Not for Sale or Public Distribution

3 downloads 0 Views 2MB Size Report
Neurocardiology and neuropulmonary. Am Acad. Neurol 2003; Honolulu 3FC.001: 1-96. [53]. Larner AJ, Janssen JC, Cipolotti L, Rossor MN. Cognitive profile.
163

Send Orders for Reprints to [email protected] Current Nutrition & Food Science, 2017, 13, 163-175

REVIEW ARTICLE ISSN: 1573-4013 eISSN: 2212-3881

Homocysteine in Neurology: From Endothelium to Neurodegeneration BENTHAM SCIENCE

Rita Morettia,*, Matteo Dal Benb,c, Silvia Gazzinb and Claudio Tiribellib,c a

Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy; bItalian Liver Foundation, Centro Studi Fegato, SS14, Km 163.5, Trieste, 34149, Italy; cDepartment of Medical, Surgical, and Health Sciences, University of Trieste, Trieste, Italy

Received: September 23, 2016 Revised: January 17, 2017 Accepted: February 02, 2017

fo

DOI: 10.2174/1573401313666170213155338

rS o ic r a le D is tri bu tio n

A R T I C L E H I S T O R Y

Abstract: Vitamin B12 and folate are supplied via two major pathways, the conversion of homocysteine to methionine and the conversion of methyl malonyl coenzyme A to succinyl coenzyme A. Therefore, the defect in both the vitamins results in an increase in both serum homocysteine and methylmalonic acid. Hence, homocysteine, vitamin B12, and folate are closely linked together in the so-called one-carbon cycle, making vitamin B12 the necessary co-enzyme for the methyl donation from 5-methyl-tetra-hydrofolate in tetra-hydro-folate, necessary for methionine synthetase. Folate is a cofactor in one-carbon metabolism, and it promotes the remethylation of homocysteine, which can cause DNA strand breakage, oxidative stress and apoptosis. Vitamin B12 and folate are involved in nucleic acid synthesis and in the methylation reactions, and their deficit causes the inhibition of S-adenosylmethionine mediated methylation reactions, and through the related toxic effects of homocysteine, a possible direct alteration of the vascular endothelium and inhibition of N-methyl-D-Aspartate receptors take place. We discussed the possible and still controversial role of homocysteine accumulation in cerebral pathologies, starting from vascular events to neurodegeneration and to endothelium damage mechanism.

1. INTRODUCTION

ot

Keywords: Homocysteine, folate, vitamin B12-cobalamin, dementias, psychiatric symptoms, vascular endothelium, folate-vitamin B12 supplementation, therapy.

N

2. BIOCHEMICAL LINKS BETWEEN COBALAMIN (VITAMIN B12), FOLATE AND HOMOCYSTEINE

The pathway of one-carbon metabolism activates onecarbon units, normally from serine, tighten to tetrahydrofolate. The produced 5,10-methylentetrahydrofolate is employed for the production of thymidylate and purines (fundamental for nucleic acid synthesis) and of methionine, fundamental for methylation-process [7]. The methionine synthesis is anticipated by the reduction of 5,10-methylentetrahydrofolate to 5-methyltetrahydrofolate (5-methylTHF) catalysed by the flavin-containing methylenetetrahydrofolate reductase [7]. 5-methyltetrahydrofolate is the substrate to methylate homocysteine, employing vitamin B12 as a catalyzer [7]. Methionine adenosyltransferase (MAT) catalyzes S-adenosylmethionine (AdoMet), in a reaction involving methionine and ATP [8, 9], and every reaction made by methyltransferases produces S-adenosylhomocysteine (AdoHcy) which is a potent inhibitor of most of them [9-11].

bl

When one discuss homocysteine, he should declare that he will also speak about folate and vitamin B12 [1-5]. Homocysteine is not involved in protein synthesis, rather as an intermediate in methionine metabolism [5] and it is either degraded via the remethylation pathway or converted, via the transsulfuration pathway into cysteine [5] (Fig. 1).

Pu

Current Nutrition & Food Science



Being that folate and vitamin B12 are necessary for the methylation process, and that these reactions are fundamental to the biochemical basis of brain metabolism, the deficiency of vitamin B12 and folate can have possible consequences in neurological diseases [4, 5]. Vitamin B12 transfers the methyl group to homocysteine and is determinant in the synthesis of methionine, which is needed for the syn-thesis of S-adenosylmethionine (SAM), the sole donor in numerous methylation reactions involving proteins, phospholipids and biogenic amines [6]. We discuss different results from many experimental studies on the topic.

*Address correspondence to this author at the Department of Medical, Surgical, and Health Sciences, University of Trieste, Trieste, Italy; Tel: +39-040-399-4572, E-mail: [email protected]

2212-3881/17 $58.00+.00

In general, AdoHcy hydrolase (SAHH) acts on AdoHcy, producing adenosine and homocysteine [10, 11], and they need to be metabolized or transported out of the cell, to prevent their accumulation [11, 12]. This hydrolysis is a reversible reaction that favors S-Adenosyl-L-homocysteine (SAH) synthesis. This event occurs regularly in the case of

© 2017 Bentham Science Publishers

164 Current Nutrition & Food Science, 2017, Vol. 13, No. 3

Moretti et al.

Homocysteine accumulation Psychiatric symptoms (schizophrenia, depression, mental retardation) Dementias Arterial and venous occlusive disease Damage to the vascular endothelium Inhibition/enhancement of NMDA (depending on glycine availability) DNA strand breakage Oxidative stress Apoptosis

Vit B12 deficiency Dementias (debated) Peripheral Neuropathy

Folate deficiency Developmental delay Cognitive deterioration Motor & gait abnormalities Behavioral & psychiatric symptoms Seizures Demyelination defects Vascular dementia & stroke Cerebral atrophy

CYSTEINE CBS

SAM

Nucleic Acid Synthesis SAM

HOMOCYSTEINE MTHF

SAH

Met-THF Proteins from diet

MTR

Inactive folates from diet

rS o ic r a le D is tri bu tio n

Methylation Reactions (Proteins Phospholipids Biogenic Amines)

MTHFR

Glycine

M-B12

SAM

&

Methionine

A-B12

MMA-CoA

S-CoA

fo

AA & Odd chain FA

Serine

THF

N

ot

Fig. (1). The one carbon cycle with special emphasis to the brain. Blue circle: Methionine cycle. MTR: Methionine Synthase; M-B12: Methyl Cobalamine. SAM: S-Adenosyl-Methionine; SAH: S-AdenosylL-Homocysteine. Purple Circle: Trans Sulfuration Pathway. CBS: Cystathionine  Synthase. Pink circle: Folate Cycle. MTHF: MethlyTetraHydroFolate; MTHFR: Methylene TetraHydroFolate Reducatse; Met-THF: Methylene-TetraHydroFolate. THF: Tetrahydrofolate. Yellow circle: AA: Amino Acids; FA: Fatty Acids; MMA-CoA: Methtyl-malonyl Coenzyme A. A-B12: Adenosyl Cobalamine. S-CoA: Succinyl-CoA. Green circles: Key players involved in homocysteine recycling.

Pu

bl

folate or vitamin B12 deficiency, leading to SAH intracellular accumulation [7, 12, 13]. If homocysteine is allowed to accumulate in normal conditions, it will be rapidly metabolized to SAH, which competes with SAM, for the active site on the methyltransferase system [6, 13-16]. The oxidation of homocysteine to homocysteic acid is one of the potential explanations with many dangerous effects of homocysteine, due to the fact that it is a mixed excitatory agonist of NMDA receptors [17]. Homocysteine is also methylated by betaine [6, 12,18], not operative in the brain [19-21]. Homocysteine remethylation is catalyzed by the methionine synthase (MTR) enzyme [19] and this is the keypoint of anchorage between folate, vitamin B12, and homocysteine. MTR requires vitamin B12 (Cbl) [9, 19, 22, 23]. During the transsulfuration pathway, homocysteine is irreversibly degraded to cysteine [6-11]. Cysteine is a precursor of glutathione, the most fundamental endogenous antioxidant [6, 11, 24]. In most tissues, homocysteine is either remethylated or exported out of the cell [6]. The liver is the main organ of

degradation of excess methionine and in maintaining homocysteine at adequate levels [11]. In the liver, there is a direct and univocal correspondence, between the increase of methionine and a concomitant increment of AdoMet [6, 7, 11], and due to an intrinsic auto regulatory system, AdoMet inhibits methylenetetrahydrofolate reductase (MTHFR) and activates CBS activity [25-28].

Excess methionine causes an important homocysteine degradation, via the transsulfuration pathway. Lack of methionine conserves homocysteine, via remethylation back to methionine [11, 21, 24]. Therefore, 5-methylTHF functions as a methyl donor for homocysteine remethylation [26-29]. The resulting tetrahydrofolate (THF) can directly be converted into 5,10methyleneTHF by the action of serine hydroxymethyltransferase (SHMT) [27-29]. The conversion of THF into 5,10methyleneTHF is catalyzed by methylenetetrahydrofolate dehydrogenase (MTHFD1) [26-29]; MTHFR enzyme, therefore, regulates the passage from 5-methyl THF to homocysteine remethylation [29].

Homocysteine in Neurology

Current Nutrition & Food Science, 2017, Vol. 13, No. 3

3. FOLIC ACID Dietary folic acid predominantly exists as polyglutamate, which have to be hydrolyzed to monoglutamates in order to be available [6]. To hydrolyze folic acid, in the gut, the folylpoly-gamma-glutamate carboxypeptidase (FGCP) is fundamental, anchored to the intestinal apical brush border and encoded by the glutamate carboxypeptidase II (GCPII) gene [6, 30, 31, 33]. Mono-glutamylated folates are then captured in the duodenum and in the upper part of the jejunum, by the high-affinity proton-coupled folate receptor (PCFT1) [32-34].

