Musanga cecropioides (Urticaceae) stem-bark mitigates sodium valproate –induced pantoxicity derangement in albino rats

Authors

  • Lucky Legbosi Nwidu Experimental Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, University of Port Harcourt, Choba, East West Road., PMB. 5323, Rivers State, Nigeria.
  • Yibala Ibor Oboma Medical Laboratory Science Department, Faculty of Basic Medical Sciences, College of Health Sciences, Niger Delta University, Amassoma, PMB 071 Yenegoa, Bayelsa State, Nigeria.

DOI:

https://doi.org/10.30574/gscbps.2019.7.1.0050

Keywords:

Nauclea latifolia stem-bark, Valproic acid, Vinpocetin, Hepatotoxicity, Nephrotoxicity, Pulmonary-toxicity

Abstract

Sodium valproate (SVPA) is indicated for the management of partial and generalized epilepsy with major limitation of pan-toxicity. Musanga cecropioides stem-bark (MCS) is used in ethnomedicine for multiple health benefits. The aim of the study was to test the ameliorative effect of MCS extract on SVPA-induced damage in rodent. The rats were administered with SVPA (500 mg/kg b.w. followed by MCS (50, 100, and 200 mg/kg b.w.) and reference drug, vinpocetine (25 mg/kg b.w.) orally and sub-acutely. The protective effects of MCS extract on their weights, hematological, biochemical, lipid, kidney and electrolytes profiles parameters were examined. Following rats sacrifice, the liver, kidney and lungs were subjected to histopathological analysis. The study indicated that SVPA significantly up-regulated the liver function enzymes (P< 0.001), lipid profile (P< 0.01-0.001), kidney function (P< 0.05–0.01) and electrolytes (P< 0.01–0.001) biomarkers and elicited gross alterations of measured indices. However, this effects were dose dependently reversed by MCS extract with higher hepatoprotective percentages, for liver enzymes (77-261%), lipid profiles (74–133%), electrolytes (59-169%) and kidney function (82–154%) compare to vinpocetine values of 63–103%, 80- 127%, 70–161%  and  27-78%,  respectively. No significant alteration in hematology and relative organ weights. The effect on histopathology corroborated biochemical study. Vinpocetine exhibit no therapeutic effect on the histopathological alteration of liver and kidney but only on the lungs. The presence of potential active ingredients in MCS extract confirms it as an alternative adjunctive therapy in abrogating SVPA – induced pan-toxicity derangement in rats.

Metrics

Metrics Loading ...

References

Fan HC, Lee HS, Chang KP, Lee YY, Lai HC and Hung PL. (2016). The impact of anti-epileptic drugs on growth and bone metabolism. International Journal of Molecular Sciences, 17, 1–22.

Tomson T, Battino D and Perucca E. (2016). The remarkable story of valproic acid. Lancet Neurology, 15,141.

Burton BS. (1882). On the propyl derivatives and decomposition products of ethyl acetoacetate. American Chemical Journal, 3, 385-395.

Meunier H, Carraz G, Meunier Y, Eymard P and Aimard M. (1963). Pharmacodynamic properties of N-dipropylacetic acid. Therapie 18, 435-438.

Terbach N, Shah R, Kelemen R, Klein PS, Gordienko D, Brown NA, Wilkinson CJ and Williams RS. (2011). Identifying an uptake mechanism for the antiepileptic and bipolar disorder treatment valproic acid using the simple biomedical model Dictyostelium. Journal of Cell Science, 124 (Pt 13), 2267-76.

Calabresi P, Galletti F, Rossi C, Sarchielli P and Cupini LM. (2007). Antiepileptic drugs in migraine: from clinical aspects to cellular mechanisms. Trends in PharmacologicalScience 28, 188-195

Ghodke-Puranik Y, Thorn CF, Lamba JK, Leeder JS, Song W, Birnbaum AK, Altman RB and Klein TE. (2013). Valproic acid pathway: pharmacokinetics and pharmacodynamics. Pharmacogenetic and Genomics, 23, 236–241.

Terbach N and Williams RS. (2009). Structure-function studies for the panacea, valproic acid. Biochemical Society Transaction, 37, 1126-1132.

Tseng JH, Chen CY, Chen PC, Hsiao SH, Fan CC, Liang YC and Chen CP. (2017). Valproic acid inhibits glioblastoma multiforme cell growth via paraoxonase 2 expression. Oncotarget, 8, 14666-14679.

Bokelmann I and Mahlknecht U. (2008). Valproic acid sensitizes chronic lymphocytic leukemia cells to apoptosis and restores the balance between pro- and antiapoptotic proteins. Molecular Medicine, 14, 20 – 27.

Venkataramani V, Rossner C, Iffland L, Schweyer S, Tamboli IY, Walter J, Wirths O and Bayer TA. (2010). Histone deacetylase inhibitor valproic acid inhibits cancer cell proliferation via down-regulation of the alzheimer amyloid precursor protein. Journal of Biological Chemistry, 285, 10678-1089.

Davidson DC, Schifitto G and Maggirwar SB. (2013). Valproic acid inhibits the release of soluble CD40L induced by non-nucleoside reverse transcriptase inhibitors in human immunodeficiency virus infected individuals. PLoS One, 8, e59950.

Crespillo AJ, Praena B, Bello-Morales R, Lerma L, Vázquez-Calvo A, Martín-Acebes MA, Tabarés E, Sobrino F and López-Guerrero JA.(2016). Inhibition of herpes virus infection in oligodendrocyte cultured cells by valproic acid. Virus Research, 214, 71- 79.

Williams RS and Bate C. (2016). An in vitro model for synaptic loss in neurodegenerative diseases suggests a neuroprotective role for valproic acid via inhibition of cPLA2 dependent signalling. Neuropharmacology. 101, 566-575.

Zhao L, Zhu L and Guo X. (2018). Valproic acid attenuates Aβ (25-35)-induced neurotoxicity in PC12 cells through suppression of mitochondria-mediated apoptotic pathway. Biomedicine and Pharmacotherapy,106, 77-82

Acharya S and Bussel JB. (2000). Hematologic toxicity of sodium valproate. Journal of Pediatrics Hematology and Oncology, 22, 62-65.

Jin J, Xiong T, Hou X, Sun X, Liao J, Huang Z, Huang M and Zhao Z. (2014). Role of Nrf2 activation and NF-κB inhibition in valproic acid induced hepatotoxicity and in diammonium glycyrrhizinate induced protection in mice. Food and Chemical Toxicology, 73, 95-104.

Gad AM. (2018). Study on the influence of caffeic acid against sodium valproate-induced nephrotoxicity in rats. Journal of Biochemistry and Molecular Toxicology, 32, e22175.

Vorhees CV. (1987). Teratogenicity and developmental toxicity of valproic acid in rats. Teratology, 35, 195 -202.

Ehashi T, Suzuki N, Ando S, Sumida K and Saito K. (2014). Effects of valproic acid on gene expression during human embryonic stem cell differentiation into neurons. Journal of Toxicological Science, 39(3), 383- 390.

Morsy BM, Safwat G M, Hussein DA and Samy R. (2017). The protective effect of Nigella sativa oil extract against neurotoxicity induced by Valproic acid. International Journal of Bioassays, 6, 5474 - 5484.

Schwarz K, Romanski A, Puccetti E, Wietbrauk S, Vogel A, Keller M, Scott JW, Serve H and Bug G. (2011). The deacetylase inhibitor LAQ824 induces notch signaling in hematopoietic progenitor cells. Leukemia Research, 35, 119 -125.

Jones MR, Hall OM, Kaye AM and Kaye AD. (2015). Drug-induced acute pancreatitis: a review. Ochsner Journal, 15, 45-51.

Main SL and Kulesza RJ. (2017). Repeated prenatal exposure to valproic acid results in cerebellar hypoplasia and ataxia. Neuroscience, 40, 34-47.

Cofini M, Quadrozzi F, Favoriti P, Favoriti M and Cofini G. (2015). Valproic acid-induced acute pancreatitis in pediatric age: case series and review of literature. Giornale Di Chirurgia, 36, 158-160.

Chateauvieux S, Morceau F, Dicato M and Diederich M. (2010). Molecular and therapeutic potential and toxicity of valproic acid. Journal of Biomedicine and Biotechnology, 4793641–18.

Chen S, Wu H, Klebe D, Hong Y and Zhang J. (2014). Valproic acid: a new candidate of therapeutic application for the acute central nervous system injuries. Neurochemistry Research, 39, 1621–1633.

Tomson T, Battino D and Perucca E. (2016). Valproic acid after five decades of use in epilepsy: time to reconsider the indications of time-honoured drug. Lancet Neurology, 15, 210–218.

Belcastro V, D’Egidio C, Striano P and Verrotti A. (2013). Metabolic and endocrine effects of valproic acid chronic treatment. Epilepsy Research, 107, 1–8.

Lheureux PER, Penaloza A, Zahir S and Gris M. (2005). Science review: carnitine in the treatment of valproic acid-induced toxicity – what is the evidence? Critical Care, 9, 431–440.

Bereczki D and Fekete I. (2008). Vinpocetine for acute ischemic stroke. Cochrane Database Systematic Review, 1, CD000580.

Cholnoky E and Dömök LI. (1976). Summary of safety tests of ethyl apovincaminate. Drug Research, 26, 1938-1944.

Vereczkey L, Szentirmay Z and Szporny L. (1979a). Kinetic metabolism ofvinpocetine in the rat. Drug Research, 29, 953-956.

Zaki HF and Abdelsalam RM. (2013). Vinpocetine protects liver against ischemia-reperfusion injury. Canadian Journal of Physiology and Pharmacology 91, 1064-70.

Fattori V, Borghi SM, Guazelli CFS, Giroldo AC, Crespigio J, Bussmann AJC, Coelho-Silva L, Ludwig NG, Mazzuco TL, Casagrande R and Verri WA Jr. (2017). Vinpocetine reduces diclofenac-induced acute kidney injury through inhibition of oxidative stress, apoptosis, cytokine production, and NF-κB activation in mice. Pharmacological Research, 120, 10-22.

Wu MP, Zhang YS, Xu X, Zhou Q, Li JD and Yan C. (2017). Vinpocetine attenuates pathological cardiac remodeling by inhibiting cardiac hypertrophy and fibrosis. Cardiovascular Drugs and Therapy, 31, 157-166.

Nosálová V, Machová J and Babulová A. (1993). Protective action of vinpocetine against experimentally induced gastric damage in rats. Drug Research, 43, 981-5.

Ruiz-Miyazawa KW, Zarpelon AC, Pinho-Ribeiro FA, Pavão-de-Souza GF, Casagrande R and Verri WA Jr. (2005). Vinpocetine reduces carrageenan-induced inflammatory hyperalgesia in mice by inhibiting oxidative stress, cytokine production and NF-κB activation in the paw and spinal cord. PLoS One, 10, e0118942.

Etuk EU. (2006). A review of medicinal plants with hypotensive or antihypertensive effects. Journal Medical Sciences, 6, 894-900

Adeneye AA. (2009). Protective activity of the stem-bark aqueous extract of Musanga cecropioides in carbon tetrachloride- and acetaminophen-induced acute hepatotoxicity in rats. African Journal of Traditional and Complementary and Alternative Medicine, 6, 131-8

Adeneye AA, Ajagbonna OP, Mojiminiyi FB, Odigie IP, Ojobor PD, Etarrh RR and Adeneye AK. (2006). The hypotensive mechanisms for the aqueous stem bark extract of Musanga cecropioides in Sprague-Dawley rats. Journal of Ethnopharmacology, 106, 203-7.

Adeneye AA, Ajagbonna OP, Adeleke TI and Bello SO. (2006). Preliminary toxicity and phytochemical studies of the stem bark aqueous extract of Musanga cecropioides in rats. Journal of Ethnopharmacology,105, 374-379.

Adeneye AA, Ajagbonna OP and Ayodele OW. (2007). Hypoglycemic and antidiabetic activities on the stem bark aqueous and ethanol extracts of Musanga cecropioides in normal and alloxan-induced diabetic rats. Fitoterapia, 78, 502-505.

Owolabi OJ, Ayinde BA, Nworgu ZA and Ogbonna OO. (2010). Antidiarrheal evaluations of the ethanol extract of Musanga cecropioides stem bark. Methods and Findings in Experimental and Clinical Pharmacology, 32, 407-11.

Nwidu LL, Oboma YI, Elmorsy E and Carter WG. (2018). Hepatoprotective effect of hydromethanolic leaf extract of Musanga cecropioides (Urticaceae) on carbon tetrachloride-induced liver injury and oxidative stress. Taibah Medical University Journal, 13, 344 – 354.

Trease GE and Evans WC. (200). A textbook of pharmacognosy. London: Baillie`re Tindall; 600.

Lorke D. (1983).A new approach to practical acute toxicity testing. Archives of Toxicology, 54, 275-289.

Niaraki MS, Nabavizadeh F, Vaezi GH, Alizadeh AM, Nahrevanian H, Moslehi A and Azizian S. (2013). Protective effect of ghrelin on sodium valproate-induced liver injury in rat. Journal of Stress Physiology and Biochemistry, 9, 97 -105.

Udobi C and Umoh B. (2017). Effects of the Ethanol Extract of the Stem Bark of Nauclea latifolia Smith [Rubiaceae] on Certain Biochemical and Hematological Indices of Swiss Albino Mice. Asian Journal of Medicine and Health, 6, 1–9.

Roy AV. (1970). Rapid method for determining alkaline phosphatase activity in serum with thymolphthalein monophosphate. Clinical Chemistry, 16, 431-436.

Reitman S and Frankel S. (1957). A colorimetric method for the determination of serum glutamic oxalacetic and glutamic pyruvic transaminases. American Journal of. Clinical Pathology28, 56 - 63.

Jendrassik L and Grof P. (1938). Estimation of total serum bilirubin level by spectrophotometrically in serum and plasma. Biochemische Zeitschrift, 297, 81-89.

Doumas BW and Watson WA. (1971). Albumin standards and the measurement of serum albumin with bromocresol green. International Journal of Clinical Chemistry and Diagnostic Laboratory Medicine, 31, 87-96.

Flack CP and Woolen JW. (1984). Prevention of interference by dextran with biuret-type assay of serum proteins”. Clinical Chemistry, 30, 559 – 561.

Lopes-Virella MF, Stone P, Ellis S and Colwell JA. (1977). Cholesterol determination in high-density lipoproteins separated by three different methods. Clinical Chemistry, 23, 882–884.

Allain CC, Poon LS, Chan CS, Richmond W and Fu PC. (1974). Enzymatic determination of total serum cholesterol. Clinical Chemistry, 20, 470- 475.

Roeschlau P, Bernt E and Grub W. (1974). Enzymatic determination of total cholesterol in serum. Z Klin Chem Klin Biochem, 12, 226.

Burtis CA. (2005). Tietz textbook of clinical chemistry andmolecular diagnosis. 4th ed. St. Louis, Missouri: Elsevier Saunders, 2293.

Maruna RFL. (1958). Quantitative estimation of sodium (Na+), potassium (K+) in human serum by colorimetric method”. Clinica Chemica Acta2, 581-585.

