Next Article in Journal
In Vitro Investigation of Six Antioxidants for Pig Diets
Previous Article in Journal
RP-HPLC/MS/MS Analysis of the Phenolic Compounds, Antioxidant and Antimicrobial Activities of Salvia L. Species
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Vitamin E, Turmeric and Saffron in Treatment of Alzheimer’s Disease

1
Koc University Medical School, Istanbul 34450, Turkey
2
Chair of Internal Medicine & Pediatrics, Alabama College of Osteopathic Medicine, Dothan, AL 36303, USA
*
Author to whom correspondence should be addressed.
Antioxidants 2016, 5(4), 40; https://doi.org/10.3390/antiox5040040
Submission received: 16 August 2016 / Revised: 2 October 2016 / Accepted: 17 October 2016 / Published: 25 October 2016

Abstract

:
Alzheimer’s disease (AD) is a growing epidemic and currently there is no cure for the disease. The disease has a detrimental effect on families and will strain the economy and health care systems of countries worldwide. The paper provides a literature review on a few ongoing possible antioxidant therapy treatments for the disease. The paper highlights use of vitamin E, turmeric and saffron for an alternative antioxidant therapy approach. Clinical studies report their therapeutic abilities as protective agents for nerve cells against free radical damage, moderating acetylcholinesterase (AChE) activity and reducing neurodegeneration, which are found as key factors in Alzheimer’s. The paper suggests that future research, with more clinical trials focused on more natural approaches and their benefits for AD treatment could be worthwhile.

1. Introduction

Alzheimer’s disease (AD) is a neurodegenerative disease, characterized by the variable deterioration of the cognitive abilities of those affected. Those who have Alzheimer’s disease begin to lose their cognitive abilities, including memory, speech, emotions, and personality. Alzheimer’s disease is a growing epidemic. In 2016, 44 million people suffer from Alzheimer’s and related dementia worldwide [1]. It is estimated that by 2050, 1 in 85 people worldwide will be afflicted with Alzheimer’s disease [2]. Alzheimer’s disease is currently the sixth leading cause of death in the United States [3]. In 2013, the journal Neurology published a study that estimated that by 2050, the number of people diagnosed with Alzheimer’s disease will be approximately 14 million in the US [3]. The AD epidemic strains the economy and health care systems of countries worldwide. Moreover, the disease has a detrimental effect on families physically, mentally, and financially. Scientists estimate the cost to be $226 billion in 2015 and up to $1.1 trillion by 2050 [4]. Ultimately, a cure for Alzheimer’s disease would be ideal; but if not, it is crucial that an efficient and cost-effective treatment plan be developed.
AD is a progressive neurodegenerative disease characterized by brain cell death that happens over a variable course of time. Although the pathogenesis of the disease is still not fully understood there is a growing consensus that behind the complex molecular mechanism appears to be the accumulation and aggregation of protein fragments. Amyloid-Beta (Aβ), known as amyloid plaques on the blood vessels and the accumulation of intracellular neurofibrillary tangles (tau) that block neurotransmitters and alter metabolism of iron, cause the destruction of nerve cells that accompanies Alzheimer’s [5,6,7,8,9,10,11,12]. It is reported that increased Aβ levels in AD patients reduce plasmalogen levels [13], increase the formation of reactive oxidative species, and slowly obstruct cerebral function [14,15]. Plasmalogens protect cells against the damaging effects of reactive oxygen species. Plasmalogens are susceptible to oxidative stress and function as antioxidants [16]. It has also been detected that plasmalogens, key structural phospholipids in neuronal membranes, are decreased in the brain of AD patients [16,17].
Clinical studies also indicate that oxidative stress is a crucial factor in AD [18,19]. Oxidative stress is a pathological state that indicates an imbalance between the production of reactive oxygen species (ROS) and the antioxidant defenses. Oxidative stress precedes Abeta production, promotes the production and aggregation of Abeta, and enhances the polymerization of intracellular neurofibrillary tangles [20]. Oxidative stress increases during ageing and is considered as a critical pathogenic factor for the onset and progression of AD [21,22,23,24,25]. Oxidative stress triggers oxidation of biomolecules leading to cellular damage [26] and has been suggested as one of the biochemical markers for the diagnosis of AD. Products of oxidative stress may diffuse into the blood and evaluating them can contribute to diagnosis of AD [27]. Although the sources of free radicals that cause oxidative stress are not fully known, some clinical studies suggest that mitochondria play an important role in the generation of free radicals, leading to oxidative damage to lipids, proteins, and nucleic acids [28]. Studies also suggest that an increase in polyunsaturated fatty acids (PUFAs) and high levels in redox metal ions could cause oxidative stress [19]. Elevated concentration of lipid peroxidation markers are detected in the brain tissue, cerebrospinal fluid and plasma of AD patients. Enhanced lipid peroxidation is linked to oxidative stress and Abeta formation [19,29,30].
A clinical study to determine the relationship between oxidative stress and cognitive performance found that high lipid peroxidation and decreased antioxidant defenses may be present early in cognitive disorders [31]. Due to the importance of the oxidative stress in AD, studies on the administration of antioxidants in AD treatment have been in progress. Antioxidants help to enhance cholinergic neuronal function or promote neuroprotective effects [32]; to convert free radicals into less reactive compounds, and provide protection for cellular components critical in the functioning of the body system [33]. PubMed, Scopus and Web of Science review on the oxidative stress studies indicate that markers of lipid peroxidation increased in AD while copper metabolism is dysregulated and total antioxidant capacity is decreased, resulting in oxidative stress that triggers neurodegeneration [34]. Applying antioxidant therapy (especially curcumin and lipid soluble vitamin E) is suggested as one of the various neuroprotective strategies in responding to oxidative stress especially in primary prevention stage [34,35].
Currently Alzheimer’s is considered an incurable disease [36]. Recovering the terminally damaged neuronal networks may not be possible because it is challenging to rejuvenate or replace dead nerve cells. Though a cure for the disease has not been found there are treatments that may help with both cognitive and behavioral symptoms of the disease [37] as well as prevention. Neuroprotective antioxidant treatment is one of the suggested treatment strategies in AD.