N

ot

fo

Additionally, in hematological conditions, where there is a defect in homocysteine methyltransferase, or when there is a combined deficiency of B12 and hcy-methyltransferase, a specific, though rare, combined effect occurs, defined as "methyl-trap" of tetrahydrofolate; THF is converted to a reservoir of methyl-THF, and therefore folic acid is trapped and cannot be employed [42]. 4. VITAMIN B12

Only in the most severe cases of gastric atrophy does intrinsic factor secretion become a rate-limiting factor for vitamin B12 absorption [7]. Most frequently, atrophic gastritis enhances the impaired release of vitamin B12 from food proteins and peptides due to impaired acid secretion [6, 7]. Vitamin B12 deficiency might be common in the frailest population including children and elderly [44]. There are different causes which can produce cobalamin deficiency: an inadequate intake (vegans, and also vegetarians but to a lesser degree, see the next chapter), malabsorption (a. inadequate production of intrinsic factor; b. disorders of the terminal ileum, Crohn’s disease and small bowel syndrome; c. competition for cobalamin, in particular in the so-called blind loop syndrome and celiac disease) [2, 3-6, 41], drugs (such as ranitidine, metformin, anticonvulsivants, chemotherapy agents, alcohol etc.), genetic deficiency of transcobalamin II [2, 41-44]. 5. FOLIC ACID AND VITAMIN B12: THEIR INTRINSIC CLINICAL RELATIONSHIP There is an intimate, biochemically known but not completely clinically understood, relationship between folic acid and vitamin B12, as highlighted in some clinical observations. Treating a B12 deficient patient with folate or conversely a folate deficient patient with B12 may exacerbate the neurologic consequences or either deficiency [6, 45-47]. The finding of such few “pure” cases reported from the literature of dementia due to vitamin B12 defect (even considering the wide-spectrum of possible causes of its defects) may be suggestive of different possible interactions of this defect to another causative role, such as folate exposure. We have reported four cases [46], where the onset of cognitive impairment due to a lack of vitamin B12 has been determined by a concomitant and erroneous assumption of folate; even the other case [47] was “spurious” in the sense that the B12 defect was enriched with a concomitant intake of folate, and not supplemented directly by vitamin B12 [6, 46, 47].

rS o ic r a le D is tri bu tio n

There are many physiological conditions which require more folate in human life than necessary, such as the growing process, recovery from systematic illness, and pregnancy [35-39]. Pathological deficiency of folate may be determined by inadequate dietary intake, infectious status [6-38], and alcoholism [6-40]. Diffuse inflammatory or degenerative diseases of the small intestine, such as Crohn's disease and coeliac disease, may reduce the activity of pteroyl polyglutamase (PPGH), a specific hydrolase required for folate absorption, and thereby lead to folate deficiency [41]. Some other situations that increase the need for folate include the following: systemic or occult hemorrhage, kidney dialysis and chronic liver disease [40-43]. Medications might interfere with folate absorption, such as many anti-epileptic drugs (phenytoin, primidone, carbamazepine or valproate), or metformin; methotrexate; sulfasalazine; oral contraceptives; triamterene; many chemotherapeutic agents [6, 42-44].

165

Pu

bl

The central nervous system requires a constant supply of glucose, for the maintenance of ATP Na/K pumps, but also needs all the essential nutrients [29]. Some of them are B vitamins, which participate in one-carbon metabolism; their importance is so fundamental that congenital deficiency of them is associated with severe impairment of brain function [29]. The most frequent clinical conditions for B12 deficiencies is the aging process [2-29].

There are different causes which can produce B12 deficiency (inadequate intake, malabsorption, drugs interaction, genetic deficiency of transcobalamin II) [6-7]; however, the effective number of vitamin B12 defect-dementia is extremely small [1-6]. There are different cases reported in the literature of cognitive disruption (the formerly called “treatable dementia”) generically attributed only to the lack of B12 [45-47]. It has been hypothesized that the increase of vitamin B12 defect in aging patients might be related to the age-related changes in gastric histology, with atrophic gastritis with hypochlorhydria or achlorhydria, along with consequently decreased secretion of intrinsic factor [6, 7].

It has also been suggested that low levels of vitamin B12 reduce methylation reactions in brain tissue [6, 7]. Moreover, the lack of SAM, largely dependent on folate and vitamin B12, through the methylation reaction [6-18] might contribute to cognitive impairment derived from vitamin B12 and folate deficiency. Not fully understood is the rule that treating a B12 deficient patient with folate may exacerbate the neurological consequences of either deficiency, and as the good clinical practice recommends, cyanocobalamin deficiencies should be excluded before folate supplementation is commenced, or if necessary, it should be appropriate to supplement folate and vitamin B12 together [7, 45-56].

Very recently [44] the National Institutes of Health has confirmed, in general practice, that the supplementation of large amounts of folic acid can mask the damaging effects of vitamin B12 deficiency. This problem can be determined by masking effects of laboratory evidence of megaloblastic anemia, originally caused by vitamin B12 deficiency [44]. Due to the fact that consumers may be ingesting more vitamins (folic acid and vitamin B12 can be obtained without a clinical prescription) than they realize, the NIH recommends

166 Current Nutrition & Food Science, 2017, Vol. 13, No. 3

that folic acid intake from fortified food and supplements should not exceed 1,000 g daily in healthy adults [44]. 6. UPDATES OF NUTRITIONAL ASPECTS OF B12 AND FOLATE Since vascular factors and neurodegenerative problems have at the moment not therapeutic options, and nutrients may be modifiable risk factors, B complex, folate, Hcy and many others have received attention [57]. Recent data, [58, 59] showed a high prevalence of low dietary intakes for vitamin B (29-41%) in old patients and estimated 9-12% of older people in the UK who suffer from folate deficiency, due to their dietary intake. Data from other communities [60] reported that ischemic stroke patients had unfavorable dietary behaviors, with lower intake of fruit, vegetables, anti-oxidant and the intake of vegetables, fruits, vitamin C and folate were significantly associated with the risk of ischemic stroke, after adjusting for confounders.

fo

ot

N

works have failed to confirm the optimistic results [68, 74, 75] and one of the prominent author on the topic declared that subnormal serum vitamin B12 levels are not an important cause of reversible dementia [75]. When taken into account the works which considered the role of the combined low serum levels of vitamin B12 and folic acid [4, 76], it can be evicted that in old people who are not demented, neither low level of vitamin B12 nor folate alone significantly affected the risk of developing AD [4, 76]. On the contrary, it can be observed that, compared with subjects with normal levels of vitamin B12 and folate, the very old patients with the lack of these vitamins showed high risk of developing AD [4]. Other reports [77] did not evidence cognitive and executive alterations. no significant modif Hassing et al. [78] showed that AD patients, correctly supplied with vitamin B12 and folic acid did not improve, but observed that they performed worse in neuropsychological tests when maintained with low folic acid levels [78]. One of the most significant and well-conducted studies on this topic, the so-called SENECA [79], showed no correlation between mental health and low levels of vitamin B12/folate status. The most interesting aspect in this riddle, is homocysteine; there are some studies, like the one by Nilsson et al. [80], which evidenced that patients affected by AD, who had elevated homocysteine plasmatic levels, and have been supplied by vitamin B12, perform better in neuropsychological tests [80]; on the contrary, if dementia’s scores are worse, no significant effect emerged from vitamin B12 supplementation [80]. After this study, Bryan et al. [81] obtained a significant improvement in memory recall, speediness of thought, of executive functions, and linguistic appropriateness, and any effect on mood, when more than 200 healthy middle-aged or older women have been supplied with folate, B12 and B6 vitamins [81].

rS o ic r a le D is tri bu tio n

The other emerging problem is the diet habits. Several authors [61], claimed that there are potential risks for vegetarians and vegans who should be advised to carefully plan their diets to monitor their vitamin B and folate levels on regular basis. In general, in the USA, the good clinical practice stated by the Institute of Medicine and recommended daily by the Harvard TH Chan School of Public Health [62, 63] recommended a general intake of 400 micrograms per day of folate, suggesting that people, who regularly drink alcohol, should intake at least 600 micrograms per day. Food sources of folate are fruits and vegetables, whole grains, beans, cereals, and fortified grains product. The recommended intake of vitamin B12 is 2.4 micrograms per day. It is found in fish, poultry, meat, eggs, dairy, fortified cereals, enriched soy or rice milk.

Moretti et al.

7. FOLIC ACID AND VITAMIN B12 DEFECTS: WHAT DO THEY DO IN REAL CLINICAL PRACTICE?

Pu

bl

Even if it has been reported in different papers [6, 50, 52] that vitamin B12 is a determinant factor causing one of the so-called treatable dementia, and Larner et al. [53, 54] reported an overview of literature suggesting that there are cases of dementia determined by vitamin B12 defect, the number of vitamin B12 defect-dementia is extremely small [6, 46, 47, 64-66]. Many studies evidenced lower serum vitamin B12 levels in subjects with Alzheimer Disease (AD) or other dementias [6, 67, 68]. One of the most profound studies [69] on the topic examined the relationship between vitamin B12 serum levels and cognitive and neuropsychiatric symptoms in dementia, and it is not clear if the defect should be related to the age of the population examined from the neurodegenerative aspects [6, 69]. However, there are many different studies which have documented the effectiveness of vitamin B12 supplementation, in improving cognition in demented patients [6, 69-72]. Another well-conducted study demonstrated that vitamin B12 treatment may improve frontal lobe and language function in patients with cognitive impairment, but rarely reverses dementia [6, 73]. Many other

Reynolds et al. [82] focused on the importance of folic acid deprivation as determinant factors for depression and worsening of demented patients, Recent studies [83-84] determined that errors in the folate transport system and the consequent lack of folic acid might cause in a development delay, cognitive alterations, motor clumsiness, behavioral alterations and signs of demyelination More recently [85], a large international double-blind placebo-controlled randomized trial, focused on many thousand women, who have previously experienced Neural Tube Defect (NTD) pregnancy; these women have been supplied by large doses of folic acid, and a reduction of NTD of 72% in the following pregnancy was demonstrated [85, 86]. In 1998, the obligatory folic acid fortification of cereal grain products had a direct consequence of a NTD decline rate of 20%. Data have been recently reproduced in larger studies [87]. Several mechanisms have been suggested to explain the possible link between folic acid lack and NTD [11]. As above reported, folic acid is related to DNA methylation, a fundamental regulating system in precocious embryogenesis [88, 89], and this might be related to the regulation of gene expression [89]. Folic acid relays on the correct functioning of the MTHFR enzyme, which acts as a donor of methyl groups, necessary for purines and pyrimidine synthesis [11]. Therefore, genetic micro-alterations of MTHFR (for example 677TT genotype) decrease the methylation capabilities [90], with a consequent

Homocysteine in Neurology

Current Nutrition & Food Science, 2017, Vol. 13, No. 3

reduction of global DNA methylation [91, 92], related to NTDs [24]. NTD has been demonstrated in vitro, by the inactivation of DNA methyltransferase (DNMT3B) [93-95]. A reduced AdoMet/AdoHcy ratio, which causes an increase of Ado Hcy, and therefore inhibits DNA methylation has been found also in a single case of reported co-existence of trisomy 21 and spina bifida [96]. These studies lead to the conclusion that folic acid supplementation during pregnancy might prevent NTDs by donating methyl groups, thereby improving methylation and therefore it is strongly recommended [86-87]. Moreover, a lack of folic acid has been associated with an increase of homocysteine, in experimental mice, treated by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), supporting the possible link between folic acid poorness and Parkinson’s Disease [97].

fo

ot

The Canadian Study of Health and Ageing [102], a 5 years population-based survey, studied the potential risk of any cerebrovascular event, including vascular dementia, vascular cognitive impairment or fatal stroke, during a strickt follow-up of serum folate levels [102]. After adjusting for normal vascular risk factors (such as smoking, nutrition, diabetes, etc.), it emerged that the risk for a vascular event, associated with the lowest folate quartile was OR 2.42 (95% CI; 1.04-5.61) [102]. The same study demonstrated that a low folate level represents a higher risk for the development of vascular events, even in female patients (OR 4.02, 95% CI; 1.37-11.81) [102].

