全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Distinct and Additive Effects of Alcohol and Thiamine Deficiency in the Developing Brain: Relevance to Fetal Alcohol Spectrum Disorder

DOI: 10.4236/jbbs.2024.146011, PP. 161-186

Keywords: Fetal Alcohol Spectrum Disorder, Thiamine Deficiency, Cerebellum, Slice Culture, Pyrithiamine

Full-Text   Cite this paper   Add to My Lib

Abstract:

Background: Neurodevelopmental abnormalities in fetal alcohol spectrum disorder (FASD) are linked to brain insulin resistance and oxidative stress. However, the role of thiamine deficiency as a distinct or additive factor in the pathogenesis of the neurodevelopmental and metabolic derangements in FASD has not been determined. Methods: Control and ethanol-exposed human PNET2 cerebellar neuronal cells and rat cerebellar slice cultures were treated with vehicle or pyrithiamine (Pyr) to assess independent and additive effects of thiamine deficiency on ethanol-mediated neurotoxicity, mitochondrial dysfunction, insulin resistance, inhibition of neuronal and glial genes, and oxidative stress. Results: Pyr treatments (0 - 200 μM) caused dose-dependent cell loss (Crystal Violet assay) and reduced mitochondrial function (MTT assay) in PNET2 neuronal cultures. Ethanol alone (100 mM) significantly reduced PNET2 neuronal viability, MTT activity, and ATP production. Over the broad dose range of Pyr treatment, ethanol significantly reduced ATP content and cell number and increased mitochondrial mass (MitoTracker Green). Ex vivo cerebellar slice culture studies revealed ethanol-induced developmental architectural disruption that was substantially worsened by Pyr. The adverse effects of ethanol were linked to increased lipid peroxidation and inhibition of asparatyl-asparaginyl-β-hydroxylase (ASPH) expression. The independent and additive effects of Pyr were associated with increased cytotoxicity, lipid peroxidation, Caspase 3 activation, and Tau accumulation. Conclusions: During development, alcohol exposure and thiamine deficiency exert distinct but overlapping molecular pathologies that ultimately impair the structure and function of cerebellar neurons. While both insults drive cell loss and mitochondrial dysfunction with increased lipid peroxidation, ethanol’s additional inhibitory effects on ASPH reflect impairments in insulin and IGF signaling. In contrast, Pyr’s main adverse effects were likely due to neurotoxicity and the activation of apoptosis cascades. The findings suggest that FASD severity may be reduced by thiamine supplementation, but without additional support for insulin/IGF signaling networks, FASD would not be prevented.

References

[1]  Mattson, S.N., Crocker, N. and Nguyen, T.T. (2011) Fetal Alcohol Spectrum Disorders: Neuropsychological and Behavioral Features. Neuropsychology Review, 21, 81-101.
https://doi.org/10.1007/s11065-011-9167-9
[2]  Mattson, S.N., Schoenfeld, A.M. and Riley, E.P. (2001) Teratogenic Effects of Alcohol on Brain and Behavior. Alcohol Research & Health, 25, 185-191.
[3]  Riley, E.P., Infante, M.A. and Warren, K.R. (2011) Fetal Alcohol Spectrum Disorders: An Overview. Neuropsychology Review, 21, 73-80.
https://doi.org/10.1007/s11065-011-9166-x
[4]  de la Monte, S.M. and Wands, J.R. (2010) Role of Central Nervous System Insulin Resistance in Fetal Alcohol Spectrum Disorders. Journal of Population Therapeutics and Clinical Pharmacology, 17, e390-e404.
[5]  Banerje, K., Mohry, L., Wands, J.R. and de la Monte, S.M. (1998) Ethanol Inhibition of Insulin Signaling in Hepatocellular Carcinoma Cells. Alcoholism: Clinical and Experimental Research, 22, 2093-2101.
https://doi.org/10.1111/j.1530-0277.1998.tb05921.x
[6]  Mohr, L., Tanaka, S. and Wands, J.R. (1998) Ethanol Inhibits Hepatocyte Proliferation in Insulin Receptor Substrate 1 Transgenic Mice. Gastroenterology, 115, 1558-1565.
https://doi.org/10.1016/s0016-5085(98)70036-8
[7]  Xu, J., Yeon, J.E., Chang, H., Tison, G., Chen, G.J., Wands, J., et al. (2003) Ethanol Impairs Insulin-Stimulated Neuronal Survival in the Developing Brain: Role of PTEN Phosphatase. Journal of Biological Chemistry, 278, 26929-26937.
https://doi.org/10.1074/jbc.m300401200
[8]  Soscia, S.J., Tong, M., Xu, X.J., Cohen, A.C., Chu, J., Wands, J.R., et al. (2006) Chronic Gestational Exposure to Ethanol Causes Insulin and IGF Resistance and Impairs Acetylcholine Homeostasis in the Brain. Cellular and Molecular Life Sciences CMLS, 63, 2039-2056.
https://doi.org/10.1007/s00018-006-6208-2
[9]  He, J., de la Monte, S. and Wands, J.R. (2007) Acute Ethanol Exposure Inhibits Insulin Signaling in the Liver. Hepatology, 46, 1791-1800.