2. Vitamin E

Vitamin E is a potent antioxidant that may have beneficial effects in Alzheimer’s in dealing with oxidative stress [38,39] and Aβ-associated free radicals [40]. Vitamin E is a group of eight different compounds that consist four forms of tocopherols and four forms of tocotrienols attached with alpha (α), beta (β), gamma (γ), and delta (δ) vitamers which collectively support antioxidation in the body. Clinical studies reported various findings on the benefits of different forms of vitamin E. When considering eight different forms of vitamin E, some clinical studies suggest that α-tocopherols and γ-tocopherols are the two forms that are most associated with a slower rate of cognitive decline [41]. It was reported that α-tocopherol is the most bioavailable antioxidant form of vitamin E and is helpful in AD [21,42,43,44]. Alpha-tocopherol was found as protective against lipid peroxidation [44] and decreased in plasma in patients with mild AD [43].
A clinical research study investigated the influence of α-, γ- and δ-tocopherol compounds on Aβ production and degradation in neuronal cell lines [45]. Researchers found that all forms of tocopherol enhanced the Aβ production and decreased the Aβ degradation. Tocopherols increase their protein levels. Researchers reported significant differences between the tocopherol compounds. They found while α-tocopherol had minor effects on Aβ production δ-tocopherol was the most effective in increasing Aβ generation [45]. In summary, researchers concluded that vitamin E has antioxidative effects and tocopherol helps increase the amyloid-β level [45]. Researchers suggested further studies on the potential role of various vitamin E forms in treating AD, and identification of the specific vitamin form without amyloidogenic potential. Although there are relatively fewer studies on tocotrienols, it has been found that tocotrienol molecules offer more powerful antioxidant capacity and blood cholesterol-lowering properties compared to tocopherol molecules due to the differences of their molecular structure [46]. Tocopherol molecules have a long tail with no double bonds whereas tocotrienol molecules have a short tail with three double bonds which enables tocotrienol move efficiently around cells, better cleanse the arteries of accumulated cholesterol and remove the plaque in arteries [46]. Some studies found that γ-tocotrienol has a higher antioxidant effect than α-tocotrienol [47,48]. A clinical study investigated the relations of α- and γ-tocopherol and the amyloid load and neurofibrillary tangle severity in AD patients [47]. Researchers found that γ-tocopherol concentrations were related with lower amyloid load (β = −2.10; p = 0.002) and lower neurofibrillary tangle severity (β = −1.16; p = 0.02) whereas α-tocopherol was associated with higher amyloid load. Researchers concluded that γ-tocopherol could be important for the neuroprotection of the brain [47].
A number of clinical studies cited the benefits of vitamin E therapy in delaying the progression of AD [49,50,51,52,53,54,55]. To determine the effect of vitamin E (alpha tocopherol) on AD, a clinical trial on patients with mild to moderate AD was conducted. Patients who were given 2000 IU/day of alpha tocopherol compared with a placebo showed a slower functional decline. The researchers reported that vitamin E (alpha tocopherol) could be beneficial in slowing functional decline in mild to moderate AD [54]. A high intake of vitamin E is linked to preventing or slowing the progression of the disease as vitamin E has been found to be a powerful antioxidant and have neuroprotective effects [49,51,52,53,55]. Antioxidants reduce neuronal death.
Scientists recognize oxidative stress to be a hallmark of AD, as it has been shown to play a role in altering cell signaling. Oxidative stress disrupts the signaling pathway, which causes tau hyperphosphorylation [52,56]. Thus agents possessing the ability to prevent oxidative damage may be a promising approach to treating AD [52]. Also, most prior research confirms that vitamin E treatment could prevent or reduce the oxidative stress-dependent brain damage [53]. Alphatocopherol, a major form of vitamin E, may possess the ability to reduce free radical mediated damage in the brain [54]. For instance, in a clinical study conducted on patients with mild to moderate AD, participants were administered either 20 mg/day of alphatocopherol (a form of vitamin E), Memantine, both, or a placebo [54]. The researchers found that participants given solely alphatocopherol had a slower deterioration than those given the placebo. There were however, no noteworthy differences between the memantine and the combination (memantine and alphatocopherol) groups. Participants using memantine or the combination group had a greater frequency of severe adverse events constituting mainly of serious infections [54]. Researchers concluded that alphatocopherol may have benefits for patients with mild to moderate AD in slowing the progression of the disease.
Lipid oxidation and oxidized DHA are suggested to be harmful to human health. Oxidized lipid intake or lipid peroxidation can contribute to the development of tumors and atherosclerosis. Clinical studies suggest lipid peroxidation can be partially counteracted by vitamin E intake [57,58,59,60].
Numerous studies show the efficacy of antioxidants such as vitamin E as a therapeutic agent [52,61,62,63]. Vitamin E is also found effective in managing acetylcholinesterase activity [53,64,65,66]. Ahmed (2012), tested the treatment of vitamin E, acetyl-l-carnitine, and alpha-lipoic acid on AD induced rats [53]. Brains with AD are characterized by increased acetylcholinesterase levels and reduced folic acid and vitamin B12 levels. The treatment was found to restore those levels to normal, like donepezil. They found the treatments might possess potential restoring effects. There is significant evidence that dietary supplementation may delay the progression of AD and other dementias, as the vitamin E treatment significantly restored acetylcholinesterase activity and increased the Na+/K+ ATPase activity. Though donepezil was shown to have the most prominent effect, vitamin E was not far behind. Both treatments increased acetylcholinesterase inhibitor and vitamin B12 levels. Some studies found that low levels of vitamin B12 might be linked to AD [66,67,68]. Vitamin E gave the most significant rise in insulin levels by 6.4% as compared to donepezil [52]. It is found that insulin may contribute to AD [69,70] as insulin may regulate the amyloid precursor protein and the production of amyloid-beta, one of the key factors that trigger AD. A clinical trial reported that increased levels of insulin influence learning and memory processes [70] and insulin resistance may increase AD. Some clinical studies suggest that high-dose supplementation of B12 might contribute to AD by lowering the homocysteine concentration, which was found as a risk factor in AD [71,72,73].
A population-based study was conducted on the relation between the consumption of antioxidants and long-term risk of dementia. The researchers used a sample of 5000 participants, 50 years and older, without dementia. The participants were given a dietary and lifestyle guideline and they were followed up for 9.6 years. Scientists found those who had a higher intake of vitamin E were associated with lower long-term risk of dementia. Thus vitamin E rich foods may possibly have the ability to reduce long-term risk of AD. It was found that those in the highest tertile, who had an average intake of 18.5 mg/day of vitamin E, were 25% less likely to develop dementia than those in the lowest tertile who took 9 mg/day [50].
High vitamin E levels in plasma have also linked to reduced risk of Alzheimer’s in advanced age [74]. Research on the effect of vitamin E on cholesterol metabolism also has promising results [75]. Although the impact of cholesterol in neurodegeneration was not conclusive in some studies [76,77] a number of studies found cholesterol as a risk factor for Alzheimer’s disease [78,79,80]. It has been reported that cholesterol causes higher levels of amyloid plaque deposits in the brain which contributes to Alzheimer’s [79].
It has been found that vitamin E reduces cholesterol-related oxidation in the heart, liver and kidneys [81]. To determine the relationship between high cholesterol-induced oxidation and vitamin E supplements in the heart, liver and kidney, researchers gave high cholesterol with and without vitamin E supplement and regular diet, over two-month and four-month periods to four different groups of rabbits. The hearts, livers and kidneys of the sample were tested for malondialdehyde (MDA)—a biomarker for oxidative stress—and chemiluminescence (CL)—a measure of oxidative stress and antioxidant reserve.
Researchers found that the increase in the heart MDA levels in the vitamin E group was 2.42-fold compared to the 3.16-fold increase in the two-month cholesterol diet and 14.65-fold increase in the four-month cholesterol diet. Researchers found that vitamin E helped to reduce the MDA in the heart by more than 83 percent; in the kidney up to 71 percent. The liver MDA levels were similar to those of the heart. As for CL, researchers found that vitamin E helped to keep antioxidant reserve at control levels in the heart and increased the antioxidant reserve in the liver. Researchers concluded that vitamin E slows the progression of oxidative stress [81].
Research studied the type of oxidation and the duration and timing of use in affecting vitamin E’s ability to offset LDL (low density lipoprotein) oxidation [75]. It was found that vitamin E could provide an effective treatment for atherosclerosis caused by LDL oxidation if the oxidation is caused by free radical damage and if the vitamin E supplement is taken early on [75].
Although high plasma vitamin E is linked to better cognitive performance [32] there are still some studies that report some disadvantages [82,83,84,85,86,87,88,89]. For instance, the factors that may be involved with the intestinal absorption of vitamin E are not accurately available [82]. Its bioavailability, for instance, may be affected by various factors such as proteins and fat soluble micronutrients in the diet that may impact the absorption of vitamin E [82]. Still there is an ongoing research about the metabolization of vitamin E in the intestinal lumen and factors that may play a role in the absorption of vitamin E [82].
Also a number of studies found the potential pro-oxidative effects of vitamin E [83,84]. For instance, the pro-oxidative effects of high dose of vitamin E are linked with increased mortality [85] and incidents of heart failure [84,85]; α- and γ-tocopherol have pro-oxidant activity toward HDL (high density lipoprotein) [86,87] and α-tocopherols on type 2 diabetes patients, with respect to acute hyperglycaemia [88]. Clinical studies that involve vitamin E supplements also found that tocopherol could facilitate the formation of free radicals and this pro-oxidant effect may impact the increase in fatal myocardial infarctions [85,89].
Some studies on the effect of vitamin E were inconclusive. In a study over a one-year period, vitamin C (1000 mg/day), and vitamin E (400 IU/day), were given to AD patients taking cholinesterase inhibitors. The supplementation decreased lipid peroxidation markers in CSF (cerebrospinal fluid); however, no significant difference was observed in cognition of the patients [51]. In a clinical study on 769 subjects, researchers found no significant differences in the progression of AD between the placebo group and the group supplemented vitamin E [90]. The study did not report any specific form of vitamin E or the composition of vitamin E supplement.
The literature reveals a lack of consensus on the level of efficacy of vitamin E treatment. Some studies claim that it only benefits patients with mild to moderate Alzheimer’s disease [44,54]. Several studies reveal that the dosage level has no effect in delaying the neurodegeneration in patients with more advanced AD [49,91]. Also, scientists dispute whether the effect of vitamin E may differ based on the patient’s response to its antioxidant properties [51]. Therefore vitamin E acts as an antioxidant and prevents or slows decline in only some patients [51].
Some vitamin E studies reported no clinical benefit. Over the three years of the study, the scientists studied to assess the benefit of vitamin E in AD patients. For the first six weeks 1000 IU/daily initial dose of vitamin E was given and the dose was increased to 2000 IU/daily after six weeks. The study demonstrated that no significant effect of vitamin E on AD patients [91].
Various meta-analyses informed on the controversial effects of the relationship between vitamin E and mortality. A meta-analysis of 19 papers that included 135,967 patients aged 47–84 years informed that vitamin E supplementation at doses above 400 IU/day is associated with a small increase in mortality from all causes [85]. Another meta-analysis between 1988 and 2009 that included 246,371 subjects, informed that supplementation with vitamin E appears to have no effect on mortality from all causes at doses up to 5500 IU/day [92]. In a 1997 clinical study over two years, 341 AD patients were given a placebo, vitamin E (2000 IU/day dl-alpha-tocopherol), a monoamine oxidase inhibitor (selegiline), or vitamin E and selegiline in random groups. Although deterioration slowed down in those taking vitamin E separately or together, mortality increased in those who take vitamin E [93]. Another clinical study aimed to assess the association of vitamin E (at a dose of 2000 IU) alone, or in combination with a cholinesterase inhibitor (ChEI), with mortality from all causes on 847 AD patients [49]. The research showed that patients who have daily intake of vitamin E tended to survive longer than those taking acetylcholinesterase inhibitors. The scientists administered 2000 IU/day of vitamin E, a combination of vitamin E with cholinesterase inhibitors, or a placebo to the patients [49]. Researchers found the hazard ratio of vitamin E to be around 0.71 and 1.2 for the cholinesterase inhibitor. The findings did not indicate that a high dose vitamin E supplementation was linked to increased mortality.
Some scientists have questioned the safety of the vitamin E dose level as some studies may have found it linked to increased mortality [94,95]. Vitamin E may have adverse reactions such as nausea, diarrhea, fatigue, headache or bleeding. Normally an adult may consume up to 1500 IU/day of vitamin E. Though the dosage level has been shown to significantly delay the progression in patients with mild to moderate AD, the given dosage of vitamin E had no significant effect on delaying the neurodegeneration of patients with more severe AD [49,55,74]. The vitamin E treatment may not have been effective for more severe AD because of the dosage level, as it was not drastically higher than the average adult intake. Nevertheless, antioxidant vitamins are currently commonly used for patients with AD. Researchers found the intake of a vitamin E and C supplementation for more than a month caused an increase in the cerebrospinal fluid and a decrease in the cerebrospinal fluid lipid peroxidation levels. The scientists performed a clinical trial studying the effect of vitamin E and C supplementation for a year. The patients were either taking a cholinesterase inhibitor with 1000 mg/day vitamin C and 400 IU/day vitamin E or solely cholinergic medication [51]. The results indicate no significant difference between the group taking the supplementation and the control group. This finding may be attributed to the low dosage level administered. There was however a small antioxidant effect observed in the cerebrospinal fluid [51].