Pu

bl

N

development, as indicated by the observation that Hcy accumulation delays the closure of the neural tube, due to an inhibition of the transmethylation pathway [94]. A link between Hcy and neurological alterations has been described in patients with severe CBS deficiency [11-29], and in genetic metabolic alterations, which induced severe hyperhomocysteinemia (HHCY) (total homocysteine >50 uM) or homocystinuria, determined by genetic alterations of remethylation or transsulfuration pathways [11-29]. When there is an MTR deficiency or dysfunction, 5-methylTHF cannot cycle resulting in the accumulation of 5-methylTHF; folate does not circulate, limiting the synthesis of purines and thymidine [6-29]. These problems have been associated with congenital/genetic hyperhomocysteinemia condition, and they occur if the defect is located in the remethylation or in the transsulfuration cascade [29]. Numerous methylation reactions are strongly needed in the brain, for the synthesis and for the degradation of many neurotransmitters, and for the assemblage of many membrane phospholipids [29]. On the other hand, the transsulfuration pathway is a key point for Hcy catabolism; it accounts for glutathione synthesis in the liver but no clear data are available for the brain [29]. Many papers focus on this point, suggesting that some enzyme reactions, such as cystathionine beta synthase and cystathionase, act in the brain [29, 107-108], and could promote the transsulfuration of Hcy into cysteine, producing the precursor of glutathione [29-109]. Only a paper documented in an in vitro model production of glutathione by astroglia by cysteine and cystathionine [29-110].

rS o ic r a le D is tri bu tio n

It is known that a lack of folic acid is related to a higher percentage of depression, but the reason is not clear [98]. Since the folic acid is the substrate for SAM, and the role of methionine in monoamine metabolism is crucial, a reduction of the monamine-neurotransmitters synthesis, i.e. serotonine and gamma-amino butyric acid [99], may be related to depression. Folic acid level and homcysteine are indirectly related to years of age, they have been related to aging process [100, 101], and, therefore, to cerebrovascular pathologies, whose main unmodifiable risk factor remains aging process [100, 101].

167

In a relatively small sample of 30 elderly Catholic sisters, affected by AD, a low folic acid level directly correlates with the severity of atrophy of the neocortex and with the amyloid deposition in the neocortex [78-103]. In the Kingsholmen aging and dementia project, in 250 old (75 to 96 years) and in 71 very old (90 to 101 years), otherwise healthy subjects, the altered memory recall has been related to folic acid low levels [104]. These findings have been reproduced with a significant correlation between lower cognitive function and raised homocysteine level and low folic acid levels [105]. 8. HOMOCYSTEINE: HOW DOES IT RELATE WITH FOLIC ACID AND VITAMIN B12 AND WHAT DOES IT CAUSES? Hcy is a sulfur-containing aminoacid, tightly related to methionine metabolism [29]. It seems involved in neural

Therefore, Hcy transsulfuration is not the fundamental reaction for the induction of the synthesis of in the brain [29, 111, 112]. It seems more probable that cysteine can be transported by a Na- dependent glutamate transporter in astrocytes. Cysteine is the rate-limiting substrate for the synthesis of glutathione [29, 113], and somehow, different cysteine precursors (cystathionine, homocysteine, and methionine) may have a major role in the production of glutathione in the brain [29, 113]. Animal models identify a specific saturable receptor for Hcy in the brain, which can implement a diffusion process [29, 114-116]; in humans, the diffusion process is the only known system of Hcy transport [29]. Human neuronal cells produce Hcy [117], but probably with different regional quantities, not investigated at the moment [29, 117]. As above mentioned, an increase of Hcy occurs in the brain and CSF within the aging process and in several neurological diseases, either in CSF either in plasma [29, 118122]. Severe HHCY (>100 lM) in children with CBS defect correlates with a 10-fold increase in concentrations of Hcy in CSF [123]; it is hypothesized also for human being, even if the finding is proved only in animal models, that Hcy might compromise the integrity of the blood-brain barrier [124]. Hcy is toxic for neurons [29, 125, 126]. Neurological damages have been reported in mice deficient in CBS enzyme (Cbs -/+ or Cbs -/-), where Hcy increased by approximately 2-50-fold in comparison to wild-type mice [125-131].

168 Current Nutrition & Food Science, 2017, Vol. 13, No. 3

The mechanisms of damage, mediated by homocysteine, are various, and not always linked to plasma HHcy’ levels as high, but also a lower level of HHcy can act as a promoter of neurodegeneration, or potential inflammatory inducer. 9. HCY AND NEURODEGENERATION The mechanism of damage evoked for neurodegeneration is the link between Hcy and endogenous glutamate receptor, where Hcy acts as an agonist [29, 132, 133], and links NMDA receptors [134, 135].

fo

Another study suggested that Hcy can activate the group I metabotrophic glutamate receptors [29, 138]. This interaction can be demonstrated by blocking NMDA, non NMDA and metabotrophic glutamate receptor, via specific antagonists: a reduction of calcium influx can be observed, when binding these receptors [29, 136].

ot

A strong number of new works concentrate themselves on the active role of Hcy; it has been demonstrated that there is a direct relation between Hcy increase and Abeta 1-40 deposition, in aged brain and in brain of patients [140]. A specific endoplasmic protein, Hcy related (HERP) has been detected, and it potentiates the c-secretase activity, and consequently Ab1-40 accumulation in the brain [29, 141]. It has been described that Hcy increases the vulnerability of neurons to Abeta deposition damage [29, 142, 143] and that Hcy might potentiate the intracellular and extracellular accumulation of Abeta 42 [29, 144].

Pu

bl

N

ing a failure in post-translational modifications or in the stability of the enzyme [29, 152]. Low concentration of SAM (or an increase in SAH) determines a lower activity of PPA2 and, even indirectly, an elevation of P-tau [29, 153, 154]. The induced depletion of folic acid in neuroblastoma cultured cells induced directly (or as a consequence of the induced hyper-Hcy and ROS- this point is not clarified) an increase of P-tau by 66% [29, 117]. Hcy increases the toxicity of Abeta on the vascular smooth muscle cells of brain small arteries [155]. Therefore, due to their role in autoregulation, small arteries can be a potential site of damage of hyper Hcy, causing alteration of the cerebral blood flow regulatory system, a rupture of blood brain barrier, and therefore hyper Hcy might potentiate its direct neurotoxic effects [29, 142, 156]. 10. HCY, ROS, INFLAMMATION The activities of methionine synthase, CBS, that mediate the clearance of Hcy, is linked to the redox potential of the cells [29, 142, 157, 158], with an observed efficacy in oxidative stress process; in this situation, more Hcy is converted into cysteine and glutathione. A disruption of the CBS causes an altered redox homeostasis, and by an induced limitation of the cysteine and glutathione levels, it causes an alteration of oxidative repairing process [29, 157-162]. The endothelial damage is mediated by one of the precursors, hydrogen sulfide (H2S), which is formed during the transsulfuration process [29, 163, 164]. H2S serves as a vasodilator, it might contribute to the control of the vessel diameter and it regulates the intracellular signaling process; it might induce a defense against redox stress on the endothelium, (see above) and it probably acts by reducing the cascade of inflammation on endothelium [163, 164]. The disruption of the redox system in vascular and neuronal cells [29, 161, 162] (where it has been linked to an increment of ROS and deactivation of nitric oxide, with the well-known inflammation cascade) induces and accelerates the lipid peroxidation sequel of events [29, 165, 166]. In fact, different studies demonstrate that the employment of antioxidants, such as N-acetyl cysteine, vitamin E or C might reduce the potential pro-inflammatory response of Hcy in animal models [117, 167-169]. In vivo studies point on the controversial role in the damage of the endothelium. Different reports documented the Th1-activity induced the Hcy inflammation response [170], and it appears that HHcy can be detected in chronic inflammatory conditions, even if vitamin B12 and folate are in range. In a double-blind interventional study [171], the role of Hcy in atherogenesis in coronary diseases was shown; they found an association between neopterin’s and Il-6’ levels and HHcy at baseline. In this study the Authors found that the implementation with vitamin B complex does not affect inflammatory markers, such as neopterin, Il-6, CRP, etc.; on the contrary, the implementation of folate caused a disappearance of the relationship between HHcy and Neopterin, found at baseline, suggesting a possible modulating role of folic acid in the inflammatory cascade. Thus, they suggest that “an optimal folate status override the influence of immunostimulation on Th1 by Hcy” [171].

rS o ic r a le D is tri bu tio n

Through Hcy-NMDA binding, Hcy indirectly enhances calcium influx [132, 135, 136]. Glycine is the variable which conditions the Hcy effects [29]; when glycine is in normal concentration (10 umol/L), Hcy acts as a partial antagonist of the glycine site of the NMDA receptor, and it inhibits the receptor-mediated activity, acting as a neuroprotective factor [29, 132]. If glycine levels are normal, only HHcy is toxic (i.e., Hcy = 100umol/L) [29, 132]. When glycine levels increase in the nervous system (more than 10 umol/L), such as reported after brain ischemia, head trauma, or migraine cluster [29, 137], a relatively low concentration of Hcy (i.e., Hcy = 10 umol/L) can act as an agonist on NMDA [29, 132, 138, 139].

Moretti et al.

