1
|
Dou X, Lee JY, Charness ME. Neuroprotective Peptide NAPVSIPQ Antagonizes Ethanol Inhibition of L1 Adhesion by Promoting the Dissociation of L1 and Ankyrin-G. Biol Psychiatry 2020; 87:656-665. [PMID: 31640849 PMCID: PMC7056560 DOI: 10.1016/j.biopsych.2019.08.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/21/2019] [Accepted: 08/21/2019] [Indexed: 01/14/2023]
Abstract
BACKGROUND Ethanol causes developmental neurotoxicity partly by blocking adhesion mediated by the L1 neural cell adhesion molecule. This action of ethanol is antagonized by femtomolar concentrations of the neuropeptide NAPVSIPQ (NAP), an active fragment of the activity-dependent neuroprotective protein (ADNP). How femtomolar concentrations of NAP antagonize millimolar concentrations of ethanol is unknown. L1 sensitivity to ethanol requires L1 association with ankyrin-G; therefore, we asked whether NAP promotes the dissociation of ankyrin-G and L1. METHODS L1-ankyrin-G association was studied using immunoprecipitation, Western blotting, and immunofluorescence in NIH/3T3 cells transfected with wild-type and mutated human L1 genes. Phosphorylation of the ankyrin binding motif in the L1 cytoplasmic domain was studied after NAP treatment of intact cells, rat brain homogenates, and purified protein fragments. RESULTS Femtomolar concentrations of NAP stimulated the phosphorylation of tyrosine-1229 (L1-Y1229) at the ankyrin binding motif of the L1 cytoplasmic domain, leading to the dissociation of L1 from ankyrin-G and the spectrin-actin cytoskeleton. NAP increased the association of L1 and EphB2 and directly activated EphB2 phosphorylation of L1-Y1229. These actions of NAP were reproduced by P7A-NAP, a NAP variant that also blocks the teratogenic actions of ethanol, but not by I6A-NAP, which does not block ethanol teratogenesis as potently. Finally, knockdown of EPHB2 prevented ethanol inhibition of L1 adhesion in NIH/3T3 cells. CONCLUSIONS NAP potently antagonizes ethanol inhibition of L1 adhesion by stimulating EphB2 phosphorylation of L1-Y1229. EphB2 plays a critical role in synaptic development; its potent activation by NAP suggests that ADNP may mediate synaptic development partly by activating EphB2.
Collapse
Affiliation(s)
- Xiaowei Dou
- Veterans Affairs Boston Healthcare System; Department of Neurology, Harvard Medical School, West Roxbury, MA 02132
| | - Jerry Y. Lee
- Veterans Affairs Boston Healthcare System; Department of Neurology, Harvard Medical School, West Roxbury, MA 02132
| | - Michael E. Charness
- Veterans Affairs Boston Healthcare System; Department of Neurology, Harvard Medical School, West Roxbury, MA 02132,Department of Neurology, Boston University, School of Medicine, Boston, MA 02119, To whom correspondence should be addressed. Michael E. Charness, M.D., VA Boston Healthcare System, 1400 VFW Parkway, West Roxbury, MA 02132, Phone: 857-203-6011,
| |
Collapse
|
2
|
Cerrizuela S, Vega-Lopez GA, Aybar MJ. The role of teratogens in neural crest development. Birth Defects Res 2020; 112:584-632. [PMID: 31926062 DOI: 10.1002/bdr2.1644] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/11/2019] [Accepted: 12/22/2019] [Indexed: 12/13/2022]
Abstract
The neural crest (NC), discovered by Wilhelm His 150 years ago, gives rise to a multipotent migratory embryonic cell population that generates a remarkably diverse and important array of cell types during the development of the vertebrate embryo. These cells originate in the neural plate border (NPB), which is the ectoderm between the neural plate and the epidermis. They give rise to the neurons and glia of the peripheral nervous system, melanocytes, chondrocytes, smooth muscle cells, odontoblasts and neuroendocrine cells, among others. Neurocristopathies are a class of congenital diseases resulting from the abnormal induction, specification, migration, differentiation or death of NC cells (NCCs) during embryonic development and have an important medical and societal impact. In general, congenital defects affect an appreciable percentage of newborns worldwide. Some of these defects are caused by teratogens, which are agents that negatively impact the formation of tissues and organs during development. In this review, we will discuss the teratogens linked to the development of many birth defects, with a strong focus on those that specifically affect the development of the NC, thereby producing neurocristopathies. Although increasing attention is being paid to the effect of teratogens on embryonic development in general, there is a strong need to critically evaluate the specific role of these agents in NC development. Therefore, increased understanding of the role of these factors in NC development will contribute to the planning of strategies aimed at the prevention and treatment of human neurocristopathies, whose etiology was previously not considered.
Collapse
Affiliation(s)
- Santiago Cerrizuela
- Área Biología Experimental, Instituto Superior de Investigaciones Biológicas (INSIBIO, CONICET-UNT), Tucumán, Argentina.,Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Tucumán, Argentina
| | - Guillermo A Vega-Lopez
- Área Biología Experimental, Instituto Superior de Investigaciones Biológicas (INSIBIO, CONICET-UNT), Tucumán, Argentina.,Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Tucumán, Argentina
| | - Manuel J Aybar
- Área Biología Experimental, Instituto Superior de Investigaciones Biológicas (INSIBIO, CONICET-UNT), Tucumán, Argentina.,Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Tucumán, Argentina
| |
Collapse
|
3
|
Cellular and animal models of skin alterations in the autism-related ADNP syndrome. Sci Rep 2019; 9:736. [PMID: 30679581 PMCID: PMC6346103 DOI: 10.1038/s41598-018-36859-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 11/26/2018] [Indexed: 12/28/2022] Open
Abstract
Mutations in ADNP have been recently associated with intellectual disability and autism spectrum disorder. However, the clinical features of patients with this syndrome are not fully identified, and no treatment currently exists for these patients. Here, we extended the ADNP syndrome phenotype describing skin abnormalities in both a patient with ADNP syndrome and an Adnp haploinsufficient mice. The patient displayed thin dermis, hyperkeratotic lesions in periarticular areas and delayed wound healing. Patient-derived skin keratinocytes showed reduced proliferation and increased differentiation. Additionally, detection of cell cycle markers indicated that mutant cells exhibited impaired cell cycle progression. Treatment of ADNP-deficient keratinocytes with the ADNP-derived NAP peptide significantly reduced the expression of differentiation markers. Sonography and immunofluorescence staining of epidermal layers revealed that the dermis was thinner in the patient than in a healthy control. Adnp haploinsufficient mice (Adnp+/−) mimicked the human condition showing reduced dermal thickness. Intranasal administration of NAP significantly increased dermal thickness and normalized the levels of cell cycle and differentiation markers. Our observations provide a novel activity of the autism-linked ADNP in the skin that may serve to define the clinical phenotype of patients with ADNP syndrome and provide an attractive therapeutic option for skin alterations in these patients.
Collapse
|
4
|
Petrelli B, Weinberg J, Hicks GG. Effects of prenatal alcohol exposure (PAE): insights into FASD using mouse models of PAE. Biochem Cell Biol 2018; 96:131-147. [PMID: 29370535 PMCID: PMC5991836 DOI: 10.1139/bcb-2017-0280] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The potential impact of prenatal alcohol exposure (PAE) varies considerably among exposed individuals, with some displaying serious alcohol-related effects and many others showing few or no overt signs of fetal alcohol spectrum disorder (FASD). In animal models, variables such as nutrition, genetic background, health, other drugs, and stress, as well as dosage, duration, and gestational timing of exposure to alcohol can all be controlled in a way that is not possible in a clinical situation. In this review we examine mouse models of PAE and focus on those with demonstrated craniofacial malformations, abnormal brain development, or behavioral phenotypes that may be considered FASD-like outcomes. Analysis of these data should provide a valuable tool for researchers wishing to choose the PAE model best suited to their research questions or to investigate established PAE models for FASD comorbidities. It should also allow recognition of patterns linking gestational timing, dosage, and duration of PAE, such as recognizing that binge alcohol exposure(s) during early gestation can lead to severe FASD outcomes. Identified patterns could be particularly insightful and lead to a better understanding of the molecular mechanisms underlying FASD.