3. Turmeric and Saffron

Turmeric (curcumin) and saffron, natural spices commonly used in South Asian cuisine such as curry, can be considered as promising alternative treatments [35,96,97].
These spices have minor adverse effects and several studies show them to be equally as effective as Donepezil [98,99,100,101]. A review of the benefits and adverse effects of the two treatments reveals the alternative treatment, saffron and turmeric, to be the more promising solution to treating those with Alzheimer’s disease [102].
Alzheimer’s disease is an epidemic in industrialized western nations. The incidence rates for Alzheimer’s disease is much higher in the US and European nations than in East Asia [103,104]. Ballabgarh, a rural town in Northern India, is reported to have the lowest rate of Alzheimer’s disease in the world, with only 0.62% of people above the age of 55 and 1.07% above the age of 65 with the disease [103]. Two studies that tested the Alzheimer’s disease incidence rate reported the incidence rate of people above the age of 65 in Ballabgarh with Alzheimer’s disease to be only 4.7 per 1000 people as compared to 17.5 in 1000 people in Monongahela Valley, Pennsylvania, US [103,104]. There is significant evidence that the spices consumed in the Indian diet may be the reason for the low incidence rates of Alzheimer’s disease.

3.1. Turmeric Studies

Turmeric’s healing ability can be attributed to the chemical composition of turmeric. Though the spice contains over 235 compounds, the major bioactive ingredients are curcuminoids, the most common being curcumin [100]. In traditional South Asian medicine, turmeric (curcumin) is used as medicine to relieve a plethora of ailments such as wounds, gallstones, cramps, and Alzheimer’s disease due to its anti-inflammatory and antioxidant properties [96,105]. A study on the relationship between curry consumption and cognitive function in the elderly found that elderly Asians who “often” (once a month or more) or “occasionally” (one or more in six months) consumed curry had superior cognitive function to those who “rarely” (less than one in six months) consumed curry [98]. Their considerably higher MMSE (Mini-Mental State Examination) scores proved their superior cognitive function [98]. Those who “often” or “occasionally” consumed curry had the odds ratio of 0.51 and 0.62 for Alzheimer’s disease whereas those who “rarely/never” had the odds ratio of 1 [98]. Scientists reported the incidence of Alzheimer’s disease for Indians aged 70–79 to be four times lower than the rate in the US (98). Thus they concluded that turmeric enhances neurocognitive functions of Alzheimer’s patients.
A growing body of clinical research suggested curcumin is effective in AD as an antioxidant, anti-inflammatory therapeutic agent that improves the cognitive functions [97,106,107,108]. Clinical studies suggest that curcumin could be effective in decreasing oxidative damage [109,110], reducing inflammation in brain microglial cells [97,111], reversing neurodegeneration resulting from Aβ production [112,113], preventing β-amyloid protein formation [114,115], decreasing beta-amyloid plaques [107,114,116], inhibiting Aβ aggregation [117,118] and blocking cholesterol formation [119]. A clinical study on AD mice showed that AD mice that were fed low doses of curcumin had a 40% reduction in the level of beta-amyloid compared to those not fed curcumin [114]. Another clinical vitro study has found curcumin to possess the ability to prevent neuronal damage in the brain by impeding the formation of the amyloid beta plaques, thus reducing the number of plaques in the brain [104]. In a four-week clinical trial with healthy subjects [120], 80 mg/day of lipidated curcumin was given to assess the health benefits. The study reported the level of Aβ(1–40) in plasma was significantly reduced.
Altered metabolism of iron is also considered a critical factor in AD pathology. The scientists suggested that cholesterol-enriched diet may increase body iron deposition that may lead AD. Administering various metal chelators that will help prevent free radical reactions could be helpful in treating the disease [8,121,122]. Curcumin was found to have a high binding affinity for iron and copper, that may work as an iron chelator in AD [123,124]. In a clinical study mice were fed curcumin-supplemented diet and researchers observed a decline in levels of ferritin protein in the liver. The study reported that curcumin has properties of an iron chelator as it modulates proteins of iron metabolism in cells [125]. In a two-month clinical study on rats the effect of curcumin on iron overload-induced lipid peroxidation and anti-oxidant depletion was analyzed [126]. The study reported a significant reduction in both iron accumulation and endogenous anti-oxidant activities in liver and spleen of the curcumin fed rats. The researchers concluded that curcumin was effective in restoring the decrease in the hepatic and splenic antioxidant activities [126].
In a study to measure the effect of curcumin (turmeric) on focal ischemia of rats, scientists found that an injection of 1–2 mg/kg, i.v. of curcumin bettered the neurological deficit and reduced the water content in the brain [99]. Also, another study done using transgenic mice with several beta-amyloid plaques found that after administering 5 mg/day of turmeric extract for six months, the accumulation of beta amyloid plaques and tau phosphorylation was significantly reduced [100].

3.2. Saffron Studies

Saffron (Crocus sativus), a spice that also possesses therapeutic abilities, has been identified as a memory-enhancing agent [101]. A number of studies have investigated saffron’s therapeutic effects. Virgo and clinical studies reported that saffron demonstrated effective antioxidant [127] and anti-inflammatory [128,129] and antiamyloidgenic abilities.
A 22-week double blind controlled trial was done to compare Crocus sativus (Saffron extract) with the cholinesterase inhibitor donepezil (Donepezil) as treatments for subjects with mild to moderate Alzheimer’s disease. Participants in the clinical trial were randomly administered either a 30 mg/day capsule of saffron or 10 mg/day of donepezil [101]. After the 22-week study, it was found that saffron had a similar effect in the improvement of cognitive function in subjects with Alzheimer’s disease as donepezil (Donepezil), due to the acetylcholinesterase frequency being at a similar level [101]. The side effects of both treatments did not differ drastically but overall more vomiting, dizziness, fatigue, and nausea occurred with subjects taking donepezil. Subjects taking saffron had slightly more cases of dry mouth and hypomania. The study concluded that Crocus sativus was effective in treating mild-to-moderate Alzheimer’s disease due to its “antioxidant and antiamyloidgenic” abilities, thus being able to inhibit the aggregation and deposition of the beta-amyloid plaques [101].
Saffron is also reported to be benefical as a protective agent against toxicities [130] and in moderating acetylcholinesterase (AChE). AChE is associated with β-amyloid plaques and neurofibrillary tangles, a hallmark of AD [131]. In an in vitro enzymatic and molecular docking study on natural remedies that inhibit AChE, saffron extract was found to moderate AChE inhibitory activity by 30% [131]. In another study saffron extract treated rats over a period of 21 days reported that they had significantly higher levels of lipid peroxidation products and antioxidant enzymes and decreased plasma levels of corticosterone. The study concluded that saffron could be effective in impairment of learning and memory and improving the oxidative stress damage to the hippocampus caused by chronic stress [132].
In another study on the effect of saffron, on learning and memory loss and the induction of oxidative stress animals were treated with two doses of saffron extract (5 and 10 μg/rat) for a week [132]. After the seven days, treatment oxidative stress markers were measured. It was found that in the saffron treated group spatial learning and memory impairment (p < 0.05) increased and the overall antioxidant reactivity capacity and antioxidant enzymes activity were improved (p < 0.01). Researchers concluded that that saffron extract can improve the degeneration in learning and memory and the oxidative stress parameters [132].
Using turmeric and saffron as a treatment for Alzheimer’s disease may be controversial, as they may not be considered as a conventional pharmaceutical drug. However, they have been shown to be an effective treatment. Numerous studies have found saffron and turmeric to possess the ability to treat patients with Alzheimer’s disease as effectively as the conventional treatment, Donepezil. These spices used, as treatments are a safer alternative because they are natural and have less adverse effects. Yet it is suggested longitudinal studies and more clinical trials to better underpin the therapeutic effect of curcumin on AD patients [102].

