HOME JOURNALS CONTACT

Pakistan Journal of Biological Sciences

Year: 2013 | Volume: 16 | Issue: 6 | Page No.: 281-286
DOI: 10.3923/pjbs.2013.281.286
Effect of Maternal Morphine Sulfate Exposure on Neuronal Plasticity of Dentate Gyrus in Balb/c Mice Offspring
M.J. Golalipour, S. Ghafari, S. Kaboli Kafshgiri, M.H. Latifi Moghadam and A.R. Moharri

Abstract: This study carried out to evaluate the effects of maternal morphine exposure during gestational and lactation period on the neuronal cells of dentate gyrus in 18 and 32 days Balb/c mice offspring. In this experimental study 10 female mice were randomly allocated into cases and controls. In experimental group, animals were received morphine sulfate 10 mg/kg/body weight intraperitoneally during 7 days before mating, gestational period (GD0-21), 18 and 32 days after delivery. The control animals were received an equivalent volume normal saline. Cerebrum of six infant for each group were removed and stained with cresyl violet and monoclonal anti-neuronal nuclei (NeuN) antibody. Quantitative computer-assisted morphometric study was done on dentate gyrus of hippocampus. In the P18 mice , the numbers of granular cells in dentate gyrus medial blade and dentate gyrus lateral blade significantly reduced from 171.45±4.2 and 174.51±3.1 cells in control group to 153.32±2.8 and 151.23±3.2 cells in 10000 μm2 area of granular layer in treated group (p<0.001). In P32 mice the numbers of granular cells in mb and lb of dentate gyrus significantly decreased from 155.31±4.1 and 153.77±3.4 in control group to 138.33±4.5 and 135.13±4.3 in treated group, respectively (p<0.001). The granular layer thickness in mb and lb area of dentate gyrus significantly reduced in treated mice in compared to controls in P18 and P32 mice (p<0.05). This study revealed that morphine administration before, during pregnancy and lactation period causes neuronal cells loss of dentate gyrus in 18 and 32 days old infant mice.

Fulltext PDF Fulltext HTML

How to cite this article
M.J. Golalipour, S. Ghafari, S. Kaboli Kafshgiri, M.H. Latifi Moghadam and A.R. Moharri, 2013. Effect of Maternal Morphine Sulfate Exposure on Neuronal Plasticity of Dentate Gyrus in Balb/c Mice Offspring. Pakistan Journal of Biological Sciences, 16: 281-286.

Keywords: Morphine sulfate, dentate gyrus, granular cells, prenatal exposure and Balb/c mouse

INTRODUCTION

The number of opiate users has risen in worldwide particularly in young people (United Nations Office on Drugs and Crime, 2006; Nestler, 2004). It is estimated that 1 in 1000 people, have been exposed to opiates in early life in the United States (Zagon and MacLaughlin, 1992). Morphine (C17 H19 O3 N) is one of the strongest known analgesic compounds (Zhang et al., 2008) and as one of the addictive drugs leads to increase cause of death, morbidity and lost productivity (Nestler, 2004).

Several studies have shown that infants of opioid dependent mothers had several behavioral abnormalities including hyperactivity, lower Mental Development Index and Lower Motor Development Index (NIDA, 1996; Ornoy et al., 1996; Wilson et al., 1979).

In the other hands, several investigations are founded that morphine has toxic effects on neurons in brain and spinal cord in animal model (Mao et al., 2002; Atici et al., 2004; Turchan-Cholewo et al., 2006; Bekheet et al., 2010; Ghafari et al., 2011).

Also, prenatal morphine exposure impairs learning and memory in juvenile rats (Yang et al., 2003, 2006). In the other hand, synaptic plasticity in the hippocampus is critical for learning and memory processes (Morris et al., 1986; Morris 1989; Silva et al., 1992).

Hippocampus is implicated in the control of several brain functions such as memory and learning and represents a neuronal structure with a high degree of functional and cellular complexity.