https://doi.org/10.1002/hep.21904
[10]  Yeon, J.E., Califano, S., Xu, J., Wands, J.R. and De La Monte, S.M. (2003) Potential Role of PTEN Phosphatase in Ethanol-Impaired Survival Signaling in the Liver. Hepatology, 38, 703-714.
https://doi.org/10.1053/jhep.2003.50368
[11]  Camp, M.C., Mayfield, R.D., McCracken, M., McCracken, L. and Alcantara, A.A. (2006) Neuroadaptations of Cdk5 in Cholinergic Interneurons of the Nucleus Accumbens and Prefrontal Cortex of Inbred Alcohol-Preferring Rats Following Voluntary Alcohol Drinking. Alcoholism: Clinical and Experimental Research, 30, 1322-1335.
https://doi.org/10.1111/j.1530-0277.2006.00160.x
[12]  de la Monte, S.M., Ganju, N., Banerjee, K., Brown, N.V., Luong, T. and Wands, J.R. (2000) Partial Rescue of Ethanol-Induced Neuronal Apoptosis by Growth Factor Activation of Phosphoinositol-3-Kinase. Alcoholism: Clinical and Experimental Research, 24, 716-726.
https://doi.org/10.1111/j.1530-0277.2000.tb02044.x
[13]  de la Monte, S.M., Neely, T.R., Cannon, J. and Wands, J.R. (2001) Ethanol Impairs Insulin-Stimulated Mitochondrial Function in Cerebellar Granule Neurons. Cellular and Molecular Life Sciences, 58, 1950-1960.
https://doi.org/10.1007/pl00000829
[14]  Hallak, H., Seiler, A.E.M., Green, J.S., Henderson, A., Ross, B.N. and Rubin, R. (2001) Inhibition of Insulin-Like Growth Factor-I Signaling by Ethanol in Neuronal Cells. Alcoholism: Clinical and Experimental Research, 25, 1058-1064.
https://doi.org/10.1111/j.1530-0277.2001.tb02317.x
[15]  Rajgopal, Y. and Vemuri, M.C. (2001) Ethanol Induced Changes in Cyclin-Dependent Kinase-5 Activity and Its Activators, P35, P67 (munc-18) in Rat Brain. Neuroscience Letters, 308, 173-176.
https://doi.org/10.1016/s0304-3940(01)02011-0
[16]  Zhang, F.X., Rubin, R. and Rooney, T.A. (1998) Ethanol Induces Apoptosis in Cerebellar Granule Neurons by Inhibiting Insulin-Like Growth Factor 1 Signaling. Journal of Neurochemistry, 71, 196-204.
https://doi.org/10.1046/j.1471-4159.1998.71010196.x
[17]  del Campo, M. and Jones, K.L. (2017) A Review of the Physical Features of the Fetal Alcohol Spectrum Disorders. European Journal of Medical Genetics, 60, 55-64.
https://doi.org/10.1016/j.ejmg.2016.10.004
[18]  Wozniak, J.R., Riley, E.P. and Charness, M.E. (2019) Clinical Presentation, Diagnosis, and Management of Fetal Alcohol Spectrum Disorder. The Lancet Neurology, 18, 760-770.
https://doi.org/10.1016/s1474-4422(19)30150-4
[19]  Cherian, P.P., Schenker, S. and Henderson, G.I. (2008) Ethanol-Mediated DNA Damage and PARP-1 Apoptotic Responses in Cultured Fetal Cortical Neurons. Alcoholism: Clinical and Experimental Research, 32, 1884-1892.
https://doi.org/10.1111/j.1530-0277.2008.00769.x
[20]  Chu, J., Tong, M. and de la Monte, S.M. (2007) Chronic Ethanol Exposure Causes Mitochondrial Dysfunction and Oxidative Stress in Immature Central Nervous System Neurons. Acta Neuropathologica, 113, 659-673.
https://doi.org/10.1007/s00401-007-0199-4
[21]  de la Monte, S.M. and Wands, J.R. (2001) Mitochondrial DNA Damage and Impaired Mitochondrial Function Contribute to Apoptosis of Insulin-Stimulated Ethanol-Exposed Neuronal Cells. Alcoholism: Clinical and Experimental Research, 25, 898-906.
https://doi.org/10.1111/j.1530-0277.2001.tb02296.x
[22]  Acheson, S.K., Stein, R.M. and Swartzwelder, H.S. (1998) Impairment of Semantic and Figural Memory by Acute Ethanol: Age-Dependent Effects. Alcoholism: Clinical and Experimental Research, 22, 1437-1442.
https://doi.org/10.1111/j.1530-0277.1998.tb03932.x
[23]  Markwiese, B.J., Acheson, S.K., Levin, E.D., Wilson, W.A. and Swartzwelder, H.S. (1998) Differential Effects of Ethanol on Memory in Adolescent and Adult Rats. Alcoholism: Clinical and Experimental Research, 22, 416-421.
https://doi.org/10.1111/j.1530-0277.1998.tb03668.x
[24]  White, A.M., Matthews, D.B. and Best, P.J. (2000) Ethanol, Memory, and Hippocampal Function: A Review of Recent Findings. Hippocampus, 10, 88-93.
