1
|
HOXD9 promotes the growth, invasion and metastasis of gastric cancer cells by transcriptional activation of RUFY3. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:412. [PMID: 31547840 PMCID: PMC6755711 DOI: 10.1186/s13046-019-1399-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 08/28/2019] [Indexed: 12/12/2022]
Abstract
Background The transcription factor HOXD9 is one of the members of the HOX family, which plays an important role in neoplastic processes. However, the role of HOXD9 in the growth and metastasis of gastric cancer (GC) remains to be elucidated. Methods In vitro functional role of HOXD9 and RURY3 in GC cells was determined using the TMA-based immunohistochemistry, western blot, EdU incorporation, gelatin zymography, luciferase, chromatin Immunoprecipitation (ChIP) and cell invasion assays. In vivo tumor growth and metastasis were conducted in nude mice. Results HOXD9 is overexpressed in GC cells and tissues. The high expression of HOXD9 was correlated with poor survival in GC patients. Functionally, HOXD9 expression significantly promoted the proliferation, invasion and migration of GC cells. Mechanically, HOXD9 directly associated with the RUFY3 promoter to increase the transcriptional activity of RUFY3. Inhibition of RUFY3 attenuated the proliferation, migration and invasiveness of HOXD9-overexpressing GC cells in vitro and in vivo. Moreover, both HOXD9 and RUFY3 were highly expressed in cancer cells but not in normal gastric tissues, with their expressions being positively correlated. Conclusions The evidence presented here suggests that the HOXD9-RUFY3 axis promotes the development and progression of human GC. Electronic supplementary material The online version of this article (10.1186/s13046-019-1399-1) contains supplementary material, which is available to authorized users.
Collapse
|
2
|
Miller MW. p53-Mediated Activities in NS-5 Neural Stem Cells: Effects of Ethanol. Alcohol Clin Exp Res 2019; 43:655-667. [PMID: 30748015 DOI: 10.1111/acer.13976] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 02/05/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Transforming growth factor (TGF) β1 and ethanol (EtOH) powerfully inhibit the proliferation, DNA repair, and survival of neural stem cells (NSCs). The present study tests the hypothesis that the EtOH-induced DNA damage response is mediated through p53 pathways and influenced by growth factor signals. METHODS Cultures of nonimmortalized NSCs, NS-5 cells, were transfected with p53 siRNA, exposed to either the mitogenic fibroblast growth factor (FGF) 2 or antimitogenic TGFβ1, and to EtOH. Stage-specific cellular and genomic responses were examined. RESULTS p53 status, EtOH exposure, and growth factor significantly affected the expression of transcripts related to the DNA damage response (including those coding for excision repair proteins), mitotic promoters, and regulators of cell death via the tumor necrosis factor pathway. There were significant compensatory increases in p53 family members, p63 and p73, notably in regard to the regulation of cell cycle restriction and apoptosis. Treatment with p53 siRNA potentiated EtOH- and TGFβ1-induced changes in the numbers of proliferating NSCs and increased the proportion of NSCs expressing the apoptotic marker annexin V. CONCLUSIONS Thus, it appears that EtOH and TGFβ1 affect proliferation, DNA repair, and survival of NSCs via p53-mediated activities.
Collapse
Affiliation(s)
- Michael W Miller
- Department of Neuroscience and Physiology, State University of New York-Upstate Medical University, Syracuse, New York.,Touro College of Osteopathic Medicine, Middletown, New York.,Research Service, Veterans Affairs Medical Center, Syracuse, New York
| |
Collapse
|
3
|
Hicks SD, Miller MW. Ethanol-induced DNA repair in neural stem cells is transforming growth factor β1-dependent. Exp Neurol 2019; 317:214-225. [PMID: 30853389 DOI: 10.1016/j.expneurol.2019.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/13/2018] [Accepted: 02/07/2019] [Indexed: 12/18/2022]
Abstract
Following neurotoxic damage, cells repair their DNA, and survive or undergo apoptosis. This study tests the hypothesis that ethanol induces a DNA damage response (DDR) in neural stem cells (NSCs) that promotes excision repair (ER) and this repair is influenced by the growth factor environment. Non-immortalized NSCs treated with fibroblast growth factor 2 or transforming growth factor (TGF) β1 were exposed to ethanol. Ethanol increased total DNA damage, reactive oxygen species, and oxidized DNA bases. TGFβ1 potentiated these toxic effects. Transcriptional analyses of cultured NSCs revealed ethanol-induced increases in transcripts related to the DDR (e.g., Hus1 and p53), base ER (e.g., Mutyh and Nthl1), and nucleotide ER (e.g., Xpc), particularly in the presence of TGFβ1. Expression and activity of ER proteins were affected by ethanol. Similar changes occurred in proliferating cells of ethanol-treated mouse fetuses. Ethanol-induced DNA repair in NSCs depends on the ambient growth factors. Gene products for DNA repair in stem cells are among the first biomarkers identifying fetal alcohol-induced damage.
Collapse
Affiliation(s)
- Steven D Hicks
- Department of Neuroscience and Physiology, State University of New York - Upstate Medical University, Syracuse, NY 13210, USA; Developmental Exposure Alcohol Research Center, Binghamton NY 13902, Cortland NY 13045, and Syracuse, NY 13210, USA
| | - Michael W Miller
- Department of Neuroscience and Physiology, State University of New York - Upstate Medical University, Syracuse, NY 13210, USA; Developmental Exposure Alcohol Research Center, Binghamton NY 13902, Cortland NY 13045, and Syracuse, NY 13210, USA; Department of Anatomy, Touro College of Osteopathic Medicine, Middletown, NY 10940, USA; Research Service, Veterans Affairs Medical Center, Syracuse, NY 13210, USA.
| |
Collapse
|
4
|
Foxworthy WA, Medina AE. Overexpression of Serum Response Factor in Neurons Restores Ocular Dominance Plasticity in a Model of Fetal Alcohol Spectrum Disorders. Alcohol Clin Exp Res 2015; 39:1951-6. [PMID: 26342644 DOI: 10.1111/acer.12844] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/14/2015] [Indexed: 01/06/2023]
Abstract
BACKGROUND Deficits in neuronal plasticity underlie many neurobehavioral and cognitive problems presented in fetal alcohol spectrum disorder (FASD). Our laboratory has developed a ferret model showing that early alcohol exposure leads to a persistent disruption in ocular dominance plasticity (ODP). For instance, a few days of monocular deprivation results in a robust reduction of visual cortex neurons' responsiveness to stimulation of the deprived eye in normal animals, but not in ferrets with early alcohol exposure. Previously our laboratory demonstrated that overexpression of serum response factor (SRF) exclusively in astrocytes can improve neuronal plasticity in FASD. Here, we test whether neuronal overexpression of SRF can achieve similar effects. METHODS Ferrets received 3.5 g/kg alcohol intraperitoneally (25% in saline) or saline as control every other day between postnatal day 10 to 30, which is roughly equivalent to the third trimester of human gestation. Animals were given intracortical injections of a Herpes Simplex Virus-based vector to express either green fluorescent protein or a constitutively active form of SRF in infected neurons. They were then monocularly deprived by eyelid suture for 4 to 5 days after which single-unit recordings were conducted to determine whether changes in ocular dominance had occurred. RESULTS Overexpression of a constitutively active form of SRF by neurons restored ODP in alcohol-treated animals. This effect was observed only in areas near the site of viral infection. CONCLUSIONS Overexpression of SRF in neurons can restore plasticity in the ferret model of FASD, but only in areas near the site of infection. This contrasts with SRF overexpression in astrocytes which restored plasticity throughout the visual cortex.
Collapse
Affiliation(s)
- W Alex Foxworthy
- Department of Pediatrics (WAF, AEM), University of Maryland, Baltimore, Maryland
| | - Alexandre E Medina
- Department of Pediatrics (WAF, AEM), University of Maryland, Baltimore, Maryland
| |
Collapse
|
5
|
Duncan J, Wang N, Zhang X, Johnson S, Harris S, Zheng B, Zhang Q, Rajkowska G, Miguel-Hidalgo JJ, Sittman D, Ou XM, Stockmeier CA, Wang JM. Chronic Social Stress and Ethanol Increase Expression of KLF11, a Cell Death Mediator, in Rat Brain. Neurotox Res 2015; 28:18-31. [PMID: 25739536 DOI: 10.1007/s12640-015-9524-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 01/20/2015] [Accepted: 02/19/2015] [Indexed: 02/06/2023]
Abstract
Major depressive disorder and alcoholism are significant health burdens that can affect executive functioning, cognitive ability, job responsibilities, and personal relationships. Studies in animal models related to depression or alcoholism reveal that the expression of Krüppel-like factor 11 (KLF11, also called TIEG2) is elevated in frontal cortex, which suggests that KLF11 may play a role in stress- or ethanol-induced psychiatric conditions. KLF11 is a transcriptional activator of monoamine oxidase A and B, but also serves other functions in cell cycle regulation and apoptotic cell death. In the present study, immunohistochemistry was used to quantify intensity of nuclear KLF11, combined with an unbiased stereological approach to assess nuclei in fronto-limbic, limbic, and other brain regions of rats exposed chronically to social defeat or ethanol. KLF11 immunoreactivity was increased significantly in the medial prefrontal cortex, frontal cortex, and hippocampus of both stressed rats and rats fed ethanol. However, expression of KLF11 protein was not significantly affected in the thalamus, hypothalamus, or amygdala in either treatment group compared to respective control rats. Triple-label immunofluorescence revealed that KLF11 protein was localized in nuclei of neurons and astrocytes. KLF11 was also co-localized with the immunoreactivity of cleaved caspase-3. In addition, Western blot analysis revealed a significant reduction in anti-apoptotic protein, Bcl-xL, but an increase of caspase-3 expression in the frontal cortex of ethanol-treated rats compared to ethanol-preferring controls. Thus, KLF11 protein is up-regulated following chronic exposure to stress or ethanol in a region-specific manner and may contribute to pro-apoptotic signaling in ethanol-treated rats. Further investigation into the KLF11 signaling cascade as a mechanism for neurotoxicity and cell death in depression and alcoholism may provide novel pharmacological targets to lessen brain damage and maximize neuroprotection in these disorders.
Collapse
Affiliation(s)
- Jeremy Duncan
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Niu J, Wang L, Liu S, Li C, Kong J, Shen HY, Xiao L. An efficient and economical culture approach for the enrichment of purified oligodendrocyte progenitor cells. J Neurosci Methods 2012; 209:241-9. [PMID: 22687939 DOI: 10.1016/j.jneumeth.2012.05.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 05/29/2012] [Accepted: 05/30/2012] [Indexed: 10/28/2022]
Abstract
Oligodendrocyte progenitor cell (OPC) culture has provided a powerful approach to mechanistically investigate the proliferation and differentiation of oligodendroglia. However, existing culture methods (including the traditional shake-off method) have limitations, particularly their low productivities. Therefore, we developed a simplified and highly efficient method to produce a large yield of OPCs with low expense by using specialised modified media, in which B104-conditioned medium (B104-CM) instead of growth factors was used as a mitogenic source for OPC propagation, while a modified OPC isolation-medium was applied to improve the isolation of OPCs. First, we withdrew foetal bovine serum when primary mixed glial cultures were 65-75% confluent and substituted with modified oligodendrocyte growth medium to enrich OPCs. Second, we employed a chemical-based method to isolate and purify OPCs from mixed glial cultures using a modified oligodendrocyte isolation medium. As a result, our approach produced a high yield of purified OPCs, approximately 90-fold higher than that produced via the traditional shake-off method. Importantly, the purified OPCs produced via our modified approach maintained typical capacities of proliferation and differentiation observed in oligodendrocyte lineage cells. Together, our modified method provides a highly efficient approach to OPC culture for oligodendrocyte research.
Collapse
Affiliation(s)
- Jianqin Niu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | | | | | | | | | | | | |
Collapse
|
7
|
Utreras E, Keller J, Terse A, Prochazkova M, Iadarola MJ, Kulkarni AB. Transforming growth factor-β1 regulates Cdk5 activity in primary sensory neurons. J Biol Chem 2012; 287:16917-29. [PMID: 22451679 DOI: 10.1074/jbc.m111.329979] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In addition to many important roles for Cdk5 in brain development and synaptic function, we reported previously that Cdk5 regulates inflammatory pain signaling, partly through phosphorylation of transient receptor potential vanilloid 1 (TRPV1), an important Na(+)/Ca(2+) channel expressed in primary nociceptive afferent nerves. Because TGF-β regulates inflammatory processes and its receptor is expressed in TRPV1-positive afferents, we studied the cross-talk between these two pathways in sensory neurons during experimental peripheral inflammation. We demonstrate that TGF-β1 increases transcription and protein levels of the Cdk5 co-activator p35 through ERK1/2, resulting in an increase in Cdk5 activity in rat B104 neuroblastoma cells. Additionally, TGF-β1 enhances the capsaicin-induced Ca(2+) influx in cultured primary neurons from dorsal root ganglia (DRG). Importantly, Cdk5 activity was reduced in the trigeminal ganglia and DRG of 14-day-old TGF-β1 knock-out mice, resulting in reduced Cdk5-dependent phosphorylation of TRPV1. The decreased Cdk5 activity is associated with attenuated thermal hyperalgesia in TGF-β1 receptor conditional knock-out mice, where TGF-β signaling is significantly reduced in trigeminal ganglia and DRG. Collectively, our results indicate that active cross-talk between the TGF-β and Cdk5 pathways contributes to inflammatory pain signaling.
Collapse
Affiliation(s)
- Elias Utreras
- Functional Genomics Section, Laboratory of Cell and Developmental Biology, NIDCR, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
8
|
Fukuda M, Kusama K, Sakashita H. Molecular insights into the proliferation and progression mechanisms of the oral cancer: Strategies for the effective and personalized therapy. JAPANESE DENTAL SCIENCE REVIEW 2012. [DOI: 10.1016/j.jdsr.2011.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
9
|
Vangipuram SD, Lyman WD. Ethanol affects differentiation-related pathways and suppresses Wnt signaling protein expression in human neural stem cells. Alcohol Clin Exp Res 2011; 36:788-97. [PMID: 22150777 DOI: 10.1111/j.1530-0277.2011.01682.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Prenatal exposure of the fetus to ethanol (EtOH) can be teratogenic. We previously showed that EtOH alters the cell fate of human neural stem cells (NSC). As Wnt signaling plays an important role in fetal brain development, we hypothesized that EtOH suppresses Wnt signaling protein expression in differentiating NSC and thereby contributes to fetal alcohol spectrum disorder. METHODS NSC isolated from fetal human brains were cultured in mitogenic media to induce neurospheres, which were dissociated into single-cell suspensions and used for all experiments. Equal numbers of NSC were cultured on lysine/laminin-coated plates for 96 hours in differentiating media containing 0, 20, or 100 mM EtOH. Total mRNA was isolated from samples containing 0 or 100 mM EtOH and changes in expression of 263 genes associated with neurogenesis and NSC differentiation were determined by Oligo GEArray technology. The biological impact of gene changes was estimated using a systems biology approach with pathway express software and KEGG database. Based on the pathways identified, expression of Wnt proteins (Wnt3a and Wnt5a), Wnt-receptor complex proteins (p-LRP6, LRP6, DVL2, and DVL3), Wnt antagonist Naked-2 (NKD-2), and downstream Wnt proteins (β-catenin, Tyr-p-GSK3β, Ser-p-GSK3β) were analyzed by Western blot. RESULTS Of the 263 genes examined, the expressions of 22 genes in differentiating NSC were either upwardly or downwardly affected by EtOH. These genes are associated with 5 pathways/cellular processes: axon guidance; hedgehog signaling; TGF-β signaling; cell adhesion molecules; and Wnt signaling. When compared to controls, EtOH, at both 20 and 100 mM concentrations, suppressed the expression of Wnt3a and Wnt5a, receptor complex proteins p-LRP6, LRP6 and DVL2, and cytoplasmic proteins Ser-p-GSK3β and β-catenin. Expression of NKD-2 and DVL3 remained unchanged and the expression of active Tyr-p-GSK3β increased significantly. CONCLUSIONS EtOH can significantly alter neural differentiation pathway-related gene expression and suppress Wnt signaling proteins in differentiating human NSC.
Collapse
Affiliation(s)
- Sharada D Vangipuram
- Children's Research Center of Michigan, The Carman and Ann Adams Department of Pediatrics, Wayne State University School of Medicine, Detroit, USA.
| | | |
Collapse
|
10
|
Tang N, Farah B, He M, Fox S, Malouf A, Littner Y, Bearer CF. Ethanol causes the redistribution of L1 cell adhesion molecule in lipid rafts. J Neurochem 2011; 119:859-67. [PMID: 21884525 DOI: 10.1111/j.1471-4159.2011.07467.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Fetal alcohol spectrum disorder is estimated to affect 1% of live births. The similarities between children with fetal alcohol syndrome and those with mutations in the gene encoding L1 cell adhesion molecule (L1) implicates L1 as a target of ethanol developmental neurotoxicity. Ethanol specifically inhibits the neurite outgrowth promoting function of L1 at pharmacologic concentrations. Emerging evidence shows that localized disruption of the lipid rafts reduces L1-mediated neurite outgrowth. We hypothesize that ethanol impairment of the association of L1 with lipid rafts is a mechanism underlying ethanol's inhibition of L1-mediated neurite outgrowth. In this study, we examine the effects of ethanol on the association of L1 and lipid rafts. We show that, in vitro, L1 but not N-cadherin shifts into lipid rafts following treatment with 25 mM ethanol. The ethanol concentrations causing this effect are similar to those inhibiting L1-mediated neurite outgrowth. Increasing chain length of the alcohol demonstrates the same cutoff as that previously shown for inhibition of L1-L1 binding. In addition, in cerebellar granule neurons in which lipid rafts are disrupted with methyl-beta-cyclodextrin, the rate of L1-mediated neurite outgrowth on L1-Fc is reduced to background rate and that this background rate is not ethanol sensitive. These data indicate that ethanol may inhibit L1-mediated neurite outgrowth by retarding L1 trafficking through a lipid raft compartment.
Collapse
Affiliation(s)
- Ningfeng Tang
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland 21209, USA
| | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
Eukaryotic cells comprise a set of organelles, surrounded by membranes with a unique composition, which is maintained by a complex synthesis and transport system. Cells also synthesize the proteins destined for secretion. Together, these processes are known as the secretory pathway or exocytosis. In addition, many molecules can be internalized by cells through a process called endocytosis. Chronic and acute alcohol (ethanol) exposure alters the secretion of different essential products, such as hormones, neurotransmitters and others in a variety of cells, including central nervous system cells. This effect could be due to a range of mechanisms, including alcohol-induced alterations in the different steps involved in intracellular transport, such as glycosylation and vesicular transport along cytoskeleton elements. Moreover, alcohol consumption during pregnancy disrupts developmental processes in the central nervous system. No single mechanism has proved sufficient to account for these effects, and multiple factors are likely involved. One such mechanism indicates that ethanol also perturbs protein trafficking. The purpose of this review is to summarize our understanding of how ethanol exposure alters the trafficking of proteins in different cell systems, especially in central nervous system cells (neurons and astrocytes) in adult and developing brains.
Collapse
|
12
|
Hicks SD, Miller MW. Effects of ethanol on transforming growth factor Β1-dependent and -independent mechanisms of neural stem cell apoptosis. Exp Neurol 2011; 229:372-80. [PMID: 21419121 DOI: 10.1016/j.expneurol.2011.03.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 02/11/2011] [Accepted: 03/07/2011] [Indexed: 01/24/2023]
Abstract
Stem cell vitality is critical for the growth of the developing brain. Growth factors can define the survival of neural stem cells (NSCs) and ethanol can affect growth factor-mediated activities. The present study tested two hypotheses: (a) ethanol causes the apoptotic death of NSCs and (b) this effect is influenced by the ambient growth factor. Monolayer cultures of non-immortalized NS-5 cells were exposed to fibroblast growth factor (FGF) 2 or transforming growth factor (TGF) β1 in the absence or presence of ethanol for 48 h. Ethanol killed NSCs as measured by increases in the numbers of ethidium bromide+ and annexin V+ cells and decreases in the number of calcein AM+ (viable) cells. These toxic effects were promoted by TGFβ1. A quantitative polymerase chain reaction array of apoptosis-related mRNAs revealed an ethanol-induced increase (≥2-fold change; p<0.05) in transcripts involved in Fas ligand (FasL) and tumor necrosis factor (TNF) signaling. These effects, particularly the FasL pathway, were potentiated by TGFβ1. Immunocytochemical analyses of NS-5 cells showed that transcriptional alterations translated into consistent up-regulation of protein expression. Experiments with the neocortical proliferative zones harvested from fetal mice exposed to ethanol showed that ethanol activated similar molecular systems in vivo. Thus, ethanol induces NSC death through two distinct molecular mechanisms, one is initiated by TGFβ1 (FasL) and another (through TNF) which is TGFβ1-independent.
Collapse
Affiliation(s)
- Steven D Hicks
- Department of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
| | | |
Collapse
|
13
|
Mooney SM, Miller MW. Role of neurotrophins on postnatal neurogenesis in the thalamus: prenatal exposure to ethanol. Neuroscience 2011; 179:256-66. [PMID: 21277941 DOI: 10.1016/j.neuroscience.2011.01.046] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 01/20/2011] [Accepted: 01/21/2011] [Indexed: 12/23/2022]
Abstract
A second wave of neuronal generation occurs in the ventrobasal nucleus of the rat thalamus (VB) during the first three postnatal weeks. The present study tested the hypotheses (1) that postnatal neurogenesis in the VB is neurotrophin-regulated and (2) that ethanol-induced changes in this proliferation are mediated by neurotrophins. The first studies examined the effects of neurotrophins on the numbers of cycling cells in ex vivo preparations of the VB from 3-day-old rats. The proportion of cycling (Ki-67-positive) VB cells was higher in cultured thalamic slices treated with neurotrophins than in controls. Interestingly, this increase occurred with nerve growth factor (NGF) alone or with a combination of NGF and brain-derived neurotrophic factor (BDNF), but not with BDNF alone. Based on these data, the VBs from young offspring of pregnant rats fed an ethanol-containing or an isocaloric non-alcoholic liquid diet were examined between postnatal day (P) 1 and P31. Studies used enzyme-linked immunosorbent assays and immunoblots to explore the effects of ethanol on the expression of neurotrophins, their receptors, and representative signaling proteins. Ethanol altered the expression of neurotrophins and receptors throughout the first postnatal month. Expression of NGF increased, but there was no change in the expression of BDNF. The high affinity receptors (TrkA and TrkB) were unchanged but ethanol decreased expression of the low affinity receptor, p75. One downstream signaling protein, extracellular signal-regulated kinase (ERK), decreased but Akt expression was unchanged. Thus, postnatal cell proliferation in the VB of young rat pups is neurotrophin-responsive and is affected by ethanol.
Collapse
Affiliation(s)
- S M Mooney
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY 13210, USA.
| | | |
Collapse
|
14
|
Hicks SD, Middleton FA, Miller MW. Ethanol-induced methylation of cell cycle genes in neural stem cells. J Neurochem 2010; 114:1767-80. [PMID: 20626555 DOI: 10.1111/j.1471-4159.2010.06886.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ethanol inhibits the proliferation of neural precursors by altering mitogenic and anti-mitogenic growth factor signaling and can affect global methylation activity in the fetus. We tested the hypothesis that epigenetic modification of specific cell cycle genes underlies the ethanol-induced inhibition of growth factor-regulated cell cycle progression. Monolayer cultures of neural stem cells (NSCs) were treated with fibroblast growth factor 2 or transforming growth factor (TGF) β1 in the absence or presence of ethanol. Ethanol increased the total length of the cell cycle by elongating the amount of time spent in the gap 1 (G1) and synthesis (S) phases of the cell cycle. Ethanol induced the hypermethylation of multiple cell cycle genes associated with the G1/S and gap 2/mitotic phase (G2/M) checkpoints and increased the expression and activity of DNA methyltransferases. These changes were most pronounced in the presence of TGFβ1. Epigenetic alterations paralleled the down-regulation of associated transcripts and other checkpoint-related mRNAs both in vitro (NS-5 cell culture) and in vivo (fetal mouse cortex). Ethanol-induced hypermethylation was accompanied by decreases in the proportion of NSCs expressing associated cell cycle proteins. Thus, ethanol disrupts growth factor-related cell cycle progression by inducing checkpoint restriction at the G1/S transition through a feed-forward system involving the methylation of G2/M regulators.
Collapse
Affiliation(s)
- Steven D Hicks
- Department of Neuroscience and Physiology, State University of New York - Upstate Medical University, Syracuse, NY 13210, USA
| | | | | |
Collapse
|
15
|
Lindke AL, Middleton FA, Miller MW. Regulating the availability of transforming growth factor ß1 in B104 neuroblastoma cells. Exp Neurol 2010; 225:123-32. [PMID: 20547156 DOI: 10.1016/j.expneurol.2010.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Revised: 05/27/2010] [Accepted: 06/07/2010] [Indexed: 10/19/2022]
Abstract
Transforming growth factor (TGF) beta1 is a key player in early brain development, hence, its availability (i.e., synthesis and release) affects neuronogenesis. TGFbeta1 moves proliferating cells out of the cell cycle and promotes their subsequent migration. The present study tested the hypothesis that neural progenitors self-regulate TGFbeta1. B104 neuroblastoma cells which can grow in the absence of serum or growth factors were used in systematic studies of transcription, translation, release, and activation. These studies relied on quantitative enzyme-linked immunosorbent assays and real-time polymerase chain reactions. TGFbeta1 positively upregulated its own intracellular expression and promoted increased release of TGFbeta1 from cells. The induction of TGFbeta1 was independent of a change in transcription, but it depended on cycloheximide-inhibited translation. Signaling mediated by downstream Smad2/3 through the TGFbeta receptors and intracellular protein transport were also required for release of TGFbeta1 from B104 cells. Thus, TGFbeta1 production and release were mediated through a feed-forward mechanism and were pivotally regulated at the level of translation. These activities appear to be key for the role of TGFbeta1 in the proliferation and migration of young neurons.
Collapse
Affiliation(s)
- Amanda L Lindke
- Department of Neuroscience and Physiology, State University of New York-Upstate Medical University, Syracuse NY 13210, USA
| | | | | |
Collapse
|
16
|
Ou XM, Stockmeier CA, Meltzer HY, Overholser JC, Jurjus GJ, Dieter L, Chen K, Lu D, Johnson C, Youdim MB, Austin MC, Luo J, Sawa A, May W, Shih JC. A novel role for glyceraldehyde-3-phosphate dehydrogenase and monoamine oxidase B cascade in ethanol-induced cellular damage. Biol Psychiatry 2010; 67:855-63. [PMID: 20022592 PMCID: PMC2854240 DOI: 10.1016/j.biopsych.2009.10.032] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Revised: 10/14/2009] [Accepted: 10/17/2009] [Indexed: 12/29/2022]
Abstract
BACKGROUND Alcoholism is a major psychiatric condition at least partly associated with ethanol (EtOH)-induced cell damage. Although brain cell loss has been reported in subjects with alcoholism, the molecular mechanism is unclear. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and monoamine oxidase B (MAO B) reportedly play a role in cellular dysfunction under stressful conditions and might contribute to EtOH-induced cell damage. METHODS Expression of GAPDH and MAO B protein was studied in human glioblastoma and neuroblastoma cell lines exposed to physiological concentrations of EtOH. Expression of these proteins was also examined in the prefrontal cortex from human subjects with alcohol dependence and in rats fed with an EtOH diet. Coimmunoprecipitation, subcellular fractionation, and luciferase assay were used to address nuclear GAPDH-mediated MAO B activation. To test the effects of inactivation, RNA interference and pharmacological intervention were used, and cell damage was assessed by terminal deoxynucleotidyl transferase (TdT)-mediated dUTP Nick End Labeling (TUNEL) and hydrogen peroxide measurements. RESULTS Ethanol significantly increases levels of GAPDH, especially nuclear GAPDH, and MAO B in neuronal cells as well as in human and rat brains. Nuclear GAPDH interacts with the transcriptional activator, transforming growth factor-beta-inducible early gene 2 (TIEG2), and augments TIEG2-mediated MAO B transactivation, which results in cell damage in neuronal cells exposed to EtOH. Knockdown expression of GAPDH or treatment with MAO B inhibitors selegiline (deprenyl) and rasagiline (Azilect) can block this cascade. CONCLUSIONS Ethanol-elicited nuclear GAPDH augments TIEG2-mediated MAO B, which might play a role in brain damage in subjects with alcoholism. Compounds that block this cascade are potential candidates for therapeutic strategies.
Collapse
Affiliation(s)
- Xiao-Ming Ou
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA.
| | - Craig A. Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, Departments of Psychiatry (CAS, GJJ and LD) and Psychology (JCO), Case Western Reserve University, Cleveland, OH 44106
| | - Herbert Y. Meltzer
- Department of Psychiatry, Psychiatric Hospital at Vanderbilt University, Nashville, TN 37212
| | - James C. Overholser
- Departments of Psychiatry (CAS, GJJ and LD) and Psychology (JCO), Case Western Reserve University, Cleveland, OH 44106
| | - George J. Jurjus
- Departments of Psychiatry (CAS, GJJ and LD) and Psychology (JCO), Case Western Reserve University, Cleveland, OH 44106
| | - Lesa Dieter
- Departments of Psychiatry (CAS, GJJ and LD) and Psychology (JCO), Case Western Reserve University, Cleveland, OH 44106
| | - Kevin Chen
- Department of Molecular Pharmacology and Toxicology, School of Pharmacy, University of Southern California, Los Angeles, CA 90033
| | - Deyin Lu
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216
| | - Chandra Johnson
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216
| | - Moussa B.H. Youdim
- Eve Topf and USA National Parkinson Foundation Centers of Excellence for Neurodegenerative Diseases Research and Department of Pharmacology, Rappaport Family Research Institute, Technion-Faculty of Medicine, Haifa, Israel
| | - Mark C. Austin
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216
| | - Jia Luo
- Departments of Internal Medicine, University of Kentucky College of Medicine, Lexington, KY 40536
| | - Akira Sawa
- Departments of Psychiatry and Neuroscience, Johns Hopkins University School of Medicine, Baltimore MD 21287
| | - Warren May
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216
| | - Jean C. Shih
- Department of Molecular Pharmacology and Toxicology, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| |
Collapse
|
17
|
Overexpression of serum response factor restores ocular dominance plasticity in a model of fetal alcohol spectrum disorders. J Neurosci 2010; 30:2513-20. [PMID: 20164336 DOI: 10.1523/jneurosci.5840-09.2010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Neuronal plasticity deficits underlie many of the neurobehavioral problems seen in fetal alcohol spectrum disorders (FASD). Recently, we showed that third trimester alcohol exposure leads to a persistent disruption in ocular dominance (OD) plasticity. For instance, a few days of monocular deprivation results in a robust reduction of cortical regions responsive to the deprived eye in normal animals, but not in ferrets exposed early to alcohol. This plasticity deficit can be reversed if alcohol-exposed animals are treated with a phosphodiesterase type 1 (PDE1) inhibitor during the period of monocular deprivation. PDE1 inhibition can increase cAMP and cGMP levels, activating transcription factors such as the cAMP response element binding protein (CREB) and the serum response factor (SRF). SRF is important for many plasticity processes such as LTP, LTD, spine motility, and axonal pathfinding. Here we attempt to rescue OD plasticity in alcohol-treated ferrets using a Sindbis viral vector to express a constitutively active form of SRF during the period of monocular deprivation. Using optical imaging of intrinsic signals and single-unit recordings, we observed that overexpression of a constitutively active form of SRF, but neither its dominant-negative nor GFP, restored OD plasticity in alcohol-treated animals. Surprisingly, this restoration was observed throughout the extent of the primary visual cortex and most cells infected by the virus were positive for GFAP rather than NeuN. This finding suggests that overexpression of SRF in astrocytes may reduce the deficits in neuronal plasticity seen in models of FASD.
Collapse
|
18
|
Liu L, Cao JX, Sun B, Li HL, Xia Y, Wu Z, Tang CL, Hu J. Mesenchymal stem cells inhibition of chronic ethanol-induced oxidative damage via upregulation of phosphatidylinositol-3-kinase/Akt and modulation of extracellular signal-regulated kinase 1/2 activation in PC12 cells and neurons. Neuroscience 2010; 167:1115-24. [PMID: 20153405 DOI: 10.1016/j.neuroscience.2010.01.057] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Revised: 01/03/2010] [Accepted: 01/27/2010] [Indexed: 01/01/2023]
Abstract
It is well known that chronic ethanol consumption damages CNS through oxidative stress which results in many dysfunctions. Recently, it has been demonstrated that as a promising strategy to treat several neurological diseases, transplanted bone marrow-derived mesenchymal stem cells (MSCs) can secrete lots of protective factors that in turn promote function recovery. In the present study, we assessed the potential effects of MSCs conditioned medium (MSC-CM) against chronic ethanol-associated damage on PC12 cells and primary cortical neurons. We found that pretreatment with MSC-CM notably improved cell survival, prevented chronic ethanol-associated apoptosis and abolished the robust deterioration in oxidative status. In addition, we also discovered that chronic ethanol exposure induced an inactivation of phosphatidylinositol-3-kinase (PI3K)/Akt and a lasting activation of extracellular signal-regulated kinase 1/2 (ERK1/2) in both PC12 cells and primary cortical neurons which were able to be reversed by MSC-CM. The PI3K inhibitor (LY294002) was able to reduce the antioxidative and cytoprotective effects conferred by MSC-CM, in part, and the ERK1/2 inhibitor (PD98059) was able to elicit significant protection from chronic ethanol cytotoxicity but not rescue the deterioration in oxidative status induced by chronic ethanol. Taken together, these findings provide the first evidence that MSCs might have potent antioxidant action to shield the apoptotic impairment from chronic ethanol exposure in PC12 cells and neurons, which is involved in upregulation of PI3K/Akt and modulation of ERK1/2 activation, at least partially.
Collapse
Affiliation(s)
- L Liu
- Department of Psychiatry, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, PR China
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Kim YK, Lee BC, Ham BJ, Yang BH, Roh S, Choi J, Kang TC, Chai YG, Choi IG. Increased transforming growth factor-beta1 in alcohol dependence. J Korean Med Sci 2009; 24:941-4. [PMID: 19794996 PMCID: PMC2752781 DOI: 10.3346/jkms.2009.24.5.941] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2008] [Accepted: 10/01/2008] [Indexed: 01/01/2023] Open
Abstract
Ethanol and its metabolite acetaldehyde increase transforming growth factor beta1 (TGF-beta1) expression in animal studies. TGF-beta1 is related with the hepatic stellate cell (the key element of hepatic fibrogenesis) and the radial glia (the key element of neuronal migration). Blood samples were collected from 41 patients with alcohol dependence, TGF-beta1 levels measured by ELISA were compared with 41 normal subjects. Plasma TGF-beta1 levels in the patients with alcohol dependence (1,653.11+/-532.45 pg/mL) were significantly higher than those of healthy subjects (669.87+/-366.53 pg/mL) (P=0.000). Patients with or without liver pathology showed no difference in TGF-beta1 (P=0.36). Increased TGF-beta1 may mediate deleterious effect of alcohol such as hepatic fibrosis and suppressed neuronal developments in alcohol dependence patients.
Collapse
Affiliation(s)
- Yong-Ku Kim
- Department of Psychiatry, College of Medicine, Korea University, Ansan Hospital, Ansan, Korea
| | - Boung Chul Lee
- Department of Neuropsychiatry, Hallym University Hangang Sacred Heart Hospital, Seoul, Korea
| | - Byung Joo Ham
- Department of Neuropsychiatry, Hallym University Hangang Sacred Heart Hospital, Seoul, Korea
| | - Byung-Hwan Yang
- Department of Neuropsychiatry, Hanyang University Hospital, Seoul, Korea
| | - Sungwon Roh
- Department of Neuropsychiatry, Hanyang University Hospital, Seoul, Korea
| | - Joonho Choi
- Department of Neuropsychiatry, Hanyang University Hospital, Seoul, Korea
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, Korea
| | - Young-Gyu Chai
- Division of Molecular & Life Science, Hanyang University College of Sciences, Seoul, Korea
| | - Ihn-Geun Choi
- Department of Neuropsychiatry, Hallym University Hangang Sacred Heart Hospital, Seoul, Korea
| |
Collapse
|
20
|
Ethanol inhibition of aspartyl-asparaginyl-beta-hydroxylase in fetal alcohol spectrum disorder: potential link to the impairments in central nervous system neuronal migration. Alcohol 2009; 43:225-40. [PMID: 19393862 DOI: 10.1016/j.alcohol.2008.09.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2008] [Revised: 08/20/2008] [Accepted: 09/17/2008] [Indexed: 12/30/2022]
Abstract
Fetal alcohol spectrum disorder (FASD) is caused by prenatal exposure to alcohol and associated with hypoplasia and impaired neuronal migration in the cerebellum. Neuronal survival and motility are stimulated by insulin and insulin-like growth factor (IGF), whose signaling pathways are major targets of ethanol neurotoxicity. To better understand the mechanisms of ethanol-impaired neuronal migration during development, we examined the effects of chronic gestational exposure to ethanol on aspartyl (asparaginyl)-beta-hydroxylase (AAH) expression, because AAH is regulated by insulin/IGF and mediates neuronal motility. Pregnant Long-Evans rats were pair-fed isocaloric liquid diets containing 0, 8, 18, 26, or 37% ethanol by caloric content from gestation day 6 through delivery. Cerebella harvested from postnatal day 1 pups were used to examine AAH expression in tissue, and neuronal motility in Boyden chamber assays. We also used cerebellar neuron cultures to examine the effects of ethanol on insulin/IGF-stimulated AAH expression, and assess the role of GSK-3beta-mediated phosphorylation on AAH protein levels. Chronic gestational exposure to ethanol caused dose-dependent impairments in neuronal migration and corresponding reductions in AAH protein expression in developing cerebella. In addition, prenatal ethanol exposure inhibited insulin and IGF-I-stimulated directional motility in isolated cerebellar granule neurons. Ethanol-treated neuronal cultures (50mMx96h) also had reduced levels of AAH protein. Mechanistically, we showed that AAH protein could be phosphorylated on Ser residues by GSK-3beta, and that chemical inhibition of GSK-3beta and/or global Caspases increases AAH protein in both control- and ethanol-exposed cells. Ethanol-impaired neuronal migration in FASD is associated with reduced AAH expression. Because ethanol increases the activities of both GSK-3beta and Caspases, the inhibitory effect of ethanol on neuronal migration could be mediated by increased GSK-3beta phosphorylation and Caspase degradation of AAH protein.
