homocysteine induces cytotoxicity and proliferation inhibition in neural stem cells via dna...

9
Homocysteine induces cytotoxicity and proliferation inhibition in neural stem cells via DNA methylation in vitro Ningning Lin, Shanchun Qin, Suhui Luo, Shanshan Cui, Guowei Huang and Xumei Zhang Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, China Keywords DNA methylation; DNA methyltransferases; homocysteine; neural stem cells; proliferation Correspondence G. Huang and X. Zhang, Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China Fax: +86 22 83336606 Tel: +86 22 83336606; +86 22 83336615 E-mails: [email protected]; [email protected] (Received 21 October 2013, revised 12 January 2014, accepted 20 February 2014) doi:10.1111/febs.12764 Mild to moderate hyperhomocysteinemia has been implicated in neurode- velopmental disorders and neurodegenerative diseases in human studies. Although the molecular mechanisms underlying the effects of homocysteine (Hcy) neurotoxicity on the nervous system are not yet fully understood, inhibition of neural stem cell (NSC) proliferation and alterations in DNA methylation may be involved. The aim of the present study was to charac- terize the effects of Hcy on DNA methylation in NSCs, and to explore how Hcy-induced changes in DNA methylation patterns affect NSC prolif- eration. We found that D,L-Hcy (301000 lM) but not L-cysteine inhibited cell proliferation and reduced levels of global DNA methylation in NSCs from neonatal rat hippocampus and increased cell injury. High levels of Hcy also induced an increase in S-adenosylhomocysteine (SAH), a decrease in the ratio of S-adenosylmethionine (SAM) to SAH, and a reduction in protein expression of the DNA methyltransferases DNMT1, DNMT3a and DNMT3b and their enzymatic activity. Moreover, the DNMT inhibitor zebularine reduced the global DNA methylation level and inhibited NSC proliferation. Our results suggest that alterations in DNA methylation may be an important mechanism by which high levels of Hcy inhibit NSC via- bility in vitro. Hcy-induced DNA hypomethylation may be caused by a reduction in the DNMT activity which is regulated by the cellular concen- trations of SAM and SAH, or their protein expression levels. Our results also suggest that Hcy may play a role in the pathogenesis of certain ner- vous system diseases via a molecular mechanism that involves negative reg- ulation of NSC proliferation and alterations in DNA methylation. Introduction Epidemiological studies show a positive, dose-depen- dent relationship between mild to moderate increases in plasma total homocysteine (Hcy) concentrations and the risk of neurodegenerative diseases such as Alzhei- mer’s disease, vascular dementia, cognitive impairment and stroke [1,2]. Increased maternal Hcy is also associ- ated with neural tube defects. Several placebo-con- trolled trials have shown that therapies that lower Hcy levels reduce the frequency of neural tube defects in both women who have a history of such pregnancies and women who do not [3]. Furthermore, a cross-sec- tional MRI study showed that elevated Hcy levels are associated with more hippocampal and cortical atrophy in healthy elderly individuals [4]. Hcy may increase the permeability of the blood-brain barrier suggesting that functional disturbances of the bloodbrain barrier may lead to increased exposure of the brain to Hcy and the neurotoxic effects of hyperhomocysteinemia [5]. How- Abbreviations BrdU, 5 0 -bromo-2 0 -deoxyuridine; DNMT, DNA methyltransferase; ERK1/2, Rxtracellular signal-regulated kinase 1/2; GFAP, glial fibrillary acidic protein; Hcy, homocysteine; LDH, lactate dehydrogenase; NSC, neural stem cell; SAH, S-adenosylhomocysteine; SAM, S-adenosylmethionine; Sox 2, SRY (sex-determining region Y)-box 2. 2088 FEBS Journal 281 (2014) 2088–2096 ª 2014 FEBS

Upload: xumei

Post on 16-Feb-2017

212 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Homocysteine induces cytotoxicity and proliferation inhibition in neural stem cells via DNA methylation               in vitro

Homocysteine induces cytotoxicity and proliferationinhibition in neural stem cells via DNA methylation in vitroNingning Lin, Shanchun Qin, Suhui Luo, Shanshan Cui, Guowei Huang and Xumei Zhang

Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, China

Keywords

DNA methylation; DNA methyltransferases;

homocysteine; neural stem cells;

proliferation

Correspondence

G. Huang and X. Zhang, Department of

Nutrition and Food Science, School of Public

Health, Tianjin Medical University, Tianjin

300070, China

Fax: +86 22 83336606

Tel: +86 22 83336606; +86 22 83336615

E-mails: [email protected];

[email protected]

(Received 21 October 2013, revised 12

January 2014, accepted 20 February 2014)

doi:10.1111/febs.12764

Mild to moderate hyperhomocysteinemia has been implicated in neurode-

velopmental disorders and neurodegenerative diseases in human studies.

Although the molecular mechanisms underlying the effects of homocysteine

(Hcy) neurotoxicity on the nervous system are not yet fully understood,

inhibition of neural stem cell (NSC) proliferation and alterations in DNA

methylation may be involved. The aim of the present study was to charac-

terize the effects of Hcy on DNA methylation in NSCs, and to explore

how Hcy-induced changes in DNA methylation patterns affect NSC prolif-

eration. We found that D,L-Hcy (30–1000 lM) but not L-cysteine inhibited

cell proliferation and reduced levels of global DNA methylation in NSCs

from neonatal rat hippocampus and increased cell injury. High levels of

Hcy also induced an increase in S-adenosylhomocysteine (SAH), a decrease

in the ratio of S-adenosylmethionine (SAM) to SAH, and a reduction in

protein expression of the DNA methyltransferases DNMT1, DNMT3a and

DNMT3b and their enzymatic activity. Moreover, the DNMT inhibitor

zebularine reduced the global DNA methylation level and inhibited NSC

proliferation. Our results suggest that alterations in DNA methylation may

be an important mechanism by which high levels of Hcy inhibit NSC via-

bility in vitro. Hcy-induced DNA hypomethylation may be caused by a

reduction in the DNMT activity which is regulated by the cellular concen-

trations of SAM and SAH, or their protein expression levels. Our results

also suggest that Hcy may play a role in the pathogenesis of certain ner-

vous system diseases via a molecular mechanism that involves negative reg-

ulation of NSC proliferation and alterations in DNA methylation.

Introduction

Epidemiological studies show a positive, dose-depen-

dent relationship between mild to moderate increases in

plasma total homocysteine (Hcy) concentrations and

the risk of neurodegenerative diseases such as Alzhei-

mer’s disease, vascular dementia, cognitive impairment

and stroke [1,2]. Increased maternal Hcy is also associ-

ated with neural tube defects. Several placebo-con-

trolled trials have shown that therapies that lower Hcy

levels reduce the frequency of neural tube defects in

both women who have a history of such pregnancies

and women who do not [3]. Furthermore, a cross-sec-

tional MRI study showed that elevated Hcy levels are

associated with more hippocampal and cortical atrophy

in healthy elderly individuals [4]. Hcy may increase the

permeability of the blood-brain barrier suggesting that

functional disturbances of the blood–brain barrier may

lead to increased exposure of the brain to Hcy and the

neurotoxic effects of hyperhomocysteinemia [5]. How-

Abbreviations

BrdU, 50-bromo-20-deoxyuridine; DNMT, DNA methyltransferase; ERK1/2, Rxtracellular signal-regulated kinase 1/2; GFAP, glial fibrillary acidic

protein; Hcy, homocysteine; LDH, lactate dehydrogenase; NSC, neural stem cell; SAH, S-adenosylhomocysteine; SAM,

S-adenosylmethionine; Sox 2, SRY (sex-determining region Y)-box 2.

2088 FEBS Journal 281 (2014) 2088–2096 ª 2014 FEBS

Page 2: Homocysteine induces cytotoxicity and proliferation inhibition in neural stem cells via DNA methylation               in vitro

ever, the exact mechanism for direct neurotoxic effects

of Hcy has yet to be elucidated.

Self-renewing neural stem cells (NSCs) that are capa-

ble of differentiating into neurons, astrocytes and oligo-

dendrocytes play a crucial role in the developing brain

as well as the adult brain. NSCs have been the subject

of intense study in recent years with regard to their

therapeutic potential in treating neurodegenerative dis-

eases and traumatic injuries of the nervous system.

Most studies investigating the consequences of Hcy tox-

icity have mainly focused on injury to mature neurons.

Elevated levels of Hcy exert a variety of adverse effects

on neural cells, including oxidative stress, activation of

caspases, mitochondrial dysfunction, and an increase in

cytosolic calcium [6,7]. As NSCs are necessary for brain

development and neural repair, elevated Hcy may cause

brain injury by inhibiting neurogenesis through its

effects on NSCs. Only few experimental data are avail-

able on Hcy neurotoxicity which may be involved in

neuronal progenitor cell proliferation. Rabaneda et al.

[8] found evidence in vitro and in vivo suggesting that

homocysteine inhibits the proliferation of neuronal pre-

cursors in the sub-ventricular zone by impairing basic

fibroblast growth factor-induced proliferation. A recent

study showed that Hcy decreases Extracellular signal-

regulated kinase 1/2 (ERK1/2) protein phosphorylation

and NSC proliferation in fetal rat brain in vitro [9].

However, evidence for the direct effect of Hcy on NSC

viability has been lacking. Here, we examined the

effects of elevated Hcy on proliferation and viability of

NSCs from neonatal rat hippocampus, and explored

the molecular mechanisms of neural cell toxicity

induced by Hcy.

Previous studies have demonstrated an essential role

for epigenetics in NSC development, and that inhibiting

DNA methylation prevents NSC differentiation [10,11].

Hcy in the body is solely of metabolic origin, ultimately

derived from metabolic cycling of methionine. Methio-

nine is converted through S-adenosylmethionine (SAM)

and S-adenosylhomocysteine (SAH) to Hcy. SAM and

SAH are metabolites of Hcy. SAM is the substrate and

SAH is the product of methyltransferase reactions.

SAM and SAH levels are important metabolic indica-

tors of cellular methylation status. Thus, Hcy may

influence global and gene promoter-specific DNA meth-

ylation. To date, no studies have examined the relation-

ship between Hcy and total DNA methylation in NSCs.

In the present study, the levels of the methylation inter-

mediates SAM and SAH, DNA methyltransferase

activity, and global DNA methylation in NSCs were

explored to test the hypothesis that Hcy-induced inhibi-

tion of NSC proliferation is caused by modifications in

DNA methylation.

Results

Effects of Hcy on neurosphere growth and NSC

proliferation

NSCs derived from the neonatal rat hippocampus

began to form suspended neurospheres after 2 days

in vitro. As shown in Fig. 1, undifferentiated NSCs in

neurospheres were characteristically spherical [12].

Most floating spheres increased in size over time, and

reached a diameter of ~ 150 lm after 5–6 days in vitro.

The neurosphere size reflects the proliferative capac-

ity of cells. Dramatic reductions in the mean diameters

of the neurospheres were observed after exposure to

all three concentrations of Hcy (P < 0.05). Further-

more, the effect appeared to be dose-dependent; the

diameter was gradually reduced with increasing con-

centration (30 > 300 > 1000 lM). The mean diameter

A B

Fig. 1. Inhibition of NSC neurosphere formation. (A) Representative images of live floating neurospheres prepared from neonatal rat

hippocampus under various Hcy treatments (C, control; L, Hcy-L; M, Hcy-M; H, Hcy-H). Scale bar = 100 lm. (B) The diameter of

neurospheres was measured using cellSens Dimension software. Eight bright-field images were randomly captured under a phase-contrast

microscope. aP < 0.05 versus control; bP < 0.05 versus Hcy-L; cP < 0.05 versus Hcy-M.

FEBS Journal 281 (2014) 2088–2096 ª 2014 FEBS 2089

N. Lin et al. Neurotoxicity of homocysteine

Page 3: Homocysteine induces cytotoxicity and proliferation inhibition in neural stem cells via DNA methylation               in vitro

of the neurospheres after treatment with 1000 lM Hcy

was significantly lower than those seen after treatment

with 30 or 300 lM Hcy (P < 0.05) (Fig. 1).

SRY (sex-determining region Y)-box 2 (Sox 2) is

essential to maintain self-renewal of undifferentiated

stem cells, and is widely used to identify multipotent

neural stem/progenitor cells in vitro. We performed

double labeling with 50-bromo-20-deoxyuridine (BrdU)

and Sox2 to assess the proliferative potential in NSCs.

Almost all the cells in the neurospheres expressed Sox2

protein, which appeared as green cytoplasmic staining

(Fig. 2A). The percentage of BrdU-positive/Sox2-posi-

tive cells was significantly lower in the Hcy-exposed

groups compared to the control (Fig. 2B). These

results suggest that the smaller size of the neurospheres

formed in the presence of Hcy may be a consequence

of the decreased proliferative capacity of the NSCs.

To rule out a possible contribution to NSC prolifer-

ation of redox action of the thiol group of Hcy,

300 lM L-cysteine, a structurally related sulfur amino

acid, was used to treat NSCs instead. As shown in

Fig. 2B, no significant difference in NSC proliferation

was detected between L-cysteine-treated and untreated

cells over the 6-day cysteine incubation period

(P > 0.05).

The toxic effects of Hcy on NSCs

Neurotoxicity was assessed by measuring the amount

of lactate dehydrogenase (LDH) released from the cells

following Hcy treatment. The results show that Hcy

increased LDH release into the cell culture medium.

Comparisons between Hcy treatment groups showed

that the LDH levels after treatment with 300 and

1000 lM Hcy were significantly higher than that after

treatment with 30 lM Hcy (P < 0.05, Fig. 2C).

SAM, SAH and global DNA methylation levels in

Hcy-treated NSCs

Previous studies have reported changes in SAM, SAH

and DNA methylation during NSC proliferation and

differentiation [10]. Our results showed a significant

decrease in global DNA methylation in Hcy-treated

A B

C

Fig. 2. Cytotoxicity and cell proliferation analysis of NSC cultures. (A) NSCs were double-labeled with Sox2 (green) and BrdU (red). Scale

bar = 100 lm. Cell proliferation was revealed by BrdU labeling. After 6 days of exposure to 30–1000 lM Hcy, 300 lM L-cysteine and/or

150 lM zebularine in vitro, the cultured cells were incubated with BrdU for 24 h and then double-labeled. Defined fields of each culture dish

were photographed, and the numbers of BrdU-positive/Sox2-positive and Sox2-positive cells were calculated using SCOPEPHOTO 3.0 software

(Scopetek, Inc., Hangzhou, China). Left column, Sox2-positive cells; center column, BrdU-positive cells; right column, Sox2/BrdU double -

positive cells (merged images). C/C1/C2, control; L/L1/L2, Hcy-L; M/M1/M2, Hcy-M; H/H1/H2, Hcy-H; Y/Y1/Y2, 300 lM L-cysteine; Z/Z1/Z2,

150 lM zebularine; Z+M/Z+M1/Z+M2, zebularine + Hcy-M. (B) Percentage of BrdU-positive versus Sox2-positive cells. (C) Cytotoxicity in

Hcy-treated NSCs. Neural cell injury was quantified by measurement of LDH released from damaged cells into the extracellular fluid. Values

are means � SD from three separate experiments. aP < 0.05 versus control; bP < 0.05 versus Hcy-L.

2090 FEBS Journal 281 (2014) 2088–2096 ª 2014 FEBS

Neurotoxicity of homocysteine N. Lin et al.

Page 4: Homocysteine induces cytotoxicity and proliferation inhibition in neural stem cells via DNA methylation               in vitro

groups compared with the control group (Fig. 3A).

Moreover, no significant difference in the global DNA

methylation level in NSCs was observed between cells

treated with 300 lM cysteine and control cells

(P > 0.05) (Fig. 3A).

To understand the mechanism underlying the change

in global DNA methylation caused by Hcy treatment,

the cellular concentrations of Hcy-related metabolites

(SAM and SAH) were measured by HPLC. We veri-

fied that treatment with Hcy did not significantly alter

the concentration of SAM in NSCs (Fig. 3B). How-

ever, SAH and Hcy concentrations increased in tan-

dem (Fig. 3C). Consequently the SAM : SAH ratio

was decreased by Hcy in a concentration-dependent

manner (Fig. 3D).

DNMT expression and activity in Hcy-treated

NSCs

A family of DNMTs, including DNMT3a, DNMT3b

and the maintenance methyltransferase DNMT1,

mediates the methylation reaction, in which a methyl

group (-CH3) is added to the 50 position of the pyrimi-

dine ring of cytosine residues, primarily at CpG dinu-

cleotides. Our results showed that protein expression

of all DNMT isoforms decreased in NSCs with

increasing Hcy concentration (Fig. 4A). The total

activity of all DNMT enzymes was also significantly

decreased after Hcy treatment (P < 0.05) (Fig. 4B).

Thus, the Hcy treatment-induced changes in total

DNMT activity may be associated with changes in

DNMT1, DNMT3a and DNMT3b protein expression.

Effects of zebularine on NSC proliferation and

global DNA methylation level

Zebularine, a DNA methylation inhibitor that acts by

forming a covalent complex with DNMTs [13], was

used to determine whether the decrease in DNMT

activity resulted in reduced cell proliferation or not.

Our results show that zebularine decreased the global

DNA methylation in NSCs to lower levels than in the

control group (P < 0.05) (Fig. 2A). Inhibition of

DNMT activity also significantly decreased the per-

centage of proliferative cells in the zebularine treat-

ment group compared with the control group

(P < 0.05). A zebularine concentration of 150 lM,which inhibited DNMT activity by 50% (data not

shown), was chosen to ensure minimal cytotoxicity

in vitro, and had a maximal inhibitory effect on NSC

proliferation.

Discussion

NSCs are crucial for both brain development and

adult neurogenesis. Because of their capacity to

undergo self-renewal and to differentiate into multiple

cell types, stem cells have the potential to serve as

A B

C D

Fig. 3. Global DNA hypomethylation, and

determination of SAM and SAH levels and

the SAM : SAH ratio in NSCs. (A) The

cells were treated with Hcy at various

concentrations, L-cysteine or zebularine for

6 days. Global methylation levels were

measured using the Methylamp Global

DNA Methylation Quantification Ultra kit.

(B–D) Intracellular SAM and SAH levels

and the SAM/SAH ratio were determined

by HPLC. Values are means � SD from

three separate experiments. aP < 0.05

versus control; bP < 0.05 versus Hcy-L.

FEBS Journal 281 (2014) 2088–2096 ª 2014 FEBS 2091

N. Lin et al. Neurotoxicity of homocysteine

Page 5: Homocysteine induces cytotoxicity and proliferation inhibition in neural stem cells via DNA methylation               in vitro

valuable sources of neural cells during attempts to

induce regeneration and tissue repair in response to

the devastating effects of neurodegenerative disorders.

In this study, Hcy decreased NSC proliferation and

increased cell injury in vitro. These results suggest that

the inhibitory effects of Hcy on NSC proliferation

may further hinder neural regeneration and repair fol-

lowing neurodegeneration. This finding may help to

explain why hyperhomocysteinemia often coincides

with some neurological diseases.

DNA methylation is important in cellular processes

during embryonic development and stem cell differen-

tiation. Recently, DNA methylation has been shown

to be closely associated with fate specification in NSCs

[10]. On the other hand, Hcy disrupts the DNA meth-

ylation cycle inducing DNA damage, cell death, and

alterations in gene expression. Perturbations in methyl

group metabolism and Hcy balance have emerged over

the past few decades as playing defining roles in a

number of pathological conditions [2]. The present

results showed that Hcy decreases global DNA meth-

ylation in NSCs. Moreover, our findings suggest that

these changes in DNA methylation may be at least

partially responsible for the inhibitory effects of Hcy

on NSC proliferation.