Increasing attention has been given to the gene expression of Presenilin 1 (PS1), which is tightly related to methylation process in brain [29, 145, 146]; it promotes the APP production [29, 146]. DNA hypomethylation up-regulates PS1 gene, and increase APP [29, 146]. Moreover, tau protein, is equally related to methylation process; the protein phosphatase 1 (PPM1), whose methylation is SAM dependent, regulates the activity of the protein phosphatase 2A, which acts as a dephosphorylating system for tau protein, fundamental for its correct function [147, 148]; hyperphosphorylation of tau inhibits its normal function; it contrasts the assembly of microtubules and promotes the precipitation of neurofibrillary tangles: hence the reduced methylation capacity can increase P-tau [29, 147, 148]. The protein phosphatase 2A (PP2A) can dephosphorylate P-tau in paired helical filaments, making tau accessible to proteolysis, to avoid the accumulation of tangles [29, 149151]. The expression of the PP2A protein is low in AD patients [152] although mRNA PP2A is increased, suggest-

Homocysteine in Neurology

Current Nutrition & Food Science, 2017, Vol. 13, No. 3

Ploder et al. [172] investigated the modification of Hcy plasma levels, in patients with multiple traumatic lesions, a consequent sepsis, and a systemic inflammatory response. The results showeda constant HHcy in sepsis patient with poor outcome (non-survivors), without evidence of any other factorial causes (such as folate or B12 poorness). The authors [172] hypothesized that the pro-inflammatory condition of these patients leads to a strong activation of macrophagesystem cascade by Hcy, with a consequent release of ample amounts of ROS, potentiating the oxidative stress. More recent results [173] demonstrate in vitro a definite activation of B lymphocyte induced by Hcy; this process seems to determine an increase of pyruvate kinase muscle isozyme 2 (PKM-2) in B cells. Its inhibition, employing shikonin, causes the restore of the metabolic changes induced by Hcy. PKM-2 seems to suggest the so-called metabolic accelerated initiation of atherosclerosis cascade mediated by HHcy, in vivo and in vitro [173].

fo

ot

Pu

bl

N

ROS released, of up-regulation of the Nuclear Factor Kappa B, considered as one of “the master regulator of the expression of inflammatory genes” [180, 181]. The lack of folic acid, associated with an increase of Hcy, might potentiate the oxidative induction of the transsulfuration reaction process [142-144]. This fact has been demonstrated also in animal experiments: if we consider apolipoprotein E-deficient mice or cultured neuronal cells deprived of folate, we can observe that the amelioration of the conditions with antioxidant agents (like SAM, or vitamin E) determines a reduction of the oxidative cascade [143, 182]. It should be mentioned the direct correlation between Hcy and SAH, in the reaction mediated by SAH-hydrolase [29]; therefore, a higher level of SA reduces the SAM concentrationand even the higher SAH level (or the lower SAM level) induces an oxidative reaction, and might explain Hcy neurotoxicity [13, 29, 129, 182]. Increasing SAM levels, or employing enriched SAM pabulum for cells neuronal tissues, might reduce apoptosis by 50% [29, 183-185]. 11. HCY AND VASCULAR DAMAGE

rS o ic r a le D is tri bu tio n

Li et al. [174] showed that in animal models, an induced hyper-Hcy produces a higher plasma level of Tumor Necrosis Factor alpha (TNF-) and of Interleukin 1 beta (IL-1) andan evident decrease of plasma levels of H2S and of cystathionine gamma-lyase expression in the peritoneal macrophages [174]. It has been demonstrated that hyper-Hcy inhibits cystathionine -lyase expression and H2S production in macrophages; HHcy is related to an increase of the DNA expression of methyltransferase and of hypermethylation process in promoter regions [174], therefore inducing a promoter trigger of inflammation. These results have been confirmed by other reports [175-177] which showed that higher levels of Hcy promote, in many different experimental conditions, the activity of specific genes, implicated in the methylation process, and therefore [175-177] HHcy induces inflammation of the endothelium matrix and triggers atherosclerosis process. A single study [178] confirmed the previously reported study, and showed that Hcy alters the transcriptional repression of fibroblast growth factor 2 [178]. Li et al. [174] definitively demonstrated that cultured macrophages cells exposed to Hcy showed a memory response, probably induced by epigenetic mutations, that influences the expression of promoter genes inflammatory response and endothelium atherogenesis [174].

169

There is a possible link between the excitotoxic effect of Hcy and the pro-inflammatory role of Hcy: the NMDA receptors are found not only in neurons (see above in the text), but also on neutrophils and macrophages. The activation of these receptors in the immune-competent cells induces an increase in the cytoplasmatic calcium influx, and an activation of a pro-inflammatory cascade, with an accumulation of ROS species [179]. To confirm these aspects, a new series of studies have been conducted in many systemic inflammatory diseases, where multi-factorial aspects (genetic, epigenetic, auto-immunitary) are involved, and where the risk of concomitant cerebral and cardio-vascular pathologies is very high. A good example is the Reumatoid Arthritis [180] where HHcy is two times more frequent than in the general population [180]. In Rheumatoid Arthritis, HHcy creates a chronic condition of oxidative stress, of prothrombotic induction, and, indirectly, by the excess of

Increased levels of Hcy, both in CSF and in plasmatic concentration, as far as aging increment [29, 183-185]; that seems quite thought-provoking, considering the tight relationship between aging and the incidence of neurodegenerative disorders [29, 183-185]. Moreover, the most antique link between Hcy and brain altered function is the white matter damage (vascular micro-alterations) evidenced by neuroimaging, in many population studies [29, 186]. Then, another, not independent association, between brain aging, Hcy and cortical and subcortical atrophy [29, 187] has been demonstrated. But even more compelling is the association between Hcy, vascular effects and its neurotoxic intrinsic potential; this triad of events occurs in vascular dementia, where amyloid cerebropathy is linked to small vessel disease and to Ach deregulation [129, 184-190]. Different studies have focused on the pure vascular damage related to hyper Hcy [29, 191, 192]. But the very first description of Hcy damages on endothelium derived from McCully [193]. He described for the first time two inborn errors of Hcy metabolism, with severe vascular damage and fatal circulation problems [194]. A recent study found that HHCy is related to high levels of fibrinogen and Abeta deposits in the blood vessels of AD patients [194]. Chung et al. [194] demonstrate that Hcy has the possibility to speed and to reinforce the interactions between fibrinogen and Aeta, and can even delay the fibrinolytic process in the AD mouse model; there is a higher burden of Abeta intra-parenchymal deposition in these mice, [195]. Many studies have focused on the data that HHcy can be considered a potential vascular risk factor [191, 192, 195], but final conclusions are still under debate. Homocysteine Studies Collaboration meta-analysis [196] suggests that HHcy can be considered an independent predictor of ischemic heart disease and stroke risk in healthy population

170 Current Nutrition & Food Science, 2017, Vol. 13, No. 3

[196]. Another cross-sectional case-control study has been conducted in order to investigate the contribution of Hcy to cerebral white matter changes in patients with AD and controls [197]. Hcy has been considered a significant risk factor for the development of leukoaraoisis, comparable with age, systolic pressure, ApoE4 genotype and smoking [197]. Four years later, the conclusive results of HOPE 2 [198] demonstrated that a supplementary intake of folic acid, vitamin B6, and vitamin B12 decreased mean plasma homocysteine levels in patients with vascular disease and diabetes, but the decrease of Hcy did not interfere with death risk due to vascular pathologies or with a significant decrease in the prevention of new events [198]. Cochrane Database of Systematic Reviews [199] showed that Hcy lowering therapy did not reduce the incidence of myocardial infarction, stroke, or death by any vascular cause [199]. However, when considering the stroke incidence as an isolate event homocysteine-lowering therapy may relate to a reduction of stroke, like the one described in other studies [6, 55, 191, 192, 195, 199].

N

ot

fo

The damage can be summarized as an altered regulation of the small arteries and an alteration of endothelium, principally related to a reduction in nitric oxide synthesis, involved in vessel autoregulation, and to a potentiation of atherothrombotic events [29, 193, 195]. CONCLUSION

Hcy remains, at the moment, a biological marker, with all its limits and without the key of solution for all the neuronal pathologies; EFSA [200] maintains a cautious position: it clearly stated that the maintenance of normal Hcy metabolism has a beneficial physiological effect. All the other protective, antioxidant, anti-inflammatory effects should be proven. Apart from the experimentally mediated suggestive role of reducing inflammation by diminishing Hcy, and the evidence of the indirect endothelium damage (mediated by H2S), there is a lack of other clinical evidence of benefits which has been obtained by reducing HHcy, contradicted by the brilliant results obtained in animal models or cells culture. To summarize, more and more evidence suggests that there should be a superimposition of microvascular damage in neurodegeneration by itself, and during ischemic events in the brain; in these conditions glutamate altered currents, NMDA receptor modifications and apoptosis occur simultaneously. So practically, the two processes (vascular and neurodegenerative) co-exist, at the same time, in probably different proportions, and it could be difficult to distinguish the effect of a compound, which might act on both the aspects. Large scale controlled studies are needed before a definite conclusion can be drawn.

rS o ic r a le D is tri bu tio n

HHcy due to inborn errors causes important and often fatal vascular pathologies [29, 194, 195]. There are strong studies which demonstrated that augmentation of Hcy is associated with increased cardio/cerebrovascular disease risk [6, 55, 191, 192, 195, 197]. However, in apparent contrast, no risk reduction has been found in homocysteine-lowering trials [7, 29, 196].

Moretti et al.

Pu

bl

Hcy remains a mystery: many experimental studies demonstrate, beyond any reasonable doubt, that Hcy has a causative role in the determination of neurological damages, due to its neurotoxic effect, to its direct or indirect vascular and endothelium induced pro-inflammatory effect. On the other hand, clinical studies have been inconclusive.

LIST OF ABBREVIATIONS AA

=

Amino Acids

A-B12

=

Adenosyl Cobalamine

AD

=

Alzheimer’s Disease

CBS

=

Cystathionine  Synthase

FA

=

Fatty Acids

Hcy

=

Homcysteine

HHCY

=

Hyperhomcysteinemia

MAT

=

Methionine Adenosyl Trasnferase

M-B12

=

Methyl Cobalamine

Met-THF

=

Methylene-TetraHydroFolate

However, Hcy might be related to neurodegenerative disorders by potentiating the effects of Abeta deposition, augmenting its toxic effects, by modifying presenilin functions, and occasionally interfering with hyperphosphorylation of tau protein. On the other hand, it might also have a direct effect on smooth muscle vascular structures, potentiating the damage induced by Abeta deposition and activating caspase activity.

MMA-CoA =

Methtyl-malonyl Coenzyme A

MTHF

=

Methly-TetraHydroFolate

MTHFR

=

Methylene TetraHydroFolate Reducatse

MTR

=

Methionine Synthase

NTD

=

Neural Tube Defect

SAH

=

S-Adenosyl-L-Homocysteine=AdoHcy

Hcy is strenuously related to vascular risk factors, too. Hcy has been described as an independent risk factor for the development of many neurological vascular events, and recent data have demonstrated a reduction in stroke mortality, after folate fortifications. The association between hyperHCY and worsening of cognitive performance, Amyloid burden and white matter hyperintensities, has been confirmed in the majority the clinical studies considered.