4. Conclusions

A growing body of clinical study shows vitamin E, saffron, and turmeric treatment as potent antioxidants in reducing neurodegeneration and treating AD patients. Clinical studies report their therapeutic abilities as protective agent in protecting nerve cells from free radical damage and in moderating acetylcholinesterase (AChE) activity, associated with β-amyloid plaques and neurofibrillary tangles, which are found as key factors in Alzheimer’s. However, there is still more research to be done. For instance, additional research testing higher dosage ranges for vitamin E treatments needs to be conducted. Research testing the long-term efficacy of vitamin E treatments in AD patients with different levels of severity needs to be done since previous studies that administered low dosages of vitamin E did not show any effect on the neurodegeneration of AD.
Therefore, in future research turmeric and saffron therapy and various antioxidant therapies should be tested using different methods such as in vivo experiments and clinical trials while testing various dosage levels of the antioxidants. There should be more testing of both synthetic drugs and the natural substances for AD treatments. However, research should be directed towards more natural substances as they have a beneficial safety profile. It is noteworthy to be aware of side effect profiles of the natural substances and differences in manufacturing, as they may be synthetic, like the pharmaceutical drugs. In summary, the objective is to find a worldwide easily accessible and inexpensive treatment as the rate of AD is exponentially increasing.

Author Contributions

The authors equally contributed to the paper. Nur Adalier conceived the idea, conducted the study and wrote the manuscript. Heath Parker provided guidance and direction for the study and assisted for the critical revision of the manuscript for the intellectual content.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. 2016 Alzheimer’s Statistics. Available online: http://www.alzheimers.net/resources/alzheimers-statistics/ (accessed on 20 September 2016).
  2. Brookmeyer, R.E.; Johnson, K.; Ziegler-Graham, H. Forecasting the global burden of Alzheimer’s Disease. Alzheimer’s Dement. J. Alzheimer’s Assoc. 2007, 3, 186–191. [Google Scholar] [CrossRef] [PubMed]
  3. De la Torre, J. Alzheimer’s Disease is incurable but Preventable. J. Alzheimer’s Dis. 2010, 20, 861–870. [Google Scholar] [CrossRef] [PubMed]
  4. 2016 Alzheimer’s Disease Facts and Figures. Available online: http://www.alz.org/facts/ (accessed on 21 May 2016).
  5. Baranello, R.J.; Bharani, K.L.; Padmaraju, V.; Chopra, N.; Lahiri, D.K.; Greig, N.H.; Pappolla, M.A.; Sambamurti, K. Amyloid-beta protein clearance and degradation (ABCD) pathways. Curr. Alzheimer Res. 2015, 12, 32–46. [Google Scholar] [CrossRef] [PubMed]
  6. Swomley, A.M.; Förster, S.; Keeney, J.T.; Triplett, J.; Zhang, Z.; Sultana, R.; Butterfield, D.A. Abeta, oxidative stress in Alzheimer disease: Evidence based on proteomics studies. Biochim. Biophys. Acta. 2014, 1842, 1248–1257. [Google Scholar] [CrossRef] [PubMed]
  7. Stargardt, A.; Gillis, J.; Kamphuis, W.; Wiemhoefer, A.; Kooijman, L.; Raspe, M.; Benckhuijsen, W.; Drijfhout, J.W.; Hol, E.M.; Reits, E. Reduced amyloid-β degradation in early Alzheimer’s disease but not in the APPswePS1dE9 and 3xTg-AD mouse models. Aging Cell 2013, 12, 499–507. [Google Scholar] [CrossRef] [PubMed]
  8. Prasanthi, J.R.; Schrag, M.; Dasari, B.; Marwarha, G.; Dickson, A.; Kirsch, W.M.; Ghribi, O. Deferiprone reduces amyloid-β and tau phosphorylation levels but not reactive oxygen species generation in hippocampus of rabbits fed a cholesterol-enriched diet. J. Alzheimer’s Dis. 2012, 30, 167–182. [Google Scholar] [CrossRef] [PubMed]
  9. Proctor, C.J.; Pienaar, I.S.; Elson, J.L.; Kirkwood, T.B.L. Aggregation, impaired degradation and immunization targeting of amyloid-beta dimers in Alzheimer’s disease: A stochastic modelling approach. Mol. Neurodegener. 2012, 7, 32. [Google Scholar] [CrossRef] [PubMed]
  10. Murphy, M.P.; LeVine, H. Alzheimer’s Disease and the β-Amyloid Peptide. J. Alzheimer’s Dis. 2010, 19, 311. [Google Scholar] [CrossRef] [PubMed]
  11. Wollen, K.A. Alzheimer’s disease: The pros and cons of pharmaceutical, nutritional, botanical, and stimulatory therapies, with a discussion of treatment strategies from the perspective of patients and practitioners. Altern. Med. Rev. 2010, 15, 223–244. [Google Scholar] [PubMed]
  12. Wang, D.S.; Dickson, D.W.; Malter, J.W. β-Amyloid Degradation and Alzheimer’s Disease. Biomed. Biotechnol. 2006, 2006, 58406. [Google Scholar] [CrossRef] [PubMed]
  13. Grimm, M.O.; Kuchenbecker, J.; Rothhaar, T.L.; Grösgen, S.; Hundsdörfer, B.; Burg, V.K.; Friess, P.; Müller, U.; Grimm, H.S.; Riemenschneider, M.; et al. Plasmalogen synthesis is regulated via alkyl-dihydroxyacetonephosphate-synthase by amyloid precursor protein processing and is affected in Alzheimer’s disease. J. Neurochem. 2011, 116, 916–925. [Google Scholar] [CrossRef] [PubMed]
  14. Abdul, H.M.; Wenk, G.L.; Gramling, M.; Hauss-Wegrzyniak, B.; Butterfield, D.A. APP and PS-1 mutations induce brain oxidative stress independent of dietary cholesterol: Implications for Alzheimer’s disease. Neurosci. Lett. 2004, 368, 148–150. [Google Scholar] [CrossRef] [PubMed]
  15. Butterfield, D.A.; Lauderback, C.M. Lipid peroxidation and protein oxidation in Alzheimer’s disease brain: Potential causes and consequences involving amyloid β-peptide-associated free radical oxidative stress. Free Radic. Biol. Med. 2002, 32, 1050–1060. [Google Scholar] [CrossRef]
  16. Rothhaar, T.L.; Grösgen, S.; Haupenthal, V.J.; Burg, V.K.; Hundsdörfer, B.; Mett, J.; Riemenschneider, M.; Grimm, H.S.; Hartmann, T.; Grimm, M.O. Plasmalogens inhibit APP processing by directly affecting γ-secretase activity in Alzheimer’s disease. Sci. World J. 2012, 2012, 141240. [Google Scholar] [CrossRef] [PubMed]
  17. Wood, P.L.; Mankidy, R.; Ritchie, S.; Heath, D.; Wood, J.A.; Flax, J.; Goodenowe, D.B. Circulating Plasmalogen Levels and Alzheimer Disease Assessment Scale-Cognitive Scores in Alzheimer Patients. J. Psychiatry Neurosci. 2010, 35, 59–62. [Google Scholar] [CrossRef] [PubMed]
  18. Perry, G.; Cash, A.D.; Smith, M.A. Alzheimer Disease and Oxidative Stress. J. Biomed. Biotechnol. 2002, 2, 120–123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Paolo, G.D.; Kim, T.W. Linking lipids to Alzheimer’s disease: Cholesterol and beyond. Nat. Rev. Neurosci. 2011, 12, 284–296. [Google Scholar] [CrossRef] [PubMed]
  20. Zhao, Y.; Zhao, B. Oxidative Stress and the Pathogenesis of Alzheimer’s Disease. Oxid. Med. Cell. Longev. 2013, 316523. [Google Scholar] [CrossRef] [PubMed]