Also, several researches have shown that the process of neurogenesis, including cell proliferation, survival, migration and differentiation continues in the dentate gyrus well into adulthood in rodents, non human primates as well as humans (Jackson-Guilford et al., 2000; Gould et al., 2000; Gould and Gross, 2002; Cameron and Gould, 1994; Gould and Tanapat, 1997). Several studies have shown the toxic effects of morphine on neuronal cells in different parts of CNS ether in adult or fetal period (Mao et al., 2002; Svensson et al., 2008; Seatriz and Hammer, 1993; Mei et al., 2009; Emeterio et al., 2006; Eisch et al., 2000; Niu et al., 2009).

Several studies have shown that opiates reduce hippocampal neurons (Svensson et al., 2008), sematosensory neurons of 6 day old (Seatriz and Hammer, 1993), neurons in layer II/III in lateral secondary visual cortex of rats (Mei et al., 2009), inhibits neurogenesis in the adult rat hippocampus (Eisch et al., 2000) and loss of GABA-containing neurons in the Dentate Gyrus (DG) area of rat offespring (Hauser et al., 1994).

Regarding to the high prevalence of opioid abuse in worldwide especially in young adult and rare study about the effect of morphine sulphate on neuronal development of dentate gyrus, the present study was carried out to clarify the neurotoxic effects of prenatal morphine sulphate administration on neuronal density of dentate gyrus in mice offspring.

MATERIALS AND METHODS

This experimental study was performed at the Golestan University of medical sciences, Gorgan, Iran. Guidelines on the care and use of laboratory animals and approval of the ethic committee of Golestan University of Medical Sciences were obtained before study.

Experimental animals: Balb/c mice, weighing 28-30 g (8-9 weeks old) were used in this study. The animals were maintained in a climate-controlled room under a 12 h alternating light/dark cycle, 20-22°C temperature. Dry food pellets and water were provided ad libitum.

Drug: Each vial contained l mL of morphine sulphate (Darou Pakhsh Co., Iran) dissolved in 3.3 mL sterile saline solution (0.85%) to give 10 mg morphine sulphate dose intraperitoneally injected into mice.

Treated groups: After 2 weeks of acclimation to the diet and the environment, females were randomly divided into control and treated groups. Ten female mice in treated group received 10 mg kg-1 b.wt. of morphine sulphate intraperitoneally (IP) during 7 days before mating, gestational period (GD 0-21) 18 days after delivery in experimental group I and 32 days after delivery in experimental group II.

Twelve female mice in control groups received an equivalent volume normal saline intraperitoneally (IP) during 7 days before mating, gestational period (GD0-21) 18 days after delivery in experimental group 3 and 32 days after delivery in experimental group IV.

After parturition, in each group, six postnatal days 18 and 32 (P18, P32) were randomly selected and were scarified after anesthesia. The brain was exposed and fixed by immersion into the fixative solutions (10% neutral-buffered formalin). After techniques processing, brains sectioned at 6 micrometer thickness using a microtome (Microm HM 325 Germeny). The coronal sections (serial sections of anterior to posterior cerebrum) serially were selected according to anatomical landmarks corresponding to bregma -1.055 to -3.30 mm of the hippocampal formation with an interval of 24 μm between every two consecutive sections. The sections were used for immunohistochemistry. Adjacent sections stained with cresyl violet for morphometrical examination.

Immunohistochemistry: Immunocytochemical labeling to detect the neuronal marker was performed by monoclonal anti-neuronal nuclei (NeuN) antibody (Millipore corporation Billerica, MA 0 1821 USA) on 5 μm thick hippocampal coronal sections.

In brief, deparaffinized sections were preincubated with citrate buffer and were washed for 9 min in 0.01 M phosphate-buffered saline (PBS, pH 7.4) and treated with 0.3% hydrogen peroxide in 0.01 M PBS including 10% methanol. The brain sections were preincubated with blocking reagent and washed in 0.01 M PBS. Then, brain sections were incubated with anti-NeuN antibody (1:100) in a humidified chamber for 1 h at room temperature.

