ArticlePDF AvailableLiterature Review

Hyperhomocysteinemia: Clinical Insights

SAGE Publications Inc
Journal of Central Nervous System Disease
Authors:
  • King Abdullah Specialized Children’s Hospital

Abstract and Figures

ABSTRACT: Homocysteine (Hcy) is a sulfhydryl-containing amino acid, and intermediate metabolite formed in metabolising methionine (Met) to cysteine (Cys); defective Met metabolism can increase Hcy. The effect of hyperhomocysteinemia (HHcy) on human health, is well described and associated with multiple clinical conditions. HHcy is considered to be an independent risk factor for common cardiovascular and central nervous disorders, where its role in folate metabolism and choline catabolism is fundamental in many metabolic pathways. HHcy induces inflammatory responses via increasing the pro-inflammatory cytokines and downregulation of anti-inflammatory cytokines which lead to Hcy-induced cell apoptosis. Conflicting evidence indicates that the development of the homocysteine-associated cerebrovascular disease may be prevented by the maintenance of normal Hcy levels. In this review, we discuss common conditions associated with HHcy and biochemical diagnostic workup that may help in reaching diagnosis at early stages. Furthermore, future systematic studies need to prove the exact pathophysiological mechanism of HHcy at the cellular level and the effect of Hcy lowering agents on disease courses.
This content is subject to copyright.
https://doi.org/10.1177/1179573520962230
Creative Commons Non Commercial CC BY-NC: This article is distributed under the terms of the Creative Commons Attribution-NonCommercial
4.0 License (https://creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial use, reproduction and distribution of the work without
further permission provided the original work is attributed as specified on the SAGE and Open Access pages (https://us.sagepub.com/en-us/nam/open-access-at-sage).
Journal of Central Nervous System Disease
Volume 12: 1–8
© The Author(s) 2020
Article reuse guidelines:
sagepub.com/journals-permissions
DOI: 10.1177/1179573520962230
Introduction
Homocysteine (Hcy) is a sulfhydryl-containing amino acid, a
homolog of cysteine with one additional methylene group.1,2
Hcy is not acquired through the diet, but is synthesized as an
intermediate metabolite from methionine (Met) metabolism. It
is converted to cysteine via the transsulfuration pathway or
resynthesized back to methionine via the re-methylation path-
way.3,4 In plasma, 99% of the Hcy is bound to proteins, including
cysteine, and cysteinylglycine via disulfide linkages, while only
1% is found in a free reduced form.5 For the estimation of the
Hcy level, plasma tissue homogenate samples are analyzed using
various methodologies such as immunoassay, capillary electro-
phoresis, enzymatic assay, liquid chromatography-mass spec-
trometry (LCMS) and high-pressure liquid chromatography
(HPLC).6 The normal Hcy levels is ranging between 5 and
15 μmol/L, while a mildly increased level is 15 to 30 μmol/L,
moderate from 30 to 100 μmol/L, and a value >100 μmol/L is
classified as severe hyperhomocysteinemia.7,8
The levels of Hcy can be increased through a defective
metabolism of Met, due to genetic defects of the transcription
of enzymes responsible for Hcy metabolism or deficiencies of
cofactors involved in these pathways such as vitamins B6, B12,
and folate.9 The effect of hyperhomocysteinemia (HHcy) on
human health was first described in the mid-20th century by
Kilmer S. McCully.10 Since then, many epidemiologic reports
indicated that HHcy is associated with multiple clinical condi-
tions, while controlled Hcy level in high risk group associated
with improved physical and mental health.11-14 It is considered
as an independent risk factor for cardiovascular disease, as well
as for stroke and myocardial infarction by the American Heart
Association.15,16 Although it is not directly involved in protein
synthesis, the exposure to a toxic effect of Hcy, induced cyto-
toxicity that lead to a reduction of cultured endothelial cells
viability through a direct and indirect effect on the pathway of
apoptosis.17,18
Studies have identified a strong association between HHcy
and induction of inflammatory determinants including the
expression of adhesion molecules, leukocyte adhesion, endothe-
lial dysfunction, oxidative stress, and reduced nitric oxide bio-
availability in both human and experimental models.19,20 In
HHcy state, NFκB, a transcription factor that regulates the
transcription of various genes involved in inflammatory and
immune responses is activated, additionally, marked increase in
pro-inflammatory cytokines and downregulation of anti-
inflammatory cytokines were observed.19
In HHcy patients, supporting evidence indicate that the
development of homocysteine-associated vascular disease may
be prevented by the maintenance of normal Hcy levels, with
conventional treatment of folate supplementation and vitamin
B6 and possibly vitamin B12.21 However, despite lowered Hcy
levels, the clinical picture of pathophysiological conditions
caused by an elevated Hcy level may not be reversible for cer-
tain conditions.22
Several studies were conducted to investigate the treatment
of HHcy in patients with a history of arteriosclerotic vascular
disease (ASVD).23,24 A large study was conducted with more
than a 1000 individuals with HHcy, to determine the effect of
supplementation with vitamins B12, B6, and folic acid for
6 weeks. The study reported that there was a proportional
reduction in the plasma Hcy level caused by the folic
Hyperhomocysteinemia: Clinical Insights
Fuad Al Mutairi1,2,3
1Medical Genetics Division, Department of Pediatrics, King Abdulaziz Medical City, Riyadh, Saudi
Arabia. 2King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia. 3King
Abdullah International Medical Research Centre, Riyadh, Saudi Arabia.
ABSTRACT: Homocysteine (Hcy) is a sulfhydryl-containing amino acid, and intermediate metabolite formed in metabolising methionine (Met)
to cysteine (Cys); defective Met metabolism can increase Hcy. The effect of hyperhomocysteinemia (HHcy) on human health, is well described
and associated with multiple clinical conditions. HHcy is considered to be an independent risk factor for common cardiovascular and central
nervous disorders, where its role in folate metabolism and choline catabolism is fundamental in many metabolic pathways. HHcy induces inflam-
matory responses via increasing the pro-inflammatory cytokines and downregulation of anti-inflammatory cytokines which lead to Hcy-induced
cell apoptosis. Conflicting evidence indicates that the development of the homocysteine-associated cerebrovascular disease may be prevented
by the maintenance of normal Hcy levels. In this review, we discuss common conditions associated with HHcy and biochemical diagnostic
workup that may help in reaching diagnosis at early stages. Furthermore, future systematic studies need to prove the exact pathophysiological
mechanism of HHcy at the cellular level and the effect of Hcy lowering agents on disease courses.
KEYWORDS: Homocysteine, folate, cobalamin, vitamin B -12, methylenetetrahydrofolate reductase, homocystinurea
RECEIVED: June 23, 202 0. REVISED ARTICLE ACCEPTED: September 6, 20 20.
TYPE: Review
FUNDING: The author r eceived n o nanci al suppo rt for the r esearch , authors hip, and /or
publication of this article.
DECLARATION OF CONFLICTING INTERESTS: The author dec lared no po tential
conic ts of inter est with re spect to t he resear ch, autho rship, an d/or publ icatio n of this
article.
CORRESPONDING AUTHOR: Fuad Al Mutairi, Division of Genetics, Department of
Pediatri cs, Kin g Abdula ziz Medi cal City, Mi nistry o f Nationa l Guard- Health A ffairs
(MNGHA), P. O. Box 22490, Ma il Code 1490, R iyadh 11426, Saudi Ar abia.
Email: almutairifu@ngha.med.sa
962230CNS0010.1177/1179573520962230Journal of Central Nervous System DiseaseAl Mutairi
review-article2020
2 Journal of Central Nervous System Disease
acid treatment and that the level of the reduction was higher in
participants with pre-treated Hcy levels.25 The dosage required
for the treatment of HHcy may vary according to the underly-
ing condition, however, the minimum effective dose of folic
acid to obtain the maximal lowering of Hcy is 400 μg.26,27
Prolonged folic acid therapy is associated with decreased vita-
min B12 blood levels and worsening symptoms of B12 defi-
ciency, so a supplement vitamin B12 is usually taken in the
form of the cyanocobalamin.28 In a randomized controlled
trial, 104 patients were divided in 2 groups, the study group
received a combined nutraceutical containing 400 μg folate-
6-5-methyltetrahydrofolate, 3 mg vitamin B6, 5 μg vitamin
B12, 2.4 mg vitamin B2, 12.5 mg zinc and 250 mg betaine once
daily for 2 months, and the control group received a supple-
ment of folic acid (5 mg/day). The results indicated that the
Hcy reduction was significantly higher in the treatment group
(P < .035).28 This may support the available evidences that
vitamin supplements significantly reduce the Hcy levels in a
sustained but suboptimal way, even if supraphysiological doses
are used.29,30 Recent study revealed the protective role of vita-
min E as antioxidant and melatonin may alleviate Hcy-induced
cell apoptosis, which may add insight into therapeutic
approaches to Hcy-induced damages in endothelial cells.31
The review will discuss Hcy and its biological functions in
the body, conditions that induce by or are related to HHcy, and
biochemical investigations that may ease the recognition of
suspected cases at early stage of disease course.
Biosynthesis and Metabolism of Homocysteine
Hcy is produced in all cells and biosynthesized from methio-
nine through multiple steps, initiated by the demethylation of
methionine (Met) as well as 3 subsequent steps.1,3 The first
step is the transfer of an adenosine group from ATP to methio-
nine by S-adenosyl methionine (SAM or AdoMet) synthetase
(also called methionine adenosyltransferase, MAT), resulting
in the formation of S-Adenosyl-L-methionine (AdoMet or
SAM)32-34 (Figure 1).
In the second step, the universal methyl donor, SAM,
donates a methyl group to acceptor molecules such as DNA,
RNA, proteins, and neurotransmitters.35 The resulting com-
pound S-adenosyl homocysteine (AdoHCys or SAH), lack-
ing the methyl group, can function as an inhibitor of most
methyltransferases and is subsequently cleaved via a revers-
ible reaction by S-adenosyl homocysteine hydrolase
(SAHH) to produce adenosine and L-homocysteine.4,36,37
Finally, the L-homocysteine can be metabolized by 2
Figure 1. The homocysteine metabolic cycle.