  21. Persson, T.; Popescu, B.O.; Cedazo-Minguez, A. Oxidative stress in Alzheimer’s disease: Why did antioxidant therapy fail? Oxid. Med. Cell. Longev. 2014, 427318. [Google Scholar] [CrossRef] [PubMed]
  22. Bradley, M.A.; Xiong-Fister, S.; Markesbery, W.R.; Lovell, M.A. Elevated 4-hydroxyhexenal in Alzheimer’s disease (AD) progression. Neurobiol. Aging 2012, 33, 1034–1044. [Google Scholar] [CrossRef] [PubMed]
  23. Ansari, M.A.; Scheff, S.W. Oxidative Stress in the Progression of Alzheimer Disease in the Frontal Cortex. J. Neuropathol. Exp. Neurol. 2010, 69, 155–167. [Google Scholar] [CrossRef] [PubMed]
  24. Gella, A.; Durany, N. Oxidative stress in Alzheimer disease. Cell. Adhes. Migr. 2009, 3, 88–93. [Google Scholar] [CrossRef]
  25. Markesbery, W.R.; Kryscio, R.J.; Lovell, M.A.; Morrow, J.D. Lipid peroxidation is an early event in the brain in amnestic mild cognitive impairment. Ann. Neurol. 2005, 58, 730–735. [Google Scholar] [CrossRef] [PubMed]
  26. Viña, J.; Lloret, A.; Giraldo, E.; Badia, M.C.; Alonso, M.D. Antioxidant pathways in Alzheimer’s disease: Possibilities of intervention. Curr. Pharm. Des. 2011, 17, 3861–3864. [Google Scholar] [CrossRef] [PubMed]
  27. Skoumalová, A.; Hort, J. Blood markers of oxidative stress in Alzheimer’s disease. J. Cell. Mol. Med. 2012, 16, 2291–2300. [Google Scholar] [CrossRef]
  28. Dumont, M.; Beal, M.F. Neuroprotective strategies involving ROS in Alzheimer disease. Free Radic. Biol. Med. 2011, 51, 1014–1026. [Google Scholar] [CrossRef] [PubMed]
  29. Chang, Y.-T.; Chang, W.; Tsai, N.-W.; Huang, C.-C.; Kung, C.-T.; Su, Y.-J.; Lin, W.-C.; Cheng, B.-C.; Su, C.-M.; Chiang, Y.-F.; et al. The Roles of Biomarkers of Oxidative Stress and Antioxidant in Alzheimer’s Disease: A Systematic Review. Biomed. Res. Int. 2014, 182303. [Google Scholar] [CrossRef] [PubMed]
  30. Kontush, A. Lipid Peroxidation and Alzheimer’s Disease: Key Role of Amyloid-Beta. Available online: http://www.ocl-journal.org/articles/ocl/pdf/2006/01/ocl2006131p46.pdf (accessed on 19 September 2016).
  31. Torres, L.L.; Quaglio, N.B.; Souza, G.T.; Garcia, R.T.; Dati, L.M.; Moreira, W.L.; Loureiro, A.P.; de Souza-Talarico, J.N.; Smid, J.; Porto, C.S.; et al. Peripheral oxidative stress biomarkers in mild cognitive impairment and Alzheimer’s disease. Alzheimer’s Dis. 2011, 26, 59–68. [Google Scholar] [CrossRef]
  32. Fata, G.L.; Weber, P.; Mohari, M.H. Effects of Vitamin E on Cognitive Performance during Ageing and in Alzheimer’s Disease. Nutrients 2014, 6, 5453–5472. [Google Scholar] [CrossRef] [PubMed]
  33. Zingg, J.M. Modulation of signal transduction by vitamin E. Mol. Asp. Med. 2007, 28, 481–506. [Google Scholar] [CrossRef] [PubMed]
  34. Schrag, M.; Mueller, C.; Zabel, M.; Crofton, A.; Kirsch, W.M.; Ghribi, O.; Squitti, R.; Perry, G. Oxidative stress in blood in Alzheimer’s disease and mild cognitive impairment: A meta-analysis. Neurobiol. Dis. 2013, 59, 100–110. [Google Scholar] [CrossRef] [PubMed]
  35. Ringman, M.; Frautschy, S.A.; Cole, G.; Masterman, D.L.; Cummings, J.L. A Potential Role of the Curry Spice Curcumin in Alzheimer’s Disease. Curr. Alzheimer Res. 2005, 2, 131–136. [Google Scholar] [CrossRef] [PubMed]
  36. De la Torre, J. A Turning Point for Alzheimer’s Disease? Biofactors 2012, 38, 78–83. [Google Scholar] [CrossRef] [PubMed]
  37. Cranwell-Bruce, L. Drugs for Alzheimer’s Disease. MEDSURG Nurs. 2010, 19, 51–53. [Google Scholar] [PubMed]
  38. Guan, J.Z.; Guan, W.-P.; Maeda, T.; Makino, N. Effect of vitamin E administration on the elevated oxygen stress and the telomeric and subtelomeric status in Alzheimer’s disease. Gerontology 2011, 58, 62–69. [Google Scholar] [CrossRef] [PubMed]
  39. Kaneai, N.; Arai, M.; Takatsu, H.; Fukui, K.; Urano, S. Vitamin E inhibits oxidative stress-induced denaturation of nerve terminal proteins involved in neurotransmission. J. Alzheimer’s Dis. 2012, 28, 183–189. [Google Scholar] [PubMed]
  40. Yatin, S.M.; Varadarajan, S.; Butterfield, D.A. Vitamin E prevents Alzheimer’s amyloid β-peptide (1–42)-induced neuronal protein oxidation and reactive oxygen species production. J. Alzheimer’s Dis. 2000, 2, 123–131. [Google Scholar] [PubMed]
  41. Morris, M.C.; Evans, D.A.; Tangney, C.C.; Bienias, J.L.; Wilson, R.S.; Aggarwal, N.T.; Scherr, P.A. Relation of the tocopherol forms to incident Alzheimer disease and to cognitive change. Am. J. Clin. Nutr. 2005, 81, 508–514. [Google Scholar] [PubMed]
  42. Joshi, Y.B.; Praticò, D. Vitamin E in aging, dementia, and Alzheimer’s disease. Biofactors 2012, 38, 90–97. [Google Scholar] [CrossRef] [PubMed]
  43. Baldeiras, I.; Santana, I.; Proença, M.T.; Garrucho, M.H.; Pascoal, R.; Rodrigues, A.; Duro, D.; Oliveira, C.R. Peripheral oxidative damage in mild cognitive impairment and mild Alzheimer’s disease. J. Alzheimer’s Dis. 2008, 15, 117–128. [Google Scholar] [PubMed]
  44. Brigelius-Flohé, R.; Traber, M.G. Vitamin E: Function and metabolism. FASEB J. 1999, 13, 1145–1155. [Google Scholar] [PubMed]
  45. Grimm, M.O.; Stahlmann, C.P.; Mett, J.; Haupenthal, V.J.; Zimmer, V.C.; Lehmann, J.; Hundsdörfer, B.; Endres, K.; Grimm, H.S.; Hartmann, T. Vitamin E: Curse or Benefit in Alzheimer’s Disease? A Systematic Investigation of the Impact of α-, γ- and δ-Tocopherol on Aβ Generation and Degradation in Neuroblastoma Cells. J. Nutr. Health Aging 2015, 19, 646–656. [Google Scholar] [CrossRef] [PubMed]
  46. Sen, C.K.; Khanna, S.; Roy, S. Tocotrienols: Vitamin E beyond Tocopherols. Life Sci. 2006, 78, 2088–2098. [Google Scholar] [CrossRef] [PubMed]
  47. Morris, M.C.; Schneider, J.A.; Li, H.; Tangney, C.C.; Nag, S.; Bennett, D.A.; Honer, W.J.; Barnas, L. Brain Tocopherols Related to Alzheimer Disease Neuropathology in Humans. Alzheimer’s Dement. 2015, 11, 32–39. [Google Scholar] [CrossRef] [PubMed]
  48. Seppanen, C.M.; Qinghua, S.A.; Saari, C. The Antioxidant Functions of Tocopherol and Tocotrienol Homologues in Oils, Fats, and Food Systems. J. Am. Oil Chem. Soc. 2010, 87, 469–481. [Google Scholar] [CrossRef]
  49. Pavlik, V.N.; Doody, R.S.; Rountree, S.D.; Darby, E.J. Vitamin E Use Is Associated with Improved Survival in an Alzheimer’s Disease Cohort. Dement. Geriatr. Cogn. Disord. 2009, 28, 536–540. [Google Scholar] [CrossRef] [PubMed]
  50. Devore, E.; Grodstein, F.; Breteler, M. Dietary antioxidants and long-term risk of dementia. Arch. Neurol. 2010, 67, 819–825. [Google Scholar] [CrossRef] [PubMed]
  51. Arlt, S.; Müller-Thomsen, T.; Beisiegel, U.; Kontush, A. Effect of One-Year Vitamin C- and E-Supplementation on Cerebrospinal Fluid Oxidation Parameters and Clinical Course in Alzheimer’s Disease. Neurochem. Res. 2012, 37, 2706–2714. [Google Scholar] [CrossRef] [PubMed]