Collapse
Affiliation(s)
- Berardino Petrelli
- Department of Biochemistry & Medical Genetics; Regenerative Medicine Program, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Joanne Weinberg
- Department of Cellular & Physiological Sciences, Faculty of Medicine, Life Sciences Institute, University of British Columbia, UBC Institute of Mental Health, Vancouver, British Columbia, Canada
| | - Geoffrey G. Hicks
- Department of Biochemistry & Medical Genetics; Regenerative Medicine Program, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
5
|
Goodlett CR, Horn KH, Zhou FC. Alcohol Teratogenesis: Mechanisms of Damage and Strategies for Intervention. Exp Biol Med (Maywood) 2016; 230:394-406. [PMID: 15956769 DOI: 10.1177/15353702-0323006-07] [Citation(s) in RCA: 250] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
There are multiple mechanisms by which alcohol can damage the developing brain, but the type of damage induced will depend on the amount and developmental timing of exposure, along with other maternal and genetic factors. This article reviews current perspectives on how ethanol can produce neuroteratogenic effects by its interactions with molecular regulators of brain development. The current evidence suggests that alcohol produces many of its damaging effects by exerting specific actions on molecules that regulate key developmental processes (e.g., L1 cell adhesion molecule, alcohol dehydrogenase, catalase), interfering with the early development of midline serotonergic neurons and disrupting their regulatory-signaling function for other target brain structures, interfering with trophic factors that regulate neurogenesis and cell survival, or inducing excessive cell death via oxidative stress or activation of caspase-3 proteases. The current understanding of pathogenesis mechanisms suggests several strategic approaches to develop rational molecular prevention. However, the development of behavioral and biologic treatments for alcohol-affected children is crucial because it is unlikely that effective delivery of preventative interventions can realistically be achieved in ways to prevent prenatal damage in at-risk pregnancies. Toward that end, behavioral training that promotes experience-dependent neuroplasticity has been effective in a rat model of cerebellar damage induced by alcohol exposure during the period of brain development that is comparable to that of the human third trimester.
Collapse
Affiliation(s)
- Charles R Goodlett
- Department of Psychology, Indiana University-Purdue University at Indianapolis, 402 North Blackford Street, Indianapolis, Indiana 46202, USA.
| | | | | |
Collapse
|
6
|
Warren KR. A Review of the History of Attitudes Toward Drinking in Pregnancy. Alcohol Clin Exp Res 2016; 39:1110-7. [PMID: 26137906 DOI: 10.1111/acer.12757] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 04/22/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND It is now well accepted in pediatrics and obstetrics that prenatal alcohol is a teratogenic agent and the primary causative factor underlying fetal alcohol spectrum disorders (FASDs), although for the majority of the 20th century that knowledge was either unknown or ignored. At least 2 factors contributed to the delay in recognizing alcohol's role in teratogenicity: the rejection of earlier evidence pertaining to alcohol and pregnancy following the repeal of Prohibition in the United States, Canada, and several European countries; and misinterpretation of earlier research findings in a eugenic rather than toxicological context. The pervasive belief held well into the 1970s that there was no risk to either mother or fetus from prenatal alcohol posed a major challenge to changing physician and public attitudes on alcohol and pregnancy. This review provides insight on key events that occurred in changing physician and public understanding of the risks posed by prenatal alcohol use in pregnancy. METHODS Historical review of events primarily in the U.S. federal government, found in referenced documents. RESULTS The transition in physician and public understanding of the risks posed by prenatal alcohol use was aided by the existence of National Institute on Alcohol Abuse and Alcoholism (NIAAA) which was created in 1971. This government agency was able to support research on alcohol and pregnancy immediately following the 1973 published clinical reports calling attention to a proposed fetal alcohol syndrome (FAS). These early research studies provided the foundation for the first government health advisory on alcohol and pregnancy, issued by NIAAA in 1977. Subsequently, the U.S. Food and Drug Administration (FDA) used this new knowledge on FAS in their effort to add alcoholic beverages to the range of products with ingredient and consumer information labeling. The ensuing hearings and actions resulted in a new health advisory under the auspices of the Surgeon General, encouraging avoidance of alcohol consumption in pregnancy. In subsequent years, Congressional attention to the FAS issue resulted in the Alcoholic Beverage Labeling Law. CONCLUSIONS The pace at which understanding of the risks of prenatal alcohol moved forward from a total misunderstanding to acceptance was aided by both the efforts of the NIAAA in its support of research, and the FDA in its efforts to improve consumer information. Today, many women in the United States as well as other countries continue to ignore advisories on avoiding alcohol consumption in pregnancy, emphasizing the need for persistence in education on these health risks.
Collapse
Affiliation(s)
- Kenneth R Warren
- National Institute on Alcohol Abuse and Alcoholism , National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
7
|
Abstract
BACKGROUND Alcohol regulates the expression and function of protein kinase C epsilon (PKCε). In a previous study we identified an alcohol binding site in the C1B, one of the twin C1 subdomains of PKCε (Das et al., Biochem. J., 421, 405-13, 2009). METHODS In this study, we investigated alcohol binding in the entire C1 domain (combined C1A and C1B) of PKCε. Fluorescent phorbol ester, SAPD and fluorescent diacylglycerol (DAG) analog, dansyl-DAG were used to study the effect of ethanol, butanol, and octanol on the ligand binding using fluorescence resonance energy transfer (FRET). To identify alcohol binding site(s), PKCεC1 was photolabeled with 3-azibutanol and 3-azioctanol, and analyzed by mass spectrometry. The effects of alcohols and the azialcohols on PKCε were studied in NG108-15 cells. RESULTS In the presence of alcohol, SAPD and dansyl-DAG showed different extent of FRET, indicating differential effects of alcohol on the C1A and C1B subdomains. Effects of alcohols and azialcohols on PKCε in NG108-15 cells were comparable. Azialcohols labeled Tyr-176 of C1A and Tyr-250 of C1B. Inspection of the model structure of PKCεC1 reveals that these residues are 40Å apart from each other indicating that these residues form two different alcohol binding sites. CONCLUSIONS The present results provide evidence for the presence of multiple alcohol-binding sites on PKCε and underscore the importance of targeting this PKC isoform in developing alcohol antagonists.
Collapse
Affiliation(s)
- Satyabrata Pany
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, United States
| | - Joydip Das
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, United States.
| |
Collapse
|
8
|
Sulik KK. Fetal alcohol spectrum disorder: pathogenesis and mechanisms. HANDBOOK OF CLINICAL NEUROLOGY 2014; 125:463-75. [PMID: 25307590 DOI: 10.1016/b978-0-444-62619-6.00026-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
This chapter provides an overview of animal model-based studies that have generated information critical to our understanding of the pathogenesis and mechanisms underlying alcohol-induced birth defects, in particular those involving the brain. Focus is placed on the developing organism itself, rather than the mother, placenta, or other extraembryonic tissues. Components of the cascades of alcohol-induced damage that are considered herein are excessive cell death, changes in the cell cycle and proliferation, cell migration, cell morphogenesis, and gene expression as well as free radical damage and interference with cell signaling. The roles played by one or more of these various factors in the genesis of structural and functional birth defects are dependent upon alcohol exposure patterns and dosage, the involved tissue, and the prenatal stage(s) at the time of exposure. Technologic advances and rapidly increasing knowledge in the fields of genetics, cell, developmental, and neurobiology are critical to accurately piecing together experimental evidence in refining our understanding of the genesis of alcohol-induced birth defects, to the planning and execution of future studies, and to applying the knowledge gained to diminish the severity or occurrence of fetal alcohol spectrum disorder.