SAM

=

S-Adenosyl-Methionine=AdoMet

S-CoA

=

Succinyl-CoA

THF

=

Tetrahydrofolate

CONFLICT OF INTEREST The authors confirm that this article content has no conflict of interest.

Homocysteine in Neurology

Current Nutrition & Food Science, 2017, Vol. 13, No. 3 [22]

ACKNOWLEDGEMENTS RM design and wrote the manuscript, analyzed the literature. MDB, SG and CT participated to the final writing and critically drafted the manuscript. MDB was supported by a U12GPFIRB11-CUP: J91J11000450001 fellowship.

[23]

[24]

REFERENCES

[7]

[8] [9]

[10] [11]

[12] [13]

[14] [15]

[16] [17]

[18] [19]

[20]

[21]

[25]

[26]

[27]

[28]

[29]

Leclerc D, Wilson A, Dumas R, et al. Cloning and mapping of a cDNA for methionine synthase reductase, a flavoprotein defective in patients with homocystinuria. Proc Natl Acad Sci USA 1998; 95: 3059-64. Sunden SL, Renduchintala MS, Park EI, Miklasz SD, Garrow TA. Betaine-homocysteine methyltransferase expression in porcine and human tissues and chromosomal localization of the human gene. Arch Biochem Biophys 1997; 345: 171-4. Quéré I, Paul V, Rouillac C, et al. Spatial and temporal expression of the cystathionine beta-synthase gene during early human development. Biochem Biophys Res Commun 1999; 254: 127-37. Finkelstein JD. Metabolic regulatory properties of S-adenosylmethionine and S-adenosylhomocysteine. Clin Chem Lab Med 2007; 45: 1694-9. Blom HJ, Shaw GM, Den-Heijer M, Finnell RH. Neural tube defects and folate: Case far from closed. Nat Rev Neurosci 2006; 7: 724-31. Garrow TA, Brenner AA, Whitehead VM, et al. Cloning of human cDNAs encoding mitochondrial and cytosolic serine hydroxymethyltransferases and chromosomal localization. J Biol Chem 1993; 268: 11910-6. Hum DW, Bell AW, Rozen R, MacKenzie RE. Primary structure of a human trifunctional enzyme. Isolation of a cDNA encoding methylenetetrahydrofolate dehydrogenase-methenyltetrahydro-folate cyclohydrolase-formyltetrahydrofolate synthetase. J Biol Chem 1988; 263: 15946-50. Obeid R, Herrmann W. Mechanisms of homocysteine neurotoxicity in neurodegenerative diseases with special reference to dementia. FEBS Lett 2006; 580: 2994-3005. Chandler CJ, Harrison DA, Buffington CA, Santiago NA, Halsted CH. Functional specificity of jejunal brush-border pteroylpolyglutamate hydrolase in pig. Am J Physiol 1991; 260: G865-72. Qiu A, Jansen M, Sakaris A, et al. Identification of an intestinal folate transporter and the molecular basis for hereditary folate malabsorption. Cell 2006; 127: 917-28. Barber RC, Lammer EJ, Shaw GM, Greer KA, Finnell RH. The role of folate transport and metabolism in neural tube defect risk. Mol Genet Metab 1999; 66: 1-9. Choi IJH, Yates Z, Veysey M, Heo YR, Lucock M. Contemporary Issues surronding folic acid fortifications initiatives. Prev Nutr Food Sci 2014; 19(4): 247-60. Kamen BA, Smith AK. A review of folate receptor alpha cycling and 5-methyltetrahydrofolate accumulation with an emphasis on cell models in vitro. Adv Drug Deliv Rev 2004; 56: 1085-97. Wang X, Qin X, Demirtas H, et al. Efficacy of folic acid supplementation in stroke prevention: a meta-analysis. Lancet 2007; 369: 1876-82. Oakley GP, Adams MJ, Dickinson CM. More folic acid for everyone, now. J Nutr 1996; 126: 751S-5S. McNulty H. Folate requirements for health in different population groups. Br J Biomed Sci 1995; 52: 110-9. Stolzenberg R. Possible folate deficiency with postsurgical infection. Nutr Clin Pract 1994(6); 9: 247-50. Pietrzik KF, Thorand B. Folate economy in pregnancy. Nutrition 1997; 13(11-12): 975-7. Cravo ML, Glória LM, Selhub J, et al. Hyperhomocysteinemia in chronic alcoholism: Correlation with folate, vitamin B-12, and vitamin B-6 status. Am J Clin Nutr 1996; 63: 220-4. Tamparo CD, Marcia LA. Diseases of the Human Body. 5th ed. F.A. Davis Company: Philadelphia, PA 2011. Hoffbrand AV, Weir DG. The history of folic acid. Br J Haematol 2001; 113: 579-89. Folate Evidence-Mayo Clinic n.d. Available at: http://www. mayoclinic.org/drugs-supplements/folate/evidence/hrb-20059475 (Accessed September 15, 2016). Ods.od.nih.gov. Dietary supplement fact sheet: Vitamin B12-health professional fact sheet n.d. Avaialble at https://ods.od.nih.gov/factsheets/Vita-minB12-HealthProfessional/(Accessed September 15, 2016). Moretti R, Torre P, Antonello R, Cazzato G. Deficit cognitivi reversibili da carenza di vitamina B12: un confronto neuropsicologico con AD. Demenze, Roma: CIC Edizioni Internazionali 2001: pp. 42-5.

rS o ic r a le D is tri bu tio n

[6]

bl

[5]

fo

[4]

Pu

[3]

ot

[2]

Selhub J, Bagley LC, Miller J, Rosenberg IH. B vitamins, homocysteine, and neurocognitive function in the elderly. Am J Clin Nutr 2000; 71: 614S-20S. Nilsson-Ehle H. Age-related changes in cobalamin (vitamin B12) handling. Implications for therapy. Drugs Aging 1998; 12: 277-92. Bottiglieri T. Folate, vitamin B12 , and neuropsychiatric disorders. Nutr Rev 1996; 54: 382-90. Wang HX, Wahlin A, Basun H, et al. Vitamin B12 and folate in relation to the development of Alzheimer’s disease. Neurology 2001; 56: 1188-94. Smith AD, Refsum H. Homocysteine, B vitamins, and cognitive impairment. Annu Rev Nutr 2016; 36: 211-39. Moretti R, Torre P, Antonello RM, Cattaruzza T, Cazzato G, Bava A. Vitamin B12 and folate depletion in cognition: A review. Neurol India 2004; 52 (3): 310-8. Miles Lm, Allen E, Mills K, Clarke R, Uauy R, Dangour AD. Vitamin B12 status and neurologic function in older people: A crosssectional analysis of baseline trial data from the older people and enhanced neurological function (OPEN) study. Am J Clin Nutr 2016; 104(3): 790-6. Mudd SH, Cantoni GL. Activation of methionine for transmethylation. III. The methionine-activating enzyme of Bakers’ yeast. J Biol Chem 1958; 231: 481-92. Kotb M, Mudd SH, Mato JM, et al. Consensus nomenclature for the mammalian methionine adenosyltransferase genes and gene products. Trends Genet 1997; 13: 51-2. Mato JM, Alvarez L, Ortiz P, Pajares MA. S-adenosylmethionine synthesis: Molecular mechanisms and clinical implications. Pharmacol Ther 1997; 73: 265-80. Blom HJ, Smulders Y. Overview of homocysteine and folate metabolism. With special references to cardiovascular disease and neural tube defects. J Inherit Metab Dis 2011; 34: 75-81. Parnetti L, Bottiglieri T, Lowenthal D. Role of homocysteine in age-related vascular and non-vascular diseases. Aging (Milano) 1997; 9: 241-57. Enk C, Hougaard K, Hippe E. Reversible dementia and neuropathy associated with folate deficiency 16 years after partial gastrectomy. Scand J Haematol 1980; 25: 63-6. Bottiglieri T. Ademetionine (S-adenosylmethionine) neuropharmacology: Implications for drug therapies in psychiatric and neurological disorders. Expert Opin Investig Drugs 1997; 6: 417-26. Weir DG, Keating S, Molloy A, et al. Methylation deficiency causes vitamin B12-associated neuropathy in the pig. J Neurochem 1988; 51: 1949-52. Surtees R, Leonard J, Austin S. Association of demyelination with deficiency of cerebrospinal-fluid S-adenosylmethionine in inborn errors of methyl-transfer pathway. Lancet 1991; 338: 1550-4. Shaw PJ. Excitatory amino acid receptors, excitotoxicity, and the human nervous system. Curr Opin Neurol Neurosurg 1993; 6: 414-22. Pennypacker LC, Allen RH, Kelly JP, et al. High prevalence of cobalamin deficiency in elderly outpatients. J Am Geriatr Soc 1992; 40: 1197-204. McKeever MP, Weir DG, Molloy A, Scott JM. Betainehomocysteine methyltransferase: organ distribution in man, pig and rat and subcellular distribution in the rat. Clin Sci 1991; 81(4): 551-6. Chadwick LH, McCandless SE, Silverman GL, et al. Betainehomocysteine methyltransferase-2: cDNA cloning, gene sequence, physical mapping, and expression of the human and mouse genes. Genomics 2000; 70: 66-73. Li YN, Gulati S, Baker PJ, et al. Cloning, mapping and RNA analysis of the human methionine synthase gene. Hum Mol Genet 1996; 5: 1851-8.