Abbreviations: ADK, adenosine kinase; AdoHCys or SAH, S-adenosyl homocysteine; AdoMet or SAM, adenosyl methionine; BHMT, betaine-homocysteine
methyltransferase; Cbl, cobalamin—vitamin B12; CBS, cystathionine beta-synthase; DMG, dimethylglycine; GNMT, glycine N-methyltransferase; Hcy, homocysteine; MAT,
methionine adenosyltransferase; 5-methyl-THF, 5-N-methyl tetrahydrofolate; 5,10-methylene-THF, 5,10-methylenetetrahydrofolate; MS, methionine synthase; MTHFR,
methylenetetrahydrofolate reductase; SAHH, S-adenosylhomocysteine hydrolase; SHMT, serine hydroxymethyltransferase.
Al Mutairi 3
reactions, transsulfuration or re-methylation, ultimately
producing L-methionine and L-cysteine, respectively.2,38
Remethylation
Remethylation involves recycling Hcy to methionine, by
using vitamin B12 as a cofactor. The methionine synthase
(MS) catalyzes the re-methylation reaction, restoring Met
by transferring the methyl group from 5-N-methyl tetrahy-
drofolate (5-methyl-THF) to Hcy.39-41 In this cycle, folate is
reduced to tetrahydrofolate, which is an important key
player in folate metabolism as a folate acceptor molecule,
which is then converted to 5,10-methylenetetrahydrofolate
(5,10-methylene-THF) by the pyridoxal phosphate (PLP)-
dependent serine hydroxymethyltransferase (SHMT).42
Methylenetetrahydrofolate reductase (MTHFR) reduces
5,10-methylene-THF to 5-methyl-THF.43
There is another pathway of remethylating Hcy that uses an
enzyme called betaine-homocysteine methyltransferase (BHMT).
The betaine pathway is restricted to the liver and kidney where
betaine can serve as methyl donor molecules. In this reaction, the
methyl group is transferred from betaine to Hcy to produce
methionine and dimethylglycine (DMG).41,44,45 Here choline
plays a significant role in Met regeneration, as it is oxidized to
betaine, which can be used in this conversion of Hcy.46,47
Transsulfuration
In the transsulfuration process, Hcy is irreversibly converted
to cysteine by cystathionine b-synthase (CBS), which is fol-
lowed by the catalysis done by cystathionine c-lyase (CTL).
Both enzymes need the cofactor pyridoxal-50-phosphate
(vitamin B6) to function.48 Serine can be enzymatically
added to homocysteine by CBS and vitamin B6, to form cys-
tathionine, which can be cleaved by CTL to form cysteine.49
Once cysteine is formed, it can be used in protein synthesis
and glutathione (GSH) production and cannot be converted
to back to Hcy.50
Causes of Hyperhomocysteinemia
Enzyme defects associated with Hcy metabolism are consid-
ered the most prevalent cause of HHcy. The enzyme defects
has been researched, especially the polymorphisms of the main
enzymes involved in Hcy metabolism such as Cystathionine
b-synthase (CBS) deficiency, Methylenetetrahydrofolate
reductase (MTHFR) deficiency, Methionine synthase defi-
ciency, and Methionine adenosyltransferase deficiency
(Table 1).37,51,52 In addition to genetic causes, many other fac-
tors related to age, lifestyle such as cigarette smoking, alcohol
consumption and nutritional deficiencies in folic acid, vitamin
B6, vitamin B12, and betaine are as responsible for HHcy.53-56
In this review, we will discuss the main enzymatic defects in
this pathway.
Cystathionine beta-synthase (CBS) deficiency or
classical homocystinuria
Classical homocystinuria (HCU) (OMIM 236200), is an auto-
somal recessive disease caused by biallelic pathogenic variations
in the CBS gene.57 Deficiency of the CBS enzyme causes ele-
vated tissue and plasma levels of Hcy and its precursor, methio-
nine.58 Typically, patients can manifest a wide range of
symptoms with variable severity involving the ocular, skeletal,
vascular, and central nervous systems.59,60
The prevalence of CBS deficiency has been reported as
1:200 000 to 1:335 000 and >200 pathogenic variants have
Table 1. Causes of hyperhomocysteinemia.
Severe >100 μmol/L
Cystathionine synthase (CBS) deciency
Untreated methylenetetrahydrofolate reductase (MTHFR)
deciency
Moderate 60 to 100 μmol/L
Methylenetetrahydrofolate reductase (MTHFR) deciency
Methionine synthase (MS) deciency
Moderate 30 to 60 μmol/L
Intracellular cobalamin metabolism, for example, cblC, cblD,
cblE, cblF, and cblG
Methionine adenosyltransferase I/III deciency
Glycine N-methyltransferase (GNMT) deciency
S-adenosylhomocysteine hydrolase (SAHH) deciency
Adenosine kinase (ADK) deciency
Sever nutritional inadequacy, for example, folate and vitamin B12
Compound heterozygosity of MTHFR
Mild 16 to 30 μmol/L
Impaired folate or vitamin B12 absorption
Mild to moderate nutritional inadequacy, for example, folate and
vitamin B12
Vegetarian diet (low vitamin B12 intake)
Chronic renal failure
Hypothyroidism
Anemia
Malignant tumors
Certain drugs affecting Hcy metabolism: cholestyramine,
metformin, methotrexate, nicotinic acid (niacin), bric acid
derivatives, and oral contraceptive pills (OCPs)
Advanced age
Lifestyle conditions such as excessive coffee or alcohol
consumption, cigarette smoking
4 Journal of Central Nervous System Disease
been described in the CBS gene, however, mutations such as
p.Ile278Thr, p.Thr191Met, p.Gly307Ser, and p.Trp323Ter are
the most prevalent mutations and represent half of all HCU
alleles.61-63 Considering the significant effect of genetic poly-
morphisms on the increase of the HCys level, current studies
are investigating the correlation between the polymorphisms
and stroke events, however, the results are still conflicting.56 Not
all polymorphisms in CBS have an effect on enzyme activity,
however a T833C polymorphism in CBS, caused mild HHcy in
different ethnic groups.64-66 Current treatment options for CBS
are very limited and often inefficient, partially due to low patient
compliance with a very strict dietary regimen.67 However recent
studies shows the efficacy of some novel therapies including
enzyme replacement and gene therapy approaches.68
Methylenetetrahydrofolate reductase (MTHFR)
Methylenetetrahydrofolate reductase (MTHFR) deficiency
(OMIM 236250), an autosomal-recessive inheritance disease, is
caused by a mutation in the MTHFR gene which encode
MTHFR, a key enzyme of folate metabolism in the process of
one-carbon metabolism.69 Polymorphisms of MTHFR would
cause impaired methylation as well as a deficiency of folate, and
a wide range of diseases including cardiovascular, tumors, neuro-
logic, and psychiatric disorders.70,71 One of the most studied
polymorphisms in MTHFR is C677T.72-74 This polymorphism
is responsible for an increase of Hcy concentration and folate
deficiency compared to a normal genotype individual.75 It is esti-
mated that 10% of the global population is homozygous (TT
genotype), which may vary in different populations reaching
25%.42 The treatment in MTHFR is symptomatic including the
treatment of associated neurological symptoms.76 Vitamin sup-
plementation should be considered in these patients including
vitamin B12, folic acid, vitamin B6, betaine, and methionine.77
Methionine synthase
Methionine synthase promotes the methyl group transfer from
methylated folate to homocysteine to yield methionine, and
Cbl act as a cofactor in this catalytic reaction. This enzyme is
encoded by the MTR gene. Mutations in this gene are the
underlying cause of methylcobalamin deficiency cblG-type.78,79
In patients with a methionine synthase deficiency (CbIG),
complementation studies on cultured fibroblasts indicated a
cblG defect.80 This deficiency is rarely reported in literature,
and the patients do not have specific neurological symptoms
for example, blindness or leukoencephalopathy associated with
normal vitamin B12 and folate, hyperhomocysteinemia with
hypomethioninemia in the absence of methylmalonic acid.74
Acquired and inherited disorders of cobalamin
Cobalamin (Cbl-Vitamin B12) is an essential cofactor for MS
in the folate cycle to ultimately produce the 5-Methyl THF
which provides a methyl group to convert homocysteine to
methionine.69,81 Inborn errors of cobalamin metabolism can
affect its absorption (intrinsic factor deficiency, Imerslund-
Gräsbeck syndrome), transportation (transcobalamin
deficiency), as well as genetic defects of the intracellular cobal-
amin metabolism such as CblC, CblD, CblE, CblF, and
CblG.82-84 Megaloblastic anemia, pancytopenia and failure to
thrive are the main manifestation of Cbl deficiency. However,
if the diagnosis is delayed, it may be accompanied by irreversi-
ble neurological deficits.83 Cbl deficiency rarely requires instant
therapy, however, treatment should be started timeously to pre-
vent severe neurologic symptoms (eg, seizures, gait distur-
bances, mental changes and extensive sensory defects) due to
the risk of irreversibility.85
Methylation disorders
Inherited methylation disorders are a group of disorders affecting
the transmethylation processes in the metabolic pathway between
methionine and Homocysteine, including methionine adenosyl-
transferase I/III, glycine N-methyltransferase, Sadenosylhomo-
cysteine hydrolase and adenosine kinase deficiencies.86 Although
isolated hypermethioninemia is the biochemical hallmark of this
group of disorders, mild to moderate HHcy can be present in all
patients.87 Three of these directly affect the reactions in the
methionine pathway. The first is the conversion of methionine to
AdoMet, catalyzed by methionine adenosyltransferase. The sec-
ond enzyme is glycine N-methyltransferase (GNMT), which
transfers a methyl group to glycine producing sarcosine. The third
enzyme, S-adenosylhomocysteine hydrolase (SAHH), is a homo-
tetrameric enzyme, which converts S adenosylhomocysteine to
homocysteine and adenosine.88-90 In methylation disorders, a low
methionine diet can be beneficial in patients with MAT I/III defi-
ciency, and to a lesser extend in SAHH and AKD.
S-adenosylmethionine supplementation may specifically be useful
in patients with MAT I/III deficiency.86
Conditions Associated With Hyperhomocysteine
Hcy function in folate metabolism and choline catabolism is
fundamental for the synthesis of different sulfur-containing
amino acids and methylated compounds which are important
for may cellular pathways.42,91 Numerous studies demonstrated
that the disruption of the Hcy metabolism which lead to HHcy
by common MTHFR gene polymorphism, increases the risk
for several complex disorders and it plays an important role in
the pathogenicity of such disorders, including the cardiovascu-
lar system for example, congestive heart disease and arthroscle-
rosis.92-94 In the central nervous system, the disorders include
cognitive impairment, Parkinson’s disease, Alzheimer’s disease
(AD), multiple sclerosis and epilepsy.95 In addition, an elevated
Hcy was linked to osteoporosis, chronic renal failure, hypothy-
roidism, insulin resistant diabetes, polycystic ovarian syndrome,
and gastrointestinal disorders.96-100 Although the molecular
mechanism in this role has not been fully defined, age related
Al Mutairi 5
disorders such as acoustic dysfunction, and age related macular
degeneration has been reported.101-104
Stroke and cardiovascular diseases
Literature support the theory of a correlation between HHcy
and the risk for peripheral vascular diseases, including stroke,
venous thromboembolism and cardiovascular disease, for
example congestive cardiomyopathy, myocardial infarction and
coronary artery disease.105 A potential mechanism is the
thrombotic activity of Hcy and its direct effect of endothelial
dysfunction, where the Hcy acts as an inhibitor of endothelial
nitric oxide synthase (eNOS), which will cause reduced bioa-
vailability of NO, through the inhibitory effect of asymmetric
dimethylarginine (ADMA).106 Findings from a clinical study
investigating patients with heart failure, support through pre-
clinical evidence that the myocardium is especially vulnerable
to damage by HHcy, which is associated with the production of
reactive oxygen species and cause the progression of cardiovas-
cular disease and left ventricle remodeling.107,108
Cognitive impairment, Alzheimer’s disease,
Parkinsons disease and epilepsy
Several case control studies confirmed a positive correlation
between HHcy as a neurotoxic condition and both vascular
dementia and AD. However, it remains unclear whether an
elevated blood homocysteine level is a direct risk factor for AD
or possibly, poor vitamin nutrition in the elderly.101 The mech-
anism through which high levels of Hcy cause AD is still being
investigated, however, in an experimental study, HHcy resulted
in increased gene expression of proinflammatory markers such
as IL1b and TNFa in microglia and an increased expression of
kinases in neuronal cells.109 Studies have also shown an increase
in neurodegeneration due to homocysteine-related oxidative
stress, causing an increase in the production of superoxide and
other reactive oxygen species, and apoptosis.110 Another pro-
posed mechanism for Hcy neurodegeneration involves its role
as an agonist for AMPA (both metabotropic and ionotropic)
and NMDA receptors. All those changes in vascular smooth
muscle cells, provide further neurotoxic peculiarity to HHcy as
a risk factor for neurodegenerative diseases.111 Recently, an
experimental model, linked the HHcy to increased oxidative
stress, upregulated expression of proteins that promote blood
coagulation, exacerbated blood-brain barrier dysfunction and
promoted the infiltration of inflammatory cells into the cortex
in traumatic brain injury (TBI).112
Gastrointestinal disorders
There is growing evidence that HHcy is associated with inflam-
matory bowel disease (IBD) and many autoimmune dis-
eases.113,114 In a meta-analysis of 28 studies, the Hcy levels were
significantly higher in IBD patients, compared to the controls.115
The pathophysiological mechanisms leading to vascular damage
in hyperhomocysteinemia are multifactorial, and still poorly
understood. Studies have also shown that the colonic mucosa of
patients with IBD has a higher level of Homocysteine and it has
been hypothesized that the lamina propria mononuclear cells
(LPMC) play an important role in homocysteine production.116
Chronic renal diseases
Hcy has been documented in patients with chronic renal fail-
ure (CRF), on dialysis or after a kidney transplant, at higher
concentrations than in individuals without kidney disease.117 A
study was conducted with 89 renal failure patients on dialysis,
to determine the frequency of the MTHFR gene mutation or
polymorphism and hyperhomocysteinemia. The study con-
firmed the high prevalence of hyperhomocysteinemia in
patients on dialysis, diagnosed in 76 patients (85.39%), as well
as the high incidence of the C677T and A1298C mutation, in
42 (47.19%) and 29 (32.58%) patients, respectively.118
A Clinical Approach for Hyperhomocysteinemia
It must be noted that HHCY, the biochemical hallmark of a
large group of diseases that characterized by variable presenta-
tion affecting many organs, however, the predominant associated
features are hematological and unexplained neurological signs
and symptoms.22 Predominantly, HHcy is associated with vita-
min B12 deficiency, where the measurements of metabolites,
such as methylmalonic acid (MMA) and Hcy, are more sensitive
in the diagnosis than the measurement of serum B12 levels
alone, with 98.4% with elevated serum MMA levels, and 95.9%
with elevated serum homocysteine levels in B12 deficiency
cases.119-121 The differing treatments for each genetic cause of
HHcy necessitate identifying a specific underlying causes, in
order to provide paradigmatic treatment. In suspected cases, a
careful clinical evaluation that using a variety of metabolites
including, total Hcy, PAA, Vitamin B6, Vitamin B12 levels, and
serum and urine levels of MMA rather than just Hcy will help
elucidate the cause of HHcy, should facilitate their exclusion
(Figure 2). The level of elevation and Hcy and the status of asso-
ciated metabolites will help in narrowing the diagnosis.
Combined sever HHcy with high methionine in PAA frequently
seen in CBS deficiency (classical homocystinuria), while in
methylation disorders (MAT I/III, GNMT, ADK, and SAHH),
HHcy is moderately elevated. In case if HHcy associated with
low methionine, MTHFR and MS should be considered. The
elevation of MMA is also important if associated with HHcy
because it is indicate inherited cobalamin disorders or vitamin
B12. After accurate interpretation of these metabolites the diag-
nosis can be confirmed by investigations at the levels of metabo-
lites, enzymatic studies and/or molecular genetic analysis.86,122
In several countries, C3-propionylcarnitine and methionine
is used as markers in newborn screening programs in asympto-
matic newborns, where C3-propionylcarnitine is used as a
6 Journal of Central Nervous System Disease
marker to detect patients with vitamin B12 deficiency, intracel-
lular Cbl disorders, while elevated methionine used for CBS
deficiency and methylation defects, such as MAT I/III and
GNMT.90 After making a diagnosis and initiating a treatment
plan, follow-up is important to determine the patient’s response
to therapy. In mild vitamin B12 deficiency, depending on the
underlying cause, frequent measurements of serum vitamin
B12, Hcy, and MAA levels is recommended for monitoring of
therapy.123-126
Conclusion
Hcy is now considered as a risk marker for cardiovascular and
cerebrovascular disease in addition to other modified and non-
modified individual factors. Simultaneous measurement of vita-
min B12, Hcy, MMA, and PAA is accepted as a sensitive method
of screening for several conditions associated with HHcy.
HHcy-induced inflammation could play a role in blood brain
barrier (BBB) dysfunction and the pathogenesis. Thus, the elim-
ination of excess homocysteine could be a potential therapeutic
intervention therefor may be value in preventative supplementa-
tion, especially folic acid, vitamin B12 and betaine, if the foods
indicated are not being consumed in sufficient quantities.
Authors’ contributions
FAM prepared and summarized the literature, designed the
figures and tables and wrote the manuscript.
Availability of data and materials
All data generated or analyzed during this study are included
in this published article, any additional data/files may be
obtained from the corresponding author.
ORCID iD
Fuad Al Mutairi https://orcid.org/0000-0002-7780-8863
REFERENCES
1. Kumar A, Pa lfrey HA, Pat hak R, Kadowitz PJ, Get tys TW, Murthy SN. e
metabol ism and signicance of homocysteine in nut rition and healt h. Nutr
Metab (Lond). 2017;14 :78 .
2. Skovierova H, Vidomanova E , Mahmood S, et a l. e molecula r and cel lular
eect of homocysteine metabolism imbala nce on huma n healt h. Int J Mol Sci.
2016 ;17:1733.
3. Selhub J. Homocysteine metabolism. Annu Rev Nutr. 1999;19:217-24 6.
4. Blom HJ, Smulders Y. Over view of homocy steine and folate metabolism. With
special references to ca rdiovascular disease and neural tube defects. J Inherit
Metab Dis. 2011;34:75-81.
5. Refsu m H, Smith AD, Uel and PM, et al. Fact s and recommendat ions about total
homocysteine determinations: an expert opinion. Clin Chem. 200 4;50: 3-32.
6. A lam SF, Kuma r S, Ganguly P. Measurement of homocysteine: a historica l per-
spective. J Clin Biochem Nutr. 2019; 65:17 1-177.
7. K ang SS, Wong PW, Mali now MR. Hyperhomocys t(e)inemia a s a risk factor for
occlusive vascular disease. Annu Rev Nutr. 199 2;1 2:279 -298.
8. Perl a-Kajan J, Twardowski T, Jak ubowsk i H. Mechanisms of homocy steine tox-
icity in humans. Amino Acids. 2 007;32:561-57 2.
9. McCully K S. Chemical pathology of homocysteine. I. Atherogenesis. Ann Clin
Lab Sci. 1993;23:477-493.
10. McCully KS. Vascu lar pathology of homocy steinemia: impl ications for the
pathogenesis of arteriosclerosis. Am J Pathol. 1969; 56:111-128.
11. Refsum H, Nurk E , Smith A D, et al. e Hordaland Homoc ysteine Study: a
community-based study of homocy steine, its determinants, and associat ions
with disease. J Nutr. 200 6;1 36:1731S -1740S.
12 . Ma linow MR, Bo stom AG, Krauss R M. Homocyst(e)ine , diet, and card iovascu-
lar diseases: a statement for healthcare professiona ls from t he Nutrition Com-
mittee , American Hea rt Association. Circulation. 1999;99:178-182.