Collapse
Affiliation(s)
- Kathleen K Sulik
- Department of Cell Biology and Physiology and Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
9
|
Seydlová G, Halada P, Fišer R, Toman O, Ulrych A, Svobodová J. DnaK and GroEL chaperones are recruited to the Bacillus subtilis membrane after short-term ethanol stress. J Appl Microbiol 2012; 112:765-74. [DOI: 10.1111/j.1365-2672.2012.05238.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
10
|
Fitzgerald DM, Charness ME, Leite-Morris KA, Chen S. Effects of ethanol and NAP on cerebellar expression of the neural cell adhesion molecule L1. PLoS One 2011; 6:e24364. [PMID: 21931691 PMCID: PMC3169602 DOI: 10.1371/journal.pone.0024364] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 08/08/2011] [Indexed: 01/16/2023] Open
Abstract
The neural cell adhesion molecule L1 is critical for brain development and plays a role in learning and memory in the adult. Ethanol inhibits L1-mediated cell adhesion and neurite outgrowth in cerebellar granule neurons (CGNs), and these actions might underlie the cerebellar dysmorphology of fetal alcohol spectrum disorders. The peptide NAP potently blocks ethanol inhibition of L1 adhesion and prevents ethanol teratogenesis. We used quantitative RT-PCR and Western blotting of extracts of cerebellar slices, CGNs, and astrocytes from postnatal day 7 (PD7) rats to investigate whether ethanol and NAP act in part by regulating the expression of L1. Treatment of cerebellar slices with 20 mM ethanol, 10−12 M NAP, or both for 4 hours, 24 hours, and 10 days did not significantly affect L1 mRNA and protein levels. Similar treatment for 4 or 24 hours did not regulate L1 expression in primary cultures of CGNs and astrocytes, the predominant cerebellar cell types. Because ethanol also damages the adult cerebellum, we studied the effects of chronic ethanol exposure in adult rats. One year of binge drinking did not alter L1 gene and protein expression in extracts from whole cerebellum. Thus, ethanol does not alter L1 expression in the developing or adult cerebellum; more likely, ethanol disrupts L1 function by modifying its conformation and signaling. Likewise, NAP antagonizes the actions of ethanol without altering L1 expression.
Collapse
Affiliation(s)
- Devon M. Fitzgerald
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts, United States of America
| | - Michael E. Charness
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts, United States of America
- Department of Neurology, Harvard Medical School, West Roxbury, Massachusetts, United States of America
- Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Kimberly A. Leite-Morris
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts, United States of America
- Departments of Psychiatry, Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Suzhen Chen
- Department of Neurology, Harvard Medical School, West Roxbury, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
11
|
Dou X, Menkari CE, Shanmugasundararaj S, Miller KW, Charness ME. Two alcohol binding residues interact across a domain interface of the L1 neural cell adhesion molecule and regulate cell adhesion. J Biol Chem 2011; 286:16131-9. [PMID: 21367865 DOI: 10.1074/jbc.m110.209254] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ethanol may cause fetal alcohol spectrum disorders (FASD) in part by inhibiting cell adhesion mediated by the L1 neural cell adhesion molecule. Azialcohols photolabel Glu-33 and Tyr-418, two residues that are predicted by homology modeling to lie within 2.8 Å of each other at the interface between the Ig1 and Ig4 domains of L1 (Arevalo, E., Shanmugasundararaj, S., Wilkemeyer, M. F., Dou, X., Chen, S., Charness, M. E., and Miller, K. W. (2008) Proc. Natl. Acad. Sci. U.S.A. 105, 371-375). Using transient transfection of NIH/3T3 cells with wild type (WT-L1) and mutated L1, we found that cysteine substitution of both residues (E33C/Y418C-L1) significantly increased L1 adhesion above levels observed for WT-L1 or the single cysteine substitutions E33C-L1 or Y418C-L1. The reducing agent β-mercaptoethanol (βME) reversibly decreased the adhesion of E33C/Y418C-L1, but had no effect on WT-L1, E33C-L1, or Y418C-L1. Thus, disulfide bond formation occurs between Cys-33 and Cys-418, confirming both the close proximity of these residues and the importance of Ig1-Ig4 interactions in L1 adhesion. Maximal ethanol inhibition of cell adhesion was significantly lower in cells expressing E33C/Y418C-L1 than in those expressing WT-L1, E33C-L1, or Y418C-L1. Moreover, the effects of βME and ethanol on E33C/Y418C-L1 adhesion were non-additive. The cutoff for alcohol inhibition of WT-L1 adhesion was between 1-butanol and 1-pentanol. Increasing the size of the alcohol binding pocket by mutating Glu-33 to Ala-33, increased the alcohol cutoff from 1-butanol to 1-decanol. These findings support the hypothesis that alcohol binding within a pocket bordered by Glu-33 and Tyr-418 inhibits L1 adhesion by disrupting the Ig1-Ig4 interaction.
Collapse
Affiliation(s)
- Xiaowei Dou
- Veterans Affairs Boston Healthcare System, Department of Neurology, Harvard Medical School, West Roxbury, Massachusetts 02132, USA
| | | | | | | | | |
Collapse
|
12
|
Thomas JD, Idrus NM, Monk BR, Dominguez HD. Prenatal choline supplementation mitigates behavioral alterations associated with prenatal alcohol exposure in rats. ACTA ACUST UNITED AC 2011; 88:827-37. [PMID: 20706995 DOI: 10.1002/bdra.20713] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Prenatal alcohol exposure can alter physical and behavioral development, leading to a range of fetal alcohol spectrum disorders. Despite warning labels, pregnant women continue to drink alcohol, creating a need to identify effective interventions to reduce the severity of alcohol's teratogenic effects. Choline is an essential nutrient that influences brain and behavioral development. Recent studies indicate that choline supplementation can reduce the teratogenic effects of developmental alcohol exposure. The present study examined whether choline supplementation during prenatal ethanol treatment could mitigate the adverse effects of ethanol on behavioral development. METHODS Pregnant Sprague-Dawley rats were intubated with 6 g/kg/day ethanol in a binge-like manner from gestational days 5-20; pair-fed and ad libitum chow controls were included. During treatment, subjects from each group were intubated with either 250 mg/kg/day choline chloride or vehicle. Spontaneous alternation, parallel bar motor coordination, Morris water maze, and spatial working memory were assessed in male and female offspring. RESULTS Subjects prenatally exposed to alcohol exhibited delayed development of spontaneous alternation behavior and deficits on the working memory version of the Morris water maze during adulthood, effects that were mitigated with prenatal choline supplementation. Neither alcohol nor choline influenced performance on the motor coordination task. CONCLUSIONS These data indicate that choline supplementation during prenatal alcohol exposure may reduce the severity of fetal alcohol effects, particularly on alterations in tasks that require behavioral flexibility. These findings have important implications for children of women who drink alcohol during pregnancy.