  52. Ahmed, H. Modulatory effects of vitamin E, acetyl-l-carnitine and α-lipoic acid on new potential biomarkers for Alzheimer’s disease in rat model. Exp. Toxicol. Pathol. 2012, 6, 549–556. [Google Scholar] [CrossRef] [PubMed]
  53. Giraldo, E.; Lloret, A.; Fuchsberger, T.; Viña, J. Aβ and Tau Toxicities in Alzheimer’s are Linked via Oxidative Stress-induced P38 Activation: Protective Role of Vitamin E. Redox Biol. 2014, 2, 873–877. [Google Scholar] [CrossRef] [PubMed]
  54. Dysken, M.W.; Sano, M.; Asthana, S.; Vertrees, J.E.; Pallaki, M.; Llorente, M.; Love, S.; Schellenberg, G.D.; McCarten, J.R.; Malphurs, J.; et al. Effect of Vitamin E and Memantine on Functional Decline in Alzheimer Disease: The TEAM-AD VA Cooperative Randomized Trial. JAMA J. Am. Med. Assoc. 2014, 311, 33–44. [Google Scholar] [CrossRef] [PubMed]
  55. Isaac, M.G.; Quinn, R.; Tabet, N. Vitamin E for Alzheimer’s disease and mild cognitive impairment. Cochrane Database Syst. Rev. 2008, 16, CD002854. [Google Scholar]
  56. McIntyre, T.M.; Hazen, S.L. Lipid Oxidation and Cardiovascular Disease: Introduction to a Review Series. Circ. Res. 2010, 107, 1167–1169. [Google Scholar] [CrossRef] [PubMed]
  57. Wefers, H.; Sies, H. The protection by ascorbate and glutathione against microsomal lipid peroxidation is dependent on vitamin E. FEBS J. 1988, 174, 353–357. [Google Scholar] [CrossRef]
  58. Princen, H.M.G.; van Duyvenvoorde, W.; Buytenhek, R.; van der Laarse, A.; van Poppel, G.; Leuven, J.A.G.; van Hinsbergh, V.W.M. Supplementation with Low Doses of Vitamin E Protects LDL from Lipid Peroxidation in Men and Women. Arterioscler. Thromb. Vasc. Biol. 1995, 15, 325–333. [Google Scholar] [CrossRef] [PubMed]
  59. Cojocel1, C.; Tolle, K.L.; El-Hajj, H.; Baumann, K. Protection against cephalosporin-induced lipid peroxidation and nephrotoxicity by (+)-cyanidanol-3 and vitamin E. Braz. J. Med. Biol. Res. 2007, 40, 867–875. [Google Scholar] [CrossRef]
  60. Sahin, K.; Sahin, N.; Onderci, M.; Yaralioglu, S.; Kucuk, O. Protective role of supplemental vitamin E on lipid peroxidation, vitamins E, A and some mineral concentrations of broilers reared under heat stress. Vet. Med. Czech. 2001, 46, 140–144. [Google Scholar]
  61. Singal, A.K.; Jampana, S.C.; Weinman, S.A. Antioxidants as Therapeutic Agents for Liver Disease. Liver Int. 2011, 31, 1432–1448. [Google Scholar] [CrossRef] [PubMed]
  62. Feng, Y.; Wang, X. Antioxidant Therapies for Alzheimer’s Disease. Oxid. Med. Cell. Longev. 2012, 472932. [Google Scholar] [CrossRef] [PubMed]
  63. Asghari, A.; Montasari, A.; Khaki, A.A. An Ultrastructural Study of the Antioxidant Effects of Vitamin E and Fennel Extract on Zona Pellucida Cell Changes of Rat Ovaries under Non-Ionizing 50 Hz Electromagnetic Fields. CJMB 2015, 2, 37–41. [Google Scholar]
  64. Kim, H.S.; Lee, B.M. Protective effect of antioxidant supplementation on plasma lipid peroxidation in smokers. J. Toxicol. Environ. Health 2001, 63, 583–598. [Google Scholar] [CrossRef] [PubMed]
  65. Bittner, D.M. Combination therapy of acetylcholinesterase inhibitor and vitamin E in Alzheimer disease. J. Clin. Psychopharmacol. 2009, 29, 511–513. [Google Scholar] [CrossRef] [PubMed]
  66. Vogiatzoglou, A.; Refsum, H.; Johnston, C.; Smith, S.M.; Bradley, K.M.; de Jager, C.; Budge, M.M.; Smith, A.D. Vitamin B12 status and rate of brain volume loss in community-dwell-ing elderly. Neurology 2008, 71, 826–832. [Google Scholar] [CrossRef] [PubMed]
  67. Tucker, K.L.; Qiao, N.; Scott, T.; Rosenberg, I.; Spiro, A., 3rd. High homocysteine and low B vitamins predict cognitive decline in aging men: The Veterans Affairs Normative Aging Study. Am. J. Clin. Nutr. 2005, 82, 627–635. [Google Scholar] [PubMed]
  68. Morris, M.S.; Jacques, P.F.; Rosenberg, I.H.; Selhub, J. Folate and vitamin B-12 status in relation to anemia, macrocytosis, and cognitive impairment in older Americans in the age of folic acid fortification. Am. J. Clin. Nutr. 2007, 85, 193–200. [Google Scholar] [PubMed]
  69. Watson, G.S.; Craft, S. The role of insulin resistance in the pathogenesis of Alzheimer’s disease: Implications for treatment. CNS Drugs 2003, 17, 27–45. [Google Scholar] [CrossRef] [PubMed]
  70. Schiöth, H.B.; Frey, W.H.; Brooks, S.J.; Benedict, C. Insulin to Treat Alzheimer’s Disease. Expert Rev. Clin. Pharmacol. 2012, 5, 17–20. [Google Scholar]
  71. Ravaglia, G.; Forti, P.; Maioli, F.; Martelli, M.; Servadei, L.; Brunetti, N.; Porcellini, E.; Licastro, F. Homocysteine and folate as risk factors for dementia and Alzheimer disease. Am. J. Clin. Nutr. 2005, 82, 636–643. [Google Scholar] [PubMed]
  72. Aisen, P.S.; Schneider, L.S.; Sano, M.; Diaz-Arrastia, R.; van Dyck, C.H.; Weiner, M.F.; Bottiglieri, T.; Jin, S.; Stokes, K.T.; Thomas, R.G.; et al. High-dose B vitamin supplementation and cognitive decline in Alzheimer disease: A randomized controlled trial. J. Am. Med. Assoc. 2008, 300, 1774–1783. [Google Scholar] [CrossRef] [PubMed]
  73. Ho, R.C.; Cheung, M.W.; Fu, E.; Win, H.H.; Zaw, M.H.; Ng, A.; Mak, A. Is high homocysteine level a risk factor for cognitive decline in elderly? A systematic review, meta-analysis, and meta-regression. Am. J. Geriatr. Psychiatry 2011, 19, 607–617. [Google Scholar] [CrossRef] [PubMed]
  74. Mangialasche, F.; Kivipelto, M.; Mecocci, P.; Rizzuto, D.; Palmer, K.; Winblad, B.; Fratiglioni, L. High plasma levels of vitamin E forms and reduced Alzheimer’s disease risk in advanced age. J. Alzheimer’s Dis. 2010, 20, 1029–1037. [Google Scholar] [CrossRef] [PubMed]
  75. Niki, E. Do free radicals play causal role in atherosclerosis? Low density lipoprotein oxidation and vitamin E revisited. J. Clin. Biochem. Nutr. 2011, 48, 3–7. [Google Scholar] [CrossRef] [PubMed]
  76. Stefani, M.; Liguri, G. Cholesterol in Alzheimer’s disease: Unresolved questions. Curr. Alzheimer Res. 2009, 6, 15–29. [Google Scholar] [CrossRef] [PubMed]
  77. Ledesma, M.D.; Dotti, C.G. The conflicting role of brain cholesterol in Alzheimer’s disease: Lessons from the brain plasminogen system. Biochem. Soc. Symp. 2005, 72, 129–138. [Google Scholar] [CrossRef] [PubMed]
  78. Sjögren, M.; Blennow, K. The link between cholesterol and Alzheimer’s disease. World J. Biol. Psychiatry 2005, 6, 85–97. [Google Scholar] [CrossRef] [PubMed]
  79. Reed, B.; Villeneuve, S.; Mack, W.; DeCarli, C.; Chui, H.C.; Jagust, W. Associations between Serum Cholesterol Levels and Cerebral Amyloidosis. JAMA Neurol. 2014, 71, 195–200. [Google Scholar] [CrossRef] [PubMed]
  80. Kivipelto, M.; Solomon, A. Cholesterol as a risk factor for Alzheimer’s disease—Epidemiological evidence. Neurologica 2006, 114, 50–57. [Google Scholar] [CrossRef] [PubMed]