https://doi.org/10.1002/(sici)1098-1063(2000)10:1<88::aid-hipo10>3.0.co;2-l
[25]  Matthews, D.B. and Silvers, J.R. (2004) The Use of Acute Ethanol Administration as a Tool to Investigate Multiple Memory Systems. Neurobiology of Learning and Memory, 82, 299-308.
https://doi.org/10.1016/j.nlm.2004.06.007
[26]  de la Monte, S.M., Tong, M., Carlson, R.I., Carter, J.J., Longato, L., Silbermann, E., et al. (2009) Ethanol Inhibition of Aspartyl-Asparaginyl-β-Hydroxylase in Fetal Alcohol Spectrum Disorder: Potential Link to the Impairments in Central Nervous System Neuronal Migration. Alcohol, 43, 225-240.
https://doi.org/10.1016/j.alcohol.2008.09.009
[27]  de Licona, H.K., Karacay, B., Mahoney, J., McDonald, E., Luang, T. and Bonthius, D.J. (2009) A Single Exposure to Alcohol during Brain Development Induces Microencephaly and Neuronal Losses in Genetically Susceptible Mice, but Not in Wild Type Mice. NeuroToxicology, 30, 459-470.
https://doi.org/10.1016/j.neuro.2009.01.010
[28]  Marquardt, K. and Brigman, J.L. (2016) The Impact of Prenatal Alcohol Exposure on Social, Cognitive and Affective Behavioral Domains: Insights from Rodent Models. Alcohol, 51, 1-15.
https://doi.org/10.1016/j.alcohol.2015.12.002
[29]  Martin, P.R., Singleton, C.K. and Hiller-Sturmhofel, S. (2003) The Role of Thiamine Deficiency in Alcoholic Brain Disease. Alcohol Research & Health, 27, 134-142.
[30]  Singleton, C. and Martin, P. (2001) Molecular Mechanisms of Thiamine Utilization. Current Molecular Medicine, 1, 197-207.
https://doi.org/10.2174/1566524013363870
[31]  Victor, M., Davis, R.D. and Collins, G.H. (1989) The Wernicke-Korsakoff Syndrome and Related Neurologic Disorders Due to Alcoholism and Malnutrition. F.A. Davis.
[32]  Sullivan, E.V. and Pfefferbaum, A. (2009) Neuroimaging of the Wernicke-Korsakoff Syndrome. Alcohol and Alcoholism, 44, 155-165.
https://doi.org/10.1093/alcalc/agn103
[33]  Bâ, A. (2009) Alcohol and B1 Vitamin Deficiency-Related Stillbirths. The Journal of Maternal-Fetal & Neonatal Medicine, 22, 452-457.
https://doi.org/10.1080/14767050802609775
[34]  Levin, S.W., Roecklein, B.A. and Mukherjee, A.B. (1985) Intrauterine Growth Retardation Caused by Dietary Biotin and Thiamine Deficiency in the Rat. Research in Experimental Medicine, 185, 375-381.
https://doi.org/10.1007/bf01851917
[35]  Roecklein, B., Levin, S.W., Comly, M. and Mukherjee, A.B. (1985) Intrauterine Growth Retardation Induced by Thiamine Deficiency and Pyrithiamine during Pregnancy in the Rat. American Journal of Obstetrics and Gynecology, 151, 455-460.
https://doi.org/10.1016/0002-9378(85)90269-8
[36]  Ba, A., N’Douba, V., D’Almeida, M. and Seri, B.V. (2005) Effects of Maternal Thiamine Deficiencies on the Pyramidal and Granule Cells of the Hippocampus of Rat Pups. Acta Neurobiologiae Experimentalis, 65, 387-398.
https://doi.org/10.55782/ane-2005-1567
[37]  Pires, R.G.W., Pereira, S.R.C., Oliveira-Silva, I.F., Franco, G.C. and Ribeiro, A.M. (2005) Cholinergic Parameters and the Retrieval of Learned and Re-Learned Spatial Information: A Study Using a Model of Wernicke-Korsakoff Syndrome. Behavioural Brain Research, 162, 11-21.
https://doi.org/10.1016/j.bbr.2005.02.032
[38]  Langlais, P.J. and Zhang, S. (1997) Cortical and Subcortical White Matter Damage without Wernicke’s Encephalopathy after Recovery from Thiamine Deficiency in the Rat. Alcoholism: Clinical and Experimental Research, 21, 434-443.
https://doi.org/10.1111/j.1530-0277.1997.tb03788.x
[39]  Hoyumpa, A.M. (1980) Mechanisms of Thiamin Deficiency in Chronic Alcoholism. The American Journal of Clinical Nutrition, 33, 2750-2761.
https://doi.org/10.1093/ajcn/33.12.2750
[40]  Shaw, S., Gorkin, B.D. and Lieber, C.S. (1981) Effects of Chronic Alcohol Feeding on Thiamin Status: Biochemical and Neurological Correlates. The American Journal of Clinical Nutrition, 34, 856-860.
https://doi.org/10.1093/ajcn/34.5.856
[41]  Harper, C. and Kril, J. (1994) An Introduction to Alcohol-Induced Brain Damage and Its Causes. Alcohol and Alcoholism, 2, 237-243.