After rinse in 0.01 MPBS, the sections were incubated with the biotinylated secondary for 10 min and then with Streptavidin HRP and rinsed in PBS. Immunoreactivity was visualized using DAB (chromogen reagent) for 30 min at room temperature. Subsequently, the tissue specimen were counterstained with Mayer’s hematoxylin and mounted with entellan (Merck, USA).

Morphometric techniques: For histomorphometric study, the sections were observed under the light microscope.

In each postnatal mouse, ten similar sections of anterior to posterior of dentate gyrus were selected and images by Olympus BX 51 microscope and DP12 digital camera attached to OLYSIA autobioreport software (Olympus Optical, Co. LTD, Tokyo, Japan). The thickness of layers of dentate gyrus included moleculare layer (mo), granule cell layer (gc) and polymorph layer (pr) were obtained from 200X magnification (Fig. 1). The granular cells density evaluated through counting numbers of neuron per 10000 μm2 area of both dentate gyrus medial blade (DGmb) and dentate gyrus lateral blade (DGlb) in 1000X magnification.

Statistical analysis: The significance of difference between the means value for the obtained data was calculated according to the Student’s t-test using SPSS 11.5 software. A significance level of 0.05 was predetermined for all statistical analyses.

RESULTS

Granular cells density: In the P18 mice the numbers of NeuN positive neurons in Dentate Gyrus medial blade (DGmb) and Dentate Gyrus lateral blade (DGlb) significantly decreased from 171.45±4.2 and 174.51±3.1 cells in control group to 153.32±2.8 and 151.23±3.2 cells in 10000 μm2 area of granular layer in treated group (p<0.001) (Fig. 1-5).

Fig. 1: Overview of dentate gyrus areas used for quantitative measurements from Balb/c mice (P18) control animal. Coronal sections stained with cresyl violet. Quantification areas are: DGmb, dentate gyrus medial blade; DGlb, dentate gyrus lateral blade (x20 magnification, Scale bar: 100 μm)

Fig. 2: Histological section of dentate gyrus in Balb/c mice (P18) control animal. Dentate gyrus lateral blade (DGlb) stained with cresyl violet (layers including: molecular layer (mo), granule cell layer (gc) and polymorph layer (po), x400 magnification, Scale bar: 50 μm)

Fig. 3: Immunohistochemical staining for granular cells of dentate gyrus in Balb/c mice (P18) control animal. Neu N Immunopositive granular cells in granule cell layer of dentate gyrus lateral blade (DGlb). (layers including: molecular layer (mo), granule cell layer (gc) and polymorph layer (po), x400 magnification, Scale bar: 50 μm)

Fig. 4: The mean number of granular cells in dentate gyrus medial blade in P18 and P32 mice of control and morphine sulphate treated mothers. The cells were expressed as the number of granule cells per 10000 μm2, (results are Means±SEM, *Compared with control animals p<0.001, n = 6)

Fig. 5: The mean number of granular cells in dentate gyrus lateral blade in P18 and P32 mice of control and morphine sulphate treated mothers. The cells were expressed as the number of granule cells per 10000 μm2, (results are Means±SEM, *Compared with control animals p<0.001, n = 6)

Table 1: Thickness of the various layers of dentate gyrus in postnatal day (P18, P32) of morphine sulphate and control mothers
Results are expressed as Mean±SEM of the mean (*compared with control group, p<0.05, n = 6)

In P32 mice the numbers of NeuN positive granular neurons in mb and lb of dentate gyrus significantly decreased from 155.31±4.1 and 153.77±3.4 cells in control group to 138.33±4.5 and 135.13±4.3 cells in treated group (p<0.001) (Fig. 1-5).

Thickness of dentate gyrus layers: The thickness of mb and lb of dentate gyrus layers in P18 and P32 mice in cases and controls offspring mice is depicted in Table 1.

The results revealed a significant reduction in the granular layer thickness in mb of dentate gyrus of treated mice (55.16±1.7, 61.38±1.1) in compared to controls (64.50±1.4, 68.35±0.9) in P18 and P32 mice (p<0.05), respectively.