Collapse
Affiliation(s)
- Jennifer D Thomas
- Center for Behavioral Teratology, Department of Psychology, San Diego State University, 6363 Alvarado Court, San Diego, CA 92120, USA.
| | | | | | | |
Collapse
|
13
|
Wiertz RWF, Marani E, Rutten WLC. Inhibition of neuronal cell–cell adhesion measured by the microscopic aggregation assay and impedance sensing. J Neural Eng 2010; 7:056003. [DOI: 10.1088/1741-2560/7/5/056003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
14
|
Thomas JD, Abou EJ, Dominguez HD. Prenatal choline supplementation mitigates the adverse effects of prenatal alcohol exposure on development in rats. Neurotoxicol Teratol 2009; 31:303-11. [PMID: 19616089 PMCID: PMC2952280 DOI: 10.1016/j.ntt.2009.07.002] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2008] [Revised: 06/30/2009] [Accepted: 07/07/2009] [Indexed: 10/20/2022]
Abstract
Prenatal alcohol exposure can lead to a range of physical, neurological, and behavioral alterations referred to as fetal alcohol spectrum disorders (FASD). Variability in outcome observed among children with FASD is likely related to various pre- and postnatal factors, including nutritional variables. Choline is an essential nutrient that influences brain and behavioral development. Recent animal research indicates that prenatal choline supplementation leads to long-lasting cognitive enhancement, as well as changes in brain morphology, electrophysiology and neurochemistry. The present study examined whether choline supplementation during ethanol exposure effectively reduces fetal alcohol effects. Pregnant dams were exposed to 6.0g/kg/day ethanol via intubation from gestational days (GD) 5-20; pair-fed and lab chow controls were included. During treatment, subjects from each group received choline chloride (250mg/kg/day) or vehicle. Physical development and behavioral development (righting reflex, geotactic reflex, cliff avoidance, reflex suspension and hindlimb coordination) were examined. Subjects prenatally exposed to alcohol exhibited reduced birth weight and brain weight, delays in eye opening and incisor emergence, and alterations in the development of all behaviors. Choline supplementation significantly attenuated ethanol's effects on birth and brain weight, incisor emergence, and most behavioral measures. In fact, behavioral performance of ethanol-exposed subjects treated with choline did not differ from that of controls. Importantly, choline supplementation did not influence peak blood alcohol level or metabolism, indicating that choline's effects were not due to differential alcohol exposure. These data indicate early dietary supplements may reduce the severity of some fetal alcohol effects, findings with important implications for children of women who drink alcohol during pregnancy.
Collapse
Affiliation(s)
- Jennifer D Thomas
- Department of Psychology, Center for Behavioral Teratology, San Diego State University, San Diego, CA 92120, USA.
| | | | | |
Collapse
|
15
|
Silva JM, Ezhkova E, Silva J, Heart S, Castillo M, Campos Y, Castro V, Bonilla F, Cordon-Cardo C, Muthuswamy SK, Powers S, Fuchs E, Hannon GJ. Cyfip1 is a putative invasion suppressor in epithelial cancers. Cell 2009; 137:1047-61. [PMID: 19524508 PMCID: PMC2754270 DOI: 10.1016/j.cell.2009.04.013] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2008] [Revised: 01/23/2009] [Accepted: 04/02/2009] [Indexed: 12/20/2022]
Abstract
Identification of bona fide tumor suppressors is often challenging because of the large number of genetic alterations present in most human cancers. To evaluate candidate genes present within chromosomal regions recurrently deleted in human cancers, we coupled high-resolution genomic analysis with a two-stage genetic study using RNA interference (RNAi). We found that Cyfip1, a subunit of the WAVE complex, which regulates cytoskeletal dynamics, is commonly deleted in human epithelial cancers. Reduced expression of CYFIP1 is commonly observed during invasion of epithelial tumors and is associated with poor prognosis in this setting. Silencing of Cyfip1 disturbed normal epithelial morphogenesis in vitro and cooperated with oncogenic Ras to produce invasive carcinomas in vivo. Mechanistically, we have linked alterations in WAVE-regulated actin dynamics with impaired cell-cell adhesion and cell-ECM interactions. Thus, we propose Cyfip1 as an invasion suppressor gene.
Collapse
Affiliation(s)
- Jose M Silva
- Watson School Biological Sciences, Howard Hughes Medical Institute, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Ethanol inhibits neuronal differentiation by disrupting activity-dependent neuroprotective protein signaling. Proc Natl Acad Sci U S A 2008; 105:19962-7. [PMID: 19047645 DOI: 10.1073/pnas.0807758105] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The mechanisms by which ethanol damages the developing and adult central nervous system (CNS) remain unclear. Activity-dependent neuroprotective protein (ADNP) is a glial protein that protects the CNS against a wide array of insults and is critical for CNS development. NAPVSIPQ (NAP), a potent active fragment of ADNP, potentiated axon outgrowth in cerebellar granule neurons by activating the sequential tyrosine phosphorylation of Fyn kinase and the scaffold protein Crk-associated substrate (Cas). Pharmacological inhibition of Fyn kinase or expression of a Fyn kinase siRNA abolished NAP-mediated axon outgrowth. Concentrations of ethanol attained after social drinking blocked NAP-mediated axon outgrowth (IC(50) = 17 mM) by inhibiting NAP activation of Fyn kinase and Cas. These findings identify a mechanism for ADNP regulation of glial-neuronal interactions in developing cerebellum and a pathogenesis of ethanol neurotoxicity.
Collapse
|
17
|
Thomas JD, Sather TM, Whinery LA. Voluntary exercise influences behavioral development in rats exposed to alcohol during the neonatal brain growth spurt. Behav Neurosci 2008; 122:1264-73. [PMID: 19045946 PMCID: PMC3164868 DOI: 10.1037/a0013271] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Children exposed to alcohol prenatally may suffer from severe brain damage, expressed as a variety of behavioral problems, including hyperactivity and learning deficits. There is a critical need to identify effective treatments for fetal alcohol effects. Physical exercise enhances cognitive ability and increases neurogenesis in the hippocampus, a brain area important for learning and memory. Thus, the present study examined whether physical exercise might reduce the severity of alcohol-induced behavioral alterations. Sprague-Dawley rats were intubated with 5.25 g/kg/day ethanol during the third trimester equivalent (postnatal days [PDs] 4-9). Intubated sham control and nontreated controls were included. From PD 21 to PD 51, half of the subjects were given access to running wheels. On PD 52, subjects were tested on the Morris water maze, and on PD 60, open field activity levels were measured. Morris maze performance was significantly impaired among ethanol-exposed subjects; exercise significantly improved performance of all groups. Similarly, ethanol-exposed subjects were overactive in the open field, an effect attenuated with exercise. In sum, these data suggest that exercise may increase neuronal plasticity not only in controls, but also in subjects exposed to alcohol during development.
Collapse
Affiliation(s)
- Jennifer D Thomas
- Department of Psychology, Center for Behavioral Teratology, San Diego State University, CA, USA.
| | | | | |
Collapse
|
18
|
Ryan SH, Williams JK, Thomas JD. Choline supplementation attenuates learning deficits associated with neonatal alcohol exposure in the rat: effects of varying the timing of choline administration. Brain Res 2008; 1237:91-100. [PMID: 18786517 PMCID: PMC2646103 DOI: 10.1016/j.brainres.2008.08.048] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 08/14/2008] [Accepted: 08/15/2008] [Indexed: 10/21/2022]
Abstract
Despite the harmful effects of fetal alcohol exposure, some pregnant women continue to drink alcohol. Thus, it is imperative to pursue safe, effective treatments for children with fetal alcohol spectrum disorders. Using an animal model, our laboratory has demonstrated that choline, an essential nutrient, effectively reduces the severity of some fetal alcohol effects, even when administered after the ethanol insult is complete. The present study investigated whether there is a critical developmental period when choline is most effective in attenuating ethanol's teratogenic effects. Sprague-Dawley rats were exposed to 5.25 g/kg/day ethanol during the third trimester equivalent brain growth spurt (postnatal days (PD) 4-9) via intubation. A non-intubation control group and a sham intubation control group were included. Following ethanol exposure, pups received subcutaneous injections of saline vehicle or choline chloride (100 mg/kg/day) from PD 11-20, PD 21-30, or PD 11-30. Beginning on PD 45, subjects were tested on a Morris water maze spatial learning task. Performance of both the ethanol-exposed group that did not receive choline and the ethanol-exposed group treated with choline from PD 21-30 was significantly impaired compared to controls during acquisition of the Morris water maze task. Performance of ethanol-exposed groups treated with choline from PD 11-20 or PD 11-30 was intermediate, not differing significantly from any other groups. However, during the probe trial, ethanol exposure produced significant deficits in spatial memory which were mitigated by all choline treatments, regardless of the timing of administration. These findings suggest that choline's therapeutic window may be very large, or spans across the two developmental periods examined in this study. Importantly, these findings indicate that choline supplementation may effectively reduce some alcohol-related learning impairments, even when administered in later childhood.