Schoenfeld RG and Lewellen CJ. (1964). A Colorimetric method for determination of serum chloride. Clinical Chemistry, 10, 533-539.

Henry RJ, Winkelman JW and Cannon DC. (1995). Clinical Chemistry, Principles and Techniques. 3rd Edn., Harper and Row, USA;1995.

Varley H and Alan HG. (1984). Tests in renal disease. In: Practical Clinical Biochemistry, Vol. 1123. William Heinemann Medical Book Ltd., London.

Kiernan JA. (2015). Histological and histochemical methods: theory and practice. 5th ed. Banbury, UK: Scion Publishing Ltd.

Johannessen CU and Johannessen SI. (2003). Valproate: past, present, and future. CNS Drug Rev, 9, 199–216.

Silva MF, Aires CC, Luis PB, Ruiter JP, Ijlst L, Duran M, Wanders RJ and Tavares de Almeida I. (2008). Valproic acid metabolism and its effects on mitochondrial fatty acid oxidation: a review. Journal Inheritable Metabolic Diseases, 31, 205–216.

Jaeri S and Islamiyah WR. (2018). The potential mechanisms of effect of valproic acid on lipid profiles: An updated review. Journal of Epileptology, 26, (1-2).

Nwidu LL, Teme RE. (2018). Hot aqueous leaf extract of Lasianthera africana (Icacinaceae) attenuates rifampicin-isoniazid-induced hepatotoxicity. J Integr Med., 16(4), 263–272.

Verrotti A, Torre RL, Trotta D, Mohn A and Chiarelli F. (2009). Valproate- induced insulin resistance and obesity in children. Hormonal Research, 71, 125–131.

Egger J and Brett EM. (1981). Effects of sodium valproate in 100 children with special reference to weight. Br. Med. J., (Clin. Res. Ed.), 283, 577–581.

Nwidu LL and Teme RE. (2018). Sub-chronic toxicity of hydromethanolic stem-bark extract of Musanga cecropioides (Cecropiaceae) in rat. EC Pharmacology and Toxicology, 6(3), 76 - 95.

Chellappan DK, Ganasen S, Btunalai S, Canasamy M, Krishnappa P, Dua K, Chellian J and Gupta G. (2016). The protective action of the aqueous extract of Auricularia polytricha in paracetamol-induced hepatotoxicity in rats. Recent Patentin Drug Delivery Formulation, 10, 72-76.

Dreifuss FE, Langer DH, Moline KA and Maxwell JE. (1989). Valproic acid hepatic fatalities. II. US experience since 1984. Neurology, 39, 201-207.

Lheureux PE and Hantson P. (2009). Carnitine in the treatment of valproic acid-induced toxicity. Clinical Toxicology (Philadelphia, Pa.). 47, 101-111.

Zhao M, Zhang T, Li G, Qiu F, Sun Y and Zhao L. (2017). Associations of CYP2C9 and CYP2A6 polymorphisms with the Concentrations of valproate and its hepatotoxin metabolites and valproate-induced hepatotoxicity. Basic Clinical Pharmacology and Toxicology, 121, 138-143.

Tang W, Borel AG, Fujimiya T and Abbott FS. (1995). Fluorinated analogues as mechanistic probes in valproic acidhepatotoxicity: hepatic microvesicular steatosis and glutathione status. Chemical Research in Toxicology, 8, 671–682.

Kesterson JW, Granneman GR and Machinist JM. (1984). The hepatotoxicity of valproic acid and its metabolites in rats. I. Toxicologic, biochemical and histopathologic studies. Hepatology, 4, 1143–1152.

Jacobi G, Thorbeck R, Ritz A, Janssen W and Schmidts HL. (1980). Fatal hepatotoxicity inchild on phenobarbitone and sodium valproate. Lancet, 1, 712–713.

Borum PR and Bennett SG. (1986). Carnitine as an essential nutrient. Journal of American College of Nutrition, 5, 177–182.