Also, the mean thickness of granular layer significantly reduced in the treated group (59.08±1.2, 63.20±2.3) in comparison with control group (67.49±1.6, 70.97±1.4) in lb of dentate gyrus in the postnatal 18 and 32 day mice (p<0.05), respectively (Table 1).

DISCUSSION

The current study found that morphine sulfate administration before, during pregnancy cause granular cells loss and reduction of dentate gyrus layer in 18 and 32 days old infant mice. Also our study showed that toxic effect of morphine continues on granular cells of hippocampus even after lactation period.

Several studies have shown the toxic effects of morphine on neuronal cells in different parts of CNS either in adult or fetal period (Mao et al., 2002; Svensson et al., 2008; Seatriz and Hammer, 1993; Mei et al., 2009; Emeterio et al., 2006; Eisch et al., 2000; Niu et al., 2009).

Another study (Mao et al., 2002) reported that morphine induces apoptosis in rat's spinal cord neurons.

Besides, other study has shown that morphine reduces number of hippocampal neurons and concluded that morphine induces apoptosis in hippocampal neuronal cells of mice fetus in in-vitro model (Svensson et al., 2008).

Several studies have reported that morphine reduces the number of neurons in sematosensory of 6 day old rats an d in layer II/III in lateral secondary visual cortex of rats (Seatriz and Hammer, 1993; Mei et al., 2009).

Furthermore, Emeterio et al. (2006) by using Double-immunofluorescence staining for the neuronal marker Neu-N and active caspase-3 and TUNEL assay combined with immunocytochemistry for the glial marker GFAP have shown that chronic morphine administration induces apoptosis in neuronal and glial cells.

Bekheet et al. (2010) showed that morphine exposure (during gestational period) orally reduces both cortical thickness and the numbers of neurons in the developing fetal frontal cerebral cortex.

Also, it is found that the long-term administration of morphine sulphate in dams significantly reduces the Purkinje cells numbers of mice offespring (Ghafari et al., 2011).

Indeed, the two studies have shown the toxic effects of morphine exposure on neuronal cells in dentate gyrus in the adult rat and rats offspring (Eisch et al., 2000; Niu et al., 2009).

Niu et al. (2009) study have reported that prenatal morphine exposure impairs the juvenile offspring's dentate synaptic plasticity and spatial memory. They concluded that decreased GABAergic inhibition may play a role in these effects.

Furthermore, long-term exposure to opiates inhibits neurogenesis in the adult rat hippocampus. Also, chronic, but not acute, morphine decreases the number of BrdUrd-positive cells in the subgranular zone of the dentate gyrus (Eisch et al., 2000).

Regarding to the effects of morphine on central nervous system, several following possible mechanisms can be considered neuronal cell loss in morphine treated animals can be due to Apoptosis or necrosis (Hauser et al., 1994). Morphine increase both Ca2+ and production of carbonyl oxidation. Ca2+ and carbonyl oxidation produce neuronal apoptosis or necrosis (Hauser et al., 1998).

Also, morphine increased Bax and Caspase-3 and reduced Bcl in rats which are indicator of apoptosis in neurons (Mao et al., 2002).

Furthermore, DNA synthesis blocking in neuroblasts can be established by morphine which this precursors arrest the neuronal cell proliferation during embryonic period. Also, this study showed that morphine (in vitro) arrests the genesis of mouse cerebellar granule neuron precursor and subsequently neuronal death (Hauser et al., 2000).

Besides, several investigations have shown that acute opioids exposure can arrested proliferation, differentiation and survival of neuroblasts and astroglia (Hauser et al., 1987; Lorber et al., 1990; Schmahl et al., 1989; Hammer et al., 1989; Zagon and MacLaughlin, 1987).

Neurotoxic effects of opioids can be induced by NMDAR-Caspase pathway. NMDA receptors are suggested to play a critical role in morphine-induced apoptosis in the superficial spinal cord dorsal horn of tolerant rats (Mao et al., 2002).

Also, prolonged morphine administration induces up-regulation of proapoptotic proteins caspase-3 and Bax as well as down-regulation of antiapoptotic protein Bcl-2. The general caspase inhibitor and caspase-3-specific inhibitor prevent morphine neurotoxicity (Mao et al., 2002).

Furthermore, mitochondrial damage (Cheng, et al., 2003; Pretorius and Bornman, 2005; Jacob, 2007) and reduction of calbindin protein as a neuroprotective agent in neurons can be considered as possible mechanism for the neuronal cells loss in hippocampus (Garcia et al., 1996).

Indeed, it was suggested that opioids block the neuronal activity, causing the neurons to receive internal signals to commit suicide (apoptosis) (Farber and Onley, 2003).

CONCLUSION

This study determined that morphine administration before and during gestational and lactation period causes the neuronal cells loss and reduction of the granular layer thickness of dentate gyrus in 18 and 32 days infant mice. Also, it can be conclude that the neurotoxic effect of morphine will be continued even with disconnect of exposure.

ACKNOWLEDGMENT

We thank the Deputy research of Golestan University of medical sciences for financial support (Grant No.: 1105).

REFERENCES

  • Atici, S., L. Cinel, I. Cinel, N. Doruk, M. Aktekin, A. Akca, H. Camdeviren and U. Oral, 2004. Opioid neurotoxicity: Comparison of morphine and tramadol in an experimental rat model. Int. J. Neurosci., 114: 1001-1011.
    CrossRef    Direct Link    


  • Bekheet, S.H., S.A. Saker, A.M. Abdel-Kader and A.E. Younis, 2010. Histopathological and biochemical changes of morphine sulphate administration on the cerebellum of albino rats. Tissue Cell, 42: 165-175.
    PubMed    


  • Cameron, H.A. and E. Gould, 1994. Adult neurogenesis is regulated by adrenal steroids in the dentate gyrus. Neuroscience, 61: 203-209.
    CrossRef    Direct Link    


  • Cheng, W.H., F.W. Quimby and X.G. Lei, 2003. Impacts of glutathione peroxidase-1 knockout on the protection by injected selenium against the pro-oxidant-induced liver aponecrosis and signaling in selenium deficient mice. Free Radic. Biol. Med., 34: 918-927.
    CrossRef    Direct Link    


  • Eisch, A.J., M. Barrot, C.A. Schad, D.W. Self and E.J. Nestler, 2000. Opiates inhibit neurogenesis in the adultrat hippocampus. PNAS, 97: 7579-7584.
    CrossRef    Direct Link    


  • Farber, N.B. and J.W. Onley, 2003. Drugs of abuse that cause developing neurons to commit suicide. Dev. Brain Res., 147: 37-45.
    CrossRef    Direct Link    


  • Garcia, M.M., J. Gilster and R.E. Harlan, 1996. Chronic morphine decreases calbindin D-28k immunoreactivity in a subset of cerebellar Purkinje neurons of rat brain. Brain Res., 734: 123-134.
    CrossRef    Direct Link    


  • Ghafari, S., D. Roshandel and M.J. Golalipour, 2011. Effect of intrauterine morphine sulfate exposure on cerebellar histomorphological changes in neonatal mice. Folia Neuropathol., 49: 328-334.
    Direct Link    


  • Gould, E. and P. Tanapat, 1997. Lesion-induced proliferation of neuronal progenitors in the dentate gyrus of the adult rat. Neuroscience, 80: 427-436.
    CrossRef    Direct Link    


  • Gould, E., P. Tanapat, T. Rydel and N. Hastings, 2000. Regulation of hippocampal neurogenesis in adulthood. Biol. Psychiatry, 48: 715-720.
    CrossRef    Direct Link    


  • Gould, E. and C.G. Gross, 2002. Neurogenesis in adult mammals: Some progress and problems. J. Neurosci., 22: 619-623.
    Direct Link    


  • Hammer, R.P.J., A.A. Ricalde and J.V. Seatriz, 1989. Effects of opiates on brain development. Neurotoxicology, 10: 475-483.
    Direct Link    


  • Hauser, K.F., J.A. Gurwell and C.S. Turbek, 1994. Morphine inhibits Purkinje cell survival and dendritic differentiation in organotypic cultures of the mouse cerebellum. Exp. Neurol., 130: 95-105.
    CrossRef    Direct Link    


  • Hauser, K.F., P.J. McLaughlin and I.S. Zagon, 1987. Endogenous opioids regulate dendritic growth and spine formation in developing rat brain. Brain. Res., 416: 157-161.
    CrossRef    Direct Link    


  • Hauser, K.F., M.E. Harris-white, J.A. Jackson, L.A.Opanashuk and J.M.Carney, 1998. Opioids disrupt Ca2+ homeostasis and induce carbonyl oxyradical production in mouse astrocytes In vitro: Transient increases and adaptation to sustained exposure. Exp. Neurol., 151: 70-76.
    CrossRef    Direct Link    


  • Hauser, K.F., A.A. Houdi, C.S. Turbeck, R.P. Elde and W. Maxson, 2000. Opioids intrinsically inhibit the genesis of mouse cerebellar granule neuron precursors In vitro: Differential impact of μ and δ receptor activation on proliferation and neurite elongation. Eur. J. Neurosci., 12: 1281-1293.
    CrossRef    Direct Link    


  • Jackson-Guilford, J., J.D. Leander and L.K. Nisenbaum, 2000. The effect of streptozotocin-induced diabetes on cell proliferation in the rat dentate gyrus. Neurosci. Lett., 293: 91-94.
    CrossRef    


  • Jacob, J.T., 2007. Effect of the cardiac glycoside digoxin, on neuronal viability, serotonin production and brain development in the embryo. MSc. Thesis, In Anatomy with Specialization in Cell Biology, School of Medicin, Faculty of Health Science, University of Pretoria, South Africa.


  • Lorber, B.A., S.K. Freitag and J.V. Bartolome, 1990. Effects of beta-endorphin on DNA synthesis in brain regions of preweanling rats. Brain Res., 531: 329-332.
    CrossRef    Direct Link    


  • Mao, J., B. Sung, RR. Ji and G. Lim, 2002. Neuronal apoptosis associated with morphine tolerance: Evidence for an opioid-induced neurotoxic mechanism. J. Neurosci., 22: 7650-7661.
    Direct Link    


  • Mei, B., L. Niu, B. Cao, D. Huang and Y. Zhou, 2009. Prenatal morphine exposure alters the layer II/III pyramidal neurons morphology in lateral secondary visual cortex of juvenile rats. Synapse, 63: 1154-1161.
    CrossRef    Direct Link    


  • Morris, R.G.M., E. Anderson, G.S. Lynch and M. Baudry, 1986. Selective impairment of learning and blockade of long-term potentiation by an N-methyl-D-aspartate receptor antagonist, AP5. Nature, 319: 774-776.
    CrossRef    Direct Link    


  • Morris, R.G., 1989. Synaptic plasticity and learning: Selective impairment of learning rats and blockade of long-term potentiation in vivo by the N-methyl-D-aspartate receptor antagonist AP5. J. Neurosci., 9: 3040-3057.
    Direct Link    


  • NIDA, 1996. National pregnancy and health survey: Drug use among women delivering live births: 1992. NIH Publication no. 96-3819. Department of Health and Human Services, Rockville, MD., USA., pp: 1-157.


  • Nestler, E.J., 2004. Historical review: Molecular and cellular mechanisms of opiate and cocaine addiction. Trends Pharmacol. Sci., 25: 210-218.
    CrossRef    


  • Niu, L., B. Cao, H. Zhu, B. Mei, M. Wang, Y. Yang and Y. Zhou, 2009. Impaired in vivo synaptic plasticity in dentate gyrus and spatial memory in juvenile rats induced by prenatal morphine exposure. Hippocampus, 19: 649-657.
    CrossRef    


  • Ornoy, A., V. Michailevskaya, I. Lukashov, R. Bar-Hamburger and S. Harel, 1996. The developmental outcome of children born to heroin-dependent mothers, raised at home or adopted. Child Abuse Neglect, 20: 385-396.
    CrossRef    


  • Emeterio, E.P.S., M. Tramullas and M.A. Hurle, 2006. Modulation of apoptosis in the mouse brain after morphine treatments and morphine withdrawal. J. Neurosci. Res., 83: 1352-1361.
    CrossRef    


  • Pretorius, E. and M.S. Bornman, 2005. Calcium-mediated aponecrosis plays a central role in the pathogenesis of estrogenic chemical-induced neurotoxicity. Med. Hypotheses, 65: 893-904.
    CrossRef    


  • Seatriz, J.V. and R.P. Hammer, 1993. Effects of opiates on neuronal development in the rat cerebral cortex. Brain Res. Bull., 30: 523-527.
    CrossRef    


  • Schmahl, W., R. Funk, U. Miaskowski and J. Plendl, 1989. Long-lasting effects of naltrexone, an opioid receptor antagonist, on cell proliferation in developing rat forebrain. Brain Res., 486: 297-300.
    CrossRef    


  • Silva, A.J., R. Paylor, J.M. Wehner and S. Tonegawa, 1992. Impaired spatial learning in alpha-calcium-calmodulin kinase II mutant mice. Science, 257: 206-211.
    PubMed    Direct Link    


  • Svensson, A.L., N. Bucht, M. Hallberg and F. Nyberg, 2008. Reversal of opiate-induced apoptosis by human recombinant growth hormone in murine foetus primary hippocampal neuronal cell cultures. Proc. Natl. Acad. Sci., 105: 7304-7308.
    CrossRef    PubMed    


  • Turchan-Cholewo, J., Y. Liu, S. Gartner, R. Reid and C. Jie et al., 2006. Increased vulnerability of ApoE4 neurons to HIV proteins and opiates: Protection by diosgenin and L-deprenyl. Neurobiol. Dis., 23: 109-119.
    CrossRef    PubMed    


  • United Nations Office on Drugs and Crime, 2006. World drug report 2006. New York: United Nations.


  • Wilson, G.S., R. McCreay, J. Kean and J.C. Baxter, 1979. The development of pre-school children of heroin-addicted mothers: A controlled study. Pediatrics, 63: 135-141.
    PubMed    Direct Link    


  • Yang, S.N., L.T. Huang, C.L. Wang, W.F. Chen and C.H. Yang et al., 2003. Prenatal administration of morphine decreases CREBSerine‐133 phosphorylation and synaptic plasticity range mediated by glutamatergic transmission in the hippocampal CA1 area of cognitive‐deficient rat offspring. Hippocampus, 13: 915-921.
    CrossRef    


  • Yang, S.N., C.A. Liu, M.Y. Chung, H.C. Huang and G.C. Yeh et al., 2006. Alterations of postsynaptic density proteins in the hippocampus of rat offspring from the morphine‐addicted mother: Beneficial effect of dextromethorphan. Hippocampus, 16: 521-530.
    CrossRef    


  • Zagon, I.S. and P.J. MacLaughlin, 1987. Endogenous opioid systems regulate cell proliferation in the developing rat brain. Brain Res., 412: 68-72.
    CrossRef    


  • Zagon, I.S. and P.J. MacLaughlin, 1992. Maternal Exposure to Opioids and the Developing Nervous System: Laboratory Findings. In: Maternal Substance Abuse and the Developing Nervous System, Zagon, I.S. and T.A. Slotkin (Eds.). Academic Press, New York, pp: 241-282


  • Zhang, Y., Q. Chen and L.C. Yu, 2008. Morphine: A protective or destructive role in neurons? Neuroscientist, 14: 561-570.
    CrossRef    Direct Link    

  • © Science Alert. All Rights Reserved