N

[1]

[30]

[31]

[32]

[33]

[34]

[35]

[36] [37] [38] [39] [40]

[41] [42] [43]

[44]

[45]

171

172 Current Nutrition & Food Science, 2017, Vol. 13, No. 3

[52] [53]

[54] [55] [56] [57]

[58]

[59]

[60]

[61]

[62]

[63]

[64]

[65] [66]

[67]

[68]

[69]

[70]

[71]

[72]

[73] [74]

[75]

[76] [77]

[78]

[79]

[80]

Meadows ME, Kaplan RF, Bromfield EB. Cognitive recovery with vitamin B12 therapy: A longitudinal neuropsychological assessment. Neurology 1994; 44: 1764-5. Eastley R, Wilcock GK, Bucks RS. Vitamin B12 deficiency in dementia and cognitive impairment: The effects of treatment on neuropsychological function. Int J Geriatr Psychiatry 2000; 15: 226-33. De La Fourniere F, Ferry M, Cnockaert X, et al. Deficience en vitamine B12 et etat dementiel etude epidemiologique multicentrique et therapeutique essai preliminaire. Sem Hop 1997; 73(5-6): 133-40. Teunisse S, Bollen AE, Van Gool WA, Walstra GJ. Dementia and subnormal levels of vitamin B12: Effects of replacement therapy on dementia. J Neurol 1996; 243: 522-9. Robins Wahlin TB, Wahlin A, Winblad B, Bäckman L. The influence of serum vitamin B12 and folate status on cognitive functioning in very old age. Biol Psychol 2001; 56: 247-65. Fioravanti M, Ferrario E, Massaia M, et al. Low folate levels in the cognitive decline of elderly patients and the efficacy of folate as a treatment for improving memory deficits. Arch Gerontol Geriatr 1998; 26: 1-13. Hassing L, Wahlin A, Winblad B, Bäckman L. Further evidence on the effects of vitamin B12 and folate levels on episodic memory functioning: A population-based study of healthy very old adults. Biol Psychiatry 1999; 45: 1472-80. Eussen SJPM, Ferry M, Hininger I, et al. Five year changes in mental health and associations with vitamin B12/folate status of elderly Europeans. J Nutr Health Aging 2002; 6: 43-50. Nilsson K, Gustafson L, Hultberg B. Improvement of cognitive functions after cobalamin/folate supplementation in elderly patients with dementia and elevated plasma homocysteine. Int J Geriatr Psychiatry 2001; 16: 609-14. Bryan J, Calvaresi E, Hughes D. Short-term folate, vitamin B12 or vitamin B-6 supplementation slightly affects memory performance but not mood in women of various ages. J Nutr 2002; 132: 1345-56. Reynolds EH. Folic acid, ageing, depression, and dementia. BMJ 2002; 324: 1512-5. Smithells RW, Sheppard S, Schorah CJ. Vitamin deficiencies and neural tube defects. Arch Dis Child 1976; 51: 944-50. Prevention of neural tube defects: Results of the medical research council vitamin study. MRC vitamin study research group. Lancet 1991; 338: 131-7. Czeizel AE, Dudás I. Prevention of the first occurrence of neuraltube defects by periconceptional vitamin supplementation. N Engl J Med 1992; 327: 1832-5. Blom HJ. Folic acid, methylation and neural tube closure in humans. Birth Defects Res A Clin Mol Teratol 2009; 85: 295-302. Pitkin RM. Folate and neural tube defects. Am J Clin Nutr 2007; 85: 285S-8S. Reik W, Dean W, Walter J. Epigenetic reprogramming in mammalian development. Science 2001; 293: 1089-93. Dean W, Lucifero D, Santos F. DNA methylation in mammalian development and disease. Birth Defects Res Part C Embryo Today Rev 2005; 75: 98-111. Frosst P, Blom HJ, Milos R, et al. A candidate genetic risk factor for vascular disease: A common mutation in methylenetetrahydrofolate reductase. Nat Genet 1995; 10: 111-3. Friso S, Choi S-W, Girelli D, et al. A common mutation in the 5,10-methylenetetrahydrofolate reductase gene affects genomic DNA methylation through an interaction with folate status. Proc Natl Acad Sci USA 2002; 99(8): 5606-11. Castro R, Rivera I, Ravasco P, et al. 5, 10-methylenetetrahydrofolate reductase (MTHFR) 677C-->T and 1298A-->C mutations are associated with DNA hypomethylation. J Med Genet 2004; 41: 454-8. Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 1999; 99: 247-57. Afman LA, Blom HJ, Drittij MJ, Brouns MR, Van Straaten HW. Inhibition of transmethylation disturbs neurulation in chick embryos. Brain Res Dev Brain Res 2005; 158(1-2): 59-65. Dunlevy LPE, Burren KA, Mills K, et al. Integrity of the methylation cycle is essential for mammalian neural tube closure. Birt Defects Res A Clin Mol Teratol 2006; 76: 544-52. Al-Gazali LI, Padmanabhan R, Melnyk S, et al. Abnormal folate metabolism and genetic polymorphism of the folate pathway in a

rS o ic r a le D is tri bu tio n

[51]

bl

[50]

fo

[49]

Pu

[48]

ot

[47]

Moretti R, Torre P, Antonello RM, Cazzato G. Is isolated vitamin B12 deficiency a sufficient causative factor of dementia? Eur J Neurol 2001; 8: 87-8. Moretti R, Torre P, Antonello RM, Cazzato G, Bava A. Vitamin B12 defect: What does it mean to cognition? Eur J Neurol 2001; 8: 731. Tucker KL, Mahnken B, Wilson PW, Jacques P, Selhub J. Folic acid fortification of the food supply. Potential benefits and risks for the elderly population. JAMA 1996; 276: 1879-85. Ubbink JB. Should all elderly people receive folate supplements? Drugs Aging 1998; 13: 415-20. Mollin DL, Ross GI. Serum vitamin B12 concentrations of patients with megaloblastic anaemia after treatment with vitamin B12 , folic acid, or folinic acid. Br Med J 1953; 2: 640-5. Dickinson CJ. No reliable evidence that folate is harmful in B-12 deficiency. BMJ 1995; 311: 949. Samuels MA. Neurocardiology and neuropulmonary. Am Acad Neurol 2003; Honolulu 3FC.001: 1-96. Larner AJ, Janssen JC, Cipolotti L, Rossor MN. Cognitive profile in dementia associated with vitamin B12 deficiency due to pernicious anaemia. J Neurol 1999; 246: 317-9. Larner AJ, Rakshi JS. Vitamin B12 deficiency and dementia. Eur J Neurol 2001; 8: 730-1. Lucock M. Is folic acid the ultimate functional food component for disease prevention? BMJ 2004; 328: 211-4. Malnick S, Goland S. Folic acid as ultimate in disease prevention: Beware of vitamin B12 deficiency. BMJ 2004; 328: 769. Porter L, Hoey L, Hughes CT, Ward M, McNulty H. Causes, consequences and public health implications of low b vitamin status in ageing. Nutrients 2016; 8(11): pii: E725. Ter Bog S, Verlaan S, Hemsworth J, et al. Micronutrients intakes and potential inadequacies of community dwelling older adults: A systematic review. Br J Nutr 2015; 113: 1195-206. Bates B, Lennox A, Bates C. NAtional and Nutritional Survey: Results from years 1-4 of the rolling programme; 2008(20092011/2012); Public Health England, London UK 2014. Choe H, Hwang JY, Yun JA, et al. Intake of antioxidants and B vitamins is inversely associated with ischemic stroke and cerebral atherosclerosis. Nutr Res Pract 2016; 10(85): 516-23. Elmadfa I, Singer I. Vitamin B12 and homocysteine status among vegetarians: A global perspective. Am J Clin Nut 2009; 89: S1693-8. Institute of Medicine. Dietary reference intakes for thiamin, riboflavin, niacin, vitamin B6, folate, vitamin B12 , panthotenic Acid, biotin, and choline. Washington DC: National Academy Press 1999. The Nutrition Source. Three of the B vitamins: Folate, vitamin B6, and vitamin B12, Available from: www.hsph.harvard.edu/nutrition source/vitamin-b/. White L, Petrovitch H, Ross GW, et al. Prevalence of dementia in older Japanese-American men in Hawaii: The Honolulu-Asia aging study. JAMA 1996; 276: 955-60. Clarfield AM. The reversible dementias: Do they reverse? Ann Intern Med 1988; 109: 476-86. Weytingh MD, Bossuyt PM, Van Crevel H. Reversible dementia: More than 10% or less than 1%? A quantitative review. J Neurol 1995; 242: 466-71. Bernard MA, Nakonezny PA, Kashner TM. The effect of vitamin B12 deficiency on older veterans and its relationship to health. J Am Geriatr Soc 1998; 46: 1199-206. Bell IR, Edman JS, Marby DW, et al. Vitamin B12 and folate status in acute geropsychiatric inpatients: affective and cognitive characteristics of a vitamin nondeficient population. Biol Psychiatry 1990; 27: 125-37. Whyte EM, Mulsant BH, Butters MA, et al. Cognitive and behavioral correlates of low vitamin B12 levels in elderly patients with progressive dementia. Am J Geriatr Psychiatry 2002; 10(3): 321-7. Healton EB, Savage DG, Brust JC, Garrett TJ, Lindenbaum J. Neurologic aspects of cobalamin deficiency. Medicine (Baltimore) 1991; 70: 229-45. Martin DC, Francis J, Protetch J, Huff FJ. Time dependency of cognitive recovery with cobalamin replacement: Report of a pilot study. J Am Geriatr Soc 1992; 40: 168-72.

N

[46]

Moretti et al.

[81]

[82] [83] [84] [85]

[86]

[87] [88]

[89] [90]

[91]

[92]

[93] [94]

[95] [96]

Homocysteine in Neurology

[103]

[104] [105]

[106]

[107] [108] [109]

[110] [111]

[112] [113]

[114]

[115]

[116]

[117] [118]

[119]

[120]

[121]

[122]

[123]

[124]

[125] [126]

[127]

[128]

[129]

[130]

[131]

[132] [133]

[134]

[135] [136]

[137] [138]

[139]

[140]

[141] [142] [143]