13. Omenn GS, Beresford SA , Motulsky AG. Preventing coronar y heart disease: B
vitam ins and homocysteine. Circulation. 1998;97:421-424.
14. Durga J, van Boxtel MP, Schouten EG, et a l. Eect of 3-year folic acid supple-
mentation on cognitive function in older adults in the FACIT trial: a ran-
domised, double blind, cont rolled t rial. Lancet. 20 07;369:2 08-216.
15. Gang uly P, Ala m SF. Role of homocy steine in the development of c ardiovascular
disease. Nutr J. 2015;14:6 .
16. Battezzati A, Bertoli S, Sa n Romerio A, Testolin G. Body composition: an
important deter minant of homocy steine a nd methionine concentrations in
healthy indiv iduals. Nutr Metab Cardiovasc Dis. 20 07;17:525 -534 .
Figure 2. Algorithm for diagnosis of hyperhomocysteinemia.
Abbreviations: ADK, adenosine kinase; AdoHCys or SAH, S-adenosyl homocysteine; AdoMet or SAM, adenosyl methionine; Cbl, cobalamin—vitamin B12; CBS,
cystathionine beta-synthase; GNMT, glycine N-methyltransferase; Hcy, homocysteine; HHcy, hyperhomocysteinemia; MAT, methionine adenosyltransferase; MS,
methionine synthase; MTHFR, methylenetetrahydrofolate reductase; SAHH, S-adenosylhomocysteine hydrolase.
Al Mutairi 7
17. Cui S, Li W, Wang P, Lv X, Gao Y, Huang G. Folic acid inhibits homocysteine-
induced cell apoptosis in human umbilical vein endot helia l cells. Mol Cell Bio-
chem. 2018;444:77-86.
18. Curro M, Gugl iandolo A, Gangemi C, R isitano R, Ienti le R, Caccamo D. Toxic
eects of mildly elevated homocysteine concentrations in neuronal-like cells.
Neurochem Res. 2014 ;39:1485-1495.
19. E lsherbiny NM, Sh arma I, Kir a D, et al. Homocysteine induces inammation in
retina and brai n. Biomolecules. 2020;10:393.
20. Mohamed R, Sha rma I, Ibrahi m AS, et a l. Hyperhomocysteinemia alters retinal
endothel ial cells ba rrier funct ion and angiogen ic potential via activation of oxi-
dative stress. Sci Rep. 2017;7:11952 .
21. Kang SS, Rosenson R S. Analyt ic approac hes for the treatment of hyperhomo-
cysteinemia a nd its impact on vasc ular disease. Cardiovasc Drugs er. 2018;32:
233 -24 0.
22. Za ric BL, Obradov ic M, Bajic V, Haidara M A, Jovanov ic M, Isenovic ER . Homo-
cysteine and hy perhomocysteinaemia. Curr Med Chem. 2019;26:2948-2961.
23. Lonn E, Yusuf S , Arnold MJ, et a l. Homocysteine loweri ng with folic acid and B
vitam ins in vascular dis ease. N Engl J Med. 2006;354:1567-1577.
2 4. Bon aa KH, Njolsta d I, Ueland PM, et a l. Homocystei ne lowering and c ardiovas-
cula r events a fter acute myocardia l infa rction. N Engl J Med. 2006;354:
1578-1588.
25. Homocy steine Lower ing Trialists’ C ollaborati on. Lowering bl ood homocyste ine
with fol ic acid based suppl ements: meta-ana lysis of randomised tr ials. Homocy s-
teine Lowering Trialists’ Collaborat ion. BMJ. 1998;316:894-898.
26 . Ma linow MR, Nieto FJ, K ruger WD, et a l. e eects of folic acid suppl ementa-
tion on plasma total homocysteine are modulated by mult ivitamin us e and meth-
ylenetet rahyd rofolate reductase genotypes. Arterioscler romb Vasc Biol. 1997; 17:
1157-1162.
27. Malinow MR, Duell PB, Hess DL, et al. Reduction of plasma homocyst(e)ine
levels by break fast cereal for tied with fol ic acid in patients w ith coronary heart
disease. N Engl J Med. 199 8;338:100 9-1015.
28. M azza A , Cicero AF, Ramazzina E, et al. Nutr aceutic al approaches to homo-
cysteine lower ing in hy pertensive subjects at low cardiovascu lar r isk: a multi-
center, randomized clinical t rial. J Biol Regul Homeost Agents. 2 016;30:
921-927.
29. Sanchez JEA, Perez LT, Hernandez D, et al. [Ecacy a nd safety of two vitamin
supplement regimens on homocy steine le vels in hemodia lysis patients. Prospec-
tive, randomized cl inica l trial]. Nefrologia. 2005; 25:288-296.
30. Aisen PS, Schneider LS, Sa no M, et al. High-dose B vitamin supplementation
and cognitive decline in Alzheimer disease: a randomized controlled tria l.
JAMA. 2 00 8;30 0:17 74-1783.
31. Aykutoglu G, Tartik M, Darendelioglu E, Ayna A , Baydas G. Melatonin and
vitam in E alleviate homocy steine-induced ox idative injur y and apoptosis in
endothelial cells. Mol Biol Re p. 2020;47:5285-5293.
32. Mudd SH, Canton i GL. Act ivation of methionine for transmet hylation. III.
e methionine-activating enzyme of Bakers’ yeast. J Biol Chem. 1958;231:
481- 49 2.
33. Cantoni GL , Durell J. Act ivation of methion ine for transmet hylation. II. e
methion ine-ac tivat ing enz yme; studies on the mechanism of the reac tion. J Biol
Chem. 1957;225:1033-1048.
34. Chia ng PK, Cantoni GL. Activation of methionine for transmethylation. Pu ri-
cation of t he S-adenosylmethion ine synthetase of bakers’ yeast and its separa-
tion into t wo forms. J Biol Chem. 1977;252:4506-4513.
35. Scott J M, Weir DG, Molloy A , McPartli n J, Daly L, Ki rke P. Folic acid met abo-
lism and mechan isms of neu ral t ube defec ts. Ciba Found Symp.
1994;181:180-187;disc ussion 7-91.
36. Castro R, Rivera I, Blom H J, Jakobs C, Tavares de Almeida I. Homocysteine
metabol ism, hyperhomoc ysteinaemia a nd vascular disease: an overvie w. J Inherit
Metab Dis. 2006;29:3-20.
37. K im J, Kim H, Roh H , Kwon Y. Causes of hy perhomocysteinemia a nd its patho-
logica l signicance. Arch Pha rm Res. 2018;41:372-383.
38. Stir zaker C, Song JZ , Ng W, et al. Methyl-CpG-binding protein MBD2 plays a
key role in maintenance and spread of DNA methylation at CpG islands and
shores in cancer. Oncogene. 2017;36:1328-1338.
39. Blom HJ, Shaw GM, den Heijer M, F innel l RH. Neural t ube defects and folate:
case fa r from closed. Nat R ev Neurosci. 2006;7:724-731.
40. Mudd SH, Ebert MH, Scriver CR. Labi le methyl group ba lances in the human:
the role of sa rcosine. Metabolism. 1980;29:707-720.
41. Piet rzik K, Bailey L, Shane B. Folic acid and L-5-met hyltetr ahydrofolate: com-
parison of clinical pharmacokinetics and pharmacodynamics. Clin Pharmacoki-
net. 2010;4 9:535-54 8.
42. Tinelli C, Di Pino A , Ficu lle E, Marcelli S, Fel igioni M. Hyperhomocy stein-
emia as a risk factor and potent ial nutraceutical ta rget for certain patholog ies.
Front Nutr. 2019 ;6:49.
43. Ju ng M, Pfeifer GP. Aging and DNA methylation. BMC Biol. 2015;13:7.
4 4. Pajares MA, Perez-Sa la D. Betaine homoc ysteine S-methyltransferase: just a
regulator of homoc ystei ne metabolism? Cell Mol Life Sci. 2006;63:2792-2803.
45. Jirac ek J, Collins ova M, Rosenber g I, et al. S-a lkylated homo cysteine der ivatives:
new inh ibitors of human beta ine-homocysteine S-methyltransferase. J Med
Chem. 2006;49:3982-3989.
46. Zeisel SH, Blusztajn J K. Choline and human nutrition. Annu R ev Nutr.
1994;14 :2 69-2 96 .
47. West AA, Caudil l MA. Applied c holine-omics: lessons from human metabolic
studies for the integration of genomics resea rch into nutrition practice. J Acad
Nutr Diet. 2014;114:1 24 2-12 50.
48. Tran C, Bonafe L , Nuoer J M, Rieger J, Berger MM. Adult classical homoc ys-
tinur ia requiring parenteral nutrition: pit falls and management. Clin Nutr.
2018; 37:1114-1120 .
49. Locasale JW. Serine, glyc ine and one-carbon units: c ancer metabolism in full
circle. Nat Rev Cancer. 2013;13:572-583.
50. Vitv itsky V, Mosharov E, Tritt M, Ataul lakhanov F, Baner jee R. Redox reg ula-
tion of homoc ysteine-dependent glutathione synthesis. Redox Rep. 2003;8:
57-6 3.
51. Ambrosino P, Lupoli R, Di Minno A, et a l. Cyclic supplementation of 5-M THF
is eect ive for the correction of hyperhomoc ysteinemia. Nutr Res. 2 015;35:
489-495.
52. Bendini MG, Lanza GA, Mazza A, et a l. [Risk factors for card iovascula r dis-
eases: what is the role for homocy steine?]. G Ital Cardiol (Rome). 2007;8:
148 -16 0.
53. Nygard O, Refsum H, Ueland PM , Vollset SE. M ajor lifestyle determ inants of
plasma tot al homocys teine distr ibution: the Horda land Homocy steine Study. Am
J Clin Nutr. 1998;67: 263 -270.
54. Panagiotakos DB, Pitsavos C, Chrysohoou C, et al. Eect of exposure to sec-
ondhand smoke on markers of in ammation: the ATTICA study. Am J Med.
20 0 4;116:145-1 50.
55. K rajcovicov a-Kudla ckova M, Blazic ek P. [Nutritional dete rminants of homo cys-
tei nemia] . Cas L ek Cesk. 2002;141:417-420.
56. Selhub J, Jacques PF, Wilson PW, Rush D, Rosenberg IH. Vita min status and
intake as primary determinants of homocysteinemia in an elderly population.
JAMA. 1993;270:2693-2698.
57. Weber Hoss GR, Sperb-Ludwig F, Schwartz IV D, Blom HJ. Classical homo-
cyst inuria: a common i nborn er ror of metabolism? A n epidemiologica l study
based on genetic databases. Mol Genet Genomic Med. 2020;8:e1214.
58. Stenson PD, Mort M, Ball EV, et al. e Human Gene Mutation Databa se:
towards a comprehensive repository of inherited mutation data for medical
research, genetic dia gnosis a nd next-generation sequencing studies. Hum Genet.
2017;136:665-677.
59. Morr is AA, Kozic h V, Santra S, et a l. Guidelines for the dia gnosis a nd manage-
ment of cystathionine beta-synthase deciency. J Inherit Metab Dis.
2017;4 0:49 -74.
60. Dutta S, Sinha S, Chattopadhyay A , et al. Cystathionine beta-synthase
T833C/8 44INS68 polymorphism: a family-based study on mental ly reta rded
children. Behav Brain Funct. 2005;1:25.
61. Mudd SH, Finkelstein JD, I rreverre F, Laster L. Homocystinuria: an enzymatic
defect. Science. 196 4;143:14 43-1445.
62. Finkelstein J D. Methionine metabolism i n mamma ls: the biochemical basis for
homocystinuria. Metabolism. 1974;23:387-398.
6 3. Sk ovby F, Gaustadnes M , Mudd SH. A revisit to t he natural h istory of homoc ys-
tinur ia due to cystath ionine beta-synthase decienc y. Mol Genet Metab.
2010;99:1-3.
64. Ding R, Lin S, Chen D. e association of cystathionine beta synthase (CBS)
T833C polymorphism and t he risk of st roke: a meta-analysis. J Neurol Sci.
2012;312:26-30.
65. Tsai M Y, Hanson NQ , Bignell MK, Schwichtenberg K A. Simultaneous detec-
tion and screeni ng of T833C and G919A mutations of t he cystathionine beta-
synthase gene by single-strand conformationa l polymorphism. Clin Biochem.
1996;29:473-477.
66. Spera ndeo MP, de Franc his R, Andria G, Sebastio G. A 68-bp insertion found
in a homocystinuric pat ient is a common variant and is skipped by alternative
splicing of the cystathionine beta-sy nthase mRNA . Am J Hum Genet.
1996;59:1391-1393.
67. Bublil EM, Majtan T. Classical homocystinuria: from cystathionine beta-sy n-
thase decienc y to novel en zyme therapies [published online ahead of print
December 16, 2019]. Biochimie. doi:10 .1016/j.bioch i.2 019.12. 007.
6 8. M ajtan T, Pey AL, Gimene z-Mascarel l P, et al. Potential pha rmacologica l chap-
erones for cystathionine beta-sy nthase-decient homocystinuria. Handb Exp
Pharmacol. 2018;245:345-383.
69. Rosenblat t DS, Whitehead VM. Cobalamin and folate deciency: acquired and
hereditary d isorders in chi ldren. Semin Hematol. 1999; 36:19-34.
70. Froese DS, Huemer M, Suormala T, et al. Mutation update and review of severe
methylenetetrahydrofolate reductase deciency. Hum Mutat. 2016;37:427-438.
71. Ferrara M, Capozzi L , Russo R. Impac t of the MT HFR C677 T polymorphism
on risk of Wilms tumor: cas e-control study. J Pediatr Hematol Oncol.
2009;31:256-258.
8 Journal of Central Nervous System Disease
72. Horigan G, McNu lty H, Ward M, Strain JJ, Pur vis J, Scott JM. Riboavin low-
ers blood pressure in card iovascula r disease patients homoz ygous for t he
677CT polymorphism in MTHFR. J Hype rtens. 2010;28:478-486.
73. Xu J, Li K, Zhou W. Relationship between genet ic polymorphism of MTHFR
C677T and lower ex tremities deep venous thrombosis. Hematology. 2019 ;24:
108-111.
74. Ar ina CA, Amir D, S iregar Y, Sembiri ng RJ. e role of polym orphism gen met h-
ylene tetra hydrofolate reductase (MTHFR) C677T in ischaemic stroke patients
with and without hyper tension. O pen Access Maced J Med S ci. 2019;7:29-32.
75. Yakub M, Moti N, Parveen S, Chaud hry B, Azam I, Iqbal M P. Polymorphisms
in MTH FR, MS and CBS genes and homoc ysteine levels in a Pakistani popula-
tion. PLoS One. 2012;7:e33222.
76. Burda P, Schafer A, Suor mala T, et al. Insights into severe 5,10-methylenetetra-
hydrofolate reductase dec iency: molecular genetic and en zymatic cha racteriza-
tion of 76 patients. Hum Mutat. 2015;36:611-621.
77. Rommer PS, Zschocke J, Fowler B, et al. Manifestations of neu rological symp-
toms and thromboembolism in adults w ith MTHFR-dec iency. J Neurol Sci.
2017;383:123-127.
78. Li YN, Gu lati S, Baker PJ, Brody LC, Banerjee R, Kruger W D. Cloning, map-
ping and R NA analys is of the human met hionine synt hase gene. Hum Mol G enet.
1996; 5:1851-1858.
79. Galbiatti AL, Ruiz MT, Bisel li-Chicote PM, et a l. 5-Methyltetra hydrofolate-
homocysteine methyltransfera se gene polymorphism (M TR) and r isk of head
and neck cancer. Braz J Med Biol Res. 2010;43:445-450.
80. Outter yck O, de Seze J, Stojk ovic T, et al. Methion ine synthas e deciency: a r are
cause of adult-onset leukoencephalopathy. Neurology. 2012;79:386-388.
81. Carmel R, Sar rai M. Diagnosis and ma nagement of clinical a nd subclinical coba l-
amin deciency: advances and controversies. C urr Hematol Rep. 20 06;5: 23-33.
82. Atk inson C, Miouss e IR, Watkins D, Rosenblatt DS, Raiman JA . Clinical, bio-
chemical, and molecular presentation i n a patient with the cblD-homocystinuria
inborn error of coba lamin metabol ism. JIMD Rep. 2014;17:7 7-81.
83. Whitehead V M. Acquired and inherited disorders of cobala min and folate in
children. Br J Haematol. 2006;134:125-136.
84. Alfadhel M, Lillquist YP, Davis C, Junker AK, Stockler-Ipsiroglu S. Eighteen-
year fol low-up of a patient with cobala min F disease (cblF): report and review.
Am J Med Genet A. 2011;155A:2571-2577.
85. Ca rmel R. How I treat cobala min (vitamin B12) deciency. Blood.
2008;112:2214-2221.
86. Bar ic I, Staufner C, Augoustides-Sav vopoulou P, et al. Consensus recommenda-
tions for the diagnosis, treatment a nd follow-up of inherited methylation disor-
ders. J Inherit Metab D is. 2017;40:5-20.
87. Chadwick S, Fitzgerald K, Weiss B, Ficicioglu C.  irteen patients with
MAT1A mutations detected through ne wborn screening: 13 years’ e xperience.
JIM D Rep. 2014 ;14:71-76.
88. Su rtees R, Leonard J, Austin S. Association of demyelinat ion with decienc y of
cerebrospinal-uid S-adenosylmethion ine in inborn errors of methyl-transfer
pat hway. Lancet. 1991; 338:1 550-155 4.
89. Strau ss KA, Ferrei ra C, Bottigl ieri T, et al. Liver t ransplanta tion for treatment of
severe S -adenos ylhomocysteine hydrol ase deciency. Mol Ge net Metab.
2015;116:44-52.
90. Mudd SH. Hy permethioninemias of genetic a nd non-genetic orig in: a rev iew.
Am J Med Genet C Semin Med Genet. 2011;157C:3-32.
91. Fowler B. Homocysteine: overview of biochemistr y, molecular biology, and role
in disease processes. Semin Vasc Med. 2005;5:77-86.
92 . Humphrey LL, Fu R , Rogers K, Freeman M, Helfand M. Homocysteine level
and corona ry heart disease incidence: a systematic rev iew and meta-ana lysis.
Mayo Clin Proc. 2008;83:1203-1212.
93. Powers RW, Majors AK , Ceru la SL, Huber HA , Schm idt BP, Roberts J M.
Change s in markers of vascula r injur y in response to transient hyperhomocyste-
inemia. Metabolism. 20 03;52:501-507.
94. Fu Y, Wang X, Kong W. Hyperhomocy steinaemia and v ascu lar injury : advances
in mecha nisms a nd dru g targets. Br J Pharmacol. 2018;175:1173-1189.
95. Ientile R , Curro M, Ferlazzo N, Condello S, Caccamo D, Pisani F. Homocyste-
ine, vit amin determinants and neurological d iseases. Front Biosci (Schol Ed).
2010;2:359-372.
96. Alsahl i S, Al Anazi A , Al Hatlani MM, et a l. Severe Crohn’s disease ma nifesta-
tions in a child with cystathionine beta-synt hase deciency. ACG Case Rep J.
2018; 5:e93.
97. Herr mann M, Widmann T, Herrmann W. Homocysteine—a newly recognised
risk fac tor for osteoporosis. Clin Chem Lab Med. 2 005;43:1111-1117.
98 . Per na AF, Sepe I, Lanz a D, Pollastro R M, De Santo NG, Ing rosso D. Hyperho -
mocysteinemia in chronic renal failu re: alternative therapeutic strategies. J Ren
Nutr. 2012 ;22:191-194.
99. Bamashmoos SA, A l-Nuzaily MA , Al-Meeri AM, Ali F H. Relationship
betw een total homoc ysteine, tota l cholesterol and c reatinine le vels in overt hy po-
thyroid patients. Springe rplus. 2013;2:423.
100. Li D, Liu HX , Fang Y Y, et al. Hyperhomocysteinemia in polycystic ovary syn-
drome: decreased betaine-homocysteine methyltransferase and cystathionine
β-synthase-mediated homocysteine metabolism. Reprod Biomed Online.
2018;37:234-241.
101. Seshadri S, Beiser A, Selhub J, et al. Plasma homocysteine as a risk factor for
dementia a nd Alzheimer ’s disease. N Engl J Med. 2002;346:476-483.
102. Morris MS. Homocysteine and Alzheimer’s disease. Lancet Neurol.
2003;2:425-428.
103. A xer-Siegel R, Bou rla D, Ehrlich R , et al. Assoc iation of neovasc ular age-re lated
macular degeneration and hyperhomocysteinemia. Am J Ophthalmol.
2004;137:84-89.
104. Martinez-Vega R, Garrido F, Partearroyo T, et al. Folic acid deciency induces
premature hearing los s through mecha nisms involving cochlear ox idative stress
and impa irment of homocysteine metabolism. FASEB J. 2015;29:418-432.
105. Wald DS, Wald NJ, Morris JK, Law M. Folic acid, homocysteine, and cardio-
vascu lar disea se: judging cau sality in t he face of inconclu sive trial ev idence. BMJ.
2006;333:1114-1117.
106. Stuhlinger MC, Tsao PS, Her JH, Kimoto M, Bal int RF, Cooke JP. Homocys-
teine impairs the nitric oxide sy nthase pathway: role of as ymmetric dimethylar-
ginine. Circulation. 2001;104:2569-2575.
107. Fourn ier P, Fourcade J, Roncalli J, Salvayre R, Galinier M, Causse E . Homocys-
teine in chronic heart fa ilure. Clin Lab. 2015; 61:1137-1145.
108. M iller A, Mujumd ar V, Palmer L, Bower JD, Tyagi SC. Reversal of endocardial
endothel ial dysfunction by fol ic acid in homocysteinemic hyper tensive rats. Am J
Hypertens. 20 02;15:157-163.
109. Weekman EM, Woolums AE, Sudduth TL , Wilcock DM. Hyperhomoc ystein-
emia-induced gene ex pression changes in the cell types of t he brain. ASN Neuro.
2017; 9:175909141774229 6.
110. Ho PI, Ashl ine D, Dhitavat S, et a l. Folate deprivation induces neurodegenera-
tion: roles of oxidat ive stress and increased homocysteine. Neurobiol Dis.
2003;14:32-42.
111. Robe rt K, Pages C, L edru A, Dela bar J, Caboche J, Janel N. Regula tion of extra-
cellu lar signal-regulated kinase by homocysteine in hipp ocampus. Neuroscience.
2005;133:925-935.
112. Tchantchou F, Goodfellow M, Li F, et al. Hyperhomoc ysteinemia-induced oxi-
dative stress e xacerbates cortical traumatic brai n injur y outcomes in rats [pub-
lished on line a head of print May 13, 2 020]. Cell Mol Neurobiol. doi:10.100 7/
s10571-020-00866-7.
113. Papa A, De Stefano V, Danese S, et al. Hyperhomocy steinemia and prevalence
of polymorphisms of homocy steine metabolism-related enzymes in pat ients with
ina mmator y bowel disease. Am J Gastroenterol. 2001;96:2677-2682.
114. Mahmud N, Molloy A, McPart lin J, et al. Inc reased preva lence of methylenetet-
rahydrofolate reductase C677T va riant in patients with inammatory bowel dis-
ease, a nd its clinica l implications. Gut. 1999;45:389-394.
115. Oussalah A, Gueant JL, Pey rin-Biroulet L . Meta-analysis: hyperhomocy steinae-
mia in in ammatory bowel diseases. Aliment Pharmacol  er. 2011;34:1173-1184.
116. Morgenstern I, Ra ijmakers MT, Peters W H, Hoensch H, K irch W. Homocystei ne,
cysteine, and glutathione in huma n colonic mucosa: ele vated levels of homocysteine
in patients with inam matory bowel disease. Dig Dis Sc i. 2003;48:2083-2090.
117. Sunder-Plassmann G, Winkelmayer WC, Fodinger M. Genetic aspec ts of
hyperhomocysteinemia in chronic kid ney disease. Semin Nephrol. 2006;26:8-13.
118. Domenici FA, Vannucc hi MT, Simoes-Ambrosio L M, Vannucch i H. Hyperho-
mocysteinemia and poly morphisms of the methylenetetrahydrofolate gene in
hemodialysis and peritoneal dialysis patients. Mol Nutr Food Res.
20 07; 51:14 30 -1436 .
119. Lindenbaum J, Savage DG, Stabler SP, Al len RH. Diagnosis of coba lamin de-
ciency : II. Relative sensitivities of serum cobala min, methylmalonic acid, and
total homocy steine concentrations. Am J Hematol. 1990;34:99-107.
120. Sumner AE, Chin MM, Abrahm JL, et al. Elevated methylmalonic ac id and
total homocysteine levels show high preva lence of vitamin B12 deciency after
gastric surgery. Ann Intern Med. 19 96;124:469-476.
121. Savage DG, Lindenbaum J, Stabler SP, Allen R H. Sensitivity of ser um methyl-
malonic acid and total homocysteine determinations for diagnosing cobalamin
and folate deciencies. Am J Med. 1994;96:239-246.
122. Huemer M, Diod ato D, Schwahn B, et al. Guidelines for diagnosis and manag e-
ment of the cobalam in-related remethylation disorders cblC, cblD, cblE, cblF,
cblG, cblJ and MTHFR deciency. J Inherit Metab Dis. 2017;40:21-48.
123. Huemer M, Burer C, Jesina P, et al. Clinical onset and course, response to treat-
ment and outcome in 24 patients wit h the cblE or cblG remethylation defect
complemented by genetic and in vitro enzyme study data. J Inherit Metab Dis.
2015;38:957-967.
124. Fisch er S, Huemer M, Baum gartner M , et al. Clini cal presentat ion and outcome in
a series of 88 patients with the cblC defect. J Inher it Metab Dis. 2 014;37: 831-84 0.
125. K nowles L , Morris AA, Walter JH. Treatment with mefolinate (5-methyltetra-
hydrofolate), but not folic acid or folinic acid, leads to measurable 5-methyltet ra-
hydrofolate in cerebrospina l uid in methylenetetra hydrofolate reductase
de cienc y. JIMD Rep. 2016;29:103-107.
1 26. Sch i M, Benoist J F, Tilea B, R oyer N, Giraudi er S, Ogier de Bau lny H. Isolated
remethylation disorders: do our treatments benet patients? J Inherit Metab Dis.
2011;34:137-145.
... 6 Normally plasma total Hcy (tHcy) level is 5-15 μmol·L −1 , but under hyperhomocysteinemia (HHcy) its value rises. 7,8 Factors that lead to HHcy include hereditary defects in Hcy metabolism, vitamins B6, B9, and B12 deficiency, chronic alcohol consumption, smoking, and taking certain medications. 7 Plasma tHcy also increases with aging through age-related vitamin deficiency and/or decline in renal function. ...
... 7,8 Factors that lead to HHcy include hereditary defects in Hcy metabolism, vitamins B6, B9, and B12 deficiency, chronic alcohol consumption, smoking, and taking certain medications. 7 Plasma tHcy also increases with aging through age-related vitamin deficiency and/or decline in renal function. 9,10 An average of 5-10% of individuals in the population have mild to moderate HHcy (15-40 μmol·L −1 ). ...
Preprint
Full-text available
Hyperhomocysteinemia (HHcy) is now being actively studied as a potential risk factor and/or biomarker for numerous pathological conditions, including brain diseases. This study aimed to analyze the proteolytic processes in the brains of rats with HHcy. Total proteolytic activity, metal-dependent, and serine proteases activities, the content of matrix metalloproteinases (MMPs), tissue inhibitor of metalloproteinases-1, cytokines, serine proteases, total protein and medium and low molecular-weight substances (MLMWS), were evaluated. HHcy was induced by DL-homocysteine thiolactone (HTL) daily intragastric administration (200 mg·kg–1 of body weight) to young and adult albino non-linear male rats for 8 weeks following rat sacrifice and brain harvesting. It was established that HHcy causes an increase in total proteolytic activity and a rise in MLMWS levels in rat brains. Serine protease activity increased to a greater extent compared to metal-dependent one, and bigger changes were observed in young rats. Rise in MMP-9 and -10 levels (in young animals), a decline in MMP-3 and -8 levels, and a decrease in the content of interleukin-1β, interferon-γ, interleukin-4 and tumor necrosis factor-α (the last two in young animals) was also detected. No significant changes were found in serine protease content. Therefore, proteolysis intensification in the brain of rats with HHcy is more likely caused by protease up-regulation through mechanisms stimulated by homocysteine, HTL, and oxidative stress, without involving pro-inflammatory signaling pathways.
... Vitamins B 6 , B 9 and B 12 are involved in homocysteine metabolism as enzymatic cofactors [13]. HHcyt is widely considered as a risk factor of vascular diseases but suspect to be associated with neurodegeneration and neurocognitive impairment, inflammatory diseases, osteoporosis [13,14]. A variety of congenital and acquired conditions lead to HHcyt, including inherited metabolic diseases of Hcyt catabolism, cofactor deficiencies and kidney or liver failure [15]. ...
... Our laboratory's reference values for homocysteinemia are between 4.40 and 13.6 μmol/L in women. Classically, HHcyt is considered mild when the plasma Hcyt level is in the 16-30 μmol/L range, moderate between 30 and 50 μmol/ L, intermediate between 50 and 100 μmol/L and severe above of 100 μmol/L [13][14][15]. Amino acids were determined in plasma samples by LC-MS/MS after AccQ-Tag derivatization using commercial kit (AccQ. ...
Article
Full-text available
Acute hepatic porphyrias are inherited metabolic disorders of heme biosynthesis characterized by the accumulation of toxic intermediate metabolites responsible for disabling acute neurovisceral attacks. Givosiran is a newly approved siRNA-based treatment of acute hepatic porphyria targeting the first and rate-limiting δ-aminolevulinic acid synthase 1 (ALAS1) enzyme of heme biosynthetic pathway. We described a 72-year old patient who presented with severe inaugural neurological form of acute intermittent porphyria evolving for several years which made her eligible for givosiran administration. On initiation of treatment, the patient developed a major hyperhomocysteinemia (>400 μmol/L) which necessitated to discontinue the siRNA-based therapy. A thorough metabolic analysis in the patient suggests that hyperhomocysteinemia could be attributed to a functional deficiency of cystathionine β-synthase (CBS) enzyme induced by givosiran. Long-term treatment with vitamin B6, a cofactor of CBS, allowed to normalize homocysteinemia while givosiran treatment was maintained. We review the recently published cases of hyperhomocysteinemia in acute hepatic porphyria and its exacerbation under givosiran therapy. We also discuss the benefits of vitamin B6 supplementation in the light of hypothetic pathophysiological mechanisms responsible for hyperhomocysteinemia in these patients. Our results confirmed the importance of monitoring homocysteine metabolism and vitamin status in patients with acute intermittent porphyria in order to improve management by appropriate vitamin supplementation during givosiran treatment.
... Loss of function mutations in MTHFR cause elevation of homocysteine levels in the body, oxidative stress, and dysregulation of methionine synthesis, since the MTHFR product is required for the conversion of homocysteine to methionine [7,8]. Hyperhomocysteinemia is associated with a multitude of clinical conditions affecting various organ systems, including cardiovascular disease, stroke, venous thrombosis, epilepsy, cognitive impairment, Alzheimer's disease, Parkinson's disease, multiple sclerosis, osteoporosis, inflammatory bowel disease, diabetes, PCOS, infertility, and chronic renal failure [9][10][11]. Two MTHFR point mutations, C677T and A1298C, are common in the American population and reduce the efficiency of MTHFR [11]. ...
Article
Full-text available
Methylenetetrahydrofolate reductase (MTHFR) is key to the metabolism of folic acid, with loss of function mutations resulting in elevated homocysteine levels, a known risk factor for cardiovascular disease. Psoriasis patients may demonstrate hyperhomocysteinemia. To assess for the association between psoriasis and MTHFR C677T and A1298C polymorphisms. A systematic literature search was conducted in MEDLINE, Embase, Cochrane CENTRAL, and Web of Science. Case reports, case–control, cohort, and cross-sectional studies with full-text availability in English were considered. Meta-analysis was conducted with pooled ORs calculated via the random effects model (I2 > 50%). Of 917 records identified, 10 studies were selected for review of 1965 psoriasis patients and 2030 controls. Meta-analysis demonstrated that for MTHFR C677T, there were positive associations between psoriasis and the allele contrast model (C vs T, pooled OR = 1.69, 95% CI = 1.10–2.59), the additive model (CC vs TT, pooled OR = 2.44, 95% CI = 1.06–5.60), the dominant model (CC vs CT + TT, pooled OR = 1.77, 95% CI = 1.06–2.98), and the recessive model (CC + CT vs TT, pooled OR = 2.08, 95% CI = 1.05–4.13). For MTHFR A1298C, there were positive associations between psoriasis and the allele contrast model (A vs C, pooled OR = 3.57, 95% CI = 1.19–10.68), the dominant model (AA vs AC + CC, pooled OR = 4.44, 95% CI = 1.12–17.66), and the overdominant model (AC vs AA + CC, pooled OR = 0.26, 95% CI = 0.07–0.91). There may be a link between the C677T and A1298C polymorphisms with psoriasis diagnosis.
... Hyperhomocysteinemia results from reduced folate levels or changes in the enzymes involved in the folate pathway. The presence of autosomal dominant and recessive inheritance patterns in epilepsy exemplifies the intricate nature of the condition, indicating that multiple genes and non-genetic variables play a role in its heritability (Al Mutairi, 2020). Seizures, involuntary movements of the limbs, transient episodes of bewilderment, and various deviations in behavior and emotions characterize epilepsy. ...
Article
Full-text available
Epilepsy is a prominent neurological disorder caused by a range of factors, including epilepsy-associated genes, hereditary variables, environmental factors such as oxidative stress and depression, and inflammatory molecules that influence it. Worldwide, more than 65 million individuals are afflicted by epilepsy. The majority of individuals with epilepsy live in nations with low to middle-income levels. In Pakistan, the incidence of epilepsy is at 10 cases per 1000 inhabitants. There are distinct classifications of epilepsy based on the frequency of seizures, which include generalized epilepsy, localized epilepsy, and epilepsy of uncertain origin. Based on the etiology of epilepsy, it is well acknowledged that this condition is characterized by a highly active network that originates from ionic transmission. Brain injury, including traumatic and ischemic injuries, leads to the production of inflammatory chemicals. The excessive production of inflammatory mediators leads to the impairment of the blood-brain barrier (BBB), which induces inflammation in both the central and peripheral regions, leading to epileptic diseases. Over a thousand genes are thought to be involved in developing epilepsy; the most extensively researched genes comprise GABRG2, SCN, CACN, KCN1A, MTHFR, MTTL1, and EFHC1 gene. Various therapeutic approaches have been devised to treat epilepsy, including neurosurgical interventions, antiepileptic medications, anticonvulsant pharmaceuticals, ketogenic dietary regimens, and herbal remedies. This review article provides a thorough analysis of epilepsy, encompassing its categorization, the inflammatory agents accountable for its onset, the genetic factors linked to its progression, and the current therapeutic approaches for this disease.
... In the present study, the assessment of serum homocysteine and CoQ10 levels was conducted to monitor health status and highlight the significance of age-related considerations within the framework of occupational health. Hyperhomocysteinemia is defined as increased homocysteine levels above 15 µmol/L, which is associated with several diseases, and predominantly an increased risk of cardiovascular disorders [40,53,54]. In the total sample, 9% of participants exhibited hyperhomocysteinemia, while 39% of participants fell within the borderline risk range (with blood homocysteine levels between 10 and 15 µmol/L). ...
Article
Full-text available
Background: This study aimed to evaluate age-specific variations in the blood levels of micronutrients, homocysteine, and CoQ10, along with physical activity (PA) patterns, among 123 Austrian adult bankers in operational and frontline roles (mean age: 43 years; 50% female). Methods: Blood analysis was conducted to assess micronutrients and the serum concentrations of homocysteine and CoQ10. The micronutrient values in whole blood were compared to sex-specific reference ranges and categorized as below, within, or above them. The Global Physical Activity Questionnaire was utilized to assess PA patterns. Participants were classified as young adults (18–34 years), middle-aged adults (35–49 years), and older adults (50–64 years). Results: Significant age-based differences were found in participants’ mean homocysteine levels (p = 0.039) and homocysteine categories (p = 0.034), indicating an increasing prevalence of hyperhomocysteinemia with age. No significant difference between age categories was observed for sex, BMI, diet types, PA levels, sedentary behavior, and CoQ10 (p > 0.05). There was no significant age-based difference in the blood concentrations of most minerals and vitamins (p > 0.05), except for magnesium among females (p = 0.008) and copper among males (p = 0.042). Conclusion: The findings offer initial evidence of the age-related differences in the health status of adult bankers, providing insights for customized approaches to occupational health that support the importance of metabolic health and overall well-being across adulthood.
Article
Full-text available
This study investigates the distribution of hyperhomocysteinemia and cardiovascular metabolic syndrome (SM) among participants, shedding light on their prevalence and co-occurrence within the study cohort. Through an analysis of demographic characteristics and health parameters, including age, gender, and body mass index (BMI), alongside nutritional data, correlations between these factors and health risks are explored. Results reveal a notable prevalence of hyperhomocysteinemia, with 45.3% of participants exhibiting this condition. Furthermore, 31.4% of the cohort does not present hyperhomocysteinemia or SM, while 23.3% shows SM without hyperhomocysteinemia. The study underscores gender-specific dietary recommendations due to significant variations in nutrient intake patterns. Additionally, inverse correlations between health risks like obesity, hypertension, and hypercholesterolemia and nutrient requirements highlight the need for tailored dietary interventions. Age-related changes in nutrient needs and the positive correlation between physical activity levels and certain nutrient demands further emphasize the importance of personalized dietary strategies. Variations in nutrient intake by gender, inverse correlations with health risks, and age-related changes underscore the need for tailored dietary strategies. These findings provide valuable insights for healthcare professionals in developing targeted nutritional interventions to mitigate disease risk and promote overall health and well-being.
Article
We undertook this review to determine if it is plausible that choline or phosphatidylcholine (PC) deficiency is a factor in necrotizing enterocolitis (NEC) after two clinical trials found a dramatic and unexpected reduction in NEC in an experimental group provided higher PC compared to a control group. Sources and amounts of choline/PC for preterm infants are compared to the choline status of preterm infants at birth and following conventional nutritional management. The roles of choline/PC in intestinal structure, mucus, mesenteric blood flow, and the cholinergic anti-inflammatory system are summarized. Low choline/PC status is linked to prematurity/immaturity, parenteral and enteral feeding, microbial dysbiosis and hypoxia/ischemia, factors long associated with the risk of developing NEC. We conclude that low choline status exists in preterm infants provided conventional parenteral and enteral nutritional management, and that it is plausible low choline/PC status adversely affects intestinal function to set up the vicious cycle of inflammation, loss of intestinal barrier function and worsening tissue hypoxia that occurs with NEC. In conclusion, this review supports the need for randomized clinical trials to test the hypothesis that additional choline or PC provided parenterally or enterally can reduce the incidence of NEC in preterm infants. IMPACT STATEMENT: Low choline status in preterm infants who are managed by conventional nutrition is plausibly linked to the risk of developing necrotizing enterocolitis.
Article
Full-text available
Background Timely diagnosis is the key factor to improve the prognosis of endometrial carcinoma (EC). To date, no particularly good markers could significantly improve the detection rate of EC. This study aimed to assess the utility of serum markers homocysteine (Hcy), human epididymal protein 4 (HE4), cancer antigen 199 (CA199), cancer antigen 125 (CA125), fibrinogen (Fib), and D-dimer (D-D) for EC diagnosis, especially Hcy of which its role in EC has not been noticed. Methods Pre-test and verification tests were performed. In Pre-test, the diagnostic value of the included markers was evaluated and the right marker was chosen to establish an efficient new risk index for screening EC. In verification tests, the applicability of the new risk index was tested. Several evaluation indices including receiver operating characteristic (ROC) curve, Youden Index, sensitivity (SN), and specificity (SP), were adopted to assess the diagnostic value of the included markers for EC. Results Hcy may be useful in the diagnosis of EC. Its diagnostic value was not significantly lower than that of HE4. Based on the diagnostic value of Hcy and HE4, a new risk index was established, which demonstrated high value in EC diagnosis (ROC, 0.801), especially among young female patients (age ≤50 years, ROC, 0.871). Furthermore, the level of Hcy, but not HE4, was notably different in normal or benign endometrial lesions, atypical endometrial hyperplasia (AEH), and EC. Conclusions The change of Hcy levels could be used to diagnose EC and when taken into consideration together with the detection of HE4, the diagnostic accuracy of EC is further improved.
Article
Full-text available
A relationship exists between hyperhomocysteinemia and cardiovascular diseases, although the underlying mechanisms are still incompletely defined. One possibility involves a homocysteine (Hcy)-induced increased oxidative stress. Melatonin (Mel) and vitamin E (vitE) are important anti-oxidants. The main purpose of this study was (1) to compare the effect of treatments with Mel, vitE or both, on Hcy-induced apoptosis in human umbilical vein endothelial cells (HUVECs), and (2) to investigate the underlying mechanisms. Cell proliferation assay was carried out by Water Soluble Tetrazolium-1 (WST-1) assay kit. Apoptotic index was calculated by TUNEL Assay. Anti-oxidant parameters were studied by measurement of reactive oxygen species (ROS) and lipid peroxidation (LPO) levels. mRNA and protein expression levels of apoptotic and anti-apoptotic genes and proteins were studied by quantitative real time polymerase chain reaction (qRT-PCR) and Western blotting experiments respectively. The results showed that treatments with Mel, vitE or Mel + vitE suppressed Hcy-induced cell death, with a higher efficiency for the Mel and Mel + vitE treatments. Our results suggests that the mechanisms by which these anti-oxidants protected endothelial cells include the decrease in ROS and LPO levels, an increase in cell migration, the downregulation of pro-apoptotic proteins Cas 3, Cas 9, Cyt C and Bax and the upregulation of anti-apoptotic protein Bcl 2. Collectively, these results revealed the protective role of vitE and Mel against Hcy-induced cell apoptosis, which may add insight into therapeutic approaches to Hcy-induced damages.
Article
Full-text available
Traumatic brain injury (TBI) is a leading cause of morbidity and mortality among military service members and civilians in the United States. Despite significant advances in the understanding of TBI pathophysiology, several clinical reports indicate that multiple genetic and epigenetic factors can influence outcome. Homocysteine (HCY) is a non-proteinogenic amino acid, the catabolism of which can be dysregulated by stress, lifestyle, aging, or genetic abnormalities leading to hyperhomocysteinemia (HHCY). HHCY is a neurotoxic condition and a risk factor for multiple neurological and cardiovascular disorders that occurs when HCY levels is clinically > 15 µM. Although the deleterious impact of HHCY has been studied in human and animal models of neurological disorders such as stroke, Alzheimer’s disease and Parkinson's disease, it has not been addressed in TBI models. This study tested the hypothesis that HHCY has detrimental effects on TBI pathophysiology. Moderate HHCY was induced in adult male Sprague Dawley rats via daily administration of methionine followed by impact-induced traumatic brain injury. In this model, HHCY increased oxidative stress, upregulated expression of proteins that promote blood coagulation, exacerbated TBI-associated blood–brain barrier dysfunction and promoted the infiltration of inflammatory cells into the cortex. We also observed an increase of brain injury-induced lesion size and aggravated anxiety-like behavior. These findings show that moderate HHCY exacerbates TBI outcomes and suggest that HCY catabolic dysregulation may be a significant biological variable that could contribute to TBI pathophysiology heterogeneity.
Article
Full-text available
Background Biallelic pathogenic variants in CBS gene cause the most common form of homocystinuria, the classical homocystinuria (HCU). The worldwide prevalence of HCU is estimated to be 0.82:100,000 [95% CI, 0.39–1.73:100,000] according to clinical records and 1.09:100,000 [95% CI, 0.34–3.55:100,000] by neonatal screening. In this study, we aimed to estimate the minimal worldwide incidence of HCU. Methods The 25 most common pathogenic alleles of HCU were identified through a literature review. The incidence of HCU was estimated based on the frequency of these common pathogenic alleles in a large genomic database (gnomAD). Results The minimum worldwide incidence of HCU was estimated to be ~0.38:100,000, and the incidence was higher in Europeans non‐Finnish (~0.72:100,000) and Latin Americans (~0.45:100,000) and lower in Africans (~0.20:100,000) and Asians (~0.02:100,000). Conclusion Our data are in accordance with the only published metanalysis on this topic. To our surprise, the observed incidence of HCU in Europeans was much lower than those described in articles exploring small populations from northern Europe but was similar to the incidence described on the basis of neonatal screening programs. In our opinion, this large dataset analyzed and its population coverage gave us greater precision in the estimation of incidence.
Article
Full-text available
Homocysteine (Hcy) is an amino acid that requires vitamins B12 and folic acid for its metabolism. Vitamins B12 and folic acid deficiencies lead to hyperhomocysteinemia (HHcy, elevated Hcy), which is linked to the development of diabetic retinopathy (DR), age-related macular degeneration (AMD), and Alzheimer’s disease (AD). The goal of the current study was to explore inflammation as an underlying mechanism of HHcy-induced pathology in age related diseases such as AMD, DR, and AD. Mice with HHcy due to a lack of the enzyme cystathionine-β-synthase (CBS) and wild-type mice were evaluated for microglia activation and inflammatory markers using immuno-fluorescence (IF). Tissue lysates isolated from the brain hippocampal area from mice with HHcy were evaluated for inflammatory cytokines using the multiplex assay. Human retinal endothelial cells, retinal pigment epithelial cells, and monocyte cell lines treated with/without Hcy were evaluated for inflammatory cytokines and NFκB activation using the multiplex assay, western blot analysis, and IF. HHcy induced inflammatory responses in mouse brain, retina, cultured retinal, and microglial cells. NFκB was activated and cytokine array analysis showed marked increase in pro-inflammatory cytokines and downregulation of anti-inflammatory cytokines. Therefore, elimination of excess Hcy or reduction of inflammation is a promising intervention for mitigating damage associated with HHcy in aging diseases such as DR, AMD, and AD.
Article
Full-text available
Elevated plasma level of homocysteine is being increasingly associated with many diseases. There is a significant interest in the development of methods to determine the total homocysteine in biologically relevant tissues. Over the years, researchers use various methods to determine the exact concentrations of homocysteine in these tissues. However, the precise method used in many studies earlier was questionable. We have reviewed various methodologies for the measurement of homocysteine. We list the commonly used methodologies currently in use to determine homocysteine levels. Through extensive literature search, we have come up with the most popular as well as the newest measurement modalities and listed them with a brief discussion of each of the methodology. In conclusion, we have presented the historical perspective of homocysteine measurement in biological fluids in this manuscript. Thus, the precise understanding of its concentration in biological fluids coupled with its importance in health and disease should justify a newer but reliable technique in the area of ongoing research in homocysteine.
Article
Full-text available
Hyperhomocysteinemia is recognized as a risk factor for several diseases, including cardiovascular and neurological conditions. Homocysteine (HCys) is a key metabolite involved in the biosynthesis and metabolism of methionine (Met), which plays a pivotal role in the physiological cell's life cycle. The biochemistry of Met is finely regulated by several enzymes that control HCys concentration. Indeed, balanced activity among the enzymes is essential for the cell's well-being, while its malfunction could raise HCys concentration which can lead to the onset of several pathological conditions. The HCys concentration increase seems to be caused mainly by the widely diffused polymorphisms of several enzymes. Nowadays, a blood test can easily detect elevated concentrations of HCys, referred to as Hyperhomocysteinemia (HHCys). Prolonged exposure to this condition can lead to the onset of cardiovascular disease and can lead to the development of atherosclerosis, stroke, inflammatory syndromes like osteoporosis and rheumatism, as well as neuronal pathologies including Alzheimer's and Parkinson's diseases. In this review, we analyzed the literature of several pathological conditions in which the molecular pathways of HHCys are involved. Interestingly, several observations indicate that the calibrated assumption of correct doses of vitamins such as folic acid, vitamin B6, vitamin B12, and betaine may control HHCys-related conditions.
Article
Genetic defects in cystathionine beta-synthase (CBS), a key enzyme of organic sulfur metabolism, result in deficiency of CBS activity and a rare inborn error of metabolism called classical homocystinuria (HCU). HCU is characterized by massive accumulation of homocysteine, an intermediate of methionine metabolism, and multisystemic clinical symptoms. Current treatment options for HCU are very limited and often inefficient, partially due to a low patient compliance with very strict dietary regimen. Novel therapeutic approaches are needed to cope with the toxic accumulation of homocysteine and restoration of a healthy metabolic balance. Human CBS is a complex intracellular multimeric enzyme that relies on three cofactors (heme, pyridoxal-5'-phosphate and S-adenosylmethionine) for proper function. Engineering and chemical modification of human CBS yielded OT-58, a first-in-class enzyme therapy candidate for HCU. Pre-clinical testing of OT-58 showed its substantial efficacy in lowering plasma and tissue concentrations of homocysteine, improving metabolic balance and correcting clinical symptoms of HCU. In addition, OT-58 showed great safety and toxicity profile when administered to non-human primates. Overwhelmingly positive and extensive pre-clinical package propelled OT-58 into a first-in-human clinical trial, which started on January 2019. In a meantime, other enzyme therapies based on modified human cystathionine gamma-lyase or erythrocyte-encapsulated bacterial methionine gamma-lyase have shown efficacy in decreasing plasma homocysteine in HCU mice. In addition, gene therapy approaches using adenovirus or minicircle DNA have been evaluated in HCU. In this review, we summarize the current efforts developing novel therapies for HCU to address a high unmet medical need among HCU patients.