[42]  Todd, K.G., Hazell, A.S. and Butterworth, R.F. (1999) Alcohol-Thiamine Interactions: An Update on the Pathogenesis of Wernicke Encephalopathy. Addiction Biology, 4, 261-272.
https://doi.org/10.1080/13556219971470
[43]  Murdock, D.S. and Gubler, C.J. (1973) Effects of Thiamine Deficiency and Treatment with the Antagonists, Oxythiamine and Pyri-Thiamine, on the Levels and Distribution of Thiamine Derivatives in Rat Brain. Journal of Nutritional Science and Vitaminology, 19, 237-249.
https://doi.org/10.3177/jnsv.19.237
[44]  McCandless, D.W., Malone, M.J. and Szoke, M. (1976) Pyrithiamine-Induced Thiamine Deficiency: Effects on Rat Myelination. International Journal for Vitamin and Nutrition Research, 46, 24-32.
[45]  Liu, J., Timm, D.E. and Hurley, T.D. (2006) Pyrithiamine as a Substrate for Thiamine Pyrophosphokinase. Journal of Biological Chemistry, 281, 6601-6607.
https://doi.org/10.1074/jbc.m510951200
[46]  Koedam, J.C. (1958) The Mode of Action of Pyrithiamine as an Inductor of Thiamine Deficiency. Biochimica et Biophysica Acta, 29, 333-344.
https://doi.org/10.1016/0006-3002(58)90192-6
[47]  Pfefferbaum, A., Adalsteinsson, E., Bell, R.L. and Sullivan, E.V. (2006) Development and Resolution of Brain Lesions Caused by Pyrithiamine-and Dietary-Induced Thiamine Deficiency and Alcohol Exposure in the Alcohol-Preferring Rat: A Longitudinal Magnetic Resonance Imaging and Spectroscopy Study. Neuropsychopharmacology, 32, 1159-1177.
https://doi.org/10.1038/sj.npp.1301107
[48]  Zhang, S.X., Weilersbacher, G.S., Henderson, S.W., Corso, T., Olney, J.W. and Langlais, P.J. (1995) Excitotoxic Cytopathology, Progression, and Reversibility of Thiamine Deficiency-Induced Diencephalic Lesions. Journal of Neuropathology and Experimental Neurology, 54, 255-267.
https://doi.org/10.1097/00005072-199503000-00012
[49]  Kopelman, M.D., Thomson, A.D., Guerrini, I. and Marshall, E.J. (2009) The Korsakoff Syndrome: Clinical Aspects, Psychology and Treatment. Alcohol and Alcoholism, 44, 148-154.
https://doi.org/10.1093/alcalc/agn118
[50]  Jia, S., VanDusen, W.J., Diehl, R.E., Kohl, N.E., Dixon, R.A., et al. (1992) Cdna Cloning and Expression of Bovine Aspartyl (Asparaginyl) Beta-Hydroxylase. Journal of Biological Chemistry, 267, 14322-14327.
https://doi.org/10.1016/s0021-9258(19)49715-9
[51]  Lavaissiere, L., Jia, S., Nishiyama, M., de la Monte, S., Stern, A.M., Wands, J.R., et al. (1996) Overexpression of Human Aspartyl(asparaginyl)beta-Hydroxylase in Hepatocellular Carcinoma and Cholangiocarcinoma. Journal of Clinical Investigation, 98, 1313-1323.
https://doi.org/10.1172/jci118918
[52]  Dinchuk, J.E., Henderson, N.L., Burn, T.C., Huber, R., Ho, S.P., Link, J., et al. (2000) Aspartyl β-Hydroxylase (Asph) and an Evolutionarily Conserved Isoform of Asph Missing the Catalytic Domain Share Exons with Junctin. Journal of Biological Chemistry, 275, 39543-39554.
https://doi.org/10.1074/jbc.m006753200
[53]  Cantarini, M.C., de la Monte, S.M., Pang, M., Tong, M., D’Errico, A., Trevisani, F., et al. (2006) Aspartyl-Asparagyl Β Hydroxylase Over-Expression in Human Hepatoma Is Linked to Activation of Insulin-Like Growth Factor and Notch Signaling Mechanisms. Hepatology, 44, 446-457.
https://doi.org/10.1002/hep.21272
[54]  Monte, S.M.d.l., Tamaki, S., Cantarini, M.C., Ince, N., Wiedmann, M., Carter, J.J., et al. (2006) Aspartyl-(asparaginyl)-β-hydroxylase Regulates Hepatocellular Carcinoma Invasiveness. Journal of Hepatology, 44, 971-983.
https://doi.org/10.1016/j.jhep.2006.01.038
[55]  Lahousse, S.A., Carter, J.J., Xu, X.J., Wands, J.R. and de la Monte, S.M. (2006) Differential Growth Factor Regulation of Aspartyl-(asparaginyl)-β-Hydroxylase Family Genes in SH-Sy5y Human Neuroblastoma Cells. BMC Cell Biology, 7, Article No. 41.
https://doi.org/10.1186/1471-2121-7-41
[56]  Lawton, M., Tong, M., Gundogan, F., Wands, J.R. and de la Monte, S.M. (2000) Aspartyl-(Asparaginyl)-β-Hydroxylase, Hypoxia-Inducible Factor-1α and Notch Cross-Talk in Regulating Neuronal Motility. Oxidative Medicine and Cellular Longevity, 3, 347-356.
https://doi.org/10.4161/oxim.3.5.13296
[57]  Silbermann, E., Moskal, P., Bowling, N., Tong, M. and de la Monte, S.M. (2010) Role of Aspartyl-(Asparaginyl)-β-Hydroxylase Mediated Notch Signaling in Cerebellar Development and Function. Behavioral and Brain Functions, 6, Article No. 68.
https://doi.org/10.1186/1744-9081-6-68
[58]  Carter, J.J., Tong, M., Silbermann, E., Lahousse, S.A., Ding, F.F., Longato, L., et al. (2008) Ethanol Impaired Neuronal Migration Is Associated with Reduced Aspartyl-Asparaginyl-β-Hydroxylase Expression. Acta Neuropathologica, 116, 303-315.
https://doi.org/10.1007/s00401-008-0377-z
[59]  Tong, M., Gao, J.S., Borgas, D. and de la Monte, S.M. (2013) Phosphorylation Modulates Aspartyl-(Asparaginyl)-Beta Hydroxylase Protein Expression, Catalytic Activity and Migration in Human Immature Neuronal Cerebellar Cells. Current Biology (Henderson, NV), 6, 133.
https://doi.org/10.4172/2324-9293.1000133
[60]  Feoktistova, M., Geserick, P. and Leverkus, M. (2016) Crystal Violet Assay for Determining Viability of Cultured Cells. Cold Spring Harbor Protocols, 2016, pdb.prot087379.
https://doi.org/10.1101/pdb.prot087379
[61]  Ghasemi, M., Turnbull, T., Sebastian, S. and Kempson, I. (2021) The MTT Assay: Utility, Limitations, Pitfalls, and Interpretation in Bulk and Single-Cell Analysis. International Journal of Molecular Sciences, 22, Article 12827.
https://doi.org/10.3390/ijms222312827
[62]  Karl, P.I. and Fisher, S.E. (1993) Ethanol Alters Hormone Production in Cultured Human Placental Trophoblasts. Alcoholism: Clinical and Experimental Research, 17, 816-821.
https://doi.org/10.1111/j.1530-0277.1993.tb00847.x
[63]  Li, Z., Zharikova, A., Vaughan, C.H., Bastian, J., Zandy, S., Esperon, L., et al. (2010) Intermittent High-Dose Ethanol Exposures Increase Motivation for Operant Ethanol Self-Administration: Possible Neurochemical Mechanism. Brain Research, 1310, 142-153.
https://doi.org/10.1016/j.brainres.2009.11.029
[64]  Silvers, J.M., Tokunaga, S., Mittleman, G., O’Buckley, T., Morrow, A.L. and Matthews, D.B. (2006) Chronic Intermittent Ethanol Exposure during Adolescence Reduces the Effect of Ethanol Challenge on Hippocampal Allopregnanolone Levels and Morris Water Maze Task Performance. Alcohol, 39, 151-158.
https://doi.org/10.1016/j.alcohol.2006.09.001
[65]  Adickes, E.D., Mollner, T.J. and Lockwood, S.K. (1988) Closed Chamber System for Delivery of Ethanol to Cell Cultures. Alcohol and Alcoholism, 23, 377-381.
https://doi.org/10.1093/oxfordjournals.alcalc.a044832
[66]  Moroz, N., Tong, M., Longato, L., Xu, H. and de la Monte, S.M. (2008) Limited Alzheimer-Type Neurodegeneration in Experimental Obesity and Type 2 Diabetes Mellitus. Journal of Alzheimers Disease, 15, 29-44.
https://doi.org/10.3233/jad-2008-15103
[67]  Stern-Straeter, J., Bonaterra, G.A., Hörmann, K., Kinscherf, R. and Goessler, U.R. (2009) Identification of Valid Reference Genes during the Differentiation of Human Myoblasts. BMC Molecular Biology, 10, Article No. 66.
https://doi.org/10.1186/1471-2199-10-66
[68]  Deochand, C., Tong, M., Agarwal, A.R., Cadenas, E. and de la Monte, S.M. (2016) Tobacco Smoke Exposure Impairs Brain Insulin/IGF Signaling: Potential Co-Factor Role in Neurodegeneration. Journal of Alzheimers Disease, 50, 373-386.
https://doi.org/10.3233/jad-150664
[69]  Tong, M., Gonzalez-Navarrete, H., Kirchberg, T., Gotama, B., Yalcin, E.B., Kay, J., et al. (2017) Ethanol-Induced White Matter Atrophy Is Associated with Impaired Expression of Aspartyl-Asparaginyl-β-Hydroxylase (ASPH) and Notch Signaling in an Experimental Rat Model. Journal of Drug and Alcohol Research, 6, 1-12.
[70]  Romero, I.A., Rist, R.J., Aleshaiker, A. and Abbott, N.J. (1997) Metabolic and Permeability Changes Caused by Thiamine Deficiency in Immortalized Rat Brain Microvessel Endothelial Cells. Brain Research, 756, 133-140.
https://doi.org/10.1016/s0006-8993(97)00127-3
[71]  Lupton, C., Burd, L. and Harwood, R. (2004) Cost of Fetal Alcohol Spectrum Disorders. American Journal of Medical Genetics Part C: Seminars in Medical Genetics, 127, 42-50.
https://doi.org/10.1002/ajmg.c.30015
[72]  May, P.A., Chambers, C.D., Kalberg, W.O., Zellner, J., Feldman, H., Buckley, D., et al. (2018) Prevalence of Fetal Alcohol Spectrum Disorders in 4 US Communities. JAMA, 319, 474-482.
https://doi.org/10.1001/jama.2017.21896
[73]  Wozniak, J.R. and Muetzel, R.L. (2011) What Does Diffusion Tensor Imaging Reveal about the Brain and Cognition in Fetal Alcohol Spectrum Disorders? Neuropsychology Review, 21, 133-147.
https://doi.org/10.1007/s11065-011-9162-1
[74]  Gundogan, F., Elwood, G., Longato, L., Tong, M., Feijoo, A., Carlson, R.I., et al. (2008) Impaired Placentation in Fetal Alcohol Syndrome. Placenta, 29, 148-157.
https://doi.org/10.1016/j.placenta.2007.10.002
[75]  Gundogan, F., Gilligan, J., Qi, W., Chen, E., Naram, R. and de la Monte, S.M. (2015) Dose Effect of Gestational Ethanol Exposure on Placentation and Fetal Growth. Placenta, 36, 523-530.
https://doi.org/10.1016/j.placenta.2015.02.010
[76]  Guttmacher, A.E., Maddox, Y.T. and Spong, C.Y. (2014) The Human Placenta Project: Placental Structure, Development, and Function in Real Time. Placenta, 35, 303-304.
https://doi.org/10.1016/j.placenta.2014.02.012
[77]  Herrick, E.J. and Bordone, B. (2024) Embryology, Placenta. StatPearls Publishing.
[78]  Lewis, P.D. (1985) Neuropathological Effects of Alcohol on the Developing Nervous System. Alcohol and Alcoholism, 20, 195-200.
[79]  West, J.R., Kelly, S.J. and Pierce, D.R. (1987) Severity of Alcohol-Induced Deficits in Rats during the Third Trimester Equivalent Is Determined by the Pattern of Exposure. Alcohol and Alcoholism, 1, 461-465.
[80]  de la Monte, S.M. and Wands, J.R. (2002) Chronic Gestational Exposure to Ethanol Impairs Insulin-Stimulated Survival and Mitochondrial Function in Cerebellar Neurons. Cellular and Molecular Life Sciences, 59, 882-893.
https://doi.org/10.1007/s00018-002-8475-x
[81]  Light, K.E., Brown, D.P., Newton, B.W., Belcher, S.M. and Kane, C.J.M. (2002) Ethanol-Induced Alterations of Neurotrophin Receptor Expression on Purkinje Cells in the Neonatal Rat Cerebellum. Brain Research, 924, 71-81.
https://doi.org/10.1016/s0006-8993(01)03224-3
[82]  Riley, E.P. and McGee, C.L. (2005) Fetal Alcohol Spectrum Disorders: An Overview with Emphasis on Changes in Brain and Behavior. Experimental Biology and Medicine, 230, 357-365.
https://doi.org/10.1177/15353702-0323006-03
[83]  de la Monte, S.M., Tong, M., Bowling, N. and Moskal, P. (2011) Si-RNA Inhibition of Brain Insulin or Insulin-Like Growth Factor Receptors Causes Developmental Cerebellar Abnormalities: Relevance to Fetal Alcohol Spectrum Disorder. Molecular Brain, 4, Article No. 13.
https://doi.org/10.1186/1756-6606-4-13
[84]  Ewenczyk, A., Ziplow, J. and Tong, M. (2012) Sustained Impairments in Brain Insulin/IGF Signaling in Adolescent Rats Subjected to Binge Alcohol Exposure during Development. Journal of Clinical & Experimental Pathology, 2, 106.
https://doi.org/10.4172/2161-0681.1000106
[85]  Tong, M., et al. (2012) Motor Function Deficits Following Chronic Prenatal Ethanol Exposure Are Linked to Impairments in Insulin/IGF, Notch and Wnt Signaling in the Cerebellum. Journal of Diabetes & Metabolism, 4, Article ID: 1000238.
https://doi.org/10.4172/2155-6156.1000238
[86]  Tong, M., Longato, L., Nguyen, Q., Chen, W.C., Spaisman, A. and de la Monte, S.M. (2011) Acetaldehyde-Mediated Neurotoxicity: Relevance to Fetal Alcohol Spectrum Disorders. Oxidative Medicine and Cellular Longevity, 2011, Article ID: 213286.
https://doi.org/10.1155/2011/213286
[87]  Papp-Peka, A., Tong, M., Kril, J.J., De La Monte, S.M. and Sutherland, G.T. (2016) The Differential Effects of Alcohol and Nicotine-Specific Nitrosamine Ketone on White Matter Ultrastructure. Alcohol and Alcoholism, 52, 165-171.
https://doi.org/10.1093/alcalc/agw067
[88]  Tong, M., Andreani, T., Krotow, A., Gundogan, F. and de la Monte, S.M. (2016) Potential Contributions of the Tobacco Nicotine-Derived Nitrosamine Ketone to White Matter Molecular Pathology in Fetal Alcohol Spectrum Disorder. Journal of Neurology and Brain Disorders, 3, 1-12.
[89]  Tong, M., Yu, R., Deochand, C. and de la Monte, S.M. (2015) Differential Contributions of Alcohol and the Nicotine-Derived Nitrosamine Ketone (NNK) to Insulin and Insulin-Like Growth Factor Resistance in the Adolescent Rat Brain. Alcohol and Alcoholism, 50, 670-679.
https://doi.org/10.1093/alcalc/agv101
[90]  Andreani, T., Tong, M., Gundogan, F., Silbermann, E. and de la Monte, S.M. (2016) Differential Effects of 3rd Trimester-Equivalent Binge Ethanol and Tobacco-Specific Nitrosamine Ketone Exposures on Brain Insulin Signaling in Adolescence. Journal of Diabetes and Related Disorders, 1, 105-114.
[91]  Zabala, V., Silbermann, E., Re, E., Andreani, T., Tong, M., et al. (2016) Potential Co-Factor Role of Tobacco Specific Nitrosamine Exposures in the Pathogenesis of Fetal Alcohol Spectrum Disorder. Gynecology and Obstetrics ResearchOpen Journal, 2, 112-125.
https://doi.org/10.17140/goroj-2-125
[92]  Legido, A. (1997) Intrauterine Exposure to Drugs. Revue Neurologique, 25, 691-702.
[93]  Blusztajn, J., Slack, B. and Mellott, T. (2017) Neuroprotective Actions of Dietary Choline. Nutrients, 9, Article No. 815.
https://doi.org/10.3390/nu9080815
[94]  Wozniak, J.R., Fink, B.A., Fuglestad, A.J., Eckerle, J.K., Boys, C.J., Sandness, K.E., et al. (2020) Four-Year Follow-Up of a Randomized Controlled Trial of Choline for Neurodevelopment in Fetal Alcohol Spectrum Disorder. Journal of Neurodevelopmental Disorders, 12, Article No. 9.
https://doi.org/10.1186/s11689-020-09312-7
[95]  Harper, C. (1982) Neuropathology of Brain Damage Caused by Alcohol. Medical Journal of Australia, 2, 277-282.
https://doi.org/10.5694/j.1326-5377.1982.tb124389.x
[96]  Harper, C. and Kril, J. (1985) Brain Atrophy in Chronic Alcoholic Patients: A Quantitative Pathological Study. Journal of Neurology, Neurosurgery & Psychiatry, 48, 211-217.
https://doi.org/10.1136/jnnp.48.3.211
[97]  Bruyn, G.W. (1989) The Wernicke-Korsakoff Syndrome and Related Neurologic Disorders Due to Alcoholism and Malnutrition 2nd Edn., by Victor, M., R. D. Adams and G. H. Collins, F. A. Davis Company, Philadelphia, PA, 1989. Journal of the Neurological Sciences, 92, 117.
https://doi.org/10.1016/0022-510x(89)90182-2
[98]  Subramanya, S.B., Subramanian, V.S. and Said, H.M. (2010) Chronic Alcohol Consumption and Intestinal Thiamin Absorption: Effects on Physiological and Molecular Parameters of the Uptake Process. American Journal of Physiology-Gastrointestinal and Liver Physiology, 299, G23-G31.
https://doi.org/10.1152/ajpgi.00132.2010
[99]  Apostolova, N., Gomez-Sucerquia, L., Moran, A., Alvarez, A., Blas-Garcia, A. and Esplugues, J. (2010) Enhanced Oxidative Stress and Increased Mitochondrial Mass during Efavirenz-Induced Apoptosis in Human Hepatic Cells. British Journal of Pharmacology, 160, 2069-2084.
https://doi.org/10.1111/j.1476-5381.2010.00866.x
[100]  Korga, A., Józefczyk, A., Zgórka, G., Homa, M., Ostrowska, M., Burdan, F., et al. (2017) Evaluation of the Phytochemical Composition and Protective Activities of Methanolic Extracts of Centaurea borysthenica and Centaurea daghestanica (Lipsky) Wagenitz on Cardiomyocytes Treated with Doxorubicin. Food & Nutrition Research, 61, Article ID: 1344077.
https://doi.org/10.1080/16546628.2017.1344077
[101]  Monte, S.M.d.l., Xu, X.J. and Wands, J.R. (2005) Ethanol Inhibits Insulin Expression and Actions in the Developing Brain. CMLS Cellular and Molecular Life Sciences, 62, 1131-1145.