173

Yanai Y, Shibasaki T, Kohno N, Mitsui T, Nakajima H. Concentrations of sulfur-containing free amino acids in lumbar cerebrospinal fluid from patients with consciousness disturbances. Acta Neurol Scand 1983; 68: 386-93. Isobe C, Murata T, Sato C, Terayama Y. Increase of total homocysteine concentration in cerebrospinal fluid in patients with Alzheimer’s disease and Parkinson’s disease. Life Sci 2005; 77: 1836-43. Surtees R, Bowron A, Leonard J. Cerebrospinal fluid and plasma total homocysteine and related metabolites in children with cystathionine beta-synthase deficiency: The effect of treatment. Pediatr Res 1997; 42: 577-82. Kamath AF, Chauhan AK, Kisucka J, et al. Elevated levels of homocysteine compromise blood-brain barrier integrity in mice. Blood 2006; 107: 591-3. Troen AM. The central nervous system in animal models of hyperhomocysteinemia. Prog Neuropsychopharmacol Biol Psychiatry 2005; 29: 1140-51. Algaidi SA, Christie LA, Jenkinson AM, et al. Long-term homocysteine exposure induces alterations in spatial learning, hippocampal signalling and synaptic plasticity. Exp Neurol 2006; 197: 8-21. Parsons RB, Waring RH, Ramsden DB, Williams AC. In vitro effect of the cysteine metabolites homocysteic acid, homocysteine and cysteic acid upon human neuronal cell lines. Neurotoxicology 1998; 19: 599-603. Kruman II, Culmsee C, Chan SL, et al. Homocysteine elicits a DNA damage response in neurons that promotes apoptosis and hypersensitivity to excitotoxicity. J Neurosci 2000; 20(18): 6920-6. Vitvitsky V, Dayal S, Stabler S, et al. Perturbations in homocysteine-linked redox homeostasis in a murine model for hyperhomocysteinemia. Am J Physiol Regul Integr Comp Physiol 2004; 287: R39-46. Streck EL, Bavaresco CS, Netto CA, Wyse AT de S. Chronic hyperhomocysteinemia provokes a memory deficit in rats in the Morris water maze task. Behav Brain Res 2004; 153: 377-81. Streck EL, Delwing D, Tagliari B, et al. Brain energy metabolism is compromised by the metabolites accumulating in homocystinuria. Neurochem Int 2003; 43: 597-602. Lipton SA, Kim WK, Choi YB, et al. Neurotoxicity associated with dual actions of homocysteine at the N-methyl-D-aspartate receptor. Proc Natl Acad Sci USA 1997; 94: 5923-8. Ito S, Provini L, Cherubini E. L-homocysteic acid mediates synaptic excitation at NMDA receptors in the hippocampus. Neurosci Lett 1991; 124: 157-61. Klancnik JM, Cuénod M, Gähwiler BH, Jiang ZP, Do KQ. Release of endogenous amino acids, including homocysteic acid and cysteine sulphinic acid, from rat hippocampal slices evoked by electrical stimulation of Schaffer collateral-commissural fibres. Neuroscience 1992; 49: 557-70. Kim JP, Koh JY, Choi DW. L-homocysteate is a potent neurotoxin on cultured cortical neurons. Brain Res 1987; 437: 103-10. Robert K, Pagès C, Ledru A, et al. Regulation of extracellular signal-regulated kinase by homocysteine in hippocampus. Neuroscience 2005; 133: 925-35. Alam Z, Coombes N, Waring RH, Williams AC, Steventon GB. Plasma levels of neuroexcitatory amino acids in patients with migraine or tension headache. J Neurol Sci 1998; 156: 102-6. Ziemiffska E, Stafiej A, Lazarewicz JW. Role of group I metabotropic glutamate receptors and NMDA receptors in homocysteine-evoked acute neurodegeneration of cultured cerebellar granule neurones. Neurochem Int 2003; 43: 481-92. Shi Q, Savage JE, Hufeisen SJ, et al. L-homocysteine sulfinic acid and other acidic homocysteine derivatives are potent and selective metabotropic glutamate receptor agonists. J Pharmacol Exp Ther 2003; 305: 131-42. Irizarry MC, Gurol ME, Raju S, et al. Association of homocysteine with plasma amyloid beta protein in aging and neurodegenerative disease. Neurology 2005; 65: 1402-8. Sai X, Kawamura Y, Kokame K, et al. Endoplasmic reticulum stress-inducible protein, Herp, enhances presenilin-mediated generation of amyloid beta-protein. J Biol Chem 2002; 277: 12915-20. Morris MS. Homocysteine and Alzheimer’s disease. Lancet Neurol 2003; 2: 425-8. Kruman II, Kumaravel TS, Lohani A, et al. Folic acid deficiency and homocysteine impair DNA repair in hippocampal neurons and

rS o ic r a le D is tri bu tio n

[102]

bl

[101]

fo

[100]

Pu

[99]

ot

[98]

child with Down syndrome and neural tube defect. Am J Med Genet 2001; 103: 128-32. Miller JW. Homocysteine, folate deficiency, and Parkinson’s disease. Nutr Rev 2002; 60: 410-3. Bottiglieri T, Laundy M, Crellin R, et al. Homocysteine, folate, methylation, and monoamine metabolism in depression. J Neurol Neurosurg Psychiatry 2000; 69: 228-32. Bottiglieri T, Crellin R, Reynolds EH. Folate and neuropsychiatry. Folate Health Disease. New York: Marcel Dekker 1995; pp. 435-62. Botez MI, Reynolds EH. Folic acid in neurology, psychiatry and internal medicine. New York: Raven Press 1979. Bottiglieri T, Reynolds EH, Laundy M. Folate in CSF and age. J Neurol Neurosurg Psychiatry 2000; 69(4): 562. Maxwell CJ, Hogan DB, Ebly EM. Serum folate levels and subsequent adverse cerebrovascular outcomes in elderly persons. Dement Geriatr Cogn Disord 2002; 13: 225-34. Snowdon DA, Tully CL, Smith CD, Riley KP, Markesbery WR. Serum folate and the severity of atrophy of the neocortex in Alzheimer disease: Findings from the Nun study. Am J Clin Nutr 2000; 71: 993-8. Clarke R, Smith AD, Jobst KA, et al. Folate, vitamin B12, and serum total homocysteine levels in confirmed Alzheimer disease. Arch Neurol 1998; 55: 1449-55. Ebly EM, Schaefer JP, Campbell NR, Hogan DB. Folate status, vascular disease and cognition in elderly Canadians. Age Ageing 1998; 27: 485-91. Yap S, Boers GH, Wilcken B, et al. Vascular outcome in patients with homocystinuria due to cystathionine beta-synthase deficiency treated chronically: a multicenter observational study. Arterioscler Thromb Vasc Biol 2001; 21: 2080-5. Griffiths R, Tudball N. Observations on the fate of cystathionine in rat brain. Life Sci 1976; 19: 1217-24. Awata S, Nakayama K, Suzuki I, Sugahara K, Kodama H. Changes in cystathionine gamma-lyase in various regions of rat brain during development. Biochem Mol Biol Int 1995; 35: 1331-8. Ichinohe A, Kanaumi T, Takashima S, et al. Cystathionine betasynthase is enriched in the brains of Down’s patients. Biochem Biophys Res Commun 2005; 338: 1547-50. Kranich O, Dringen R, Sandberg M, Hamprecht B. Utilization of cysteine and cysteine precursors for the synthesis of glutathione in astroglial cultures: Preference for cystine. Glia 1998; 22: 11-8. Scott JM, Molloy AM, Kennedy DG, Kennedy S, Weir DG. Effects of the disruption of transmethylation in the central nervous system: An animal model. Acta Neurol Scand Suppl 1994; 154: 27-31. Shanker G, Allen JW, Mutkus LA, Aschner M. The uptake of cysteine in cultured primary astrocytes and neurons. Brain Res 2001; 902: 156-63. Wu G, Fang Y-Z, Yang S, Lupton JR, Turner ND. Glutathione metabolism and its implications for health. J Nutr 2004; 134: 489-92. Grieve A, Butcher SP, Griffiths R. Synaptosomal plasma membrane transport of excitatory sulphur amino acid transmitter candidates: Kinetic characterization and analysis of carrier specificity. J Neurosci Res 1992; 32: 60-8. Griffiths R, Grieve A, Allen S, Olverman HJ. Neuronal and glial plasma membrane carrier-mediated uptake of L-homocysteate is not selectively blocked by beta-p chlorophenylglutamate. Neurosci Lett 1992; 147: 175-8. Zeise ML, Knöpfel T, Zieglgänsberger W. (+/-)-beta-Parachlorophenylglutamate selectively enhances the depolarizing response to L-homocysteic acid in neocortical neurons of the rat: Evidence for a specific uptake system. Brain Res 1988; 443: 373-6. Ho PI, Ashline D, Dhitavat S, et al. Folate deprivation induces neurodegeneration: Roles of oxidative stress and increased homocysteine. Neurobiol Dis 2003; 14: 32-42. Eto K, Asada T, Arima K, Makifuchi T, Kimura H. Brain hydrogen sulfide is severely decreased in Alzheimer’s disease. Biochem Biophys Res Commun 2002; 293: 1485-8. Regland B, Andersson M, Abrahamsson L, et al. Increased concentrations of homocysteine in the cerebrospinal fluid in patients with fibromyalgia and chronic fatigue syndrome. Scand J Rheumatol 1997; 26: 301-7. Regland B, Abrahamsson L, Blennow K, Grenfeldt B, Gottfries CG. CSF-methionine is elevated in psychotic patients. J Neural Transm Vienna Austria 2004; 111: 631-40.

N

[97]

Current Nutrition & Food Science, 2017, Vol. 13, No. 3

174 Current Nutrition & Food Science, 2017, Vol. 13, No. 3

[150]

[151]

[152]

[153]

[154]

[155]

[156]

[157] [158]

[159]

[160]

[161] [162]

[163] [164]

[165]

[166]

[167]

[168]

[169]

[170] [171]

[172]

[173]

[174]

[175]