Flomenbaum NE, Goldfrank LR, Hoffman RS, Howland MA, Lewin NA and Nelson LS. (2006). Goldfrank’s Toxicological Emergencies. New York: McGraw-Hill 192-212

Raskind JY and El-Chaar GM. (2000). The role of carnitine supplementation during valproic acid therapy. Annals Pharmacotherapy, 34, 630–638.

Millington DS, Bohan TP, Roe CR, Yergey AL and Liberato DJ. (1985). Valproylcarnitine: a novel drug metabolite identified by fast atom bombardment and thermospray liquid chromatographymass spectrometry. Clinica Chimica Acta, 145, 69–76.

Camina MF, Rozas I, Castro-Gago M, Paz JM, Alonso C and Rodriguez-Segade S. (1991). Alteration of renal carnitine metabolism by anticonvulsant treatment. Neurology; 41, 1444–1448.

Farkas V, Bock I, Cseko J and Sandor A. (1996). Inhibition of carnitine biosynthesis by valproic acid in rats – the biochemical mechanism of inhibition. Biochemical Pharmacology, 52, 1429–1433.

Belcastro V, D’Egidio C, Striano P and Verrotti A. (2013). Metabolic and endocrine effects of valproic acid chronic treatment.Epilepsy Research, 107, 1–8.

Abaci A, Saygi M, Yis U, Demir K, Dirik E and Bober E. (2009). Metabolic alterations during valproic acid treatment: a prospective study. Pediatric Neurology, 41, 435–439.

Evans JL, Goldfine ID, Maddux BA and Grodsky GM. (2003). Are oxidative stress-activated signaling pathways mediators of insulin resistance and beta-cell dysfunction? Diabetes, 52, 1–8.

Carraz G, Beriel H, Luu duc H and Lebreton S. (1965). Approches dans la pharmacodynamie biochimique de la structure N-dipropylacetique”. Therapie, 20, 419 – 426.

Shuto K and Nishigaki T. (1970). The pharmacological studies on sodium dipropylacetate anticonvulsant activities and general pharmacological actions. Pharmacometrics, 4, 937– 949.

Jakutiene E, Grikiniene J, Vaitkevicius A, Tschaika M, Didziapetriene J and Stakisaitis D. (2007). Sodium valproate stimulates potassium and chloride urinary excretion in rats: gender differences. BMC Pharmacology, 7, 9.

Zhang LF, Liu LS, Chu XM, Xie H, Cao LJ, Guo C, A JY, Cao B, Li MJ, Wang GJ and Hao HP. (2014). Combined effects of a high-fat diet and chronic valproic acid treatment on hepatic steatosis and hepatotoxicity in rats. Acta Pharmacologica Sinica, 35(3), 363-372.

Kadiri AB and Ajayi GO. (2009). Phyto-anatomical characteristics of the West African {umbrella tree} Musanga cercropioides M. Smithii R. Br. (Moraceae). Indian Journal Science Technology, 2, 1-5.

Buniyamin A, Ayinde D, Onwukaeme N and Eric KO. (2007). Isolation and characterization of two phenolic compounds from the stem bark of Musanga cecropioides R. Brown (Moraceae). Acta Poloniae Pharmaceutical Drug Research, 64, 183-185.

Ajazuddin, Alexander A, Qureshi A, Kumari L, Vaishnav P, Sharma M, Saraf S and Saraf S. (2014). Role of herbal bioactives as a potential bioavailability enhancer for Active Pharmaceutical Ingredients. Fitoterapia, 97, 1-14.

Downloads

Published

2019-04-30

How to Cite

Nwidu, L. L., & Oboma, Y. I. (2019). Musanga cecropioides (Urticaceae) stem-bark mitigates sodium valproate –induced pantoxicity derangement in albino rats. GSC Biological and Pharmaceutical Sciences, 7(1), 06–027. https://doi.org/10.30574/gscbps.2019.7.1.0050

Issue

Section

Original Article