https://doi.org/10.1007/s00018-005-4571-z
[102]  Goodlett, C.R., Mahoney, J.C. and West, J.R. (1989) Brain Growth Deficits Following a Single Day of Alcohol Exposure in the Neonatal Rat. Alcohol, 6, 121-126.
https://doi.org/10.1016/0741-8329(89)90036-0
[103]  Pierce, D.R. and West, J.R. (1987) Differential Deficits in Regional Brain Growth Induced by Postnatal Alcohol. Neurotoxicology and Teratology, 9, 129-141.
https://doi.org/10.1016/0892-0362(87)90089-4
[104]  Butterworth, R.F. (1989) Effects of Thiamine Deficiency on Brain Metabolism: Implications for the Pathogenesis of the Wernicke-Korsakoff Syndrome. Alcohol and Alcoholism, 24, 271-279.
https://doi.org/10.1093/oxfordjournals.alcalc.a044913
[105]  Yang, F. and Luo, J. (2015) Endoplasmic Reticulum Stress and Ethanol Neurotoxicity. Biomolecules, 5, 2538-2553.
https://doi.org/10.3390/biom5042538
[106]  Chornyy, S., Parkhomenko, J. and Chorna, N. (2007) Thiamine Deficiency Caused by Thiamine Antagonists Triggers Upregulation of Apoptosis Inducing Factor Gene Expression and Leads to Caspase 3-Mediated Apoptosis in Neuronally Differentiated Rat PC-12 Cells. Acta Biochimica Polonica, 54, 315-322.
https://doi.org/10.18388/abp.2007_3252
[107]  Ince, N., de la Monte, S.M. and Wands, J.R. (2000) Overexpression of Human Aspartyl (Asparaginyl) beta-Hydroxylase Is Associated with Malignant Transformation. Cancer Research, 60, 1261-1266.
[108]  Aihara, A., Huang, C., Olsen, M.J., Lin, Q., Chung, W., Tang, Q., et al. (2014) A Cell-Surface β-Hydroxylase Is a Biomarker and Therapeutic Target for Hepatocellular Carcinoma. Hepatology, 60, 1302-1313.
https://doi.org/10.1002/hep.27275
[109]  Luu, M., Sabo, E., de la Monte, S.M., Greaves, W., Wang, J., Tavares, R., et al. (2009) Prognostic Value of Aspartyl (asparaginyl)-β-Hydroxylase/Humbug Expression in Non-Small Cell Lung Carcinoma. Human Pathology, 40, 639-644.
https://doi.org/10.1016/j.humpath.2008.11.001
[110]  Maeda, T., Sepe, P., Lahousse, S., Tamaki, S., Enjoji, M., et al. (2003) Antisense Oligodeoxynucleotides Directed against Aspartyl (Asparaginyl) β-Hydroxylase Suppress Migration of Cholangiocarcinoma Cells. Journal of Hepatology, 38, 615-622.
https://doi.org/10.1016/s0168-8278(03)00052-7
[111]  Sepe, P.S., Lahousse, S.A., Gemelli, B., Chang, H., Maeda, T., Wands, J.R., et al. (2002) Role of the Aspartyl-Asparaginyl-β-Hydroxylase Gene in Neuroblastoma Cell Motility. Laboratory Investigation, 82, 881-891.
https://doi.org/10.1097/01.lab.0000020406.91689.7f
[112]  Lee, J. (2008) Overexpression of Humbug Promotes Malignant Progression in Human Gastric Cancer Cells. Oncology Reports, 19, 795-800.
https://doi.org/10.3892/or.19.3.795
[113]  Wang, J., de la Monte, S.M., Sabo, E., Kethu, S., Tavares, R., Branda, M., et al. (2007) Prognostic Value of Humbug Gene Overexpression in Stage II Colon Cancer. Human Pathology, 38, 17-25.
https://doi.org/10.1016/j.humpath.2006.07.009
[114]  De La Monte, S.M., Yeon, J., Tong, M., Longato, L., Chaudhry, R., Pang, M., et al. (2008) Insulin Resistance in Experimental Alcohol-Induced Liver Disease. Journal of Gastroenterology and Hepatology, 23, e477-e486.
https://doi.org/10.1111/j.1440-1746.2008.05339.x
[115]  Finotti, A., Treves, S., Zorzato, F., Gambari, R. and Feriotto, G. (2008) Upstream Stimulatory Factors Are Involved in the P1 Promoter Directed Transcription of the Abetah-J-J Locus. BMC Molecular Biology, 9, Article No. 110.
https://doi.org/10.1186/1471-2199-9-110
[116]  Sturla, L., Tong, M., Hebda, N., Gao, J., Thomas, J., Olsen, M., et al. (2016) Aspartate-β-Hydroxylase (ASPH): A Potential Therapeutic Target in Human Malignant Gliomas. Heliyon, 2, e00203.
https://doi.org/10.1016/j.heliyon.2016.e00203
[117]  Huang, C., Iwagami, Y., Aihara, A., Chung, W., de la Monte, S., Thomas, J., et al. (2016) Anti-Tumor Effects of Second Generation β-Hydroxylase Inhibitors on Cholangiocarcinoma Development and Progression. PLOS ONE, 11, e0150336.
https://doi.org/10.1371/journal.pone.0150336

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133