Chemical names of VPA and derivatives
1. Introduction
Autism spectrum disorders (ASD) have attracted public attention by its high prevalence, elevated social cost and large impact on the family [1]. Since the first descriptions of autism made by Hans Asperger in 1938 [2] and by Leo Kanner in 1943 [3, 4], much discussion has focused in the search for the triggering points of autism and identifying risk factors has become a high priority of scientists. Nevertheless, even after almost seventy years since the first reports, the etiology of autism remains unknown and its molecular basis is not well understood. Environmental factors (such as virus, bacteria, drugs, etc.) known to increase the risk of autism have critical periods of action during embryogenesis. Congenital syndromes are found in high rates in patients with autism including somatic changes originated early in the first trimester [5].
The link between rubella and autism came from epidemic rubella in which the incidence of autism diagnosis in prenatally exposed offspring was more than 10-fold higher than normal. The study describes 243 children exposed to congenital rubella, where 25% presented mental retardation, 15% had reactive behavior and 7% was included in the autism spectrum [6].
Valproic acid (VPA) has traditionally been prescribed for epilepsy, but is increasingly used for psychiatric condition, such as bipolar disease by its modulation on GABA neurotransmission [7]. Furthermore, it has been also shown to be associated with an increased prevalence of autism. In fact, prospective and retrospective studies demonstrate that exposure to VPA during pregnancy is associated with approximately three-fold increase in the rate of major anomalies and a possible set of dysmorphic features with decreased intrauterine growth [8, 9], characteristics of Fetal Valprotate Syndrome (FVS) described in item 3. Histone deacetylase (HDAC) inhibition by VPA and changes in gene expression may explain part of the teratogenicity of this drug.
Thus,
2. Valproic acid
The compound VPA (Figure 1A) is a fatty acid synthesized in 1882 [15] as an analogue of valeric acid, found naturally in valerian (
Valproic Acid | 2-Propylvaleric acid, 2-Propylpentanoic acid or Di-n-dipropylacetic acid |
Sodium Valproate | Sodium 2-propylvalerate |
Valproate semisodium | Sodium hydrogen bis(2-propylvalerate) |
Valproate Pivoxil | Hydroxymethyl 2-propylvalerate pivalate |
Valpromide | 2-Propylvaleramide |
Table 1.

Figure 1.
The molecular structure of VPA and derivatives showed in ball and stick view. A. Valproic acid. B. Valproate semisodium, C. Sodium valproate. In A is possible to compare both chemical and ball and stick structures (used also to illustrate derivatives).
The therapeutic concentration of sodium valproate (the sodium salt of VPA) during chronic oral treatment ranges from 40-100 mg/mL (280–700 mmol/L) in plasma and from 6–27 mg/g (42–190 mmol/g) in brain [18]. From this point, to simplify the reading throughout the text, the VPA abbreviation will be used when referring to valproic acid and derivatives.
The VPA is marketed under brand names including: Convulex (Pfizer-UK and Byk Madaus-South Africa), Depakene (Abbott Laboratories-USA, Brazil and Canada), Depakine (Sanofi Aventis-France and Sanofi Synthelabo-Romania), Deprakine (Sanofi Aventis-Finland), Encorate (Sun Pharmaceuticals-India), Epilim (Sanofi Synthelabo-Australia), Valcote (Abbot Laboratories-Argentina).
The VPA effects of clinical importance include GABAergic activity increase, excitatory neurotransmission decrease, and modification of monoamines [19]. The biochemical and biological effects of VPA are summarized in Table 2.
|
|
|
HDAC (inhibition) | Open DNA transcription | [20] |
Mitochondria | Energy metabolism impairment | [21] |
Lymphocytes | Modification of the epigenotype | [22] |
Neurons from |
Reduction in firing rate | [23] |
c-Jun N-terminal kinase (JNK) | Defective neurite formation | [24] |
GSK3β inhibitor | Promotion of hair re-growth | [25] |
Beta-catenin-Ras-ERK-p21Cip/WAF1 pathway | Differentiation and inhibition of proliferation in neural progenitor cells | [26] |
Constitutive androstane receptor (CAR) and pregnane X receptor (PXR) | Up-regulation of |
[27] |
Matrix metalloprotease-9 inhibitor | Attenuation of blood-spinal cord barrier (BSCB) after spinal cord injury (SCI) | [28] |
PI3K/Akt/mTOR pathway | Skeletal muscle hypertrophy | [29] |
Table 2.
Biochemical and biological effects of VPA
3. Valproic acid as an environmental risk factor in the development of autism
After the VPA license for use in 1978, the first adverse report of a fetus exposed to the drug was published in 1980 [30]. Since then. particular attention has been directed to the occurrence of neural tube defects in infants exposed to VPA
The timing for the teratogenic effect of VPA that increases the risk for autism cannot be estimated directly, as the drug is typically taken throughout the entire pregnancy [33]. Many children exposed
The classical autism was first reported to be one of the behavioral outcomes of VPA exposure [41] through several case reports [12, 39, 45]. The first epidemiological study with drugs as environmental risk factors of autism was described in 2000, with 57 offspring of women taking anticonvulsants (see ref [46], summarized in Tables 3 and 4).
|
|
Poor social interaction | 53 |
Poor communication skills | 49 |
Short attention span | 46 |
Insistence on routines | 44 |
Hand flapping | 25 |
Table 3.
Autism features in children exposed
|
|
|
Neural tube defect | 1 1 |
1000 (0-5), 800 (5-40) 1200 (1-17), 1500 (17-26), 2000 (26-40) |
Genitourinary | 1 1 5 |
1000 (0-40) 1000 (0-5), 800 (5-40) 1500 (0-40) |
Extremities | 2 1 |
1000 (0-40) 1500 (0-40) |
Eyes | 1 1 |
700 (0-40) 800 (0-40) |
Teeth | 2 1 |
1000 (0-40) 1700 (0-40) |
Diastasis recti | 1 | 1200 (0-40) |
Table 4.
Congenital malformations in children exposed
Fifty two children were ascertained through the Fetal Anticonvulsant Syndrome Association (FACS) and five were referred to the Aberdeen Medical Genetics Service (AMGS). The number of patients exposed
4. Animal model of autism induced by prenatal exposure to VPA
Considering human evidences of autism followed by early
|
|
|
Skeletal abnormalities | Mice | [48, 49] |
Rabbits | [50] | |
Rhesus monkey | [51] | |
Cardiac abnormalities | Mice | [52] |
Neural tube defects (including spina bifida) | Mice | [53] |
Cranial neural tube defects | Rats | [54] |
Behavioral abnormalities | Rats | [55] |
Table 5.
Patterns of abnormal development across species after
The use of animal models allows a wide range of research possibilities including the search for etiologic clues, molecular targets, and biomarkers. The main aspects to take into account in developing animal models, is (i) to reproduce a circumstance that would lead to a certain condition, for example, inducing a genetic disease by manipulating a specific gene; (ii) to induce similar patterns found in the studied condition, for example, observing the same behavioral alterations found in a particular impairment; (iii) to observe if the model has similarities to a human features when exposed to certain treatment [56]. The time of induction, dosage of VPA and the way of administration in rodents are variable in the literature, as demonstrated in Table 6. It is important to observe that in rats, 600 mg/Kg VPA at 12.5 days of pregnancy is the most investigated due to similarities in the features of autism. Besides the higher number of studies describing prenatal exposure to VPA, there are some protocols reporting also postnatal exposure and behavioral features of autism [57, 58].
|
Procedure (mg/Kg VPA) |
|
|
||
9 | IP (200, 400, 800) | [59] |
9 | SC (400) | [60] |
9, 12.5, 14.5 | IP (500) | [61] |
11 | OA (800) | [62, 63] |
12, 13, 14 | IP (100) | [64] |
12.5 | IP (500) | [65] |
13 | SC (600) | [66, 67] |
|
||
7, 9.5, 12, 15 | IP (400) | [68] |
8, 9, 10, 11 | OA (800) | [69] |
9 | IP (600) | [70] |
9 | OA (800) | [71-74] |
9, 11 | AO (800) | [75, 76] |
11, 12, 13 | IP (200) | [77] |
1.5 | IP (500) | [78] |
1.5, 12, 12.5 | IP (350) | [9] |
12 | IP (400) | [79] |
12 | IP (600) | [80-83] |
12.5 | IP (600) | [10, 14, 84-93] |
12.5 | SC (350) | [94] |
12.5 | IP (350) | [95] |
12.5 | IP (400, 500, 600) | [77] |
12.5 | IP (500) | [96] |
Table 6.
Prenatal exposure of VPA in rodents: Time of induction and dosage
The diagnoses of autism take into account behavioral alterations in three main areas: sociability, communication and behavioral stereotypies and narrow range of interests. Therefore, a consistent animal model must show similar behavioral abnormalities, which might indicate common neural alterations.
Our group has administrated a single intraperitoneal injection of 600 mg/kg VPA in pregnant rats at the embryonic day 12.5, observing variations in social memory, and flexibility to change strategy [84]. Females were kept separate and with free access to their own litters. Somatic aspects observed during the pups' development, included body weight, ear unfolding and eye opening which were unchanged between groups. In three-chambered-apparatus test, used to observe social memory, preferences and interests, the VPA group spent less time in the presence of a stranger rat and more time in the presence of an object, indicating a reduced place preference conditioned by conspecific and an increased preference for the object, revealing sociability impairments. As adults, they showed inappropriate social approach to a stranger rat, decreased preference for social novelty, apparently normal social recognition, no spatial learning deficits and normal resistance to change on Morris water maze.
5. Brain alterations induced by prenatal exposure to VPA
Once prenatal exposure to VPA became a reliable tool to model autism, more brain alterations were investigated in rodents exposed to this teratogen, as summarized in Table 7.
|
|
|
|
Rat | 500 | Altered distribution of 5-HT neurons in the dorsal raphe nucleus. | [71] |
350 | Reduction in the number of motor neurons from hypoglossal and oculomotor nuclei. | [9] | |
500 | Reduction in the number of putative synaptic contacts in connection between layer 5 pyramidal neurons. | [97] | |
400 | Prolonged neuronal progenitor cells proliferation in embrionary period. | [79] | |
600 | Decreased number of purkinje cells, neuronal degeneration and chromatolysis. | [98] | |
Mice | 500 | Reduction in the number of Parvalbumin -positive inhibitory neurons in the neocortex. | [99] |
500 | Nissl-positive cell loss in the middle and lower layers of the prefrontal cortex and in the lower layers of the somatosensory cortex. | [65] |
Table 7.
Brain alterations induced by
Behavioral outcomes started to be studied, demonstrating a number of anatomic and behavioral features characteristic of human cases by exposing rodents’ embryos to VPA at the time of neural tube closure. One of the affected structures in the brains is the cerebellum. Magnetic resonance imaging showed that patients with autism have reduced size of the cerebellum when compared to controls, displaying smaller vermal lobules VI and VII. This abnormality is probably an outcome of developmental hypoplasia and not likely shrinkage or deterioration after full development had been achieved [100]. Similar alterations were found in brains from rat model of autism induced by prenatal exposure to VPA [9]. Exposed rats showed a reduction in the number of motor neurons of the earliest-forming motor nuclei (V, XII), and had the VI th and III rd cranial nerve nuclei affected. In the same way, another work found diminished number of cells in the posterior lobe of the cerebellum [86]. In this context, cerebellar anatomy alterations in humans might be due to loss of neurons in the cranial nerve motor nuclei, as demonstrated in rats.
The amygdala is likely to be also linked to autism, due to its involvement in social-emotional behavior. Rats exposed to VPA
Synaptic impairments were already described in autism, which may be related to neuroligins alterations. Neuroligins are a family of proteins which play a central role in synaptic maturation and were affected in rats after
Synaptic plasticity is influenced by brain-derived neurotrophic factor (BDNF), a factor that modulates several neurochemical parameters. High levels of BDNF have been reported in the blood of patients with autism [102]. BDNF acts through TrkB-mediated activation of various signal transduction pathways, including pathways that involve PI3K, mitogen-activated kinase (MAPK), and phospholipase C-γ [103]. Infusion of BDNF in the nucleus
Several hypotheses have arisen to explain the social deficits in autism. One of these proposals points an alteration in opioidergic mechanisms as a likely causative of behavioral impairments in this disorder [107]. Opioid peptides are involved in stress responses and affective states, and blockage of their receptors causes dysphoria in humans. Enkephalins are part of the opioid family and are distributed in brain areas, like the striatum and the nucleus accumbens, involved in processing emotional information, anxiety and fear. Exposure to VPA reduced proenkephalin mRNA expression in both the core and shell of the nucleus
The monoamine system is also altered in patients with autism and their relatives. It was demonstrated that children with autism have increased 5-HT (serotonin) synthesis capacity when compared to children with typical development [108]. Besides, it is widely known that sleep disorders are common in autistic children [109]. Interestingly, increased levels of serotonin was found in pre-frontal cortex of rats prenatally exposed to VPA in association with disrupted sleep/awake rhythm. The elevated levels of 5-HT were found during light phase of animals’ circadian rhythm [74]. It was proved that serotonergic neurons have a silent firing rate during REM sleep [110], indicating that the sleep disturbance found in the animals may be related to increased levels of 5-HT found in their prefrontal cortex. In addition, higher levels of 5-HT were also reported in the left side of hippocampus and in blood from rats [111]. However, using the whole hippocampus, it was demonstrated 46% decrease in 5-HT levels from rats exposed to VPA
Recently, we observed hippocampal reactive astrogliosis in the group of rats exposed
Seven Fresh-frozen

Figure 2.
Astrocyte immunoreactive to GFAP in hippocampus from rats. A. Representative image from control group, B. Representative image from VPA group. Scale bar = 50 μm
Glutamatergic excitatory synapses are the major type of synapses in the brain and it was found that glutamate metabolism is altered in autistic CNS, particularly the glutamate receptors AMPA, NMDA and mGluR5 [114]. In agreement, rats exposed
Although social impairments are one of the most important features observed in autism, patients present several other symptoms, including motor disturbances. Motor stereotypies are part of the so called autism triad of impairments, but hypotonia, motor apraxia, toe-walking, have already been reported [116]. Evaluation of motor cortex neurons of rats exposed to VPA
The superior olivary complex (SOC) plays different roles in hearing. It is located within the lower brainstem and it is involved in encoding temporal features of sound and descending modulation of the cochlear nucleus and cochlea for listening in background noise. Rats exposed to VPA
The cerebellum have been the focus of studies involving active and chronic neuroinflammatory process in autistic patients, demonstrating the presence of proinflammatory chemokines such as MCP-1 as well as antiinflammatory cytokines such as TGF-β1 in this brain structure. These findings support the idea that a chronic state of specific cytokine activation occurs in autism [113]. Because neuroimmune responses are influenced by the genetic background of the host, the role of neuroinflammation in the context of the genetic and other factors that determine the autism phenotype remains an important issue to be investigated.
6. Concluding remarks and scientific challenges
The spectrum of autism comprises a multifactorial group of disorders, with phenotypic diversity related to the symptoms and increasing prevalence. One of the major challenges of cognitive neuroscience is to understand how changes in the structural properties of the brain affect the plasticity exhibited whenever a person develops, ages, learns a new skill, make social interaction or adapts to a disease. In ASD, it is necessary studies in this field attempting to explain and understand the trigger of autism. In this context, it is not easy to find a single animal model able to captures the entire molecular and cellular alterations observed in patients with ASD.
Studies of
The present chapter summarizes findings obtained in rodents exposed
References
- 1.
A global public health strategy for autism spectrum disorders. Autism Res.Wallace S Fein D Rosanoff M Dawson G Hossain S Brennan L et al 2012 Jun;5 3 211 7 - 2.
Das psychisch abnormale Kind [The psychically abnormal child]. Wien Klin Wochenschr.Asperger H 1938 - 3.
Autistic Disturbances of Affective Contact. Nervous Child.Kanner L 1943 1943 2 217 50 - 4.
Early infantile autism,Kanner L Eisenberg L 1943 1955 Psychiatr Res Rep Am Psychiatr Assoc. 1957 Apr(7):55-65. - 5.
Autism and environmental influences: review and commentary. Rev Environ Health.Bello S. C 2007 Apr-Jun;22 2 139 56 - 6.
Follow-up report on autism in congenital rubella. J Autism Child Schizophr.Chess S 1977 Mar;7 1 69 81 - 7.
A review of valproate in psychiatric practice. Expert Opin Drug Metab Toxicol.Haddad P. M Das A Ashfaq M Wieck A 2009 May;5 5 539 51 - 8.
Verification of the fetal valproate syndrome phenotype. Am J Med Genet.Ardinger H. H Atkin J. F Blackston R. D Elsas L. J Clarren S. K Livingstone S et al 1988 Jan;29 1 171 85 - 9.
Embryological origin for autism: developmental anomalies of the cranial nerve motor nuclei. J Comp Neurol.Rodier P. M Ingram J. L Tisdale B Nelson S Romano J 1996 Jun 24;370 2 247 61 - 10.
Class I histone deacetylase inhibition ameliorates social cognition and cell adhesion molecule plasticity deficits in a rodent model of autism spectrum disorder. Neuropharmacology.Foley A. G Gannon S Rombach-mullan N Prendergast A Barry C Cassidy A. W et al 2012 Sep;63 4 750 60 - 11.
Amelioration of behavioral aberrations and oxidative markers by green tea extract in valproate induced autism in animals. Brain Res.Banji D Banji O. J Abbagoni S Hayath M. S Kambam S Chiluka V. L 2011 Sep 2;1410 141 51 - 12.
Fetal valproate syndrome: clinical and neuro-developmental features in two sibling pairs. Dev Med Child Neurol.Christianson A. L Chesler N Kromberg J. G 1994 Apr;36 4 361 9 - 13.
Teratogenicity of sodium valproate. Expert Opin Drug Saf.Alsdorf R Wyszynski D. F 2005 Mar;4 2 345 53 - 14.
Behavioral alterations in rats prenatally exposed to valproic acid: animal model of autism. Neuropsychopharmacology.Schneider T Przewlocki R 2005 Jan;30 1 80 9 - 15.
On the propyl derivatives and decomposition products of ethylacetoacetate. American Chemistry Journal.Burton B. S 1882 18 3 385 95 - 16.
Pharmacodynamic properties of N-dipropylacetic acid]. Therapie.Meunier H Carraz G Neunier Y Eymard P Aimard M 1963 Mar-Apr;18 435 8 - 17.
Correlation of valproate plasma concentrations and dose in bipolar affective disorder. J Clin Psychopharmacol.Perry P. J Bever-stille K. A Arndt S Gundersen S 2000 Apr;20 2 277 9 - 18.
Valproic acid metabolism and its effects on mitochondrial fatty acid oxidation: A review. J Inherit Metab Dis.Silva M. F Aires C. C Luis P. B Ruiter J. P Ijlst L Duran M et al 2008 Apr 4. - 19.
Molecular and therapeutic potential and toxicity of valproic acid. J Biomed Biotechnol.Chateauvieux S Morceau F Dicato M Diederich M 2010 - 20.
Valproate administration to mice increases histone acetylation and 5-lipoxygenase content in the hippocampus. Neurosci Lett.Yildirim E Zhang Z Uz T Chen C. Q Manev R Manev H 2003 Jul 17;345 2 141 3 - 21.
Fetal valproate syndrome. J Med Genet.Clayton-smith J Donnai D 1995 Sep;32 9 724 7 - 22.
Valproate induces epigenetic modifications in lymphomonocytes from epileptic patients. Prog Neuropsychopharmacol Biol Psychiatry.Tremolizzo L Difrancesco J. C Rodriguez-menendez V Riva C Conti E Galimberti G et al 2012 Oct;39 1 47 51 - 23.
Reduction in firing rate of substantia nigra pars reticulata neurons by valproate: influence of different types of anesthesia in rats. Brain Res.Löscher W Rohlfs A Rundfeldt C 1995 Dec;702(1-2):133-44. - 24.
The mood stabilizer valproic acid improves defective neurite formation caused by Charcot-Marie-Tooth disease-associated mutant Rab7 through the JNK signaling pathway. J Neurosci Res.Yamauchi J Torii T Kusakawa S Sanbe A Nakamura K Takashima S et al 2010 Nov;88 14 3189 97 - 25.
Valproic acid induces hair regeneration in murine model and activates alkaline phosphatase activity in human dermal papilla cells. PLoS One.Lee S. H Yoon J Shin S. H Zahoor M Kim H. J Park P. J et al 2012 e34152. - 26.
Valproic acid induces differentiation and inhibition of proliferation in neural progenitor cells via the beta-catenin-Ras-ERK-Jung G. A Yoon J. Y Moon B. S Yang D. H Kim H. Y Lee S. H et al 21Cip WAF1 pathway. BMC Cell Biol.2008 - 27.
Valproic acid induces CYP3A4 and MDR1 gene expression by activation of constitutive androstane receptor and pregnane X receptor pathways. Drug Metab Dispos.Cerveny L Svecova L Anzenbacherova E Vrzal R Staud F Dvorak Z et al 2007 Jul;35 7 1032 41 - 28.
Valproic acid attenuates blood-spinal cord barrier disruption by inhibiting matrix metalloprotease-9 activity and improves functional recovery after spinal cord injury. J Neurochem.Lee J. Y Kim H. S Choi H. Y Oh T. H Ju B. G Yune T. Y 2012 Jun;121 5 818 29 - 29.
Valproic acid activates the PI3K/Akt/mTOR pathway in muscle and ameliorates pathology in a mouse model of Duchenne muscular dystrophy. Am J Pathol.Gurpur P. B Liu J Burkin D. J Kaufman S. J 2009 Mar;174 3 999 1008 - 30.
Teratogenicity of valproic acid. J Pediatr.Dalens B Raynaud E. J Gaulme J 1980 Aug;97 2 332 3 - 31.
Maternal epilepsy, valproate exposure, and birth defects. Lancet.Mastroiacovo P Bertollini R Morandini S Segni G 1983 Dec24 31 - 32.
Valproate and birth defects. Lancet.Robert E Rosa F 1983 Nov 12;2(8359):1142. - 33.
The teratology of autism. Int J Dev Neurosci.Arndt T. L Stodgell C. J Rodier P. M 2005 Apr-May;23(2-3):189-99. - 34.
Autism in fetal alcohol syndrome: a report of six cases. Alcohol Clin Exp Res.Nanson J. L 1992 Jun;16 3 558 65 - 35.
Autism in thalidomide embryopathy: a population study. Dev Med Child Neurol.Stromland K Nordin V Miller M Akerstrom B Gillberg C 1994 Apr;36 4 351 6 - 36.
Pregnancy and the risk of teratogenicity. Epilepsia.Lindhout D Omtzigt J. G 1992 Suppl 4:S41 8 - 37.
DiLiberti JH Farndon PA, Dennis NR, Curry CJ. The fetal valproate syndrome. Am J Med Genet.1984 Nov;19 3 473 81 - 38.
Foetal valproate syndrome and autism: additional evidence of an association’. Dev Med Child Neurol.Bescoby-chambers N Forster P Bates G 2001 Dec;43(12):847. - 39.
Fetal valproate syndrome and autism: additional evidence of an association. Dev Med Child Neurol.Williams G King J Cunningham M Stephan M Kerr B Hersh J. H 2001 Mar;43 3 202 6 - 40.
Fetal valproate syndrome. Am J Med Genet.Chessa L Iannetti P 1986 Jun;24 2 381 2 - 41.
Dysmorphic features: an important clue to the diagnosis and severity of fetal anticonvulsant syndromes. Arch Dis Child Fetal Neonatal Ed.Kini U Adab N Vinten J Fryer A Clayton-smith J 2006 Mar;91(2):F90 5 - 42.
Fetal valproate syndrome: is there a recognisable phenotype? J Med Genet.Winter R. M Donnai D Burn J Tucker S. M 1987 Nov;24 11 692 5 - 43.
Valproic acid embryopathy: report of two siblings with further expansion of the phenotypic abnormalities and a review of the literature. Am J Med Genet.Kozma C 2001 Jan 15;98 2 168 75 - 44.
Valproate embryopathy in three sets of siblings: further proof of hereditary susceptibility. Neurology.Malm H Kajantie E Kivirikko S Kaariainen H Peippo M Somer M 2002 Aug 27;59 4 630 3 - 45.
A male with fetal valproate syndrome and autism. Dev Med Child Neurol.Williams P. G Hersh J. H 1997 Sep;39 9 632 4 - 46.
A clinical study of 57 children with fetal anticonvulsant syndromes. J Med Genet.Moore S. J Turnpenny P Quinn A Glover S Lloyd D. J Montgomery T et al 2000 Jul;37 7 489 97 - 47.
Linking etiologies in humans and animal models: studies of autism. Reprod Toxicol.Rodier P. M Ingram J. L Tisdale B Croog V. J 1997 Mar-Jun;11(2-3):417-22. - 48.
Teratogenic potential of valproic acid. Lancet.Brown N. A Kao J Fabro S 1980 Mar 22;1 8169 660 1 - 49.
RC. Teratogenic effects of valproic acid and diphenylhydantoin on mouse embryos in culture. Teratology.Bruckner A Lee Y. J O Shea K. S Henneberry 1983 Feb;27 1 29 42 - 50.
Teratogenesis of calcium valproate in rabbits. Teratology.Petrere J. A Anderson J. A Sakowski R Fitzgerald J. E De La Iglesia F. A 1986 Dec;34 3 263 9 - 51.
Predicting the human teratogenic potential of the anticonvulsant, valproic acid, from a non-human primate model. Toxicology.Mast T. J Cukierski M. A Nau H Hendrickx A. G 1986 May;39 2 111 9 - 52.
Sodium valproate-induced cardiovascular abnormalities in the Jcl:ICR mouse fetus: peak sensitivity of gestational day and dose-dependent effect. Teratology.Sonoda T Ohdo S Ohba K Okishima T Hayakawa K 1993 Aug;48 2 127 32 - 53.
Valproic acid-induced spina bifida: a mouse model. Teratology.Ehlers K Sturje H Merker H. J Nau H 1992 Feb;45 2 145 54 - 54.
Teratogenic effects on the neuroepithelium of the CD-1 mouse embryo exposed in utero to sodium valproate. Teratology.Turner S Sucheston M. E De Philip R. M Paulson R. B 1990 Apr;41 4 421 42 - 55.
Behavioral teratogenicity of valproic acid: selective effects on behavior after prenatal exposure to rats. Psychopharmacology (Berl).Vorhees C. V 1987 92 2 173 9 - 56.
Mouse behavioral assays relevant to the symptoms of autism. Brain Pathol.Crawley J. N 2007 Oct;17 4 448 59 - 57.
VPA-induced apoptosis and behavioral deficits in neonatal mice. Brain Res.Yochum C. L Dowling P Reuhl K. R Wagner G. C Ming X 2008 Apr 8;1203 126 32 - 58.
A new neurobehavioral model of autism in mice: pre- and postnatal exposure to sodium valproate. J Autism Dev Disord.Wagner G. C Reuhl K. R Cheh M Mcrae P Halladay A. K 2006 Aug;36 6 779 93 - 59.
Genetic and maternal effects on valproic acid teratogenesis in C57BL/6J and DBA/2J mice. Toxicol Sci.Downing C Biers J Larson C Kimball A Wright H Ishii T et al 2010 Aug;116 2 632 9 - 60.
Valproic acid increases formation of reactive oxygen species and induces apoptosis in postimplantation embryos: a role for oxidative stress in valproic acid-induced neural tube defects. Mol Pharmacol.Tung E. W Winn L. M 2011 Dec;80 6 979 87 - 61.
Autism-like behaviours with transient histone hyperacetylation in mice treated prenatally with valproic acid. Int J Neuropsychopharmacol.Kataoka S Takuma K Hara Y Maeda Y Ago Y Matsuda T 2011 Nov18 1 13 - 62.
Prenatal exposure to valproic acid leads to reduced expression of synaptic adhesion molecule neuroligin 3 in mice. Neuroscience.Kolozsi E Mackenzie R. N Roullet F. I Decatanzaro D Foster J. A 2009 Nov 10;163 4 1201 10 - 63.
Behavioral and molecular changes in the mouse in response to prenatal exposure to the anti-epileptic drug valproic acid. Neuroscience.Roullet F. I Wollaston L Decatanzaro D Foster J. A 2010 Oct 13;170 2 514 22 - 64.
Transplacental genotoxicity of antiepileptic drugs: animal model and pilot study on mother/newborn cohort. Reprod Toxicol.Fucic A Stojkovic R Miskov S Zeljezic D Markovic D Gjergja R et al 2010 Dec;30 4 613 8 - 65.
Effect of prenatal valproic acid exposure on cortical morphology in female mice. J Pharmacol Sci.Hara Y Maeda Y Kataoka S Ago Y Takuma K Matsuda T 2012 118 4 543 6 - 66.
Validating gamma oscillations and delayed auditory responses as translational biomarkers of autism. Biol Psychiatry.Gandal M. J Edgar J. C Ehrlichman R. S Mehta M Roberts T. P Siegel S. J 2010 Dec 15;68 12 1100 6 - 67.
5-antagonist mediated reversal of elevated stereotyped, repetitive behaviors in the VPA model of autism. PLoS One. 2011;6(10):e26077. ,Mehta MV ,Gandal MJ .Siegel SJ mGlu R - 68.
The critical period of valproate exposure to induce autistic symptoms in Sprague-Dawley rats. Toxicol Lett.Kim K. C Kim P Go H. S Choi C. S Yang S. I Cheong J. H et al 2011 Mar 5;201 2 137 42 - 69.
Morphological abnormalities of embryonic cranial nerves after in utero exposure to valproic acid: implications for the pathogenesis of autism with multiple developmental anomalies. Int J Dev Neurosci.Tashiro Y Oyabu A Imura Y Uchida A Narita N Narita M 2011 Jun;29 4 359 64 - 70.
Le Guisquet AM, Garreau L, Ternant D, Bodard S, et al. Behavior and serotonergic disorders in rats exposed prenatally to valproate: a model for autism. Neurosci Lett.Dufour-rainfray D Vourc h P 2010 Feb 5;470 1 55 9 - 71.
Maternal administration of thalidomide or valproic acid causes abnormal serotonergic neurons in the offspring: implication for pathogenesis of autism. Int J Dev Neurosci.Miyazaki K Narita N Narita M 2005 Apr-May;23(2-3):287-97. - 72.
Swim stress exaggerates the hyperactive mesocortical dopamine system in a rodent model of autism. Brain Res.Nakasato A Nakatani Y Seki Y Tsujino N Umino M Arita H 2008 Feb 8;1193 128 35 - 73.
Nonexploratory movement and behavioral alterations in a thalidomide or valproic acid-induced autism model rat. Neurosci Res.Narita M Oyabu A Imura Y Kamada N Yokoyama T Tano K et al 2009 Jan;66 1 2 6 - 74.
Abnormality of circadian rhythm accompanied by an increase in frontal cortex serotonin in animal model of autism. Neurosci Res.Tsujino N Nakatani Y Seki Y Nakasato A Nakamura M Sugawara M et al 2007 Feb;57 2 289 95 - 75.
Observation of fetal brain in a rat valproate-induced autism model: a developmental neurotoxicity study. Int J Dev Neurosci.Kuwagata M Ogawa T Shioda S Nagata T 2009 Jun;27 4 399 405 - 76.
Valproate-induced developmental neurotoxicity is affected by maternal conditions including shipping stress and environmental change during early pregnancy. Toxicol Lett.Ogawa T Kuwagata M Hori Y Shioda S 2007 Nov 1;174(1-3):18-24. - 77.
Prenatal VPA Exposure and Changes in Sensory Processing by the Superior Colliculus. Front Integr Neurosci.Dendrinos G Hemelt M Keller A 2011 - 78.
Elevated NMDA receptor levels and enhanced postsynaptic long-term potentiation induced by prenatal exposure to valproic acid. Proc Natl Acad Sci U S A.Rinaldi T Kulangara K Antoniello K Markram H 2007 Aug 14;104 33 13501 6 - 79.
Chan Kim K, Choi CS, Jeon SJ, Kwon KJ, Han SH, et al. Prenatal exposure to valproic acid increases the neural progenitor cell pool and induces macrocephaly in rat brain via a mechanism involving the GSK-3beta/beta-catenin pathway. Neuropharmacology.Go H. S 2012 Jul 27. - 80.
Discrimination learning and reversal of the conditioned eyeblink reflex in a rodent model of autism. Behav Brain Res.Stanton M. E Peloso E Brown K. L Rodier P 2007 Jan 10;176 1 133 40 - 81.
PM. Induction of the homeotic gene Hoxa1 through valproic acid’s teratogenic mechanism of action. Neurotoxicol Teratol.Stodgell C. J Ingram J. L O Bara M Tisdale B. K Nau H Rodier 2006 Sep-Oct;28 5 617 24 - 82.
Prenatal exposure to valproic acid enhances synaptic plasticity in the medial prefrontal cortex and fear memories. Brain Res Bull.Sui L Chen M 2012 Apr 10;87 6 556 63 - 83.
Interstimulus interval (ISI) discrimination of the conditioned eyeblink response in a rodent model of autism. Behav Brain Res.Murawski N. J Brown K. L Stanton M. E 2009 Jan 23;196 2 297 303 - 84.
Animal model of autism induced by prenatal exposure to valproate: behavioral changes and liver parameters. Brain Res.Bambini-junior V Rodrigues L Behr G. A Moreira J. C Riesgo R Gottfried C 2011 Aug 23;1408 8 16 - 85.
Gestational valproate alters BOLD activation in response to complex social and primary sensory stimuli. PLoS One.Felix-ortiz A. C Febo M 2012 e37313. - 86.
Prenatal exposure of rats to valproic acid reproduces the cerebellar anomalies associated with autism. Neurotoxicol Teratol.Ingram J. L Peckham S. M Tisdale B Rodier P. M 2000 May-Jun;22 3 319 24 - 87.
Jr., Murawski NJ, Kulesza RJ, Jr. Malformation of the superior olivary complex in an animal model of autism. Brain Res.Lukose R Schmidt E Wolski T. P 2011 Jun 29;1398 102 12 - 88.
Nociceptive changes in rats after prenatal exposure to valproic acid. Pol J Pharmacol.Schneider T Labuz D Przewlocki R 2001 Sep-Oct;53 5 531 4 - 89.
Gender-specific behavioral and immunological alterations in an animal model of autism induced by prenatal exposure to valproic acid. Psychoneuroendocrinology.Schneider T Roman A Basta-kaim A Kubera M Budziszewska B Schneider K et al 2008 Jul;33 6 728 40 - 90.
Environmental enrichment reverses behavioral alterations in rats prenatally exposed to valproic acid: issues for a therapeutic approach in autism. Neuropsychopharmacology.Schneider T Turczak J Przewlocki R 2006 Jan;31 1 36 46 - 91.
Prenatal exposure to valproic acid disturbs the enkephalinergic system functioning, basal hedonic tone, and emotional responses in an animal model of autism. Psychopharmacology (Berl).Schneider T Ziolkowska B Gieryk A Tyminska A Przewlocki R 2007 Sep;193 4 547 55 - 92.
Demethylation of specific Wnt/beta-catenin pathway genes and its upregulation in rat brain induced by prenatal valproate exposure. Anat Rec (Hoboken).Wang Z Xu L Zhu X Cui W Sun Y Nishijo H et al 2010 Nov;293 11 1947 53 - 93.
Animal model of autism using GSTM1 knockout mice and early post-natal sodium valproate treatment. Behav Brain Res.Yochum C. L Bhattacharya P Patti L Mirochnitchenko O Wagner G. C 2010 Jul 11;210 2 202 10 - 94.
Immunogold study of effects of prenatal exposure to lipopolysaccharide and/or valproic acid on the rat blood-brain barrier vessels. J Neurocytol.Vorbrodt A. W Dobrogowska D. H Kozlowski P. B Rabe A Tarnawski M Lee M. H 2005 Dec;34 6 435 46 - 95.
