• Tidak ada hasil yang ditemukan

Autism spectrum disorder an omics perspe

N/A
N/A
Protected

Academic year: 2017

Membagikan "Autism spectrum disorder an omics perspe"

Copied!
10
0
0

Teks penuh

(1)

R

EVIEW

Autism spectrum disorder: An omics perspective

Alisa G. Woods

1,2

, Kelly L. Wormwood

1

, Armand G. Ngounou Wetie

1

, Roshanak Aslebagh

1

,

Bernard S. Crimmins

2

, Thomas M. Holsen

3

and Costel C. Darie

1

1

Biochemistry & Proteomics Group, Department of Chemistry & Biomolecular Science, Clarkson University,

Potsdam, NY, USA

2

SUNY Plattsburgh Neuropsychology Clinic and Psychoeducation Services, Plattsburgh, NY, USA

3

Department of Civil & Environmental Engineering, Clarkson University, Potsdam, NY, USA

Received: August 22, 2014 Revised: September 11, 2014 Accepted: October 7, 2014

Current directions in autism spectrum disorder (ASD) research may require moving beyond

genetic analysis alone, based on the complexity of the disorder, heterogeneity and

conver-gence of genetic alterations at the cellular/functional level. Mass spectrometry (MS) has been

increasingly used to study CNS disorders, including ASDs. Proteomic research using MS is

directed at understanding endogenous protein changes that occur in ASD. This review focuses

on how MS has been used to study ASDs, with particular focus on proteomic analysis. Other

neurodevelopmental disorders have been investigated using MS, including fragile X syndrome

(FXS) and Smith-Lemli-Opitz Syndrome (SLOS), genetic syndromes highly associated with

ASD comorbidity.

Keywords:

Autism spectrum disorder / Neurodevlopmental disorders

1

Autism overview

Autism spectrum disorder (ASD) consists of social deficits,

communication problems, and repetitive behaviors [1]. ASDs

were previously separated in the DSM-IV-TR into Asperger’s

syndrome, autism, and pervasive developmental disorder not

otherwise specified [2, 3], but the DSM-5 collapsed these into

one term, ASD. Clinical aspects are now described as a dyad:

(i) persistent deficits in social communication and (ii)

re-stricted, repetitive behavior, interests, and activities [1, 2]. An

estimated one in every 68 US children has ASD [4],

com-pared to year 2002 estimates of one in 150 children [5] and

worldwide prevalence rates are similar [6]. The reasons for

this increase are not known. Early detection of ASD is highly

desirable to promote early behavioral interventions and more

functional outcomes [7].

Correspondence: Dr. Costel C. Darie, Biochemistry & Proteomics Group, Department of Chemistry & Biomolecular Science Clark-son University, 8 ClarkClark-son Avenue, Potsdam, NY, 13699-5810, USA

E-mail: cdarie@clarkson.edu Fax:+1-315-268-6610

Abbreviations: ADHD, attention-deficit hyperactivity disorder; ASD, autism spectrum disorder;FMR1, fragile X mental retarda-tion;FXS, fragile X syndrome;PON1, paraoxanase/arylesterase 1;PPI, protein–protein interaction;SLOS, Smith-Lemli-Opitz syn-drome

ASD is likely a heterogeneous disorder with multiple

causes, involving genes and the environment [8–10].

Numer-ous genetic studies of ASD have been conducted [11],

support-ing not only a strong genetic link in many cases, but also a

high likely degree of heterogeneity of cause [12]. Many

impli-cated genes are involved in nervous system development and

neurotransmitter systems [13]. ASD is more than 20-times

increased in first-degree relatives [14], based on a twin study.

In identical twins, between 60 and 92% ASD concordance

occurs and from 0 to 10% in fraternal twins [14, 15]. Despite

probable genetic influences, numerous associated genes have

been suggested, without clear consistency in many instances.

[16]. Over 100 genes and 40 genomic loci appear to be

asso-ciated [17, 18]. A recent study described the first clear link in

ASD subtypes and the CHD8 gene mutation [19], indicating

that more progress may follow in the genetic understanding

of other subtypes of ASD. Genes involved in ASD have

com-mon molecular mechanisms, [20], suggesting convergence of

function at the protein level [21]. Currently genetic complexity

and genetics alone do not provide clear predictive markers,

limiting the current utility of genetic testing for ASD [22].

Clinical geneticists rule out known genetic syndromes when

evaluating ASD, and 80% of families are left without a

defini-tive diagnosis [23].

Additional corresponding author: Dr. Alisa G. Woods, E-mail:

awoods@clarkson.edu

Colour Online: See the article online to view Figs. 1–3 in colour.

(2)

In addition to genetic influences on ASD etiology,

envi-ronmental exposures likely play a major role. For example,

certain pesticides and industrial chemicals may influence

the development of ASD, particularly in susceptible

individ-uals [24]. Further studies investigating the timing, dosage or

mechanisms that induce ASD are needed [25]. Recent results

of the Childhood Autism Risks from Genetics and

Environ-ment (CHARGE) study reported that prenatal close

proxim-ity to organophosphates produced a 60% increased ASD risk.

The risk increased further for third trimester exposure [26].

Thousands of high production volume chemicals circulate

through commerce [27], and hundreds of these chemicals

have recently been detected in pregnant women’s tissues [28].

Understanding the link between environmental exposures of

these chemicals and the onset of ASD is a major knowledge

gap [24].

For example, a relatively new class of compounds that

have begun to receive a significant amount of attention is

organophosphorous flame retardants [29]. Currently slated as

a replacement for the pentabrominated diphenyl ether

mix-ture, this class of compounds is ubiquitous and exhibit

ele-vated concentrations relative to other neurotoxicants, such as,

brominated diphenyl ethers in indoor environments [30–32].

Similar to the OP pesticides, triaryl-, and trialkylphosphate

flame retardants hydrolyze in the blood resulting in

diaryl-and dialkylphosphates, respectively, [33,34] to conformations

conspicuously similar to OPs oxon metabolites, suggesting

organophosphorous flame retardants may have OP-like

neu-rotoxicities and contribute to the onset and severity of ASD

in vulnerable groups.

Whether genetically or environmentally induced (or both

in collaboration) numerous investigations indicate

biolog-ical disturbances in ASDs [35–38], but a clear diagnostic

biomarker for ASD is not available. ASDs are diagnosed

based on behavior, and despite the current existence of

gold-standard behavioral tests [39, 40], biomarkers could further

improve screening and diagnosis. ASDs can be

unrecog-nized in children and even in adults, and current screening

for ASDs can produce false positive or false negative results

[41]. Moving beyond genetic analysis alone may provide new

inroads to diagnosis and understanding of ASD biomarkers.

MS-based proteomics could provide one route for exploration.

2

MS analysis of endogenous molecules

in ASDs

2.1 Differential proteomics: qualitative and

quantitative analysis of proteins in ASD

Current analyses using MS to study ASDs have focused on

endogenously produced biomarkers that could aid in

diagno-sis or understanding of ASDs. Despite a variety of different

proteins identified, some consistency has emerged in the

liter-ature, specifically complement proteins and apolipoproteins

(apos).

One study analyzed blood serum in individuals with

ASD and comorbid attention-deficit hyperactivity disorder

(ADHD) (n

=

9 ASD

+

ADHD, ASD alone

n

=

7) compared

to age-matched controls (n

=

12). Three peaks that were

dif-ferent in ASD versus controls were identified, but the amino

acid sequence information of the peptides that corresponded

to these peaks was not identified [42], however, the

investi-gators speculated that the peptides corresponding to these

peaks may be part of an apo protein [43]. Our group obtained

the sera from these investigators, and reanalyzed them in

our lab. We confirmed increased levels of apoA1 and apoA4,

in individuals with ASD [44]. We further found significant

elevations in the high-density lipoprotein associated enzyme

[45] serum paraoxanase/arylesterase 1 (PON1) [44], which is

also involved in toxin metabolism and detoxification (such as

due to organophosphate exposure), and could help prevent

oxidative stress [46]. It interacts with the cholesterol-carrying

proteins known as apos, which bind PON1 increasing the

stability and activity of PON1 [47]. Interestingly PON1 gene

mutations have been associated with ASD [48]. Notably, one

study of 50 children with ASD found that PON1 protein

ac-tivity (but not gene polymorphisms) was associated with ASD

[49], underscoring the need to analyze biomarkers at the

pro-tein level.

Further suggesting the possibility that apos (and

lipid-associated molecules) are indeed ASD biomarkers,

eleva-tions in apo B-100 and apo A-IV were observed in an earlier

proteomic study that compared high functioning ASD with

low-functioning ASD [50]. Significantly elevated complement

factor H-related protein, complement C1q and fibronectin 1

and apoB-100 was also measured in children with ASD

com-pared to typically developing controls in the same study [50]. A

proteomic study of individuals with Asperger’s syndrome has

also implicated apo dysregulation (apoE, apoC2, and apo A1),

although this change seems to be more specific to females.

Specific proteomic studies examining Asperger’s syndrome

compared to non-Asperger’s ASD could help shed light on

this discrepancy [51].

Consistent with the possible presence of dysregulated

complement proteins in ASD, a different MS analysis of blood

plasma used surface-enhanced laser desorption/ionization

TOF MS to examine peptides in plasma of children with ASD

compared to typically developing controls. Increases in three

peptides were measured, corresponding to C3 complement

protein fragments [52,53]. Additional proteomic studies need

to be conducted to confirm the existence of complement

pro-tein and apo disturbances in ASDs. These studies may help to

identify proteomic biomarker signatures, and possible ASD

subtypes associated with specific biomarkers.

(3)

of the few published proteomic studies of salivary biomarkers

in ASD [55].

Our group has recently used 2D differential in-gel

elec-trophoresis (2D-DIGE) to investigate the differences between

the salivary proteomes of children with ASD and matched

controls. DIGE is primarily used in protein expression

pro-filing experiments of at least two samples or conditions

allowing the determination of the relative abundance of

pro-teins [56]. In 2D-DIGE, propro-teins are labeled with

fluores-cent cyanine dyes (Cy2, 3, and 5) prior to 2D-PAGE [57],

run on 2D-PAGE and then scanned for quantitative

analy-sis of the two proteomes investigated [58, 59]. In our

2D-DIGE experiments, we observed that many proteins were

differentially expressed between the ASD and control

con-ditions mostly observed as either green or red colored gel

spots; the yellow colored gel spots contained proteins with

unchanged levels (Fig. 1). Nano-LC-MS/MS and

MALDI-MS/MS analysis of the proteins contained in the red or

green-colored gel spots identified many of the

differen-tially expressed proteins. One such protein upregulated in

ASD was S100-A9 also called migration inhibitory

factor-related protein 14 (MRP-14) or calgranulin-B (Fig. 1). This

protein has not been previously reported to be associated

with ASD.

[image:3.595.55.534.311.634.2]

In considering proteomic biomarker analysis in ASDs,

specific attributes of the individuals studied, including age

and gender also need to be taken into consideration. Two

studies have emphasized the possibility of gender-specific

differences in ASD biomarkers, finding that males with

Asperger’s syndrome tend to have altered levels of cytokines

and other inflammatory molecules, whereas biomarkers in

fe-males with Asperger’s seem to be growth factors, hormones

and factors associated with lipid transport, and metabolism

[51,60]. Further investigation into gender specific biomarkers

in ASDs is warranted, particularly since there is a gender bias

in ASDs, favoring diagnosis in males. Indeed, neuroimaging

Figure 1. Difference gel electrophoresis (DIGE). (A)Analytical DIGE gel, where protein samples were labeled with fluorescent cyanine dyes (Cy2, 3, and 5) prior to 2D PAGE. The differentially expressed proteins in ASD (cy3 or green) and matched controls (cy5 or red) are either green or red; the yellow ones are unchanged between ASD and controls. (B)Preparative DIGE gel, from which the differentially expressed proteins were picked and digested by trypsin and analyzed by nano-LC-MS/MS or MALDI-MS. The spots that were specific to ASD (left) or control (right) samples are indicated. (C) MALDI-MS/MS spectrum, whose analysis led to the identification of a peptide with the sequence LGHPDTLNQGEFK which is part of protein S100-A9, also named migration inhibitory factor-related protein 14 (MRP-14) or calgranulin-B. This protein was found to be upregulated in ASD in the DIGE experiments The y- and b-ion series, the amino acid sequence of the peptide (top of the spectrum and the name of the protein (left) are indicated. Reprinted with permission from [56].

(4)

evidence has suggested that the neuroanatomy of ASD may

differ in males versus females [61].

With regard to age differences, one study has reported that

12 proteins vary with age in individuals with ASD, including

those involved with inflammation, growth, and hormonal

sig-naling. Examples include higher levels of adiponectin in ASD

with increased age and a decrease with age in typically

de-veloping individuals. Other potential markers that increase

with age in ASD include C-reactive protein, haptoglobin,

TRAIL-R3, matrix metalloproteinase, thyroglobulin, and

can-cer antigen 19–9. Many of these molecules are suggestive of

an inflammatory condition. Age-related changes in

biomark-ers and the possible impact of these molecules on behavioral

phenotypes would be a valuable focus of future studies [62].

2.2 Differential proteomics: analysis of PTMs of

proteins in ASD

Modulation of the function of many proteins is achieved

mostly by transient or definitive PTMs of proteins. Transient

modifications include phosphorylation, acetylation,

myristoy-lation, etc., while stable modifications are presented as

disul-fide bonding formation, protein truncation, and/or

glycosy-lation [63–66]. Only one study investigated PTMs in ASD by

examining salivary peptides examined (n

=

27) compared

to age-matched controls using HPLC-ESI-Ion-Trap MS. In

this study, the investigators found hypophosphorylation of

statherin, histatin 1, and acidic proline-rich proteins in

sub-jects with ASDs compared with controls [55].

2.3 Differential proteomics: analysis of

protein-protein interactions (PPIs) in ASD

Most protein biomarkers or biomarker signatures for diseases

and/or disorders are identified using qualitative and

quanti-tative proteomics, reflecting proteins that are either over- or

under-expressed due to the diagnosed condition. However,

recently, additional protein modifications have been

recog-nized. Phosphorylation, acetylation or glycosylation are

sev-eral such examples. Therefore, both protein quantitation and

protein PTMs provide a more comprehensive picture of the

state of the disease/disorder. The same principle applies to

ASD. However, to our knowledge, no research group has

con-sidered, in addition to protein quantitation and protein PTMs,

stable or transient PPIs as a possible extra indicator of a

dis-ease or disorder. Therefore, we believe that both protein levels

(quantitation), protein PTMs, types and location of PTMs, as

well as stable or transient PPIs, with or without PTMs should

be measured as part of a comprehensive approach. In fact,

our lab recently reported that ApoA1, ApoA4, and PON1

pro-teins, in addition to being dysregulated, are also predicted to

interact with each other as part of HDLs (Figs. 2 and 3) [44].

Protein interactions and networks could provide an important

component of biomarker signature identification in ASD and

take into account a systems approach in understanding the

disorder, rather than focusing on a single molecule.

3

MS-based metabolomic and small

molecule analysis in ASD

LC-MS/MS was used to analyze the urine of young adults with

severe ASD and schizophrenia versus nondiagnosed controls

(n

=

15, 18, and 18, respectively). Butofenine, a molecule

re-lated to serotonin, was elevated versus controls in individuals

with ASD and schizophrenia and correlated with hyperactivity

[67]. This corresponds with serotonin abnormalities reported

in ASD in three MS studies [68–70].

Oxidative stress, the generation of ROS causing tissue

and cellular damage [71], has been consistently implicated in

ASD [72], supported by studies using MS. GC-MS identified

higher urinary lipid peroxidation markers and endothelium

activation in individuals with ASD (n

=

26) versus

non-ASD controls (n

=

12). Lipid peroxidation correlated with

endothelium and platelet activation [73]. MS analyses at the

metabolomic and small molecule level have supported that

oxidative stress and dysregulation of serotonin may be present

in ASD. Overall, proteomic evidence uncovered by MS

sup-ports a role for altered complement proteins and

apolipopro-teins in ASDs, and further supports the presence of oxidative

stress and alterations in serotonergic signaling.

4

MS Analysis in fragile X syndrome

(5)

[95]. MALDI-TOF MS was used to examine FMR1

methy-lation in blood of 62 premutation (between 55–200 CGG

repeats in FMR1 gene) or 18 full mutation (more than 200

CGG repeats) females compared to controls (n

=

74). FMR1

intron 1 hypermethylation was predictive of verbal cognitive

impairment [96]. In this study a MS-based biochemical

mea-surement was paired with a behavioral test, underscoring the

possibility of interdisciplinary research in

neurodevelopmen-tal disorders with MS. Such an interdisciplinary approach

may increase for neurodevelopmental disorder assessment

in the future, as MS-based proteomic use increases.

5

MS Analysis in Smith-Lemli-Opitz

syndrome

Smith-Lemli-Opitz syndrome (SLOS) is a genetically

in-herited deficit in cholesterol synthesis frequently

associ-ated with ASD, as well as intellectual/learning problems

and many physical problems [97–100]. Partial or total

defi-ciency of the

Dhcr7

gene causes SLOS [101]. Tissue

choles-terol and total scholes-terol levels are substantially depleted, and

7-dehydrocholesterol levels increase as a result of the gene

mutation. High 7DH levels inhibit

Hmgcr

further causing

cellular cholesterol deficits [102]. SLOS symptoms improve

marginally with cholesterol supplementation, and cholesterol

supplementation does not cure or even largely alleviate the

disorder. Notably, there are individuals with ASD without

SLOS with low total cholesterol [100], supporting the

pos-sible association of cholesterol disturbances and some ASD

subtypes.

[image:5.595.57.537.340.667.2]

MS studies on SLOS and SLOS animal models have

fo-cused on sterols analysis, specifically 7-dehydrocholesterol

levels and cholesterol [103–107]. Recently the first MS

pro-teomic study in a SLOS model (rat retina) was published.

[108]. The model demonstrates some SLOS characteristics

of retinal degeneration and visual impairment generated by

treatment with AY9944. This drug (AY9944) inhibits DHCR7

(3 beta-hydroxysterol-Delta 7-reductase), the enzyme found

to be defective in SLOS [109]. Rat retinas from the SLOS

rat model (n

=

5) were compared to retinas from age- and

sex-matched controls (n

=

5), via nano-RPLC. Statistically

Figure 2. (A) Nano-LC-MS/MS analysis of the sera from children with ASD and matched controls. The MS/MS spectra whose interpretation led to the identification of PON1, ApoA1, and ApoA4. Them/zof the precursor ion, ions, the b and y product ions, the amino acid sequence of the identified peptides and the proteins that the peptides belong to are shown. (B)Relative quantitation of ApoA4 using the precursor ion intensity. The precursor ion investigated hadm/zof 704.23 (2+) and corresponded to the peptide with the amino acid sequence LGEVNTYAGDLQK. A1-A7 are ASD and C1-C7 are controls. Reprinted with permission from [44].

(6)
[image:6.595.52.377.63.355.2]

Figure 3. (A) Validation of the LCMS/MS data (shown in Fig. 2) by WB using anti-ApoA1 and anti-ApoA4 Ab. (B) String anal-ysis for ApoA1, ApoA4, and PON1 proteins for identification of the potential interac-tion partners for these proteins. Reprinted with permission from [44].

significant differences in 101 proteins were reported,

in-cluding those regulating lipid metabolism, oxidative stress,

vision, proteolysis, cell death, and vesicular/membrane

trans-port. Western blot and immunohistochemistry validated

spe-cific protein targets. Cathepsin D, glial fibrillary acidic

pro-tein, Stat3, and

-crystallin were elevated in the SLOS model

retina, and apoE was decreased. Further proteomic studies

of human biomaterials taken from individuals with SLOS

could help elucidate proteomic changes that occur and that

contribute to the disorder, in addition to deficits in sterols.

6

Conclusions

MS-based proteomics can be used for the analysis of

human biomaterials to further understand

neurodevelop-mental disorders. For ASD, MS analysis directed at proteomic

biomarker discovery may provide tools to understand ASD

etiology and potentially aid in ASD identification. As such,

proteomic biomarkers will likely be part of a comprehensive

biomarker signature, rather than individual identifiers.

Ulti-mately, they may provide the potential biological diagnostic

(or predictor) for ASD that does not currently exist, likely

complemented by additional measurements, such as

behav-ioral assessments [110]. Proteomic biomarkers may also be

used for treatment monitoring, and can help further

eluci-date the biological consequences of this disorder. In contrast,

FXS and SLOS, are diagnosed via genotyping. Using

MS-based proteomics methods could supplement the diagnosis

of these syndromes or provide further information, such as

indicators of symptom severity. MS-based analyses could be

employed clinically in FXS and SLOS for biomarker

identifi-cation, directed toward protein target discovery,

understand-ing the consequences of these disorders and for monitorunderstand-ing

medication effects. Although in early stages of

implementa-tion, the use of MS holds great potential in furthering the

general understanding of neurodevelopmental disorders.

The authors would like to thank Dr. Witold Winnik

(Envi-ronment Protection Agency, Research Triangle Park, Durham,

NC) for the MALDI MS analysis of the DIGE spots. This work

was supported in part by the Redcay Foundation (SUNY

Platts-burgh), the Alexander von Humboldt Foundation, SciFund

Chal-lenge, private donations (Ms. Mary Stewart Joyce & Mr. Kenneth

Sandler), and by the U.S. Army research office (DURIP grant

#W911NF-11-1-0304).

The authors have declared no conflict of interest.

7

References

[1] APA,Diagnostic and Statistical Manual of Mental Disor-ders. 5th ed. American Psychiatric Association, Arlington, VA 2013.

[2] Woods, A. G., Mahdavi, E., Ryan, J. P., Treating clients with Asperger’s syndrome and autism.Child Adolesc. Psychiatry Ment. Health2013,7, 32.

(7)

[4] Autism and Developmental Disabilities Monitoring Net-work Surveillance Year 2010 Principal Investigators, and Centers for Disease Control and Prevention, Prevalence of autism spectrum disorder among children aged 8 years— autism and developmental disabilities monitoring network, 11 sites, United States, 2010.MMWR Surveill. Summ.2014, 63, 1–21.

[5] Autism and Developmental Disabilities Monitoring Net-work Surveillance Year 2002 Principal Investigators, and Centers for Disease Control and Prevention, Prevalence of autism spectrum disorders–autism and developmental dis-abilities monitoring network, 14 sites, United States, 2002. MMWR Surveill. Summ. 2007,56, 12–28.

[6] Quaak, I., Brouns, M. R., Van de Bor, M., The dynamics of autism spectrum disorders: how neurotoxic compounds and neurotransmitters interact.Int. J. Environ. Res. Public Health, 2013,10, 3384–3408.

[7] Lai, M. C., Lombardo, M. V., Baron-Cohen, S., Autism. Lancet2014,383, 896–910.

[8] Herbert, M. R., Contributions of the environment and en-vironmentally vulnerable physiology to autism spectrum disorders.Curr. Opin. Neurol. 2010,23, 103–110.

[9] Ousley, O., Cermak, T., Autism Spectrum Disorder: defining Dimensions and Subgroups.Curr. Dev. Disord. Rep.2014, 1, 20–28.

[10] Xu, L. M., Li, J. R., Huang, Y., Zhao, M. et al., AutismKB: an evidence-based knowledgebase of autism genetics. Nu-cleic Acids Res.. 2012,40(Database issue), D1016–D1022. [11] State, M. W., Levitt, P., The conundrums of understanding

genetic risks for autism spectrum disorders.Nat. Neurosci. 2011,14, 1499–1506.

[12] Leblond, C. S., Heinrich, J., Delorme, R., Proepper, C. et al., Genetic and functional analyses of SHANK2 mutations sug-gest a multiple hit model of autism spectrum disorders. PLoS Genet. 2012,8, e1002521.

[13] Banerjee, S., Riordan, M., Bhat, M. A., Genetic aspects of autism spectrum disorders: insights from animal models. Front. Cell. Neurosci. 2014,8, 58.

[14] Bailey, A., Le Couteur, A., Gottesman, I., Bolton, P. et al., Autism as a strongly genetic disorder: evidence from a British twin study.Psychol. Med. 1995,25, 63–77. [15] Le Couteur, A., Bailey, A., Goode, S., Pickles, A. et al.,

A broader phenotype of autism: the clinical spectrum in twins.J. Child Psychol. Psychiatry1996,37, 785–801. [16] Veenstra-VanderWeele, J., Cook, E. H. Jr., Molecular

genet-ics of autism spectrum disorder.Mol. Psychiatry2004,9, 819–832.

[17] Betancur, C., Etiological heterogeneity in autism spectrum disorders: more than 100 genetic and genomic disorders and still counting.Brain Res. 2011,1380, 42–77.

[18] Nickl-Jockschat, T. and Michel, T. M., [Genetic and brain structure anomalies in autism spectrum disorders. Towards an understanding of the aetiopathogenesis?].Nervenarzt 2011,82, 618–627.

[19] Bernier, R., Golzio, C., Xiong, B., Stessman, H. A. et al., Disruptive CHD8 mutations define a subtype of autism early in development.Cell2014,158, 263–276.

[20] Lanz, T. A., Guilmette, E., Gosink, M. M., Fischer, J. E. et al., Transcriptomic analysis of genetically defined autism can-didate genes reveals common mechanisms of action.Mol. Autism2013,4, 45.

[21] Pinto, D., Delaby, E., Merico, D., Barbosa, M. et al., Con-vergence of genes and cellular pathways dysregulated in autism spectrum disorders.Am. J. Hum. Genet. 2014,94, 677–694.

[22] Gurrieri, F., Working up autism: the practical role of medical genetics.Am. J. Med. Genet. C Semin. Med. Genet. 2012, 160C, 104–110.

[23] Carter, M. T., Scherer, S. W., Autism spectrum disorder in the genetics clinic: a review.Clin. Genet. 2013,83, 399–407. [24] Landrigan, P., Lambertini, L., Birnbaum, L., A research strat-egy to discover the environmental causes of autism and neurodevelopmental disabilities.Environ. Health Perspect. 2012,120, a258–a260.

[25] Shelton, J. F., Hertz-Picciotto, I., Pessah, I. N., Tipping the balance of autism risk: potential mechanisms linking pesti-cides and autism.Environ. Health Perspect. 2012,120, 944– 951.

[26] Shelton, J. F., Geraghty, E. M., Tancredi, D. J., Delwiche, L. D. et al., Neurodevelopmental Disorders and Prenatal Res-idential Proximity to Agricultural Pesticides: the CHARGE Study.Environ. Health Perspect. 2014,122, 1103–1109. [27] Howard, P. H. and Muir, D. C., Identifying new persistent and

bioaccumulative organics among chemicals in commerce. Environ. Sci. Technol. 2010,44, 2277–2285.

[28] Woodruff, T. J., Zota, A. R., Schwartz, J. M., Environmen-tal chemicals in pregnant women in the United States: NHANES 2003–2004.Environ. Health Perspect. 2011,119, 878–885.

[29] van der Veen, I., de Boer, J., Phosphorus flame retardants: properties, production, environmental occurrence, toxicity, and analysis.Chemosphere2012,88, 1119–1153.

[30] Reemtsma, T., Quintana, J. B., Rodil, R., Garcia-Lopez, M., Rodriguez, I., Organophosphorus flame retardants and plasticizers in water and air I. Occurrence and fate.Trends Anal. Chem. 2008.27, 727–737.

[31] Stapleton, H. M., Klosterhaus, S., Eagle, S., Fuh, J. et al., Detection of organophosphate flame retardants in furniture foam and U.S. house dust.Environ. Sci. Technol. 2009,43, 7490–7495.

[32] Van den Eede, N., Dirtu, A. C., Neels, H., Covaci, A., An-alytical developments and preliminary assessment of hu-man exposure to organophosphate flame retardants from indoor dust.Environ. Int. 2011,37, 454–461.

[33] Kurebayashi, H., Tanaka, A., Yamaha, T., Metabolism and disposition of the flame retardant plasticizer, tri-p-cresyl phosphate, in the rat.Toxicol. Appl. Pharmacol. 1985,77, 395–404.

[34] Sasaki, K., Suzuki, T., Takeda, M., Uchiyama, M., Metabolism of phosphoric acid triesters by rat liver ho-mogenate.Bull. Environ. Contam. Toxicol. 1984,33, 281– 288.

[35] Nickl-Jockschat, T., Michel, T. M., The role of neurotrophic factors in autism.Mol. Psychiatry2011,16, 478–490.

(8)

[36] Theoharides, T. C., Kempuraj, D., Redwood, L., Autism: an emerging ’neuroimmune disorder’ in search of therapy. Ex-pert Opin. Pharmacother. 2009,10, 2127–2143.

[37] Veenstra-VanderWeele, J., Blakely, R. D., Networking in autism: leveraging genetic, biomarker and model sys-tem findings in the search for new treatments. Neuropsy-chopharmacology2012,37, 196–212.

[38] Woods, A. G., Sokolowska, I., Taurines, R., Gerlach, M. et al., Potential biomarkers in psychiatry: focus on the cholesterol system.J. Cell Mol. Med. 2012,16, 1184–1195.

[39] Lord, C., Risi, S., Lambrecht, L., Cook, E. H. et al., The autism diagnostic observation schedule–generic: a standard mea-sure of social and communication deficits associated with the spectrum of autism.J. Autism Dev. Disord. 2000,30, 205–223.

[40] Lord, C., Risi, S., DiLavore, P. S., Shulman, C. et al., Autism from 2 to 9 years of age.Arch. Gen. Psychiatry2006,63, 694–701.

[41] Dereu, M., Roeyers, H., Raymaekers, R., Meirsschaut, M., Warreyn, P., How useful are screening instruments for tod-dlers to predict outcome at age 4? General development, language skills, and symptom severity in children with a false positive screen for autism spectrum disorder.Eur. Child Adolesc. Psychiatry2012,21, 541–551.

[42] Taurines, R., Dudley, E., Conner, A. C., Grassl, J. et al., Serum protein profiling and proteomics in autistic trum disorder using magnetic bead-assisted mass spec-trometry.Eur. Arch. Psychiatry Clin. Neurosci. 2010,260, 249–255.

[43] Taurines, R., Dudley, E., Grassl, J., Warnke, A. et al., Pro-teomic research in psychiatry.J. Psychopharmacol.2011, 25, 151–196.

[44] Ngounou Wetie, A. G., Wormwood, K., Thome, J., Dudley, E. et al., A pilot proteomic study of protein markers in autism spectrum disorder.Electrophoresis2014,35, 2046–2054. [45] Kotani, K., Yamada, T., Gugliucci, A., Paired measurements

of paraoxonase 1 and serum amyloid A as useful disease markers.Biomed. Res. Int. 2013,2013, 481–437.

[46] Furlong, C. E., Suzuki, S. M., Stevens, R. C., Marsillach, J. et al., Human PON1, a biomarker of risk of disease and exposure.Chem. Biol. Interact. 2010,187, 355–361. [47] Gaidukov, L., Viji, R. I., Yacobson, S., Rosenblat, M. et al.,

ApoE induces serum paraoxonase PON1 activity and sta-bility similar to ApoA-I.Biochemistry2010,49, 532–538. [48] Eskenazi, B., Huen, K., Marks, A., Harley, K. G. et al., PON1

and neurodevelopment in children from the CHAMACOS study exposed to organophosphate pesticides in utero. En-viron. Health Perspect. 2010,118, 1775–1781.

[49] Pasca, S. P., Dronca, E., Nemes, B., Kaucsar, T. et al., Paraox-onase 1 activities and polymorphisms in autism spectrum disorders.J. Cell Mol. Med. 2010,14, 600–607.

[50] Corbett, B. A., Kantor, A. B., Schulman, H., Walker, W. L. et al., A proteomic study of serum from children with autism showing differential expression of apolipoproteins and complement proteins.Mol. Psychiatry2007,12, 292– 306.

[51] Steeb, H., Ramsey, J. M., Guest, P. C., Stocki, P. et al., Serum proteomic analysis identifies sex-specific differences in lipid metabolism and inflammation profiles in adults di-agnosed with Asperger syndrome.Mol. Autism2014,5, 4. [52] Momeni, N., Brudin, L., Behnia, F., Nordstrom, B. et al., High complement factor I activity in the plasma of children with autism spectrum disorders.Autism. Res. Treat.2012.2012, 868576.

[53] Momeni, N., Bergquist, J., Brudin, L., Behnia, F. et al., A novel blood-based biomarker for detection of autism spec-trum disorders.Transl. Psychiatry2012,2, e91.

[54] Loo, J. A., Yan, W., Ramachandran, P., Wong, D. T., Compar-ative human salivary and plasma proteomes.J. Dent. Res. 2010,89, 1016–1023.

[55] Castagnola, M., Messana, I., Inzitari, R., Fanali, C. et al., Hypo-phosphorylation of salivary peptidome as a clue to the molecular pathogenesis of autism spectrum disorders. J. Proteome Res. 2008,7, 5327–5332.

[56] Wetie, A. G., Dekroon, R. M., Mocanu, M., Ryan, J. P. et al., Mass spectrometry for the study of autism and neurodevel-opmental disorders.Adv. Exp. Med. Biol. 2014.806, 525– 544.

[57] Unlu, M., Morgan, M. E., Minden, J. S., Difference gel electrophoresis: a single gel method for detecting changes in protein extracts. Electrophoresis 1997, 18, 2071–2077.

[58] Lilley, K. S., Razzaq, A., Dupree, P., Two-dimensional gel electrophoresis: recent advances in sample preparation, detection and quantitation.Curr. Opin. Chem. Biol. 2002, 6, 46–50.

[59] Tonge, R., Shaw, J., Middleton, B., Rowlinson, R. et al., Val-idation and development of fluorescence two-dimensional differential gel electrophoresis proteomics technology. Pro-teomics2001,1, 377–396.

[60] Schwarz, E., Guest, P. C., Rahmoune, H., Wang, L. et al., Sex-specific serum biomarker patterns in adults with Asperger’s syndrome.Mol. Psychiatry2011,16, 1213–1220.

[61] Lai, M. C., Lombardo, M. V., Suckling, J., Ruigrok, A. N. et al., Biological sex affects the neurobiology of autism. Brain2013,136, 2799–2815.

[62] Ramsey, J. M., Guest, P. C., Broek, J. A., Glennon, J. C. et al., Identification of an age-dependent biomarker signature in children and adolescents with autism spectrum disorders. Mol. Autism2013,4, 27.

[63] Wetie, A. G., Woods, A. G., Darie, C. C., Mass spectro-metric analysis of post-translational modifications (PTMs) and protein-protein interactions (PPIs).Adv Exp. Med. Biol. 2014,806, 205–235.

[64] Ngounou Wetie, A. G., Sokolowska, I., Woods, A. G., Roy, U. et al., Protein-protein interactions: switch from classi-cal methods to proteomics and bioinformatics-based ap-proaches.Cell Mol. Life Sci. 2014,71, 205–228.

(9)

[66] Ngounou Wetie, A. G., Sokolowska, I., Woods, A. G., Roy, U. et al., Investigation of stable and transient protein-protein interactions: past, present, and future.Proteomics2013,13, 538–557.

[67] Emanuele, E., Colombo, R., Martinelli, V., Brondino, N. et al., Elevated urine levels of bufotenine in patients with autistic spectrum disorders and schizophrenia.Neuro. Endocrinol. Lett. 2010,31, 117–121.

[68] Kałuzna-Czaplinska, J., Michalska, M., Rynkowski, J., De-termination of tryptophan in urine of autistic and healthy children by gas chromatography/mass spectrometry.Med. Sci. Monit.2010,16, CR488–CR492.

[69] Lam, K. S., Aman, M. G., Arnold, L. E., Neurochemical cor-relates of autistic disorder: a review of the literature.Res. Dev. Disabil. 2006,27, 254–289.

[70] Pedersen, O. S., Liu, Y., Reichelt, K. L., Serotonin uptake stimulating peptide found in plasma of normal individuals and in some autistic urines.J. Pept. Res.53, 641–646. [71] Sokolowska, I., Woods, A. G., Wagner, J., Dorler, J. et al.,

Mass spectrometry for proteomics-based investigation of oxidative stress and heat shock proteins, in: Andreescu, S., Hepel, M. (Eds.),Oxidative Stress: Diagnostics, Preven-tion, and Therapy. American Chemical Society, Washing-ton, D.C. 2011.

[72] Ghanizadeh, A., Akhondzadeh, S., Hormozi, M., Makarem, A. et al., Glutathione-related factors and oxidative stress in autism, a review.Curr. Med. Chem. 2012,19, 4000–4005. [73] Yao, Y., Walsh, W. J., McGinnis, W. R., Pratico, D., Altered

vascular phenotype in autism: correlation with oxidative stress.Arch. Neurol. 2006,63, 1161–1164.

[74] McCary, L. M., Roberts, J. E., Early identification of autism in fragile X syndrome: a review.J. Intellect. Disabil. Res. 2012,57, 803–814.

[75] Boyle, L., Kaufmann, W. E., The behavioral phenotype of FMR1 mutations.Am. J. Med. Genet. C Semin. Med. Genet. 2010,154C, 469–476.

[76] Hagerman, R., Lauterborn, J., Au, J., Berry-Kravis, E., Frag-ile X syndrome and targeted treatment trials.Results Probl. Cell Differ. 2012,54, 297–335.

[77] McCary, L. M. and Roberts, J. E., Early identification of autism in fragile X syndrome: a review.J. Intellect. Disabil. Res. 2013,57, 803–814.

[78] Smith, L. E., Barker, E. T., Seltzer, M. M., Abbeduto, L., Greenberg, J. S., Behavioral phenotype of fragile X syn-drome in adolescence and adulthood.Am. J. Intellect. Dev. Disabil. 2012,117, 1–17.

[79] Brown, W., The molecular biology of fragile X mutation, in: Hagerman, R., Hagerman, P. J., (Eds.),Fragile X Syndrome: Diagnosis, Treatment, and Research, Johns Hopkins Uni-versity Press: Baltimore, MD 2002, pp. 110–135.

[80] De Rubeis, S., Fernandez, E., Buzzi, A., Di Marino, D., Bagni, C., Molecular and cellular aspects of mental retardation in the Fragile X syndrome: from gene mutation/s to spine dys-morphogenesis.Adv. Exp. Med. Biol. 2012,970, 517–551. [81] Hagerman, R. J., Berry-Kravis, E., Kaufmann, W. E., Ono, M.

Y. et al., Advances in the treatment of fragile X syndrome. Pediatrics2009,123, 378–390.

[82] Berry-Kravis, E. M., Hessl, D., Rathmell, B., Zarevics, P. et al., Effects of STX209 (arbaclofen) on neurobehavioral function in children and adults with fragile X syndrome: a random-ized, controlled, phase 2 trial.Sci. Transl. Med. 2012,4, 152ra127.

[83] Dziembowska, M., Pretto, D. I., Janusz, A., Kaczmarek, L. et al., High MMP-9 activity levels in fragile X syndrome are lowered by minocycline.Am. J. Med. Genet. A2013,161A, 1897–1903.

[84] Leigh, M. J., Nguyen, D. V., Mu, Y., Winarni, T. I. et al., A ran-domized double-blind, placebo-controlled trial of minocy-cline in children and adolescents with fragile x syndrome. J. Dev. Behav. Pediatr. 2013,34, 147–155.

[85] Jacquemont, S., Berry-Kravis, E., Hagerman, R., von Rai-son, F. et al., The challenges of clinical trials in frag-ile X syndrome. Psychopharmacology (Berl) 2013, 231, 1237–1250.

[86] Berry-Kravis, E., Hessl, D., Abbeduto, L., Reiss, A. L. et al., Outcome measures for clinical trials in fragile X syndrome. J. Dev. Behav. Pediatr.2013,34, 508–522.

[87] Wormwood, K., Sokolowska, I., Ryan, J. P., Russell, S. et al., The potential for proteomics in understanding neurodevel-opmental disorders.J. Proteomics Bioinform. 2013,S5. [88] Darie, C. C., Biniossek, M. L., Gawinowicz, M. A., Milgrom,

Y. et al., Mass spectrometric evidence that proteolytic pro-cessing of rainbow trout egg vitelline envelope proteins takes place on the egg.J. Biol. Chem. 2005,280, 37585– 37598.

[89] Klemmer, P., Meredith, R. M., Holmgren, C. D., Klychnikov, O. I. et al., Proteomics, ultrastructure, and physiology of hip-pocampal synapses in a fragile X syndrome mouse model reveal presynaptic phenotype.J. Biol. Chem. 2011,286, 25495–25504.

[90] Liao, L., Park, S. K., Xu, T., Vanderklish, P., Yates, J. R., 3rd, Quantitative proteomic analysis of primary neurons reveals diverse changes in synaptic protein content in fmr1 knockout mice.Proc. Natl. Acad. Sci. USA2008,105, 15281– 15286.

[91] Monzo, K., Dowd, S. R., Minden, J. S., Sisson, J. C., Pro-teomic analysis reveals CCT is a target of Fragile X men-tal retardation protein regulation in Drosophila.Dev. Biol. 2010,340, 408–418.

[92] Zhang, Y. Q., Friedman, D. B., Wang, Z., Woodruff, E., 3rd et al., Protein expression profiling of the drosophila fragile X mutant brain reveals up-regulation of monoamine syn-thesis.Mol. Cell. Proteomics2005,4, 278–290.

[93] Zhang, Y. Q., Matthies, H. J., Mancuso, J., Andrews, H. K. et al., The Drosophila fragile X-related gene regulates ax-oneme differentiation during spermatogenesis.Dev. Biol. 2004,270, 290–307.

[94] Kaufmann, W. E., Cohen, S., Sun, H. T., Ho, G., Molecular phenotype of Fragile X syndrome: FMRP, FXRPs, and pro-tein targets.Microsc. Res. Tech.2002,57, 135–144. [95] Godler, D. E., Slater, H. R., Bui, Q. M., Ono, M. et al., FMR1

intron 1 methylation predicts FMRP expression in blood of female carriers of expanded FMR1 alleles.J. Mol. Diagn. 2011,13, 528–536.

