.2004.09.002. [DOI] [PubMed] [Google Scholar]
- 106.Larrieu S, Letenneur L, Helmer C, Dartigues J, Barberger-Gateau P. Nutritional factors and risk of incident dementia in the PAQUID longitudinal cohort. J Nutr Health Aging. 2004;8:150–154. [PubMed] [Google Scholar]
- 107.Liu J, Killilea DW, Ames BN. Age-associated mitochondrial oxidative decay: improvement of carnitine acetyltransferase substrate-binding affinity and activity in brain by feeding old rats acetyl-L-carnitine and/or R-alpha-lipoic acid. Proc Natl Acad Sci U.S.A. 2002;99:1876–1881. doi: 10.1073/pnas.261709098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Abreu RV, Silva-Oliveira EM, Moraes MFD, Pereira GS, Moraes-Santos T. Chronic coffee and caffeine ingestion effects on the cognitive function and antioxidant system of rat brains. Pharmacol Biochem Behav. 2011;99:659–664. doi: 10.1016/j.pbb.2011.06.010. [DOI] [PubMed] [Google Scholar]
- 109.Kang J, Ascherio A, Grodstein F. Fruit and vegetable consumption and cognitive decline in ageing women. Ann Neurol. 2005;57:713–720. doi: 10.1002/ana.20476. [DOI] [PubMed] [Google Scholar]
- 110.Morris M, Evans D, Tangney C, Bienias J, Wilson R. Associations of vegetable and fruit consumption with age-related cognitive change. Neurology. 2006;67:1370–1376. doi: 10.1212/01.wnl.0000240224.38978.d8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Fahnestock M, Marchese M, Head E, Pop V, Michalski B, Milgram WN, et al. BDNF increases with behavioral enrichment and an antioxidant diet in the aged dog. Neurobiol Aging. 2012;33:546–554. doi: 10.1016/j.neurobiolaging.2010.03.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Nimse SB, Pal D. Free radicals, natural antioxidants, and their reaction mechanisms. RSC Advances. 2015;5:27986–28006. [Google Scholar]
- 113.Codex Alimentarius Commission. Draft revised standard for honey. Alinorm. 2001;01/25:19–26. [Google Scholar]
- 114.Lusby P, Coombes A, Wilkinson J. Honey: a potent agent for wound healing? J Wound Ostomy Continence Nurs. 2002;29:295–300. doi: 10.1067/mjw.2002.129073. [DOI] [PubMed] [Google Scholar]
- 115.Molan PC. The limitations of the methods of identifying the floral source of honeys. Bee World. 1998;79:59–68. [Google Scholar]
- 116.Ramírez-Arriaga E, Navarro-Calvo LA, Díaz-Carbajal E. Botanical characterisation of Mexican honeys from a subtropical region (Oaxaca) based on pollen analysis. Grana. 2011;50:40–54. [Google Scholar]
- 117.Chen L, Mehta A, Berenbaum M, Zangerl AR, Engeseth NJ. Honeys from different floral sources as inhibitors of enzymatic browning in fruit and vegetable homogenates. J Agric Food Chem. 2000;48:4997–5000. doi: 10.1021/jf000373j. [DOI] [PubMed] [Google Scholar]
- 118.Wang J, Li QX. Chemical composition, characterization, and differentiation of honey botanical and geographical origins. Adv Food Nutr Res. 2011;62:89–137. doi: 10.1016/B978-0-12-385989-1.00003-X. [DOI] [PubMed] [Google Scholar]
- 119.Jaganathan S, Mandal M. Honey constituents and its apoptotic effect in colon cancer cells. JAAS. 2009;1:29–36. [Google Scholar]
- 120.Khalil I, Abdul Motalib M, Anisuzzaman Sathi Z, Hye M, Shahjahan W. Biochemical analysis of different brands of unifloral honey available at the northern region of Bangladesh. Sciences. 2001;1:385–388. [Google Scholar]
- 121.Al-Waili NS. Effects of daily consumption of honey solution on hematological indices and blood levels of minerals and enzymes in normal individuals. J Med Food. 2003;6:135–140. doi: 10.1089/109662003322233549. [DOI] [PubMed] [Google Scholar]
- 122.Schramm DD, Karim M, Schrader HR, Holt RR, Cardetti M, Keen CL. Honey with high levels of antioxidants can provide protection to healthy human subjects. J Agric Food Chem. 2003;51:1732–1735. doi: 10.1021/jf025928k. [DOI] [PubMed] [Google Scholar]
- 123.Pérez RA, Iglesias MT, Pueyo E, González M, De Lorenzo C. Amino acid composition and antioxidant capacity of Spanish honeys. J Agric Food Chem. 2007;55:360–365. doi: 10.1021/jf062055b. [DOI] [PubMed] [Google Scholar]
- 124.Bogdanov S, Jurendic T, Sieber R, Gallmann P. Honey for nutrition and health: a review. J Am Coll Nutr. 2008;27:677–689. doi: 10.1080/07315724.2008.10719745. [DOI] [PubMed] [Google Scholar]
- 125.Beretta G, Gelmini F, Lodi V, Piazzalunga A, Maffei Facino R. Profile of nitric oxide (NO) metabolites (nitrate, nitrite and N-nitroso groups) in honeys of different botanical origins: Nitrate accumulation as index of origin, quality and of therapeutic opportunities. J Pharm Biomed Anal. 2010;53:343–349. doi: 10.1016/j.jpba.2010.04.010. [DOI] [PubMed] [Google Scholar]
- 126.Yao L, Jiang Y, D’Arcy B, Singanusong R, Datta N, Caffin N, et al. Quantitative high-performance liquid chromatography analyses of flavonoids in Australian Eucalyptus honeys. J Agric Food Chem. 2004;52:210–214. doi: 10.1021/jf034990u. [DOI] [PubMed] [Google Scholar]
- 127.Dimitrova B, Gevrenova R, Anklam E. Analysis of phenolic acids in honeys of different floral origin by solid-phase extraction and high-performance liquid chromatography. Phytochem Anal. 2007;18:24–32. doi: 10.1002/pca.948. [DOI] [PubMed] [Google Scholar]
- 128.Crane E. Honey. A comprehensive survey. Crane, Russak: The University of California; 1975. [Google Scholar]
- 129.Rakha MK, Nabil ZI, Hussein AA. Cardioactive and vasoactive effects of natural wild honey against cardiac malperformance induced by hyperadrenergic activity. J Med Food. 2008;11:91–98. doi: 10.1089/jmf.2006.172. [DOI] [PubMed] [Google Scholar]
- 130.Ab Wahab SZ, Hussain NHN, Zakaria R, Kadir AA, Mohamed N, Tohit NM, et al. Long-Term Effects of Honey on Cardiovascular Parameters and Anthropometric Measurements of Postmenopausal Women. Complement Ther Med. 2018;41:154–160. doi: 10.1016/j.ctim.2018.08.015. [DOI] [PubMed] [Google Scholar]
- 131.Al-Waili NS, Saloom KY, Al-Waili TN, Al-Waili AN, Akmal M, Al-Waili FS, et al. Influence of various diet regimens on deterioration of hepatic function and hematological parameters following carbon tetrachloride: a potential protective role of natural honey. Nat Prod Res. 2006;20:1258–1264. doi: 10.1080/14786410600906475. [DOI] [PubMed] [Google Scholar]
- 132.Erejuwa OO, Sulaiman SA, Wahab MS, Sirajudeen KNS, Salleh MS, Gurtu S. Hepatoprotective effect of Tualang honey supplementation in streptozotocin-induced diabetic rats. Int J Appl Res Nat Prod. 2012;4:37–41. [Google Scholar]
- 133.Erejuwa OO, Sulaiman SA, Wahab MS, Sirajudeen KNS, Salleh MS, Gurtu S. Antioxidant protection of Malaysian Tualang honey in pancreas of normal and streptozotocin-induced diabetic rats. Ann Endocrinol (Paris) 2010;71:291–296. doi: 10.1016/j.ando.2010.03.003. [DOI] [PubMed] [Google Scholar]
- 134.Erejuwa OO, Sulaiman SA, Wahab MS, Sirajudeen KNS, Salleh MS, Gurtu S. Antioxidant protective effect of glibenclamide and metformin in combination with honey in pancreas of streptozotocin-induced diabetic rats. Int J Mol Sci. 2010;11:2056–2066. doi: 10.3390/ijms11052056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Beretta G, Granata P, Ferrero M, Orioli M, Maffei Facino R. Standardization of antioxidant properties of honey by a combination of spectrophotometric/fluorimetric assays and chemometrics. Anal Chim Acta. 2005;533:185–191. [Google Scholar]
- 136.Beretta G, Orioli M, Facino RM. Antioxidant and radical scavenging activity of honey in endothelial cell cultures (EA. hy926). Planta Med. 2007;73:1182–1189. doi: 10.1055/s-2007-981598. [DOI] [PubMed] [Google Scholar]
- 137.Erejuwa OO, Sulaiman SA, Wahab MS, Salam SK, Salleh MS, Gurtu S. Comparison of antioxidant effects of honey, glibenclamide, metformin, and their combinations in the kidneys of streptozotocin-induced diabetic rats. Int J Mol Sci. 2011;12:829–843. doi: 10.3390/ijms12010829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Erejuwa OO, Sulaiman SA, Wahab MS, Sirajudeen KNS, Salleh MS, Gurtu S. Differential responses to blood pressure and oxidative stress in streptozotocin-induced diabetic wistar-kyoto rats and spontaneously hypertensive rats: effects of antioxidant (Honey) treatment. Int J Mol Sci. 2011;12:1888–1907. doi: 10.3390/ijms12031888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Zaid SS, Sulaiman SA, Sirajudeen KNS, Othman NH. The effects of Tualang honey on female reproductive organs, tibia bone and hormonal profile in ovariectomised rats-animal model for menopause. BMC Complem Altern Med. 2010;10:82–88. doi: 10.1186/1472-6882-10-82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Mohamed M, Sulaiman SA, Jaafar H, Sirajudeen KNS. Effect of different doses of Malaysian honey on reproductive parameters in adult male rats. Andrologia. 2012;44:182–186. doi: 10.1111/j.1439-0272.2010.01159.x. [DOI] [PubMed] [Google Scholar]
- 141.Erejuwa OO, Sulaiman SA, Wahab MS, Sirajudeen KNS, Salleh MS, Gurtu S. Honey supplementation in spontaneously hypertensive rats elicits antihypertensive effect via amelioration of renal oxidative stress. Oxid Med Cell Longev. 2012;2012 doi: 10.1155/2012/374037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Tan H, Rahman R, Gan SH, Halim AS, Hassan S, Sulaiman SA, et al. The antibacterial properties of Malaysian Tualang honey against wound and enteric microorganisms in comparison to Manuka honey. BMC Complem Altern Med. 2009;9:34. doi: 10.1186/1472-6882-9-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Deng J, Liu R, Lu Q, Hao P, Xu A, Zhang J, et al. Biochemical properties, antibacterial and cellular antioxidant activities of buckwheat honey in comparison to manuka honey. Food Chem. 2018;252:243–249. doi: 10.1016/j.foodchem.2018.01.115. [DOI] [PubMed] [Google Scholar]
- 144.Imtara H, Elamine Y, Lyoussi B. Honey antibacterial effect boosting using Origanum vulgare L essential oil. Evid Based Complement Alternat Med. 2018;Article ID 7842583:14 pages. doi: 10.1155/2018/7842583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Feás X, Estevinho ML. A survey of the in vitro antifungal activity of heather (Erica Sp) organic honey. J Med Food. 2011;14:1284–1288. doi: 10.1089/jmf.2010.0211. [DOI] [PubMed] [Google Scholar]
- 146.Azonwade FE, Gbaguidi B, Paraiso A, Dah-Nouvlessounon DM, Goubalan E, Baba-Moussa F, et al. Polyphenolic profile, and antioxidant and antifungal activities of honey products in Benin. Afr J Microbiol Res. 2018;12:9–18. [Google Scholar]
- 147.Zeina B, Othman O, Al-Assad S. Effect of honey versus thyme on Rubella virus survival in vitro. J Altern Complement Med. 1996;2:345–348. doi: 10.1089/acm.1996.2.345. [DOI] [PubMed] [Google Scholar]
- 148.Kassim M, Achoui M, Mustafa MR, Mohd MA, Yusoff KM. Ellagic acid, phenolic acids, and flavonoids in Malaysian honey extracts demonstrate in vitro anti-inflammatory activity. Nutr Res. 2010;30:650–659. doi: 10.1016/j.nutres.2010.08.008. [DOI] [PubMed] [Google Scholar]
- 149.Alvarez-Suarez JM, Giampieri F, Cordero M, Gasparrini M, Forbes-Hernandez TY, Mazzoni L, et al. Activation of AMPK/Nrf2 signalling by Manuka honey protects human dermal fibroblasts against oxidative damage by improving antioxidant response and mitochondrial function promoting wound healing. J Funct Foods. 2016;25:38–49. [Google Scholar]
- 150.Fazalda A, Quraisiah A, Azlina N, Fahami M. Antiulcer effect of honey in nonsteroidal anti-inflammatory drugs induced gastric ulcer model in rats: A systematic review. Evid Based Complement Alternat Med. 2018;2018:1–12. doi: 10.1155/2018/7515692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Oršolić N, Terzić S, Šver L, Bašić I. Honey-bee products in prevention and/or therapy of murine transplantable tumours. J Sci Food Agric. 2005;85:363–370. [Google Scholar]
- 152.Fukuda M, Kobayashi K, Hirono Y, Miyagawa M, Ishida T, Ejiogu EC, et al. Jungle honey enhances immune function and antitumor activity. Evid Based Complement Alternat Med. 2011;Article ID 908743:8 pages. doi: 10.1093/ecam/nen086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Fauzi AN, Norazmi MN, Yaacob NS. Tualang honey induces apoptosis and disrupts the mitochondrial membrane potential of human breast and cervical cancer cell lines. Food Chem Toxicol. 2011;49:871–878. doi: 10.1016/j.fct.2010.12.010. [DOI] [PubMed] [Google Scholar]
- 154.Afrin S, Giampieri F, Gasparrini M, Forbes-Hernandez TY, Cianciosi D, Reboredo-Rodriguez P, et al. The inhibitory effect of Manuka honey on human colon cancer HCT-116 and LoVo cell growth Part 2: Induction of oxidative stress, alteration of mitochondrial respiration and glycolysis, and suppression of metastatic ability. Food Funct. 2018;9:2158–2170. doi: 10.1039/c8fo00165k. [DOI] [PubMed] [Google Scholar]
- 155.Al-Lawati HT, Al-Ajmi HIS, Waly MI. Antioxidant and health properties of beehive products against oxidative stress-mediated carcinogenesis. In: Waly M, Rahman M, editors. Bioactive components, diet and medical treatment in cancer prevention. Cham: Springer; 2018. [Google Scholar]
- 156.Ghazali FC. Morphological characterization study of Malaysian honey - A VPSEM, EDX randomised attempt. Annals Microscopy. 2009;9:93–102. [Google Scholar]
- 157.Khalil M, Alam N, Moniruzzaman M, Sulaiman SA, Gan SH. Phenolic acid composition and antioxidant properties of Malaysian honeys. J Food Sci. 2011;76:C921–C928. doi: 10.1111/j.1750-3841.2011.02282.x. [DOI] [PubMed] [Google Scholar]
- 158.Khalil MI, Mahaneem M, Jamalullail SMS, Alam N, Sulaiman SA. Evaluation of radical scavenging activity and colour intensity of nine Malaysian honeys of different origin. JAAS. 2011;3:4–11. [Google Scholar]
- 159.Kishore RK, Halim AS, Syazana M, Sirajudeen KNS. Tualang honey has higher phenolic content and greater radical scavenging activity compared with other honey sources. Nutr Res. 2011;31:322–325. doi: 10.1016/j.nutres.2011.03.001. [DOI] [PubMed] [Google Scholar]
- 160.Moniruzzaman M, Khalil MI, Sulaiman SA, Gan SH. Physicochemical and antioxidant properties of Malaysian honeys produced by Apis cerana, Apis dorsata and Apis mellifera. BMC Complem Altern Med. 2013;13:43–54. doi: 10.1186/1472-6882-13-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Ghashm AA, Othman NH, Khattak MN, Ismail NM, Saini R. Antiproliferative effect of Tualang honey on oral squamous cell carcinoma and osteosarcoma cell lines. BMC Complem Altern Med. 2010;10:49. doi: 10.1186/1472-6882-10-49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Ahmad I, Jimenez H, Yaacob NS, Yusuf N. Tualang honey protects keratinocytes from ultraviolet radiation-induced inflammation and DNA damage. Photochem Photobiol. 2012;88:1198–1204. doi: 10.1111/j.1751-1097.2012.01100.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Mahaneem M, Sulaiman SA, Yatiban MK, Hasnan J. Effects of Malaysian honey on the male reproductive system in rats. MJMS. 2007;14:114. [Google Scholar]
- 164.Mahaneem M, Sulaiman SA, Jaafar H, Sirajudeen KNS, Ismail Z, Islam M. Effect of honey on testicular functions in rats exposed to cigarette smoke. JAAS. 2011;3:12–17. [Google Scholar]
- 165.Kapoor S. Systemic benefits and potential uses of Tualang honey in addition to its beneficial effects on postmenopausal bone structure. Clinics. 2012;67:1345–1345. doi: 10.6061/clinics/2012(11)20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Mohd-Effendy N, Mohamed N, Muhammad N, Mohamad IN, Shuid AN. The effects of Tualang honey on bone metabolism of postmenopausal women. Evid Based Complement Alternat Med. 2012;2012:938574. doi: 10.1155/2012/938574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Zaid SSM, Sulaiman SA, Othman NH, Soelaiman I-N, Shuid AN, Mohamad N, et al. Protective effects of Tualang honey on bone structure in experimental postmenopausal rats. Clinics. 2012;67:779–784. doi: 10.6061/clinics/2012(07)13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Aljadi A, Kamaruddin M. Evaluation of the phenolic contents and antioxidant capacities of two Malaysian floral honeys. Food Chem. 2004;85:513–518. [Google Scholar]
- 169.Gheldof N, Engeseth NJ. Antioxidant capacity of honeys from various floral sources based on the determination of oxygen radical absorbance capacity and inhibition of in vitro lipoprotein oxidation in human serum samples. J Agric Food Chem. 2002;50:3050–3055. doi: 10.1021/jf0114637. [DOI] [PubMed] [Google Scholar]
- 170.Shafin N, Othman Z, Zakaria R, Nik Hussain NH. Tualang honey supplementation reduces blood oxidative stress levels/activities in postmenopausal women. ISRN Oxidative Medicine. 2014;Article ID 364836:4 pages. [Google Scholar]
- 171.Al-Rahbi B, Zakaria R, Othman Z, Hassan A, Ahmad AH. Protective effects of Tualang honey against oxidative stress and anxiety-like behaviour in stressed ovariectomized rats. Int Sch Res Notices. 2014;Article ID 521065:10 pages. doi: 10.1155/2014/521065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Oyefuga OH, Ajani EO, Salau BA, Agboola F, Adebawo OO. Honey consumption and its anti-ageing potency in White Wister albino rats. Sch J Biol Sci. 2012;1:15–19. [Google Scholar]
- 173.Wan Yaacob WMH, Long I, Zakaria R, Othman Z. Protective effects of tualang honey and its methanolic fraction against lipopolysaccharide-induced oxidative damage in the rats’ hippocampus. HEJ. 2018;9(Supplement 1):36. [Google Scholar]
- 174.Mohd Sairazi NS, Sirajudeen KNS, Asari MA, Swamy M, Muzaimi M, Sulaiman SA. Effect of tualang honey against KA-induced oxidative stress and neurodegeneration in the cortex of rats. BMC Complement Altern Med. 2017;17 doi: 10.1186/s12906-016-1534-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Wan Yaacob WMH, Long I, Zakaria R, Othman Z. Tualang honey and its methanolic fraction protect against LPS-induced neuroinflammation and amyloid deposition in male rats. AJMB. 2018:Supplement 3. [Google Scholar]
- 176.Mohd Sairazi NS, Sirajudeen KNS, Mustapha M, Swamy M, Asari MA, Sulaiman SA. Tualang honey reduced neuroinflammation and caspase-3 activity in rat brain after kainic acid-induced status epilepticus. Evid Based Complement Alternat Med. 2018;2018:7287820. doi: 10.1155/2018/7287820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Al-Rahbi B, Zakaria R, Othman Z, Hassan A, Ahmad AH. The effects of Tualang honey supplement on medial prefrontal cortex morphology and cholinergic system in stressed ovariectomised rats. IJARNP. 2014;7:28–36. [Google Scholar]
- 178.Al-Rahbi B, Zakaria R, Othman Z, Hassan A, Ahmad AH. Enhancement of the brain BDNF concentration and restoration of HPA axis reduce depressive-like behaviour in stressed ovariectomised rat treated with Tualang honey. Sci World J. 2014;2014:310821. doi: 10.1155/2014/310821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Gheldof N, Wang X-H, Engeseth NJ. Identification and quantification of antioxidant components of honeys from various floral sources. J Agric Food Chem. 2002;50:5870–5877. doi: 10.1021/jf0256135. [DOI] [PubMed] [Google Scholar]
- 180.Al ML, Daniel D, Moise A, Bobis O, Laslo L, Bogdanov S. Physico-chemical and bioactive properties of different floral origin honeys from Romania. Food Chem. 2009;112:863–867. [Google Scholar]
- 181.Ferreira IC, Aires E, Barreira J, Estevinho LM. Antioxidant activity of Portuguese honey samples: Different contributions of the entire honey and phenolic extract. Food Chem. 2009;114:1438–1443. [Google Scholar]
- 182.Tomás-Barberán FA, Martos I, Ferreres F, Radovic BS, Anklam E. HPLC flavonoid profiles as markers for the botanical origin of European unifloral honeys. J Sci Food Agric. 2001;81:485–496. [Google Scholar]
- 183.Al-Mamary M, Al-Meeri A, Al-Habori M. Antioxidant activities and total phenolics of different types of honey. Nutr Res. 2002;22:1041–1047. [Google Scholar]
- 184.Meda A, Lamien CE, Romito M, Millogo J, Nacoulma OG. Determination of the total phenolic, flavonoid and proline contents in Burkina Fasan honey, as well as their radical scavenging activity. Food Chem. 2005;91:571–577. [Google Scholar]
- 185.Gheldof N, Wang X-H, Engeseth NJ. Buckwheat honey increases serum antioxidant capacity in humans. J Agric Food Chem. 2003;51:1500–1505. doi: 10.1021/jf025897t. [DOI] [PubMed] [Google Scholar]
- 186.Pérez E, Rodríguez-Malaver AJ, Vit P. Antioxidant capacity of Venezuelan honey in wistar rat homogenates. J Med Food. 2006;9:510–516. doi: 10.1089/jmf.2006.9.510. [DOI] [PubMed] [Google Scholar]
- 187.Erejuwa OO, Sulaiman SA, Ab Wahab MS. Honey: a novel antioxidant. Molecules. 2012;17:4400–4423. doi: 10.3390/molecules17044400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Zivkovic L, Bajic V, Dekanski D, Cabarkapa-Pirkovic A, Giampieri F, Gasparrini M, et al. Manuka honey attenuates oxidative damage induced by H2O2 in human whole blood in vitro. Food Chem Toxicol. 2018;119:61–65. doi: 10.1016/j.fct.2018.05.034. [DOI] [PubMed] [Google Scholar]
- 189.Akouchekian S, Omranifard V, Maracy MR, Pedram A, Zefreh AA. Efficacy of herbal combination of sedge, saffron, and Astragalus honey on major neurocognitive disorder. J Res Med Sci. 2018;23 doi: 10.4103/jrms.JRMS_949_17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Oyekunle OA, Akanmu MA, Ogundeji TP. Evaluation of anxiolytic and novelty induced behaviours following bee-honey consumption in rats. J Neurosci Behav Health. 2010;2:38–43. [Google Scholar]
- 191.Saxena AK, Phyu HP, Al-Ani IM, Talib NA. Potential protective effect of honey against chronic cerebral hypoperfusion-induced neurodegeneration in rats. J Anat Soc India. 2014;63:151–155. [Google Scholar]
- 192.Kamarulzaidi MA, Yusoff MZM, Mohamed AM. Tualang honey consumption enhanced hippocampal pyramidal count and spatial memory performance of adult male rats. Sains Malaysiana. 2016;45:15–220. [Google Scholar]
- 193.Abdul Majeed WI, Sulieman HB, Zubayr MO, Imam A, Amin A, Biliaminu SA, et al. Honey prevents neurobehavioural deficit and oxidative stress induced by lead acetate exposure in male wistar rats - A preliminary study. Metab Brain Dis. 2016;31:37–44. doi: 10.1007/s11011-015-9733-6. [DOI] [PubMed] [Google Scholar]
- 194.Jafari Anarkooli I, Barzegar Ganji H, Pourheidar M. The protective effects of insulin and natural honey against hippocampal cell death in streptozotocin-induced diabetic rats. J Diabetes Res. 2014;Article ID 491571:8 pages. doi: 10.1155/2014/491571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Navarro-Arévalo A, Cañavate C, Sánchez-Del-Pino M. Myocardial and skeletal muscle ageing and changes in oxidative stress in relationship to rigorous exercise training. Mech Ageing Dev. 1999;108:207–217. doi: 10.1016/s0047-6374(99)00013-5. [DOI] [PubMed] [Google Scholar]
- 196.Hatao H, Oh-Ishi S, Itoh M, Leeuwenburgh C, Ohno H, Ookawara T, et al. Effects of acute exercise on lung antioxidant enzymes in young and old rats. Mech Ageing Dev. 2006;127:384–390. doi: 10.1016/j.mad.2005.12.008. [DOI] [PubMed] [Google Scholar]