  81. Prasad, K.; McNair, E.D.; Qureshi, A.M.; Casper-Bell, G. Vitamin E slows the progression of hypercholesterolemia-induced oxidative stress in heart, liver and kidney. Mol. Cell. Biochem. 2012, 368, 181–187. [Google Scholar] [CrossRef] [PubMed]
  82. Borel, P.; Preveraud, D.; Desmarchelier, C. Bioavailability of vitamin E in humans: An update. Nutr. Rev. 2013, 71, 319–331. [Google Scholar] [CrossRef] [PubMed]
  83. Stepanyan, V.; Crowe, M.; Haleagrahara, N.; Bowden, B. Effects of vitamin E supplementation on exercise-induced oxidative stress: A meta-analysis. Appl. Physiol. Nutr. Metabol. 2014, 39, 1029–1037. [Google Scholar] [CrossRef] [PubMed]
  84. Saremi, A.; Arora, R. Vitamin E and cardiovascular disease. Am. J. Ther. 2010, 17, 56–65. [Google Scholar] [CrossRef] [PubMed]
  85. Miller, E.R.; Pastor-Burriuso, R.; Dalal, D.; Riemersma, R.A.; Appel, L.J.; Guallar, E. Meta-analysis: High-dosage vitamin E supplementation may increase all-cause mortality. Ann. Intern. Med. 2005, 142, 37–46. [Google Scholar] [CrossRef] [PubMed]
  86. Chapman, T.M.; Kim, H.J.; Min, D.B. Prooxidant activity of oxidized alpha-tocopherol in vegetable oils. J. Food Sci. 2009, 74, 536–542. [Google Scholar] [CrossRef] [PubMed]
  87. Nadeem, N.; Woodside, J.V.; Kelly, S.; Allister, R.; Young, I.S.; McEneny, J. The two faces of α- and γ-tocopherols: An in vitro and ex vivo investigation into VLDL, LDL and HDL oxidation. J. Nutr. Biochem. 2012, 23, 845–851. [Google Scholar] [CrossRef] [PubMed]
  88. Winterbone, M.S.; Sampson, M.J.; Saha, S.; Hughes, J.C.; Hughes, D.A. Pro-oxidant effect of alpha-tocopherol in patients with type 2 diabetes after an oral glucose tolerance test-a randomised controlled trial. Cardiovasc. Diabetol. 2007, 6, 8. [Google Scholar] [CrossRef] [PubMed]
  89. Bast, A.; Haenen, G.R.R.M. Interplay between glutathione and lipoic acid in the protection against microsomal lipid peroxidation. Biochim. Biophys. Acta. 1988, 963, 558–561. [Google Scholar] [CrossRef]
  90. Kang, J.H.; Cook, N.; Manson, J.; Buring, J.E.; Grodstein, F. A randomized trial of vitamin E supplementation and cognitive function in women. Arch. Intern. Med. 2006, 166, 2462–2468. [Google Scholar] [CrossRef] [PubMed]
  91. Petersen, R.C.; Thomas, R.C.; Grundman, M.; Bennett, D.; Doody, R.; Ferris, S.; Galasko, D.; Jin, S.; Kaye, J.; Levey, A.; et al. Vitamin E and Donepezil for the Treatment of Mild Cognitive Impairment. N. Engl. J. Med. 2005, 352, 2379–2388. [Google Scholar] [CrossRef] [PubMed]
  92. Abner, E.L.; Schmitt, F.A.; Mendiondo, M.S.; Marcum, J.L.; Kryscio, R.J. Vitamin E and all-cause mortality: A meta-analysis. Curr. Aging Sci. 2011, 4, 158–170. [Google Scholar] [CrossRef] [PubMed]
  93. Sano, M.; Ernesto, C.; Thomas, R.G.; Klauber, M.R.; Schafer, K.; Grundman, M.; Woodbury, P.; Growdon, J.; Cotman, C.W.; Pfeiffer, E.; et al. A controlled trial of selegiline, alpha-tocopherol, or both as treatment for Alzheimer’s disease: The Alzheimer’s Disease Cooperative Study. N. Engl. J. Med. 1997, 336, 1216–1222. [Google Scholar] [CrossRef] [PubMed]
  94. Hayden, K.M.; Welsh-Bohmer, K.A.; Wengreen, H.J.; Zandi, P.P.; Lyketsos, C.G.; Breitner, J.C.S. Risk of mortality with vitamin E supplements: The Cache County Study. Am. J. Med. 2007, 120, 180–184. [Google Scholar] [CrossRef] [PubMed]
  95. Bjelakovic, G.; Nikolova, D.; Gluud, L.L.; Simonetti, R.G.; Gluud, C. Mortality in randomized trials of antioxidant supplements for primary and secondary prevention: Sytematic review and meta-analysis. JAMA 2007, 297, 842–857. [Google Scholar] [CrossRef] [PubMed]
  96. Mishra, S.; Palanivelu, K. The Effect of Curcumin (Turmeric) on Alzheimer’s Disease: An Overview. Ann. Indian Acad. Neurol. 2008, 11, 13–19. [Google Scholar] [CrossRef] [PubMed]
  97. Ng, T.P.; Chiam, P.C.; Lee, T.; Chua, H.C.; Lim, L.; Kua, E.H. Curry consumption and cognitive function in the elderly. Am. J. Epidemiol. 2006, 164, 898–906. [Google Scholar] [CrossRef] [PubMed]
  98. Jiang, J.W.; Wang, Y.J.; Sun, M.; Hu, F.; Zhu, D.Y. Neuroprotective Effect of Curcumin on Focal Cerebral Ischemic Rats by Preventing Blood-Brain Barrier Damage. Eur. J. Pharmacol. 2007, 30, 54–62. [Google Scholar] [CrossRef] [PubMed]
  99. Akhondzadeh, S.; Shafiee Sabet, M.; Harirchian, M.H.; Togha, M.; Cheraghmakani, H.; Razeghi, S.; Hejazi, S.S.; Yousefi, M.H.; Alimardani, R.; Jamshidi, A.; et al. A 22-week, Multicenter, Randomized, Double-blind Controlled Trial of Crocus Sativus in the Treatment of Mild-to-Moderate Alzheimer’s Disease. Psychopharmacology 2010, 207, 637–643. [Google Scholar] [CrossRef] [PubMed]
  100. Zhang, L.; Fiala, M.; Cashman, J.; Sayre, J.; Espinosa, A.; Mahanian, M.; Zaghi, J.; Badmaev, V.; Graves, M.C.; Bernard, G.; et al. Curcuminoids enhance amyloid-beta uptake by macrophages of Alzheimer’s disease patients. J. Alzheimer’s Dis. 2006, 10, 1–7. [Google Scholar] [PubMed]
  101. Brondino, N.; Re, S.; Boldrini, A.; Cuccomarino, A.; Lanati, N.; Barale, F.; Politi, P. Curcumin as a Therapeutic Agent in Dementia: A Mini Systematic Review of Human Studies. Sci. World J. 2014, 174282. [Google Scholar] [CrossRef] [PubMed]
  102. Chandra, V.; Pandav, R.; DeKosky, S. Incidence of Alzheimer’s disease in a rural community in India. The Indo-US study. Neurology 2001, 57, 985–989. [Google Scholar] [CrossRef] [PubMed]
  103. Hishikawa, N.; Takahashi, Y.; Krishna, U. Effects of turmeric on Alzheimer’s disease with behavioral and psychological symptoms of dementia. Ayurveda 2012, 33, 499–504. [Google Scholar] [CrossRef] [PubMed]
  104. Ahmed, T.; Gilani, A. Therapeutic Potential of Turmeric in Alzheimer’s Disease: Curcumin or Curcuminoids? Phytother. Res. 2014, 28, 517–525. [Google Scholar] [CrossRef] [PubMed]
  105. Al-Rubaei, Z.M.; Mohammad, T.U.; Ali, L.K. Effects of local curcumin on oxidative stress and total antioxidant capacity in vivo study. Pak. J. Biol. Sci. 2014, 17, 1237–1241. [Google Scholar] [CrossRef] [PubMed]
  106. Yao, E.C.; Xue, L. Therapeutic Effects of Curcumin on Alzheimer’s Disease. Adv. Alzheimer’s Dis. 2014, 3, 145–159. [Google Scholar] [CrossRef]
  107. Giri, R.K.; Rajagopal, V.; Kalra, V.K. Curcumin, the active constituent of turmeric, inhibits amyloid peptide-induced cytochemokine gene expression and CCR5-mediated chemotaxis of THP-1 monocytes by modulating early growth response-1 transcription factor. J. Neurochem. 2004, 91, 1199–1210. [Google Scholar] [CrossRef] [PubMed]
  108. Suryanarayana, P.; Satyanarayana, A.; Balakrishna, N.; Kumar, P.U.; Reddy, G.B. Effect of turmeric and curcumin on oxidative stress and antioxidant enzymes in streptozotocin-induced diabetic rat. Med. Sci. Monit. 2007, 13, 286–292. [Google Scholar]