DNA methylation, which adds a methyl groups to a

cytosine base at CpG dinucleotide residues, is cata-

lyzed by DNMTs. The degree of DNA methylation

regulates gene expression patterns by altering chroma-

tin structures. Three active DNMTs have been identi-

fied in mammals: DNMT1, DNMT3a, and DNMT3b.

DNMT1 is essential for maintaining methylation pat-

terns during DNA replication, whereas DNMT3a and

DNMT3b are primarily responsible for de novo meth-

ylation in embryonic and postnatal tissues [14,15]. In

the developing mouse brain, deletion of DNMT1 in

progenitor cells impairs neuronal maturation and sur-

vival, and causes precocious astroglial differentiation

[16]. DNMT3a is expressed in postnatal NSCs and is

required for neurogenesis. In the postnatal forebrain,

DNMT3a is expressed in the subventricular zone and

the hippocampal dentate gyrus. DNMT3a mediates

methylation at non-promoter regions in genes related

to nervous system development and neurogenesis, but

methylates the proximal promoter regions of non-neu-

ronal genes such as glial fibrillary acidic protein

(GFAP)[17]. During neurogenesis, DNMT3b is pre-

dominantly expressed in proliferating neural precursor

cells within the cerebral cortex [18,19]. DNMT3b may

play an important role in the initial steps of progenitor

cell differentiation, although this function remains to

be demonstrated. Thus, it appears that the DNA

methylation status during new neural cell formation is

regulated by these DNMTs. The present study showed

A B

C D

Fig. 4. Effects of Hcy on DNMT protein

expression and enzyme activity. DNMT1,

DNMT3a and DNMT3b protein expression

were detected by western blot analysis

and normalized to b-actin expression. (A–

C) Representative western blots of

DNMT1, DNMT3a and DNMT3b, and bar

graphs of their expression levels (ratios of

DNMT1, DNMT3a and DNMT3b to b-

actin). (D) Total DNMT activity was

measured for all four treatment groups.

Values are mean labeling indices for three

samples per treatment. aP < 0.05 versus

control; bP < 0.05 versus Hcy-L.

2092 FEBS Journal 281 (2014) 2088–2096 ª 2014 FEBS

Neurotoxicity of homocysteine N. Lin et al.

Page 6: Homocysteine induces cytotoxicity and proliferation inhibition in neural stem cells via DNA methylation               in vitro

that inhibition of DNMT activity by Zebularine treat-

ment decreased the total DNA methylation level and

the percentage of proliferative NSCs. This suggests

that, by regulating these DNMTs, varying concentra-

tions of Hcy may alter global DNA methylation in

proliferating NSCs, further affecting neurogenesis in

the developing and mature central nervous system.

SAH is produced as a by-product of SAM-

dependent methyltransferase reactions. Most cellular

methyltransferases are inhibited by accumulation of

intracellular and cellular SAH. High-affinity binding

of SAH to the active site of cellular methyltransferases

results in product inhibition of the enzyme activity

[20]. The DNMT enzyme activity may be also affected

by SAM/SAH levels in the cell. The SAM : SAH ratio

is frequently used as an indicator of cellular methyla-

tion potential. A decrease in the SAM : SAH ratio is

predictive of a reduced methylation capacity. The pres-

ent results show that elevated Hcy levels are associated

with an increase in SAH and a decrease in the

SAM : SAH ratio in NSCs. We conclude that Hcy

may decrease DNMT activity by altering the concen-

tration of SAH and the SAM/SAH ratio. In addition,

our observation that Hcy decreased the protein expres-

sion of DNMT1, DNMT3a and DNMT3b suggests

that the reduction in enzymatic DNMT activity may

also be due in part to reduced protein expression.

Numerous studies in recent years have investigated

the role of Hcy as a cause of brain damage. It was

found that Hcy itself can cause disturbed redox

potentials, thus promoting calcium influx, accumula-

tion of amyloid and tau proteins, apoptosis and

neuronal death [21]. Thus, it is possible that Hcy, as

a sulfur-containing amino acid, may affect NSC via-

bility and proliferation via an alteration in the redox

status of Hcy. In this study, L-cysteine was used to

detect the effect of the redox actions of the thiol

group on NSC proliferation. A 300 lM concentration

of L-cysteine was used to reflect the moderate homo-

cysteine concentration used here. Recently, L-cysteine

at lower doses (0.5–5 lM) had been shown to improve

the cell viability of NSCs, and 1 lM L- cysteine had

the optimal effect on cell viability [22]. On the other

hand, cysteine has been found to be toxic to cultured

neurons at relatively high concentrations. It was

reported that 1 mM L-cysteine decreased neuronal sur-

vival to 30.5%, whereas we found that L-cysteine had

no cytotoxic effects on proliferative NSCs when it

was used at a concentration of 300 lM [23]. The

results suggest that redox actions of the thiol group

of Hcy may be not the major cause of Hcy-induced

cell proliferation inhibition, at least at a concentration

of 300 lM.

In summary, our study confirmed that Hcy at high

concentrations inhibited NSC proliferation and exerted

a cytotoxic effect. This may explain why high Hcy

levels result in neural tube defects and some neurode-

generative diseases. Moreover, the DNMT inhibiter

zebularine reduced cell proliferation and the levels of

global DNA methylation in NSCs. Hcy affects DNA

methylation in NSCs, which may be mediated by

decreased DNMT enzyme activity. The inhibition of

DNMT activation caused by Hcy treatment may be

associated with a high concentration of cellular SAH,

a decreased SAM : SAH ratio, and reduced DNMT

protein expression.

Experimental procedures

Materials

Dulbecco’s modified Eagle’s medium, basic fibroblast growth

factor and epidermal growth factor were obtained from

Gibco (Carlsbad, CA, USA). D,L-homocysteine, L-cysteine,

BrdU, SAM and SAH were purchased from Sigma (St Louis,

MO, USA). Zebularine was purchased from Merck Chemi-

cals (Darmstadt, Germany). The DNA methyltransferase

activity/inhibition assay kit was purchased from Active

Motif (Carlsbad, CA, USA). Mouse monoclonal antibodies

against BrdU, b-actin, DNMT1 and DNMT3a were

obtained from Cell Signaling Technology Inc. (Boston, MA,

USA). Rabbit monoclonal antibodies against DNMT3b

and Sox2 were obtained from Abcam (Cambridge, UK). All

secondary antibodies were obtained from Zhongshan

Goldbridge Biotechnology (Beijing, China).

NSC culture

The Tianjin Medical University Animal Ethics Committee

approved the experimental protocols in this study (study

number TMUaMEC 2012016). Neonatal Sprague Dawley

rats were purchased from Beijing Medical Laboratory

Animal Co. Ltd. (Beijing, China). NSCs were isolated

and maintained as previously described [24]. The cultured

cells were assigned to seven treatment groups: control

(Hcy-free), Hcy-L (low Hcy, 30 lM), Hcy-M (moderate

Hcy, 300 lM), Hcy-H (high Hcy, 1000 lM), Cys (300 lML-cysteine), Zeb (150 lM zebularine) and Zeb + Hcy-M

(150 lM zebularine + 300 lM Hcy). The duration of the

Hcy treatment was 6 days. After treatment, NSCs were

harvested for the following immunocytochemistry and

western blot analysis.

Determination of neurosphere size

To determine the size of the neurospheres after 6 days of

treatment with varying concentrations of Hcy, eight bright-

FEBS Journal 281 (2014) 2088–2096 ª 2014 FEBS 2093

N. Lin et al. Neurotoxicity of homocysteine

Page 7: Homocysteine induces cytotoxicity and proliferation inhibition in neural stem cells via DNA methylation               in vitro

field images for each group were randomly captured using

an inverted microscope and digital camera (Olympus,

Tokyo, Japan). The diameters of the neurospheres were

measured using cellSens Dimension software (Olympus).

BrdU incorporation and immunocytochemistry

NSC proliferation was determined by in vitro labeling with

the thymidine analog BrdU. On the 6th day of culture, the

cells were incubated with BrdU (10 lg�mL�1) for 24 h.

The cultures were then processed for double-labeled immu-

nofluorescence. The cells were fixed in 4% paraformalde-

hyde for 30 min at room temperature, permeabilized using

0.1% Triton X-100 in NaCl/Pi for 5 min, and incubated

with blocking buffer for 1 h with 1% fetal bovine serum.

The cells were incubated overnight at 4 °C in a mixture of

rabbit monoclonal anti-Sox2 antibody (1 : 100) and mouse

monoclonal anti-BrdU antibody (1 : 100). Subsequently,

cells were incubated for 2 h at room temperature with

fluorescein isothiocyanate-conjugated goat anti-rabbit IgG

and tetramethylrhodamine isothiocyanate-conjugated goat

anti-mouse IgG. Immunostained cells were visualized by

indirect fluorescence under a fluorescent microscope

(Olympus).

Measurement of LDH activity

LDH, an indicator of cell injury, was detected by colori-

metric (440 nm) assay with the LDH cytotoxicity detection

Kit (Nanjing Jiancheng Biochemical Reagent Co., Nanjing,

China), according to the manufacturer’s instructions. The

assay is based on measurement of LDH released into the

culture medium from damaged cells. The released LDH

can be quantified by a coupled enzymatic reaction. First,

LDH catalyzes the conversion of lactate to pyruvate via

reduction of NAD+ to NADH. Second, diaphorase uses

NADH to reduce a tetrazolium salt to a red formazan

product. Briefly, the cells were cultured in the presence of

Hcy at various concentrations (0, 30, 300 and 1000 lM) for48 h. The cell medium (50 lL) was then gently aspirated

and used for LDH determination. After the reaction, the

absorbance for each sample was read at wavelength of

440 nm.

Global DNA methylation detection

Global methylation levels were measured using a Methy-

lamp Global DNA Methylation Quantification Ultra kit

(Epigentek Group Inc., New York, NY, USA) as previ-

ously described [25]. According to the manufacturer’s

instructions, DNA is immobilized to the strip well

specifically coated with a DNA affinity substance. The

methylated fraction of DNA can then be recognized by a

5-methylcytosine antibody and quantified through an

ELISA-like reaction. The amount of methylated DNA is

proportional to the absorbance at 450 nm.

HPLC analysis

Cell cultures were rinsed twice with NaCl/Pi and frozen at

�80 °C. After thawing, cells were scraped into 1 mL deion-

ized water and sonicated for 15 s on ice. The macromole-

cules were precipitated using 1.5 M perchloric acid solution

at 4 °C for 1 h, adjusting the pH to 4–5 with KOH, and

then centrifuged for 15 min at 9000 g. The supernatant was

freeze-dried and later subjected to HPLC analysis [26].

HPLC was performed on a Waters HPLC system (Waters

Corp., Milford, MA, USA) using a Venusil MP C18 col-

umn (particle size 5lm, diameter 4.6mm, length 250mm) at

30 °C. The mobile phase consisted of 50 mM NaH2PO4

(pH 4.38), 10 mM C7H15NaO3S and 20% methanol. The

flow rate was 1 mL�min�1, and detection was performed at

254 nm.

Western blotting

Protein expression of DNMT isoforms in cultured cells was

assessed by western blot analysis. The cells were washed

with ice-cold phosphate-buffered saline, and lyzed with

TNE/NP- 40 buffer (50 mM Tris-HCl pH 7.4, 100 mM

NaCl, 1 mM EDTA, 0.1% Nonidet P-40). Proteins were

separated by 12% SDS/PAGE, and transferred to polyv-

inylidene difluoride membranes. The membranes were

blocked using 5% non-fat milk, and incubated with pri-

mary antibodies anti-DNMT1, anti-DNMT3a and

anti-DNMT3b (all at a 1 : 1000 dilution) overnight at

4 °C, followed by appropriate secondary antibodies for l h

at room temperature. Proteins were detected by chemilumi-

nescence assay. Quantification of proteins was performed

by densitometric analysis using IMAGEJ software (version

1.61, National Institute of Mental Health, Bethesda, MD,

USA). The intensity of each protein band was normalized

to that of the respective b-actin band (detected using a

1 : 5000 dilution of antibody).

DNMT activity

Global DNMT activity was evaluated in nuclear fractions

using an Active Motif DNA methyltransferase activity/inhi-

bition colorimetric assay kit, according to the manufac-

turer’s instructions. The same quantity of cells from each

sample was analyzed. A lot-specific standard curve was

generated using the DNMT1 enzyme provided in the kit.

Absorbance was measured on a microplate reader at

450 nm, and DNMT activity was calculated according to

the following formula: DNMT activity (absorbance/h/

mg) = [(sample absorbance � blank absorbance)/(lg of

protein 9 hour of incubation time)] 9 1000.

2094 FEBS Journal 281 (2014) 2088–2096 ª 2014 FEBS

Neurotoxicity of homocysteine N. Lin et al.

Page 8: Homocysteine induces cytotoxicity and proliferation inhibition in neural stem cells via DNA methylation               in vitro

Statistical analysis

The data are expressed as means � SD, and were analyzed

using SPSS13.0 statistical software (SPSS Inc., Chicago, IL,

USA). One-way analysis of variance (ANOVA) followed

by the Student– Newman–Keuls test for multiple

comparisons were used to determine significant differences

among the experimental groups. The criterion for statistical

significance was P < 0.05.

Acknowledgements

This research was supported by grants from the

National Natural Science Foundation of China (num-

bers 81130053 and 81373003).

References

1 Herrmann W & Obeid R (2011) Homocysteine: a

biomarker in neurodegenerative diseases. Clin Chem

Lab Med 49, 435–441.

2 Schalinske KL & Smazal AL (2012) Homocysteine

imbalance: a pathological metabolic marker. Adv Nutr

3, 755–762.

3 Vollset SE, Refsum H, Irgens LM, Emblem BM,

Tverdal A, Gjessing HK, Monsen AL & Ueland PM

(2000) Plasma total homocysteine, pregnancy

complications, and adverse pregnancy outcomes: the

Hordaland homocysteine study. Am J Clin Nutr 71,

962–968.

4 den Heijer T, Vermeer SE, Clarke R, Oudkerk M,

Koudstaal PJ, Hofman A & Breteler MM (2003)

Homocysteine and brain atrophy on MRI of non-

demented elderly. Brain 126, 170–175.

5 Lee ES, Chen H, Soliman KF & Charlton CG (2005)

Effects of homocysteine on the dopaminergic system

and behavior in rodents. Neurotoxicology 26, 361–371.

6 Wang J, Bai X, Chen Y, Zhao Y & Liu X (2012)

Homocysteine induces apoptosis of rat hippocampal

neurons by inhibiting 14–3–3e expression and activating

calcineurin. PLoS One 7, e48247.

7 Ho PI, Ortiz D, Rogers E & Shea TB (2002) Multiple

aspects of homocysteine neurotoxicity: glutamate

excitotoxicity, kinase hyperactivation and DNA

damage. J Neurosci Res 70, 694–702.

8 Rabaneda LG, Carrasco M, L�opez-Toledano MA,

Murillo-Carretero M, Ruiz FA, Estrada C & Castro C

(2008) Homocysteine inhibits proliferation of neuronal

precursors in the mouse adult brain by impairing the

basic fibroblast growth factor signaling cascade and

reducing extracellular regulated kinase 1/2-dependent

cyclin E expression. FASEB J 22, 3823–3835.

9 Yan H, Zhang X, Luo S, Liu H, Wang X, Gao Y,

Wilson JX & Huang G (2013) Effects of homocysteine

on ERK signaling and cell proliferation in fetal neural

stem cells in vitro. Cell Biochem Biophys 66, 131–137.

10 Namihira M, Kohyama J, Abematsu M & Nakashima

K (2008) Epigenetic mechanisms regulating fate

specification of neural stem cells. Philos Trans R Soc

Lond B Biol Sci 363, 2099–2109.

11 Singh RP, Shiue K, Schomberg D & Zhou FC (2009)

Cellular epigenetic modifications of neural stem cell

differentiation. Cell Transplant 18, 1197–1211.

12 Zhang XM, Huang GW, Tian ZH, Ren DL & X

Wilson J (2009) Folate deficiency induces neural stem

cell apoptosis by increasing homocysteine in vitro.

J Clin Biochem Nutr 45, 14–19.

13 Zhou L, Cheng X, Connolly BA, Dickman MJ, Hurd

PJ & Hornby DP (2002) Zebularine: a novel DNA

methylation inhibitor that forms a covalent complex

with DNA methyltransferases. J Mol Biol 321, 591–599.

14 Jaenisch R & Bird A (2003) Epigenetic regulation of

gene expression: how the genome integrates intrinsic

and environmental signals. Nat Genet 33, 245–254.

15 Okano M, Bell DW, Haber DA & Li E (1999) DNA

methyltransferases Dnmt3a and Dnmt3b are essential

for de novo methylation and mammalian development.

Cell 99, 247–257.

16 Hutnick LK, Golshani P, Namihira M, Xue Z,

Matynia A, Yang XW, Silva AJ, Schweizer FE & Fan

G (2009) DNA hypomethylation restricted to the

murine forebrain induces cortical degeneration and

impairs postnatal neuronal maturation. Hum Mol Genet

18, 2875–2888.

17 Wu H, Coskun V, Tao J, Xie W, Ge W, Yoshikawa K,

Li E, Zhang Y & Sun YE (2010) Dnmt3a-dependent

nonpromoter DNA methylation facilitates transcription

of neurogenic genes. Science 329, 444–448.

18 Feng J, Chang H, Li E & Fan G (2005) Dynamic

expression of de novo DNA methyltransferases Dnmt3a

and Dnmt3b in the central nervous system. J Neurosci

Res 79, 734–746.

19 Martins-Taylor K, Schroeder DI, LaSalle JM, Lalande

M & Xu RH (2012) Role of DNMT3B in the

regulation of early neural and neural crest specifiers.

Epigenetics 7, 71–82.

20 Chiang PK (1998) Biological effects of inhibitors of

S–adenosyl-homocysteine hydrolase. Pharmacol Ther

77, 115–134.

21 Obeid R & Herrmann W (2006) Mechanisms of

homocysteine neurotoxicity in neurodegenerative

diseases with special reference to dementia. FEBS Lett

580, 2994–3005.

22 Wang Z, Liu DX, Wang FW, Zhang Q, Du ZX, Zhan

JM, Yuan QH, Ling EA & Hao AJ (2013) L-Cysteine

promotes the proliferation and differentiation of neural

stem cells via the CBS/H2S pathway. Neuroscience 237,

106–117.

FEBS Journal 281 (2014) 2088–2096 ª 2014 FEBS 2095

N. Lin et al. Neurotoxicity of homocysteine

Page 9: Homocysteine induces cytotoxicity and proliferation inhibition in neural stem cells via DNA methylation               in vitro

23 Wang XF & Cynader MS (2001) Pyruvate released by

astrocytes protects neurons from copper-catalyzed

cysteine neurotoxicity. J Neurosci 21, 3322–3331.

24 Luo S, Zhang X, YuM, Yan H, Liu H, Wilson JX &

Huang G (2013) Folic acid acts through DNA

methyltransferases to induce the differentiation of neural

stem cells into neurons. Cell Biochem Biophys 66, 559–566.

25 Wang BX, Yin BL, He B, Chen C, Zhao M, Zhang

WX, Xia ZK, Pan YZ, Tang JQ, Zhou XM et al.

(2012) Overexpression of DNA damage-induced 45agene contributes to esophageal squamous cell cancer

by promoter hypomethylation. J Exp Clin Cancer Res

31, 11.

26 Cavallaro RA, Fuso A, D’Anselmi F, Seminara L &

Scarpa S (1994) The effect of S–adenosylmethionine

on CNS gene expression studied by cDNA microarray

analysis. Biochem Biophys Res Commun 205,

1748–1754.

2096 FEBS Journal 281 (2014) 2088–2096 ª 2014 FEBS

Neurotoxicity of homocysteine N. Lin et al.