(10)

[96] Godler, D. E., Slater, H. R., Bui, Q. M., Storey, E. et al., Fragile X mental retardation 1 (FMR1) intron 1 methylation in blood predicts verbal cognitive impairment in female carriers of expanded FMR1 alleles: evidence from a pilot study.Clin. Chem. 2012,58, 590–598.

[97] Aneja, A., Tierney, E., Autism: the role of cholesterol in treatment.Int. Rev. Psychiatry2008,20, 165–170.

[98] Bukelis, I., Porter, F. D., Zimmerman, A. W., Tierney, E., Smith-Lemli-Opitz syndrome and autism spectrum disor-der.Am. J. Psychiatry2007,164, 1655–1661.

[99] Diaz-Stransky, A., Tierney, E., Cognitive and behavioral as-pects of Smith-Lemli-Opitz syndrome.Am. J. Med. Genet. C Semin. Med. Genet. 2012,160C, 295–300.

[100] Tierney, E., Bukelis, I., Thompson, R. E., Ahmed, K. et al., Abnormalities of cholesterol metabolism in autism spec-trum disorders. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2006,141B, 666–668.

[101] DeBarber, A. E., Eroglu, Y., Merkens, L. S., Pappu, A. S., Steiner, R. D., Smith-Lemli-Opitz syndrome. Expert Rev. Mol. Med. 2011,13, e24.

[102] Fitzky, B. U., Moebius, F. F., Asaoka, H., Waage-Baudet, H. et al., 7-Dehydrocholesterol-dependent proteolysis of HMG-CoA reductase suppresses sterol biosynthesis in a mouse model of Smith-Lemli-Opitz/RSH syndrome.J. Clin. Invest. 2001,108, 905–915.

[103] Corso, G., Gelzo, M., Barone, R., Clericuzio, S. et al., Sterol profiles in plasma and erythrocyte membranes in patients with Smith-Lemli-Opitz syndrome: a six-year experience. Clin. Chem. Lab. Med. 2011,49, 2039–2046.

[104] Griffiths, W. J., Wang, Y., Karu, K., Samuel, E. et al., Po-tential of sterol analysis by liquid chromatography-tandem mass spectrometry for the prenatal diagnosis of Smith-Lemli-Opitz syndrome.Clin. Chem. 2008,54, 1317–1324. [105] Meljon, A., Watson, G. L., Wang, Y., Shackleton, C. H.,

Griffiths, W. J., Analysis by liquid chromatography-mass spectrometry of sterols and oxysterols in brain of the newborn Dhcr7(Delta3–5/T93M) mouse: a model of Smith-Lemli-Opitz syndrome. Biochem. Pharmacol. 2013, 86, 43–55.

[106] Paglia, G., D’Apolito, O., Gelzo, M., Dello Russo, A., Corso, G., Direct analysis of sterols from dried plasma/blood spots by an atmospheric pressure thermal desorption chemical ionization mass spectrometry (APTDCI-MS) method for a rapid screening of Smith-Lemli-Opitz syndrome.Analyst 2010,135, 789–796.

[107] Patti, G. J., Shriver, L. P., Wassif, C. A., Woo, H. K. et al., Nanostructure-initiator mass spectrometry (NIMS) imag-ing of brain cholesterol metabolites in Smith-Lemli-Opitz syndrome.Neuroscience2010,170, 858–864.

[108] Tu, C., Li, J., Jiang, X., Sheflin, L. G. et al., Ion current based proteomic profiling of the retina in a rat model of Smith-Lemli-Opitz syndrome.Mol. Cell Proteomics2013,12, 3583– 3598.

[109] Fliesler, S. J., Retinal degeneration in a rat model of Smith-Lemli-Opitz Syndrome: thinking beyond cholesterol defi-ciency.Adv. Exp. Med. Biol. 2010,664, 481–489.

Gambar

Figure 1. Difference gel electrophoresis (DIGE). (A) Analytical DIGE gel, where protein samples were labeled with fluorescent cyanine dyes
Figure 2. (A) Nano-LC-MS/MS analysis of the sera from children with ASD and matched controls
Figure 3. (A) Validation of the LCMS/MSdata (shown in Fig. 2) by WB using anti-ApoA1 and anti-ApoA4 Ab

Referensi

Dokumen terkait

Tidak ada pengaruh pemberian terapi relaksasi terhadap jumlah hafalan Al- Qur‟an santriwati Pondok Pesantren Tahfizhul Qur‟an Daarul Qur‟an Colomadu.. Ada pengaruh pemberian

MICROSCOPY) KOMPOSIT BERMATRIK EBONIT DENGAN KANDUNGAN SULFUR 40 PHR YANG DIPERKUAT SERAT RAMI.. DAN BAMBU DENGAN KANDUNGAN MASING-MASING

Namun pihak keluarga menyadari bahwa motivasi dari tenaga kesehatan serta dukungan dari orang terdekat dengan klien merupakan kebutuhan yang harus dipenuhi agar

Ekstrak etanol umbi ubi jalar ungu dan oranye dilakukan uji sitotoksik dengan metode MTT, dilanjutkan dengan uji kualitatif golongan senyawa yang terkandung di dalam

Ekstrak etanol umbi ubi jalar ungu ( Ipomoea batatas L.) mengandung flavonoid jenis antosianin yang memiliki aktivitas sitotoksik dan menghambat proliferasi.

Tes Tertulis : melakukan penjumlahan dan pengurangan, perkalian dan pembagian bilangan asli , bilangan bulat dan pecahan sesuai prosedur/aturan dengan memperhatikan nilai

 Mengidentifikasi berbagai bentuk energi (panas, listrik, gerak) dan menjelaskan kegunaannya dalam kehidupan sehari-hari secara lisan dan tulis.  Menyebutkan contoh-contoh

Penelitian ini merupakan penelitian tindakan kelas (Classroom Action Research). Penelitian dilakukan di kelas II SDN Nayu Banjarsari Surakarta pada semester II tahun pelajaran