  109. Lim, G.P.; Chu, T.; Yang, F.; Beech, W.; Frautschy, S.A.; Cole, G.M. The Curry Spice Curcumin Reduces Oxidative Damage and Amyloid Pathology in an Alzheimer Transgenic Mouse. J. Neurosci. 2001, 21, 8370–8377. [Google Scholar] [PubMed]
  110. Kim, H.Y.; Park, E.J.; Joe, E.H.; Jou, I. Curcumin suppresses Janus kinase-STAT inflammatory signaling through activation of Src homology 2 domain-containing tyrosine phosphatase 2 in brain microglia. J. Immunol. 2003, 171, 6072–6079. [Google Scholar] [CrossRef] [PubMed]
  111. Begum, A.N.; Jones, M.R.; Lim, G.P.; Morihara, T.; Kim, P.; Heath, D.D.; Rock, C.L.; Pruitt, M.A.; Yang, F.; Hudspeth, B.; et al. Curcumin structure-function, bioavailability, and efficacy in models of neuroinflammation and Alzheimer’s disease. J. Pharmacol. Exp. Ther. 2008, 326, 196–208. [Google Scholar] [CrossRef] [PubMed]
  112. Garcia-Alloza, M.; Borrelli, L.A.; Rozkalne, A.; Hyman, B.T.; Bacskai, B.J. Curcumin Labels Amyloid Pa-thology in vivo, Disrupts Existing Plaques, and Partially Restores Distorted Neurites in an Alzheimer Mouse Model. J. Neurochem. 2007, 102, 1095–1104. [Google Scholar] [CrossRef] [PubMed]
  113. Yang, F.; Lim, P.; Begum, A.N.; Ubeda, O.J.; Simmons, M.R.; Ambegaokar, S.S.; Chen, P.P.; Kayed, R.; Glabe, C.G.; Frautschy, S.A.; et al. Curcumin inhibits formation of amyloid beta oligomers and fibrils, binds plaques, and reduces amyloid in vivo. Neurochem. Int. 2009, 54, 199–204. [Google Scholar]
  114. Shytle, R.D.; Bickford, P.C.; Rezai-zadeh, K.; Zeng, H.L.; Tan, J.; Sanberg, P.R.; Sanberg, C.D.; Roschek, B.; Fink, R.C.; Alberte, R.S. Optimized turmeric extracts have potent anti-amyloidogenic effects. Curr. Alzheimer Res. 2009, 6, 564–571. [Google Scholar] [CrossRef] [PubMed]
  115. Fiala, M.; Liu, P.T.; Espinosa-Jeffrey, A.; Rosenthal, M.J.; Bernard, G.; Ringman, J.M.; Sayre, J.; Zhang, L.; Zaghi, J.; Dejbakhsh, S.; et al. Innate immunity and transcription of MGAT-III and Toll-like receptors in Alzheimers disease patients are improved by bisdemethoxycurcumin. Proc. Natl. Acad. Sci. USA 2007, 104, 12849–12854. [Google Scholar] [CrossRef] [PubMed]
  116. Ono, K.; Hasegawa, K.; Naiki, H.; Yamada, M. Curcumin has potent anti-amyloidogenic effects for Alzheimer’s β-amyloid fibrils in vitro. J. Neurosci. Res. 2004, 75, 742–750. [Google Scholar] [CrossRef] [PubMed]
  117. Reddy, C.; Lokesh, B.R. Studies on spice principles as antioxidants in the inhibition of lipid peroxidation of rat liver microsomes. Mol. Cell. Biochem. 1992, 111, 117–124. [Google Scholar] [PubMed]
  118. Soni, K.B.; Kuttan, R. Effect of oral curcumin administration on serum peroxides and cholesterol in human volunteers. Indian J. Physiol. Pharmacol. 1992, 36, 273–275. [Google Scholar] [PubMed]
  119. Disilvestro, R.A.; Joseph, E.; Zhao, S.; Bomser, J. Diverse effects of a low dose supplement of lipidated curcumin in healthy middle aged people. Nutr. J. 2012, 11, 79. [Google Scholar] [CrossRef] [PubMed]
  120. Jomova, K.; Valko, M. Importance of iron chelation in free radical-induced oxidative stress and human disease. Curr. Pharm. Des. 2011, 17, 3460–3473. [Google Scholar] [CrossRef] [PubMed]
  121. Cuajungco, M.P.; Fagét, K.Y.; Huang, X.; Tanzi, R.E.; Bush, A.I. Metal chelation as a potential therapy for Alzheimer’s disease. Ann. N. Y. Acad. Sci. 2000, 920, 292–304. [Google Scholar] [CrossRef] [PubMed]
  122. Baum, L.; Ng, A. Curcumin interaction with copper and iron suggests one possible mechanism of action in Alzheimer’s disease animal models. J. Alzheimer’s Dis. 2004, 6, 367–377, discussion 443–449. [Google Scholar] [PubMed]
  123. Mandel, S.; Amit, T.; Bar-Am, O.; Youdim, M.B. Iron dysregulation in Alzheimer’s disease: Multimodal brain permeable iron chelating drugs, possessing neuroprotective-neurorescue and amyloid precursor protein-processing regulatory activities as therapeutic agents. Prog. Neurobiol. 2007, 82, 348–360. [Google Scholar] [CrossRef] [PubMed]
  124. Jiao, Y.; Wilkinson, J.; Pietsch, C.E.; Buss, J.L.; Wang, W.; Planalp, R.; Torti, F.M.; Torti, S.V. Iron chelation in the biological activity of curcumin. Free Radic. Biol. Med. 2006, 40, 1152–1160. [Google Scholar] [CrossRef] [PubMed]
  125. Badria, F.A.; Ibrahim, A.S.; Badria, A.F.; Elmarak, A.A. Curcumin Attenuates Iron Accumulation and Oxidative Stress in the Liver and Spleen of Chronic Iron-Overloaded Rats. PLoS ONE 2015, 10, e0134156. [Google Scholar] [CrossRef] [PubMed]
  126. Hosseinzadeh, H.; Shamsaie, F.; Mehri, S. Antioxidant activity of aqueous and ethanolic extracts of Crocus sativus L. stigma and its bioactive constituents crocin and safranal. Pharmacogn. Mag. 2010, 5, 419–424. [Google Scholar]
  127. Hosseinzadeh, H.; Younesi, M.H. Antinociceptive and anti-inflammatory effects of Crocus sativus L. stigma and petal extracts in mice. BMC Pharmacol. 2002, 2, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Hosseinzadeh, H.; Shariaty, V.M. Anti-nociceptive effect of safranal, a constituent of Crocus sativus (saffron), in mice. Pharmacologyonline 2007, 2, 498–503. [Google Scholar]
  129. Razavi, B.M.; Hosseinzadeh, H. DARU Saffron as an antidote or a protective agent against natural or chemical toxicities. J. Pharm. Sci. 2015, 23, 31. [Google Scholar] [CrossRef]
  130. Geromichalos, G.D.; Lamari, F.N.; Papandreou, M.A.; Trafalis, D.T.; Margarity, M.; Papageorgiou, P.; Sinakos, Z. Saffron as a Source of Novel Acetylcholinesterase Inhibitors: Molecular Docking and in vitro Enzymatic Studies. J. Agric. Food Chem. 2012, 60, 6131–6138. [Google Scholar] [CrossRef] [PubMed]
  131. Ghadrdoost, B.; Vafaei, A.A.; Rashidy-Pour, A.; Hajisoltani, R.; Bandegi, A.R.; Motamedi, F.; Haghighi, S.; Sameni, H.R.; Pahlvan, S. Protective effects of saffron extract and its active constituent crocin against oxidative stress and spatial learning and memory deficits induced by chronic stress in rats. Eur. J. Pharmacol. 2011, 667, 222–229. [Google Scholar] [CrossRef] [PubMed]
  132. Ghaffari, S.H.; Hatami, H.; Dehghan, G. Saffron ethanolic extract attenuates oxidative stress, spatial learning, and memory impairments induced by local injection of ethidium bromide. Res. Pharm. Sci. 2015, 10, 222–232. [Google Scholar] [PubMed]

Share and Cite

MDPI and ACS Style

Adalier, N.; Parker, H. Vitamin E, Turmeric and Saffron in Treatment of Alzheimer’s Disease. Antioxidants 2016, 5, 40. https://doi.org/10.3390/antiox5040040

AMA Style

Adalier N, Parker H. Vitamin E, Turmeric and Saffron in Treatment of Alzheimer’s Disease. Antioxidants. 2016; 5(4):40. https://doi.org/10.3390/antiox5040040

Chicago/Turabian Style

Adalier, Nur, and Heath Parker. 2016. "Vitamin E, Turmeric and Saffron in Treatment of Alzheimer’s Disease" Antioxidants 5, no. 4: 40. https://doi.org/10.3390/antiox5040040

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop