Browse > Article
http://dx.doi.org/10.5487/TR.2018.34.2.173

Valproic Acid Exposure of Pregnant Rats During Organogenesis Disturbs Pancreas Development in Insulin Synthesis and Secretion of the Offspring  

Komariah, Komariah (Department of Histology, Faculty of Dentistry, Trisakti University)
Manalu, Wasmen (Department of Anatomy, Physiology, and Pharmacology, Faculty of Veterinary Medicine, Bogor Agricultural University)
Kiranadi, Bambang (Department of Anatomy, Physiology, and Pharmacology, Faculty of Veterinary Medicine, Bogor Agricultural University)
Winarto, Adi (Department of Anatomy, Physiology, and Pharmacology, Faculty of Veterinary Medicine, Bogor Agricultural University)
Handharyani, Ekowati (Department of Clinic, Reproduction, and Pathology, Faculty of Veterinary Medicine, Bogor Agricultural University)
Roeslan, M. Orliando (Department of Biology Oral, Faculty of Dentistry, Trisakti University)
Publication Information
Toxicological Research / v.34, no.2, 2018 , pp. 173-182 More about this Journal
Abstract
Valproic acid (VPA) plays a role in histone modifications that eventually inhibit the activity of histone deacetylase (HDAC), and will affect the expressions of genes Pdx1, Nkx6.1, and Ngn3 during pancreatic organogenesis. This experiment was designed to study the effect of VPA exposure in pregnant rats on the activity of HDAC that controls the expression of genes regulating the development of beta cells in the pancreas to synthesize and secrete insulin. This study used 30 pregnant Sprague-Dawley rats, divided into 4 groups, as follows: (1) a control group of pregnant rats without VPA administration, (2) pregnant rats administered with 250 mg VPA on day 10 of pregnancy, (3) pregnant rats administered with 250 mg VPA on day 13 of pregnancy, and (4) pregnant rats administered with 250 mg VPA on day 16 of pregnancy. Eighty-four newborn rats born to control rats and rats administered with VPA on days 10, 13, and 16 of pregnancy were used to measure serum glucose, insulin, DNA, RNA, and ratio of RNA/DNA concentrations in the pancreas and to observe the microscopical condition of the pancreas at the ages of 4 to 32 weeks postpartum with 4-week intervals. The results showed that at the age of 32 weeks, the offspring of pregnant rats administered with 250 mg VPA on days 10, 13, and 16 of pregnancy had higher serum glucose concentrations and lower serum insulin concentrations, followed by decreased concentrations of RNA, and the ratio of RNA/DNA in the pancreas. Microscopical observations showed that the pancreas of the rats born to pregnant rats administered with VPA during pregnancy had low immunoreaction to insulin. The exposure of pregnant rats to VPA during pregnancy disturbs organogenesis of the pancreas of the embryos that eventually disturb the insulin production in the beta cells indicated by the decreased insulin secretion during postnatal life.
Keywords
Valproic acid; Histone deacetylase; Gene expression; Pancreas organogenesis; Insulin; Rat;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Pan, F.C. and Wright, C. (2011) Pancreas organogenesis: from bud to plexus to gland. Dev. Dyn., 240, 530-565.   DOI
2 Shih, H.P. and Sander, W.M. (2013) Pancreas organogenesis: from lineage determination to morphogenesis. Annu. Rev. Cell Dev. Biol., 29, 81-105.   DOI
3 Wang, X., Wei, X., Pang, Q. and Yia, F. (2012) Histone deacetylases and their inhibitors: molecular mechanisms and therapeutic implications indiabetes mellitus. Acta Pharm. Sin., 2, 387-395.   DOI
4 Sostrup, B., Gaarn, L.W., Nalla, A., Billestrup, N. and Nielsen, J.N. (2014) Co-ordinated regulation of neurogenin-3 expression in the maternal and fetal pancreas during pregnancy. Acta Obstet. Gynecol. Scand., 93, 1190-1197.   DOI
5 Maksoud, H.M.A., El-Shazly, S.M. and El Saied, M.H. (2016) Effect of antiepileptic drug (valproic acid) on children growth. Gaz. Egypt Paediatr. Assoc., 64, 69-73.   DOI
6 Vajda, F. and O'Brien, T. (2010) Valproic acid use in pregnancy and congenital malformations. N. Engl. J. Med., 363, 1771-1772.   DOI
7 Guerrini, R. (2006) Valproate as a mainstay of therapy for pediatric epilepsy. Paediatr. Drugs, 2, 113-29.
8 Gerace, D., Wilks, M.R., O'Brien, B.A. and Simpson, A.M. (2015) The use of ${\beta}$-cell transcription factors in engineering artificial ${\beta}$ cells from non-pancreatic tissue. Gene Ther., 22, 1-8.   DOI
9 Yong, J., Ansari, P.I. and Kaufman, R.J. (2016) When less is better: ER stress and beta cell proliferation. Dev. Cell, 36, 4-6.   DOI
10 Aguirre, A.M.L., Aguirre, A.A.C., Camberos, E.P., Solis, H.E. and Martinez, N.E.D. (2015) Development of the endocrine pancreas and novel strategies for b-cell mass restoration and diabetes therapy. Braz. J. Med. Biol. Res., 48, 765-776.   DOI
11 Ververis, K., Alison, H., Karagiannis, T.C. and Licciardi, P.V. (2013) Histone deacetylase inhibitors (HDACIS): multitargeted anticancer agents. Biologics, 7, 47-60.
12 Ornoy, A. and Ergaz, Z. (2010) Alcohol abuse in pregnant women: Effects on the fetus and newborn, mode of action and maternal treatment. Int. J. Environ. Res. Public Health, 7, 364-379.   DOI
13 da Costa, R.F.M., Kormann, M.L., Galina, A. and Rehen, S.K. (2015) Valproate disturbs morphology and mitochondrial membrane potential in human neural cells. Appl. In Vitro Toxicol., 1, 254-261.   DOI
14 Ximenes, J.C.M., Verde, E.C.L., Mazzacoratti, M.G.N. and Viana, G.S.B. (2012) Valproic acid, a drug with multiple molecular targets related to its potential neuroprotective action. Neurosci. Med., 3, 107-123.   DOI
15 Halsall, J.A. and Turner, B.M. (2016) Histone deacetylase inhibitors for cancer therapy: an evolutionarily ancient resistance response may explain their limited success. Bioessays, 38, 1102-1110.   DOI
16 Dahlan, M.S. (2014) Statistics for medicine and health; descriptive, bivariate, and multivariate. Epidemiology Indonesia, Jakarta, Indonesia, pp. 110-117.
17 Wlodarczyk, B.J., Palacios, A.M., Chapa, C.J., Zhu, H., George, T.M. and Finnell, R.H. (2011) Genetic basis of susceptibility to teratogen induced birth defects. Am. J. Med. Genet., 157, 215-226.   DOI
18 De Felice, A., Ricceri, L., Venerosi, A., Chiarotti, F. and Calamandrei, G. (2015) Multifactorial origin of neurodevelopmental disorders: approaches to understanding complex etiologies. Toxics, 3, 89-129.   DOI
19 Kokate, P. and Bang, R. (2017) Study of congenital malformation in tertiary care centre, Mumbai, Maharashtra, India. Int. J. Reprod. Contracept. Obstet. Gynecol., 6, 89-93.
20 Tang, O.S., Danielsson, K.G. and Ho, P.C. (2007) Misoprostol: pharmacokinetic profiles, effects on the uterus and sideeffects. Int. J. Gynaecol. Obstet., 99, 160-167.   DOI
21 Berridge, M.J. (2014) Cell cycle and proliferation. Cell Signalling Biology, 901-943.
22 Bertoli, C., Skotheim, J.M. and De Bruin, R.A.M. (2013) Control of cell cycle transcription during G1 and S phases. Nat. Rev. Mol. Cell Biol., 14, 518-528.   DOI
23 Li, Q., Foote, M. and Chen, J. (2014) Effects of histone deacetylase inhibitor valproic acid on skeletal myocyte development. Sci. Rep., 4, 1-4.
24 Gallagher, S.J., Tiffen, J.C. and Hersey, P. (2015) Histone modifications, modifiers and readers in melanoma resistance to targeted and immune therapy. Cancers, 7, 1959-1982.   DOI
25 Gomez, D.L., O'Driscoll, M., Sheets, T.P., Hruban, R.H., Oberholzer, J., McGarrigle, J.J. and Shamblott, M.J. (2015) Neurogenin 3 expressing cells in the human exocrine pancreas have the capacity for endocrine cell fate. PLoS ONE, 10, e0133862.   DOI
26 Deng, S., Vatamaniuk, M., Huang, X., Doliba, N., Lian, M., Frank, A., Velidedeoglu, E., Desai, N.M., Koeberlein, B., Wolf, B., Clyde, F.B., Ali, N., Franz, M.M. and James, F.M. (2004) Structural and functional abnormalities in the islets isolated from type 2 diabetic subjects. Diabetes, 53, 624-632.   DOI
27 Otsuka, T., Tsukahara, T. and Takeda, H. (2015) Development of the pancreas in medaka, Oryzias latipes, from embryo to adult. Dev. Growth Differ., 57, 557-569.   DOI
28 Schwartz, M.W., Guyenet, S.J. and Cirulli, V. (2010) The hypothalamus and beta-cell connection in the gene-targeting era. Diabetes, 59, 2991-2993.   DOI
29 Sanchez, A.M., Rutter, G.A. and Latreille, M. (2017) MiRNAs in ${\beta}$-cell development, identity, and disease. Front. Genet., 7, 226.
30 Pagliuca, F.W., Millman, J.R., Gurtler, M., Segel, M., Dervort, A.D., Ryu, J.H., Peterson, Q.P., Greiner, D. and Melton, D.A. (2014) Generation of functional human pancreatic ${\beta}$ cells in vitro. J. Cell, 159, 428-439.   DOI
31 Baynest, H.W. (2015) Classification, pathophysiology, diagnosis and management of diabetes mellitus. J. Diabetes Metab., 6, 541.
32 Henquin, J.C. and Rahier, J. (2011) Pancreatic alpha cell mass in European subjects with type 2 diabetes. Diabetologia, 54, 1720-1725.   DOI
33 Pscherer, S., Larbig, M., Stritsky, B., Pfutzner, A. and Forst, T. (2012) In type 2 diabetes patients insulin glargine is associated with lower postprandial release of intact proinsulin compared with sulfonylurea treatment. J. Diabetes Sci. Technol., 6, 634-640.   DOI
34 Reef, R., Ball, M.C., Feller, I.C. and Lovelock, C.E. (2010) Relationships among RNA : DNA ratio, growth and elemental stoichiometry in mangrove trees. Funct. Ecol., 24, 1064-1072.   DOI
35 Chicharo, M.A. and Chicharo, L. (2008) RNA:DNA ratio and other nucleic acid derived indices in marine ecology. Int. J. Mol. Sci., 9, 1453-1471.   DOI
36 Olivar, M.P., Diaz, M.V. and Chicharo, M.A. (2009) Tissue effect on RNA:DNA ratios of marine fish larvae. Sci. Mar., 171-182.
37 FizeIova, M., Cederberg, H., Stancakova, A., Jauhiainen, R., Vangipurapu, J., Kuusisto, J. and Laakso, M. (2014) Markers of tissue-specific insulin resistance predict the worsening of hyperglycemia, incident type 2 diabetes and cardiovascular disease. PLoS ONE, 10, e109772.
38 Ali, S.F. and Padhi, R. (2009) Optimal blood glucose regulation of diabetic patients using single network adaptive critics. Optimal Control Applications & Methods, 32, 196-214.
39 Cerf, M.E. (2013) Beta cell dysfunction and insulin resistance. Front. Endocrinol., 37, 1-12.
40 Arystarkhova, E., Liu, Y.B., Salazar, C., Stanojevic, V., Clifford, R.J., Kaplan, J.H., Kidder, G.M. and Weadner, K.J. (2013) Hyperplasia of pancreatic beta cells and improved glucose tolerance in mice deficient in the FXYD2 subunit of Na,K-ATPase. J. Biol. Chem., 288, 7077-7085.   DOI
41 Xu, W., Wang, Y., Li, Y., Wang, L., Xiong, X., Su, J. and Zhang, Y. (2012) Valproic acid improves the in vitro development competence of bovine somatic cell nuclear transfer embryos. Cell. Reprogram., 14, 138-145.   DOI
42 Kurihara, Y., Suzuki, T., Sakaue, M., Murayama, O., Miyazaki, Y. and Onuki, A. (2014) Valproic acid, a histone deacetylase inhibitor, decreases proliferation of and induces pecific neurogenic differentiation of canine adipose tissuederived stem cells. J. Vet. Med. Sci., 76, 15-23.   DOI
43 Schulpen, S.H.W., Pennings, J.L.A. and Piersma, A.H. (2015) Gene expression regulation and pathway analysis after valproic acid and carbamazepine exposure in a human embryonic stem cell-based neurodevelopmental toxicity assay. Toxicol. Sci., 146, 311-320.   DOI
44 Giavini, E. and Menegola, E. (2014) Teratogenic activity of HDAC inhibitors. Curr. Pharm. Des., 20, 1-6.
45 Rajendran, P., Kidane, A.I., Yu, T.W., Dashwood, W.M., Bisson, W.H., Lohr, C.V., Ho, E., Williams, D.E. and Dashwood, R.H. (2013) HDAC turnover, CtIP acetylation and dysregulated DNA damage signaling in colon cancer cells treated with sulforaphane and related dietary isothiocyanates. Epigenetics, 8, 612-623.   DOI
46 Shi, D. and Gu, W. (2012) Dual roles of MDM2 in the regulation of p53: ubiquitination dependent and ubiquitination independent mechanisms of MDM2 repression of p53 activity. Genes Cancer, 3, 240-248.   DOI
47 Garner, E. and Raj, K. (2008) Protective mechanisms of p53-p21-pRb proteins against DNA damage-induced cell death. Cell Cycle, 7, 1-6.
48 Shkreta, L. and Chabot, B. (2015) The RNA splicing response to DNA damage. Biomolecules, 5, 2935-2977.   DOI
49 Foleya, C.J., Bradleyc, D.L. and Hook, T.O. (2016) A review and assessment of the potential use of RNA:DNA ratios to assess the condition of entrained fish larvae. Ecol. Indic., 60, 346-357.   DOI
50 Yoon, K.H., Ko, S.H., Cho, J.H., Lee, J.M., Ahn, Y.B., Song, K.H., Yoo, S.J., Kang, M.L., Cha, B.Y. and Lee, K.W. (2003) Selective ${\beta}$-cell loss and ${\alpha}$-cell expansion in patients with type 2 diabetes mellitus in Korea. J. Clin. Endocrinol. Metab., 88, 2300-2308.   DOI