Reis EA, Zugno AI, Franzon R, et al. Pretreatment with vitamins E and C prevent the impairment of memory caused by homocysteine administration in rats. Metab Brain Dis 2002; 17: 211-7. Tchantchou F, Graves M, Ortiz D, Rogers E, Shea TB. Dietary supplementation with 3-deaza adenosine, N-acetyl cysteine, and Sadenosyl methionine provide neuroprotection against multiple consequences of vitamin deficiency and oxidative challenge: relevance to age-related neurodegeneration. Neuromolecular Med 2004; 6: 93-103. Prudova A, Bauman Z, Braun A, et al. S-adenosylmethionine stabilizes cystathionine beta-synthase and modulates redox capacity. Proc Natl Acad Sci USA 2006; 103: 6489-94. Murr C, Widner B, Wirleeitner B, Fuchs D. Neopterin as a marker for immune system activation. Curr Drug Metab 2001; 2: 175-87 Bleie O, Semb AG, Grundt H, et al. Homcysteine-lowering therapy does not affect inflammatory markers of atherosclerosis in patients with stable coronary disease. J Int Med 2007; 262(2): 244-53. Ploder M, Kurz K, Splitter A, Neurauter G, Roth E, Fuch D. Early increase of plasma Hcy in sepsis patients with poor outcome. Mol Med 2010; 16: 498-504. Deng J, Lu S, Li H, et al. Homocysteine activates B cells via regulating PKM-2 dependent metabolic reprogramming. J Immunol 2017; 198: 170-83. Li J-J, Li Q, Du H-P, et al. Homocysteine Triggers inflammatory responses in macrophages through inhibiting CSE-H2S signaling via DNA hypermethylation of CSE promoter. Int J Mol Sci 2015; 16: 12560-77. Krishna SM, Dear A, Craig JM, Norman PE, Golledge J. The potential role of homocysteine mediated DNA methylation and associated epigenetic changes in abdominal aortic aneurysm formation. Atherosclerosis 2013; 228: 295-305. Zhou S, Zhang Z, Xu G. Notable epigenetic role of hyperhomocysteinemia in atherogenesis. Lipids Health Dis 2014; 13: 134. Yi-Deng J, Tao S, Hui-Ping Z, et al. Folate and ApoE DNA methylation induced by homocysteine in human monocytes. DNA Cell Biol 2007; 26: 737-44. Chang P-Y, Lu S-C, Lee C-M, et al. Homocysteine inhibits arterial endothelial cell growth through transcriptional downregulation of fibroblast growth factor-2 involving G protein and DNA methylation. Circ Res 2008; 102: 933-41. Boldyrev A, Bryshkova E, MAshkina A, Vladychenskaya E. Why is homocysteine toxic for the nervous and immune systems? Curr Aging Sci 2013; 6(1): 29-36. Essouma M, Noubiap JJN, Therapeutic potential of folic acid supplementation for cardiovascular disease prevention through homocysteine lowering and blockade in rheumatoid arthritis patients. Biomark Res 2015; 3: 24. Ying G, Wang Y, Cen XM, Yang M, Liang Y, Xie QB. Lipid peroxidation-mediated inflammation promotes cell apoptosis through activation of NFK-B pathway in rheumatoid arthritis synovial cells. Med Infalmm 2015; 2015: 1-10. Villalobos MA, De La Cruz JP, Cuerda MA, et al. Effect of Sadenosyl-L-methionine on rat brain oxidative stress damage in a combined model of permanent focal ischemia and global ischemiareperfusion. Brain Res 2000; 883: 31-40. Matsui Y, Kubo Y, Iwata N. S-adenosyl-L-methionine prevents ischemic neuronal death. Eur J Pharmacol 1987; 144: 211-6. Ho PI, Ortiz D, Rogers E, Shea TB. Multiple aspects of homocysteine neurotoxicity: Glutamate excitotoxicity, kinase hyperactivation and DNA damage. J Neurosci Res 2002; 70: 694-702. Fuso A, Seminara L, Cavallaro RA, D’Anselmi F, Scarpa S. Sadenosylmethionine/homocysteine cycle alterations modify DNA methylation status with consequent deregulation of PS1 and BACE and beta-amyloid production. Mol Cell Neurosci 2005; 28: 195204. Wright CB, Paik MC, Brown TR, et al. Total homocysteine is associated with white matter hyperintensity volume: The Northern Manhattan study. Stroke J Cereb Circ 2005; 36: 1207-11. den Heijer T, Vermeer SE, Clarke R, et al. Homocysteine and brain atrophy on MRI of non-demented elderly. Brain J Neurol 2003; 126: 170-5.

rS o ic r a le D is tri bu tio n

[149]

bl

[148]

fo

[147]

Pu

[146]

ot

[145]

N

[144]

sensitize them to amyloid toxicity in experimental models of Alzheimer’s disease. J Neurosci J Soc Neurosci 2002; 22: 1752-62. Hasegawa T, Ukai W, Jo D-G, et al. Homocysteic acid induces intraneuronal accumulation of neurotoxic Abeta42: Implications for the pathogenesis of Alzheimer’s disease. J Neurosci Res 2005; 80: 869-76. Selkoe DJ. Presenilin, Notch, and the genesis and treatment of Alzheimer’s disease. Proc Natl Acad Sci U S A 2001; 98: 11039-41. Scarpa S, Fuso A, D’Anselmi F, Cavallaro RA. Presenilin 1 gene silencing by S-adenosylmethionine: A treatment for Alzheimer disease? FEBS Lett 2003; 541: 145-8. Leulliot N, Quevillon-Cheruel S, Sorel I, et al. Structure of protein phosphatase methyltransferase 1 (PPM1), a leucine carboxyl methyltransferase involved in the regulation of protein phosphatase 2A activity. J Biol Chem 2004; 279: 8351-8. Ferreira A, Lu Q, Orecchio L, Kosik KS. Selective phosphorylation of adult tau isoforms in mature hippocampal neurons exposed to fibrillar A beta. Mol Cell Neurosci 1997; 9: 220-34. Wang JZ, Gong CX, Zaidi T, Grundke-Iqbal I, Iqbal K. Dephosphorylation of Alzheimer paired helical filaments by protein phosphatase-2A and -2B. J Biol Chem 1995; 270: 4854-60. Vogelsberg-Ragaglia V, Schuck T, Trojanowski JQ, Lee VM. PP2A mRNA expression is quantitatively decreased in Alzheimer’s disease hippocampus. Exp Neurol 2001; 168: 402-12. Sontag E, Hladik C, Montgomery L, et al. Downregulation of protein phosphatase 2A carboxyl methylation and methyltransferase may contribute to Alzheimer disease pathogenesis. J Neuropathol Exp Neurol 2004; 63: 1080-91. Zhao W-Q, Feng C, Alkon DL. Impairment of phosphatase 2A contributes to the prolonged MAP kinase phosphorylation in Alzheimer’s disease fibroblasts. Neurobiol Dis 2003; 14: 458-69. Vafai SB, Stock JB. Protein phosphatase 2A methylation: A link between elevated plasma homocysteine and Alzheimer’s disease. FEBS Lett 2002; 518: 1-4. Tolstykh T, Lee J, Vafai S, Stock JB. Carboxyl methylation regulates phosphoprotein phosphatase 2A by controlling the association of regulatory B subunits. EMBO J 2000; 19: 5682-91 Mok SS, Turner BJ, Beyreuther K, et al. Toxicity of substratebound amyloid peptides on vascular smooth muscle cells is enhanced by homocysteine. Eur J Biochem FEBS 2002; 269: 3014-22. Nelson AR, Sweeney MD, Sagare AP, Zlokovic BV. Neurovascular dysfunction and neurodegeneration in dementia and Alzheimer’s disease. Biochim Biophys Acta 2016; 1862: 887-900. Zou C-G, Banerjee R. Homocysteine and redox signaling. Antioxid Redox Signal 2005; 7: 547-59. Banerjee R, Zou C-G. Redox regulation and reaction mechanism of human cystathionine-beta-synthase: A PLP-dependent hemesensor protein. Arch Biochem Biophys 2005; 433: 144-56. Maclean KN, Janosík M, Kraus E, et al. Cystathionine betasynthase is coordinately regulated with proliferation through a redox-sensitive mechanism in cultured human cells and Saccharomyces cerevisiae. J Cell Physiol 2002; 192: 81-92. Baydas G, Ozer M, Yasar A, Tuzcu M, Koz ST. Melatonin improves learning and memory performances impaired by hyperhomocysteinemia in rats. Brain Res 2005; 1046: 187-94. Perna AF, Ingrosso D, De Santo NG. Homocysteine and oxidative stress. Amino Acids 2003; 25: 409-17. James SJ, Cutler P, Melnyk S, et al. Metabolic biomarkers of increased oxidative stress and impaired methylation capacity in children with autism. Am J Clin Nutr 2004; 80: 1611-7. Jellinger KA. Pathology and pathogenesis of vascular cognitive impairment-a critical update. Front Aging Neurosci 2013; 5: 17. Pushpakumar S, Kundu S, Sen U. Endothelial dysfunction: The link between homocysteine and hydrogen sulfide. Curr Med Chem 2014; 21: 3662-72. Wyse ATS, Zugno AI, Streck EL, et al. Inhibition of Na(+),K(+)ATPase activity in hippocampus of rats subjected to acute administration of homocysteine is prevented by vitamins E and C treatment. Neurochem Res 2002; 27: 1685-9. Ho PI, Collins SC, Dhitavat S, et al. Homocysteine potentiates beta-amyloid neurotoxicity: role of oxidative stress. J Neurochem 2001; 78: 249-53.

Moretti et al.

[176]

[177]

[178]

[179]

[180]

[181]

[182]

[183] [184]

[185]

[186]

[187]

Homocysteine in Neurology

[194]

[195]

[196]

[197]

[198] [199]

[200]

175

Ueland PM, Refsum H, Beresford SA, Vollset SE. The controversy over homocysteine and cardiovascular risk. Am J Clin Nutr 2000; 72: 324-32. Homocysteine Studies Collaboration. Homocysteine and risk of ischemic heart disease and stroke: A meta-analysis. JAMA 2002; 288: 2015-22. Hogervorst E, Ribeiro HM, Molyneux A, Budge M, Smith AD. Plasma homocysteine levels, cerebrovascular risk factors, and cerebral white matter changes (leukoaraiosis) in patients with Alzheimer disease. Arch Neurol 2002; 59: 787-93. Lonn E, Yusuf S, Arnold MJ, et al. Homocysteine lowering with folic acid and B vitamins in vascular disease. N Engl J Med 2006; 354: 1567-77. Martí-Carvajal AJ, Solà I, Lathyris D, Salanti G. Homocysteine lowering interventions for preventing cardiovascular events. Cochrane Database Syst Rev 2009: CD006612. EFSA Panelon Dietetic products, nutrition and allergies. Guidance on the scientific requirements for health claims related to antioxidant, oxidative damage and cardiovascular health. EFSA J 2011; 9(12): 2474.

rS o ic r a le D is tri bu tio n

[193]

bl

[192]

fo

[191]

Pu

[190]

ot

[189]

Nurk E, Refsum H, Tell GS, et al. Plasma total homocysteine and memory in the elderly: The Hordaland homocysteine study. Ann Neurol 2005; 58: 847-57. Sachdev PS. Homocysteine and brain atrophy. Prog Neuropsychopharmacol Biol Psychiatry 2005; 29: 1152-61. Seshadri S, Beiser A, Selhub J, et al. Plasma homocysteine as a risk factor for dementia and Alzheimer’s disease. N Engl J Med 2002; 346: 476-83. Eikelboom JW, Lonn E, Genest J, Hankey G, Yusuf S. Homocyst(e)ine and cardiovascular disease: A critical review of the epidemiologic evidence. Ann Intern Med 1999; 131: 363-75. Bostom AG, Rosenberg IH, Silbershatz H, et al. Nonfasting plasma total homocysteine levels and stroke incidence in elderly persons: The Framingham study. Ann Intern Med 1999; 131: 352-5. McCully KS. Vascular pathology of homocysteinemia: Implications for the pathogenesis of arteriosclerosis. Am J Pathol 1969; 56: 111-28. Chung YC, Kruyer A, Yao Y, et al. Hyperhomocysteinemia exacerbates Alzheimer’s disease pathology by way of the beta-amyloid fibrinogen interaction. J Thromb Haemost 2016; 14: 1442-52.

N

[188]

Current Nutrition & Food Science, 2017, Vol. 13, No. 3