Collapse
Affiliation(s)
- S Hunter Ryan
- Department of Psychology, Center for Behavioral Teratology, San Diego State University, San Diego, CA 92120, USA
| | | | | |
Collapse
|
19
|
Abstract
Ethanol produces a wide variety of behavioral and physiological effects in the body, but exactly how it acts to produce these effects is still poorly understood. Although ethanol was long believed to act nonspecifically through the disordering of lipids in cell membranes, proteins are at the core of most current theories of its mechanisms of action. Although ethanol affects various biochemical processes such as neurotransmitter release, enzyme function, and ion channel kinetics, we are only beginning to understand the specific molecular sites to which ethanol molecules bind to produce these myriad effects. For most effects of ethanol characterized thus far, it is unknown whether the protein whose function is being studied actually binds ethanol, or if alcohol is instead binding to another protein that then indirectly affects the functioning of the protein being studied. In this Review, we describe criteria that should be considered when identifying alcohol binding sites and highlight a number of proteins for which there exists considerable molecular-level evidence for distinct ethanol binding sites.
Collapse
Affiliation(s)
- R Adron Harris
- Section of Neurobiology and Waggoner Center for Alcohol and Addiction Research, Institutes for Neuroscience and Cell & Molecular Biology, University of Texas, Austin, TX 78712, USA.
| | | | | |
Collapse
|
20
|
Wiertz RF, Rutten WC, Marani E. Electric impedance sensing during the inhibition of cell-cell adhesion. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2008; 2008:2012-2015. [PMID: 19163088 DOI: 10.1109/iembs.2008.4649585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Electric cell impedance sensing (ECIS) was used to monitor the change of in vitro neuron-neuron adhesion in response to the blocking of N-Cam, N-Cadherin and L1. ECIS is a method in which cell morphology and cell mobility can be indirectly measured by changes in intercellular resistance. Antibodies and soluble extracellular domains of the cell adhesion molecules N-Cam, N-Cadherin and L1 were used as blockers of these adhesion molecules on the cell surface. In a 96 hour aggregation assay on a low adhesive substrate, the effect of mentioned blockers on the aggregation was investigated. The N-Cadherin antibody showed effective in aggregation inhibition at concentrations of 3 and 10 micrograms/ml. Up to 96 hours no aggregation occurred. A similar effect was achieved by the N-Cadherin protein, although less distinct. Blocking of N-CAM and L1 revealed no inhibition of aggregation. Results from impedance measurements correspond to those of the aggregation assays. The neuron-neuron adhesion in monolayers was inhibited by blocking of cell adhesion molecules and monitored by ECIS. Impedances of neuron covered electrodes were significantly lower in the presence of N-Cadherin antibody and protein at concentrations of 1, 3 and 10 micrograms/ml, indicating a less profound binding between adjacent neuron.The results from both the aggregation assays and the impedance measurements demonstrate the applicability of CAM blocking for the regulation of culture topography.
Collapse
Affiliation(s)
- R F Wiertz
- University of Twente, Enschede, 7500 AE The Netherlands.
| | | | | |
Collapse
|
21
|
Abstract
Prenatal ethanol exposure causes fetal alcohol spectrum disorders (FASD) in part by disrupting the neural cell adhesion molecule L1. L1 gene mutations cause neuropathological abnormalities similar to those of FASD. Ethanol and 1-butanol inhibit L1-mediated cell-cell adhesion (L1 adhesion), whereas 1-octanol antagonizes this action. To test the hypothesis that there are alcohol binding sites on L1, we used 3-azibutanol and 3-azioctanol, the photoactivatable analogs of 1-butanol and 1-octanol, to photolabel the purified Ig1-4 domain of human L1 (hL1 Ig1-4). 3-Azibutanol (11 mM), like ethanol, inhibited L1 adhesion in NIH/3T3 cells stably transfected with hL1, whereas subanesthetic concentrations of 3-azioctanol (14 microM) antagonized ethanol inhibition of L1 adhesion. 3-Azibutanol (100-1,000 microM) and 3-azioctanol (10-100 microM) photoincorporated into Tyr-418 on Ig4 and into two adjacent regions in the N terminus, Glu-33 and Glu-24 to Glu-27. A homology model of hL1 Ig1-4 (residues 33-422), based on the structure of the Ig1-4 domains of axonin-1, suggests that Glu-33 and Tyr-418 hydrogen-bond at the interface of Ig1 and Ig4 to stabilize a horseshoe conformation of L1 that favors homophilic binding. Furthermore, this alcohol binding pocket lies within 7 A of Leu-120 and Gly-121, residues in which missense mutations cause neurological disorders similar to FASD. These data suggest that ethanol or selected mutations produce neuropathological abnormalities by disrupting the domain interface between Ig1 and Ig4. Characterization of alcohol agonist and antagonist binding sites on L1 will aid in understanding the molecular basis for FASD and might accelerate the development of ethanol antagonists.
Collapse
|
22
|
Parnell SE, Chen SY, Charness ME, Hodge CW, Dehart DB, Sulik KK. Concurrent dietary administration of D-SAL and ethanol diminishes ethanol's teratogenesis. Alcohol Clin Exp Res 2007; 31:2059-64. [PMID: 17949468 DOI: 10.1111/j.1530-0277.2007.00524.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND SAL (SALLRSIPA) is a peptide fragment of activity-dependent neurotrophic factor. Both L- and D-SAL diminish ethanol's pathogenesis, however, the D-peptide is protease resistant, and can therefore be effectively administered in a diet. The present study tested the hypothesis that D-SAL provided in a liquid diet containing ethanol will prevent ethanol-induced teratogenicity in mice. METHODS Following an ethanol acclimation period, female C57Bl/6J mice were withdrawn from the ethanol, bred, and then returned during gestational days (GD) 7 and 8 to a control liquid diet or one containing 4.8% ethanol alone or in combination with 5.6 microg/ml D-SAL. At these doses, the mice received approximately 75 microg of D-SAL on each day and achieved peak blood-alcohol concentrations on GD 8 that ranged from 148-162 mg/dl. On GD 14, the fetuses were examined for the presence of ocular abnormalities including microphthalmia and irregularly shaped pupils, teratogenic effects known to result from this ethanol exposure paradigm. RESULTS Dietary D-SAL reduced the incidence of ocular defects in ethanol-exposed fetuses from 29 to 10% in the right eyes and from 21 to 7.5% in the left eyes; levels similar to those observed in pair-fed controls. In addition to decreasing their incidence, D-SAL also reduced the severity of the ocular defects. CONCLUSIONS These results demonstrate that oral D-SAL can prevent ethanol-induced ocular defects. Because ocular defects are commonly associated with CNS damage, oral D-SAL may also prove valuable in preventing ethanol-induced brain defects.
Collapse
Affiliation(s)
- Scott E Parnell
- Bowles Center for Alcohol Studies, Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Sant'Anna LB, Tosello DO. Fetal alcohol syndrome and developing craniofacial and dental structures--a review. Orthod Craniofac Res 2007; 9:172-85. [PMID: 17101024 DOI: 10.1111/j.1601-6343.2006.00377.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Fetal alcohol syndrome (FAS) is a collection of signs and symptoms seen in children exposed to alcohol in the prenatal period. It is characterized mainly by a distinct pattern of craniofacial malformations, physical and mental retardation. However, with the increased incidence of FAS, there is a great variation in the clinical features of FAS. DESIGN Narrative review. RESULTS This review describes data from clinical and experimental studies, and in vitro models. Experimental studies have shown that alcohol has a direct toxic effect on the ectodermal and mesodermal cells of the developing embryo, particularly in the cells destined to give rise to dentofacial structures (i.e. cranial neural crest cells). Other effects, such as, abnormal pattern of cranial and mandibular growth and altered odontogenesis are described in detail. The exact mechanism by which alcohol induces its teratogenic effects remains still unknown. The possible mechanisms are outlined here, with an emphasis on the developing face and tooth. Possible future research directions and treatment strategies are also discussed. CONCLUSION Early identification of children affected by prenatal alcohol exposure leads to interventions, services, and improved outcomes. FAS can be prevented with the elimination of alcohol consumption during pregnancy. We need to provide education, target high-risk groups, and make this issue a high priority in terms of public health.
Collapse
Affiliation(s)
- L B Sant'Anna
- Faculty of Education, Vale do Paraíba University, São José dos Campos, SP, Brazil.
| | | |
Collapse
|
24
|
Gozes I, Morimoto BH, Tiong J, Fox A, Sutherland K, Dangoor D, Holser-Cochav M, Vered K, Newton P, Aisen PS, Matsuoka Y, van Dyck CH, Thal L. NAP: research and development of a peptide derived from activity-dependent neuroprotective protein (ADNP). CNS DRUG REVIEWS 2006; 11:353-68. [PMID: 16614735 PMCID: PMC6741706 DOI: 10.1111/j.1527-3458.2005.tb00053.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Activity-dependent neuroprotective protein (ADNP) is essential for brain formation. Peptide activity scanning identified NAP (NAPVSIPQ) as a small active fragment of ADNP that provides neuroprotection at very low concentrations. In cell culture, NAP has demonstrated protection against toxicity associated with the beta-amyloid peptide, N-methyl-D-aspartate, electrical blockade, the envelope protein of the AIDS virus, dopamine, H2O2, nutrient starvation and zinc overload. NAP has also provided neuroprotection in animal models of apolipoprotein E deficiency, cholinergic toxicity, closed head injury, stroke, middle aged anxiety and cognitive dysfunction. NAP binds to tubulin and facilitates microtubule assembly leading to enhanced cellular survival that is associated with fundamental cytoskeletal elements. A liquid-chromatography, mass spectrometry assay demonstrated that NAP reaches the brain after either intravenous or intranasal administration. In a battery of toxicological tests including repeated dose toxicity in rats and dogs, cardiopulmonary tests in dogs, and functional behavioral assays in rats, no adverse side effects were observed with NAP concentrations that were approximately 500-fold higher than the biologically active dose. A Phase Ia clinical trial in the US assessed the tolerability and pharmacokinetics of intranasal administration of NAP in sequential ascending doses. The results supported the safety and tolerability of a single dose of NAP administered at up to 15 mg intranasally. Furthermore, dosing was recently completed for a second Phase I clinical trial in healthy adults and elderly volunteers with an intravenous formulation of NAP. NAP is poised for further clinical development targeting several indications, including Alzheimer's disease.
Collapse
Affiliation(s)
- Illana Gozes
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Toso L, Endres M, Vink J, Abebe DT, Brenneman DE, Spong CY. Learning enhancement with neuropeptides. Am J Obstet Gynecol 2006; 194:1153-8; discussion 1158-9. [PMID: 16580319 DOI: 10.1016/j.ajog.2005.12.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2005] [Accepted: 12/15/2005] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Previous work has demonstrated that two synthetic peptides can prevent prenatal alcohol-induced damage as assessed by prevention of learning abnormalities in adult offspring as well as improve outcome from traumatic brain damage. The current studies were undertaken to evaluate whether these peptides could enhance performance in a learning and memory paradigm when administered either prenatally or to aged mice. STUDY DESIGN For prenatal treatment, C57Bl6/J mice were treated on gestational day 8 with 1 oral administration of D-NAP or D-SAL alone or D-NAP+D-SAL or a double dose of D-SAL. Control groups were same-regimen treated with vehicle alone. Learning was assessed in adult male offspring (35-50 days) by using the Morris water maze. To evaluate aged animals, 12-month-old mice were treated with D-NAP and D-SAL or vehicle alone daily and tested on the Morris water maze. RESULTS Offspring exposed prenatally to D-NAP+D-SAL learned significantly faster than controls, with an earlier onset of learning and an overall decreased latency to find the hidden platform (P < .05). Animals exposed prenatally to either D-NAP or D-SAL alone learned similar to control, with a trend toward faster latencies. Aged animals who received D-NAP+D-SAL learned significantly faster than age-matched controls, with an earlier onset of learning (P < .05). CONCLUSION Combined D-NAP+D-SAL enhanced learning in healthy young mice and aged mice. These findings suggest potential therapeutic interventions not only during a critical developmental period, but also in aged animals.
Collapse
Affiliation(s)
- Laura Toso
- Unit on Perinatal and Developmental Neurobiology, National Institute of Child Health and Development, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|
26
|
Sari Y, Gozes I. Brain deficits associated with fetal alcohol exposure may be protected, in part, by peptides derived from activity-dependent neurotrophic factor and activity-dependent neuroprotective protein. ACTA ACUST UNITED AC 2006; 52:107-18. [PMID: 16488478 DOI: 10.1016/j.brainresrev.2006.01.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2005] [Revised: 12/20/2005] [Accepted: 01/13/2006] [Indexed: 11/26/2022]
Abstract
This review discusses the effects of prenatal alcohol exposure on the developing brain and the potential use of derived peptides from activity-dependent neurotrophic factor (ADNF) and activity-dependent neuroprotective protein (ADNP) in neuroprotection against the insults of alcohol. Alcohol is known to impede the growth of the central nervous system and to induce neurodegeneration through cellular apoptosis. Sari et al. have shown that prenatal alcohol exposure reduced the fetal brain weight, the size of the brain regions and the number of serotonin (5-HT) neurons. Prenatal alcohol exposure compromises neural tube midline development. Sari et al. further suggested that the timing of alcohol exposure during pregnancy is critical to the induction of deficits in 5-HT neurons, as well as other types of neurons and consequently results in deficits in neural tube development. ADNF and ADNP are glial-derived proteins discovered to be induced by vasoactive intestinal peptide (VIP). These proteins are expressed during embryonic development. Functional assays and genetic manipulations have identified these proteins as highly important for neural tube closure and brain formation/development. The peptide derivatives of ADNF, ADNF-14 (VLGGGSALLRSIPA), ADNF-9 (or SALLRSIPA = SAL) and of ADNP, NAPVSIPQ = NAP have shown neuroprotective effects and have been proven to prevent brain damage associated with prenatal alcohol exposure in animals. Here, we discuss the many aspects of alcohol-associated growth restriction in the developing brain and the potential inhibition of this severe phenotype through the use of neuroprotective peptides.
Collapse
Affiliation(s)
- Youssef Sari
- Indiana University School of Medicine, Department of Anatomy and Cell Biology, Neuroscience Programs, 635 Barnhill Drive, MS5035, Indianapolis, IN 46202, USA.
| | | |
Collapse
|
27
|
Zhang TA, Hendricson AW, Wilkemeyer MF, Lippmann MJ, Charness ME, Morrisett RA. Synergistic effects of the peptide fragment D-NAPVSIPQ on ethanol inhibition of synaptic plasticity and NMDA receptors in rat hippocampus. Neuroscience 2005; 134:583-93. [PMID: 15963648 DOI: 10.1016/j.neuroscience.2005.04.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2004] [Revised: 04/14/2005] [Accepted: 04/16/2005] [Indexed: 11/22/2022]
Abstract
The L1 cell adhesion molecule has been implicated in ethanol teratogenesis as well as NMDAR-dependent long-term potentiation (LTP) of synaptic transmission, a process thought to be critical for neural development. Ethanol inhibits LTP at least in part by interacting with NMDA receptors. Ethanol also inhibits L1-mediated cell adhesion in a manner that is prevented by an octapeptide, D-NAPVSIPQ (D-NAP), as well as long chain alcohols such as 1-octanol. Here we analyzed the effects of D-NAP and 1-octanol on ethanol modulation of LTP induced by theta burst stimulation in two subfields of the rat hippocampus, the dentate gyrus and area CA1. When theta burst stimulation was delivered in ethanol (50 mM), LTP was inhibited by about 50%. Surprisingly, when D-NAP (10(-7) M) and ethanol were co-applied or applied sequentially, LTP was completely absent. The effects of D-NAP were persistent, since delivery of a second theta burst stimulation following washout of D-NAP and ethanol elicited minimal plasticity. Application of D-NAP alone had no effect on LTP induction or expression. The synergistic effect of D-NAP on ethanol inhibition of LTP was concentration-dependent since D-NAP (10(-10) M) had an intermediate effect, while D-NAP (10(-13) M) had no effect on ethanol suppression of LTP. These observations were also replicated with a different ethanol antagonist, 1-octanol, in area CA1. To address the mechanisms underlying this long-lasting suppression of LTP, the sensitivity of pharmacologically isolated NMDAR extracellular field potentials to combinations of D-NAP and ethanol was determined. D-NAP (10(-7)M) alone had no effect on NMDA extracellular field potentials; however, the peptide significantly increased the inhibitory action of ethanol on NMDA extracellular field potential. The findings suggest that D-NAP and 1-octanol selectively interact with NMDA receptors in an ethanol-dependent manner, further implicating the L1 cell adhesion molecule in alcohol-related brain disorders.
Collapse
Affiliation(s)
- T A Zhang
- The College of Pharmacy and The Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-1074, USA
| | | | | | | | | | | |
Collapse
|
28
|
Chen SY, Charness ME, Wilkemeyer MF, Sulik KK. Peptide-mediated protection from ethanol-induced neural tube defects. Dev Neurosci 2005; 27:13-9. [PMID: 15886480 DOI: 10.1159/000084528] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2004] [Accepted: 10/08/2004] [Indexed: 11/19/2022] Open
Abstract
Ethanol inhibition of L1-mediated cell adhesion may contribute to the spectrum of neurological, behavioral and morphological abnormalities associated with prenatal ethanol exposure. We showed previously that the neuroprotective peptides NAPVSIPQ (NAP) and SALLRSIPA (SAL) antagonize ethanol inhibition of L1 adhesion and prevent ethanol-induced growth retardation in mouse whole embryo culture. Here we ask whether NAP and SAL also prevent ethanol-induced major malformations of the nervous system. Gestational day 8.0 (3-5 somites) C57BL/6J mouse embryos were grown for 6 h in control medium, 100 mM ethanol and 10(-10) M peptides and then maintained for an additional 20 h in control medium. At the end of the culture period, only embryos having 18-19 somite pairs were examined and compared for the degree of neural tube closure. Ethanol exposure resulted in neural tube defects (NTDs) consistent with total dysraphia and anencephaly. Co-incubation with ethanol and L-NAP (all L-amino acids), D-NAP (all D-amino acids) or SAL significantly increased the percentage of embryos that had begun to close their neural folds at the level of the forebrain/midbrain junction or that had progressed beyond this stage of closure. P7A-NAP (NAPVSIAQ), which lacks neuroprotective activity, but retains activity as an antagonist of ethanol inhibition of L1 adhesion, was effective in preventing ethanol-induced NTDs. In contrast, I6A-NAP (NAPVSAPQ), which shows reduced efficacy as an ethanol antagonist but retains its neuroprotective efficacy, did not significantly diminish the induction of NTDs by ethanol. These findings demonstrate the ability of NAP and SAL to prevent ethanol-induced NTDs and support the hypothesis that ethanol teratogenesis is caused in part by ethanol inhibition of L1-mediated cell adhesion.
Collapse
Affiliation(s)
- Shao-Yu Chen
- Bowles Center for Alcohol Studies and Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, NC 27599-7178, USA.
| | | | | | | |
Collapse
|
29
|
|
30
|
Qin L, Block ML, Liu Y, Bienstock RJ, Pei Z, Zhang W, Wu X, Wilson B, Burka T, Hong JS. Microglial NADPH oxidase is a novel target for femtomolar neuroprotection against oxidative stress. FASEB J 2005; 19:550-7. [PMID: 15791005 DOI: 10.1096/fj.04-2857com] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Inflammation has been increasingly recognized to contribute to the pathogenesis of Parkinson's disease. Several compounds are neuroprotective at femtomolar concentrations through the inhibition of inflammation. However, the mechanisms mediating femtomolar-acting compounds are poorly understood. Here we show that both gly-gly-phe (GGF), a tri-peptide contained in the dynorphin opioid peptide, and naloxone are neuroprotective at femtomolar concentrations against LPS-induced dopaminergic neurotoxicity through the reduction of microglial activation. Mechanistic studies demonstrated the critical role of NADPH oxidase in the GGF and naloxone inhibition of microglial activation and associated DA neurotoxicity. Pharmacophore analysis of the neuroprotective dynorphin peptides and naloxone revealed common chemical properties (hydrogen bond acceptor, hydrogen bond donor, positive ionizable, hydrophobic) of these femtomolar-acting compounds. These results support a common high-affinity site of action for several femtomolar-acting compounds, where NADPH oxidase is the critical mechanism governing neuroprotection, suggesting a novel avenue of anti-inflammatory and neuroprotective therapy.
Collapse
Affiliation(s)
- Liya Qin
- Neuropharmacology Section, Laboratory of Pharmacology and Chemistry, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Wilkemeyer MF, Chen SY, Menkari CE, Sulik KK, Charness ME. Ethanol antagonist peptides: structural specificity without stereospecificity. J Pharmacol Exp Ther 2004; 309:1183-9. [PMID: 14762101 DOI: 10.1124/jpet.103.063818] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Increasing evidence suggests that ethanol damages the developing nervous system partly by disrupting the L1 cell adhesion molecule. Ethanol inhibits L1-mediated cell adhesion, and compounds that antagonize this action also prevent ethanol-induced embryotoxicity. Two such compounds are the small peptides NAPVSIPQ (NAP) and SALLRSIPA (SAL). We showed previously that NAP and SAL antagonize ethanol inhibition of L1 adhesion at femtomolar to picomolar concentrations. Here we demonstrate that, despite this extraordinary potency, both NAP and SAL lack stereospecificity. d-NAP, a peptide composed entirely of d-amino acids, was an effective ethanol antagonist in NIH/3T3 cells transfected with human L1 and in the NG108-15 neural cell line. Interestingly, Ala-substituted derivatives of d-NAP demonstrate the same structure-activity relation as the corresponding derivatives of l-NAP. The Ser-Ile-Pro motif was important for the ethanol antagonist activity of d-NAP, l-NAP, and l-SAL, with Ile being the most critical element in all three. Like l-NAP, d-NAP effectively reduced ethanol-induced growth retardation in mouse whole embryo culture. The potential resistance of d-peptides to proteases makes d-NAP a potentially attractive agent for the prevention of fetal alcohol syndrome.
Collapse
Affiliation(s)
- Michael F Wilkemeyer
- Neurology Service for Veterans Affairs Boston Healthcare System, 1400 VFW Parkway, West Roxbury, MA 02132, USA
| | | | | | | | | |
Collapse
|
32
|
Brenneman DE, Spong CY, Hauser JM, Abebe D, Pinhasov A, Golian T, Gozes I. Protective peptides that are orally active and mechanistically nonchiral. J Pharmacol Exp Ther 2004; 309:1190-7. [PMID: 15007105 DOI: 10.1124/jpet.103.063891] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previous reports identified two peptides that mimic the action of neuroprotective proteins derived from astrocytes. These peptides, NAPVSIPQ and SALLRSIPA, prevent neuronal cell death produced by electrical blockade, N-methyl-d-aspartate, and beta-amyloid peptide (25-35). In the present study, all d-amino acid peptides of NAPVSIPQ and SALLRSIPA were synthesized and compared respectively to the corresponding all l-amino acid peptides. In rat cerebral cortical test cultures cotreated with 1 microM tetrodotoxin, the d-amino acid peptides produced similar potency and efficacy for neuroprotection as that observed for their respective l-amino acid peptides. Since all these peptides tested individually exhibited attenuation of efficacy at concentrations of >10 pM, combinations of these peptides were tested for possible synergies. Equimolar d-NAPVSIPQ and d-SALLRSIPA combination treatment produced potent neuroprotection (EC(50), 0.03 fM) that did not attenuate with increasing concentrations. Similarly, the combination of l-NAPVSIPQ and d-SALLRSIPA also had high potency (EC(50), 0.07 fM) without attenuation of efficacy. Combined administration of peptides was tested in a model of fetal alcohol syndrome and in a model of learning impairment: apolipoprotein E knockout mice. Intraperitoneal administration of d-NAPVSIPQ plus d-SALLRSIPA to pregnant mice (embryonic day 8) attenuated fetal demise after treatment with an acute high dose of alcohol. Furthermore, oral administration of d-NAPVSIPQ plus d-SALLRSIPA significantly increased fetal survival after maternal alcohol treatment. Apolipoprotein E knockout mice injected with d-NAPVSIPQ plus d-SALLRSIPA showed improved performance in the Morris water maze. These studies suggest therapeutic potential for the combined administration of neuroprotective peptides that can act through a mechanism independent of chiral recognition.
Collapse
Affiliation(s)
- Douglas E Brenneman
- Section of Developmental and Molecular Pharmacology, Laboratory of Developmental Neurobiology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Wilkemeyer MF, Chen SY, Menkari CE, Brenneman DE, Sulik KK, Charness ME. Differential effects of ethanol antagonism and neuroprotection in peptide fragment NAPVSIPQ prevention of ethanol-induced developmental toxicity. Proc Natl Acad Sci U S A 2003; 100:8543-8. [PMID: 12808140 PMCID: PMC166265 DOI: 10.1073/pnas.1331636100] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
NAPVSIPQ (NAP), an active fragment of the glial-derived activity-dependent neuroprotective protein, is protective at femtomolar concentrations against a wide array of neural insults and prevents ethanol-induced fetal wastage and growth retardation in mice. NAP also antagonizes ethanol inhibition of L1-mediated cell adhesion (ethanol antagonism). We performed an Ala scanning substitution of NAP to determine the role of ethanol antagonism and neuroprotection in NAP prevention of ethanol embryotoxicity. The Ser-Ile-Pro region of NAP was crucial for both ethanol antagonism and protection of cortical neurons from tetrodotoxin toxicity (neuroprotection). Ala replacement of either Ser-5 or Pro-7 (P7A-NAP) abolished NAP neuroprotection but minimally changed the efficacy of NAP ethanol antagonism. In contrast, Ala replacement of Ile-6 (I6A-NAP) caused a decrease in potency (>2 logarithmic orders) with only a small reduction (<10%) in the efficacy of NAP neuroprotection but markedly reduced the efficacy (50%) and the potency (5 logarithmic orders) of NAP ethanol antagonism. Ethanol significantly reduced the number of paired somites in mouse whole-embryo culture; this effect was prevented significantly by 100 pM NAP or by 100 pM P7A-NAP, but not by 100 pM I6A-NAP. The structure-activity relation for NAP prevention of ethanol embryotoxicity was similar to that for NAP ethanol antagonism and different from that for NAP neuroprotection. These findings support the hypothesis that NAP antagonism of ethanol inhibition of L1 adhesion plays a central role in NAP prevention of ethanol embryotoxicity and highlight the potential importance of ethanol effects on L1 in the pathophysiology of fetal alcohol syndrome.
Collapse
Affiliation(s)
- Michael F. Wilkemeyer
- Neurology Service, Veterans Affairs Boston
Healthcare System, West Roxbury, MA 02132;
Department of Neurology, Harvard Medical
School, Boston, MA 02115; Department of
Neurology, Brigham and Women's Hospital, Boston, MA 02115;
Bowles Center for Alcohol Studies, University of
North Carolina, Chapel Hill, NC 27599; Department
of Cell and Developmental Biology, University of North Carolina School of
Medicine, Chapel Hill, NC 27599; and Section on
Developmental and Molecular Pharmacology, National Institute of Child Health
and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Shao-yu Chen
- Neurology Service, Veterans Affairs Boston
Healthcare System, West Roxbury, MA 02132;
Department of Neurology, Harvard Medical
School, Boston, MA 02115; Department of
Neurology, Brigham and Women's Hospital, Boston, MA 02115;
Bowles Center for Alcohol Studies, University of
North Carolina, Chapel Hill, NC 27599; Department
of Cell and Developmental Biology, University of North Carolina School of
Medicine, Chapel Hill, NC 27599; and Section on
Developmental and Molecular Pharmacology, National Institute of Child Health
and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Carrie E. Menkari
- Neurology Service, Veterans Affairs Boston
Healthcare System, West Roxbury, MA 02132;
Department of Neurology, Harvard Medical
School, Boston, MA 02115; Department of
Neurology, Brigham and Women's Hospital, Boston, MA 02115;
Bowles Center for Alcohol Studies, University of
North Carolina, Chapel Hill, NC 27599; Department
of Cell and Developmental Biology, University of North Carolina School of
Medicine, Chapel Hill, NC 27599; and Section on
Developmental and Molecular Pharmacology, National Institute of Child Health
and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Douglas E. Brenneman
- Neurology Service, Veterans Affairs Boston
Healthcare System, West Roxbury, MA 02132;
Department of Neurology, Harvard Medical
School, Boston, MA 02115; Department of
Neurology, Brigham and Women's Hospital, Boston, MA 02115;
Bowles Center for Alcohol Studies, University of
North Carolina, Chapel Hill, NC 27599; Department
of Cell and Developmental Biology, University of North Carolina School of
Medicine, Chapel Hill, NC 27599; and Section on
Developmental and Molecular Pharmacology, National Institute of Child Health
and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Kathleen K. Sulik
- Neurology Service, Veterans Affairs Boston
Healthcare System, West Roxbury, MA 02132;
Department of Neurology, Harvard Medical
School, Boston, MA 02115; Department of
Neurology, Brigham and Women's Hospital, Boston, MA 02115;
Bowles Center for Alcohol Studies, University of
North Carolina, Chapel Hill, NC 27599; Department
of Cell and Developmental Biology, University of North Carolina School of
Medicine, Chapel Hill, NC 27599; and Section on
Developmental and Molecular Pharmacology, National Institute of Child Health
and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Michael E. Charness
- Neurology Service, Veterans Affairs Boston
Healthcare System, West Roxbury, MA 02132;
Department of Neurology, Harvard Medical
School, Boston, MA 02115; Department of
Neurology, Brigham and Women's Hospital, Boston, MA 02115;
Bowles Center for Alcohol Studies, University of
North Carolina, Chapel Hill, NC 27599; Department
of Cell and Developmental Biology, University of North Carolina School of
Medicine, Chapel Hill, NC 27599; and Section on
Developmental and Molecular Pharmacology, National Institute of Child Health
and Human Development, National Institutes of Health, Bethesda, MD 20892
- To whom correspondence should be addressed at: Department of Neurology (127),
Harvard Medical School, Veterans Affairs Boston Healthcare System, 1400 VFW
Parkway, West Roxbury, MA 02132. E-mail:
| |
Collapse
|
34
|
Greenberg DA. Linking acquired neurodevelopmental disorders to defects in cell adhesion. Proc Natl Acad Sci U S A 2003; 100:8043-4. [PMID: 12835424 PMCID: PMC166177 DOI: 10.1073/pnas.1533226100] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- David A Greenberg
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945, USA.
| |
Collapse
|
35
|
Riley EP, Guerri C, Calhoun F, Charness ME, Foroud TM, Li TK, Mattson SN, May PA, Warren KR. Prenatal Alcohol Exposure: Advancing Knowledge Through International Collaborations. Alcohol Clin Exp Res 2003. [DOI: 10.1111/j.1530-0277.2003.tb02731.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
36
|
Prenatal Alcohol Exposure: Advancing Knowledge Through International Collaborations. Alcohol Clin Exp Res 2003. [DOI: 10.1097/00000374-200301000-00020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|