Altered morphology of motor cortex neurons in the VPA rat model of autism. Dev Psychobiol.Snow W. M Hartle K Ivanco T. L 2008 Nov;50 7 633 9 - 96.
La Mendola D, Sandi C, Markram H. Abnormal fear conditioning and amygdala processing in an animal model of autism. Neuropsychopharmacology.Markram K Rinaldi T 2008 Mar;33 4 901 12 - 97.
Hyperconnectivity of local neocortical microcircuitry induced by prenatal exposure to valproic acid. Cereb Cortex.Rinaldi T Silberberg G Markram H 2008 Apr;18 4 763 70 - 98.
Bacopa monniera (L.) Wettst ameliorates behavioral alterations and oxidative markers in sodium valproate induced autism in rats. Neurochem Res.Sandhya T Sowjanya J Veeresh B 2012 May;37 5 1121 31 - 99.
Common circuit defect of excitatory-inhibitory balance in mouse models of autism. J Neurodev Disord.Gogolla N Leblanc J. J Quast K. B Sudhof T. C Fagiolini M Hensch T. K 2009 Jun;1 2 172 81 - 100.
Hypoplasia of cerebellar vermal lobules VI and VII in autism. N Engl J Med.Courchesne E Yeung-courchesne R Press G. A Hesselink J. R Jernigan T. L 1988 May 26;318 21 1349 54 - 101.
The intense world syndrome--an alternative hypothesis for autism. Front Neurosci.Markram H Rinaldi T Markram K 2007 Nov;1 1 77 96 - 102.
Neuropeptides and neurotrophins in neonatal blood of children with autism or mental retardation. Ann Neurol.Nelson K. B Grether J. K Croen L. A Dambrosia J. M Dickens B. F Jelliffe L. L et al 2001 May;49 5 597 606 - 103.
Trk receptors: roles in neuronal signal transduction. Annu Rev Biochem.Huang E. J Reichardt L. F 2003 72 609 42 - 104.
Infusion of BDNF into the nucleus accumbens of aged rats improves cognition and structural synaptic plasticity through PI3K-ILK-Akt signaling. Behav Brain Res.Li M Dai F. R Du X. P Yang Q. D Zhang X Chen Y 2012 May 16;231 1 146 53 - 105.
Del Bianco P, Paolone G, Caprioli D, Modafferi AM, Nencini P, et al. Social isolation selectively reduces hippocampal brain-derived neurotrophic factor without altering plasma corticosterone. Behav Brain Res.Scaccianoce S 2006 Apr 3;168 2 323 5 - 106.
Interaction between BDNF and serotonin: role in mood disorders. Neuropsychopharmacology.Martinowich K Lu B 2008 Jan;33 1 73 83 - 107.
Biochemical aspects in autism spectrum disorders: updating the opioid-excess theory and presenting new opportunities for biomedical intervention. Expert Opin Ther Targets.Shattock P Whiteley P 2002 Apr;6 2 175 83 - 108.
Developmental changes in brain serotonin synthesis capacity in autistic and nonautistic children. Ann Neurol.Chugani D. C Muzik O Behen M Rothermel R Janisse J. J Lee J et al 1999 Mar;45 3 287 95 - 109.
An investigation on sleep disturbance of autistic children. Folia Psychiatr Neurol Jpn.Hoshino Y Watanabe H Yashima Y Kaneko M Kumashiro H 1984 38 1 45 51 - 110.
Activity of brain serotonergic neurons in the behaving animal. Pharmacol Rev.Jacobs B. L Fornal C. A 1991 Dec;43 4 563 78 - 111.
Increased monoamine concentration in the brain and blood of fetal thalidomide- and valproic acid-exposed rat: putative animal models for autism. Pediatr Res.Narita N Kato M Tazoe M Miyazaki K Narita M Okado N 2002 Oct;52 4 576 9 - 112.
Astrocytes process synaptic information. Neuron Glia Biol.Araque A 2008 Feb;4 1 3 10 - 113.
Neuroglial activation and neuroinflammation in the brain of patients with autism. Ann Neurol.Vargas D. L Nascimbene C Krishnan C Zimmerman A. W Pardo C. A 2005 Jan;57 1 67 81 - 114.
Glutamate receptor dysfunction and drug targets across models of autism spectrum disorders. Pharmacol Biochem Behav.Carlson G. C 2012 Feb;100 4 850 4 - 115.
Model of autism: increased ratio of excitation/inhibition in key neural systems. Genes Brain Behav.Rubenstein J. L Merzenich M. M 2003 Oct;2 5 255 67 - 116.
Prevalence of motor impairment in autism spectrum disorders. Brain Dev.Ming X Brimacombe M Wagner G. C 2007 Oct;29 9 565 70 - 117.
A clinicopathological study of autism. Brain.Bailey A Luthert P Dean A Harding B Janota I Montgomery M et al 1998 May;121 ( Pt 5):889-905. - 118.
Autism and hearing loss. J Autism Dev Disord.Rosenhall U Nordin V Sandstrom M Ahlsen G Gillberg C 1999 Oct;29 5 349 57 - 119.
Jr., Lukose R, Stevens LV. Malformation of the human superior olive in autistic spectrum disorders. Brain Res.Kulesza R. J 2011 Jan 7;1367 360 71