Collapse
|
21
|
Lindsley TA, Miller MW, Littner Y, Bearer CF. Signaling pathways regulating cell motility: a role in ethanol teratogenicity? Alcohol Clin Exp Res 2006; 30:1445-50. [PMID: 16899049 PMCID: PMC4199580 DOI: 10.1111/j.1530-0277.2006.00173.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This article summarizes the proceedings of a symposium presented at the 2005 annual meeting of the Research Society on Alcoholism in Santa Barbara, California. The organizer and chair was Tara A. Lindsley. The presentations were (1) Ethanol and Neuron Migration in the CNS, by Michael W. Miller; (2) Ethanol and L1-mediated Neurite Outgrowth, by Yoav Littner and Cynthia F. Bearer; and (3) Ethanol and Axon Guidance, by Tara A. Lindsley.
Collapse
Affiliation(s)
- Tara A Lindsley
- Center for Neuropharmacology & Neuroscience, MC-136, Albany Medical College, 47 New Scotland Ave., Albany NY 12208, USA.
| | | | | | | |
Collapse
|
22
|
Miller MW, Mooney SM, Middleton FA. Transforming growth factor beta1 and ethanol affect transcription and translation of genes and proteins for cell adhesion molecules in B104 neuroblastoma cells. J Neurochem 2006; 97:1182-90. [PMID: 16686695 DOI: 10.1111/j.1471-4159.2006.03858.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Transforming growth factor (TGF) beta1 and ethanol retard the migration of young, post-mitotic neurons to the developing cerebral cortex. The coordination of this migration depends upon cell adhesion proteins (CAPs). We examined the effects of TGFbeta1 and ethanol on genes related to both TGF and CAPs. Rat B104 neuroblastoma cells were treated with TGFbeta1 (0 or 10 ng/mL) and ethanol (0 or 400 mg/dL) for 6-48 h. Total RNA was purified from each sample and analyzed using the Rat U34A GeneChip (Affymetrix). Candidate genes were those up- or down-regulated by either TGFbeta1 or ethanol. Twenty transcripts of CAPs were identified as being expressed by B104 cells and as being affected by treatment with TGFbeta1 or ethanol. The expression was verified for five representative genes (neural cell adhesion molecule, L1, and integrins alpha1, alpha7, and beta1) using assays with real-time reverse transcriptase-polymerase chain reactions. Each of these genes showed time-dependent changes. The changes were reflected in increases in protein expression that appeared within 24 or 48 h. Thus, the effects of TGFbeta1 and ethanol on CAPs parallel changes described in vivo and likely underlie changes associated with ethanol-induced alterations in neuronal migration.
Collapse
Affiliation(s)
- Michael W Miller
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York 13210, USA
| | | | | |
Collapse
|
23
|
Siegenthaler JA, Miller MW. Transforming growth factor beta 1 promotes cell cycle exit through the cyclin-dependent kinase inhibitor p21 in the developing cerebral cortex. J Neurosci 2006; 25:8627-36. [PMID: 16177030 PMCID: PMC6725510 DOI: 10.1523/jneurosci.1876-05.2005] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
During cortical neurogenesis, cell proliferation and cell cycle exit are carefully regulated to ensure that the appropriate numbers of cells are produced. The antiproliferative agent transforming growth factor beta1 (TGFbeta1) and its receptors are endogenously expressed in proliferative zones of the developing cerebral cortex, thus implicating the growth factor in cell cycle regulation. The present study tested the hypothesis that TGFbeta1 promotes cell cycle exit in the cortical ventricular zone (VZ) through modulation of cell cycle protein expression, in particular cyclin D1 and the cyclin-dependent kinase inhibitors p27 and p21. Although it did not affect the length of the cell cycle, TGFbeta1 decreased the fraction of VZ-cycling cells by 21% and increased the number of VZ cells exiting the cell cycle a commensurate 24%. TGFbeta1 selectively increased the expression of p21 in the VZ. In addition, high p21 expression levels were observed in VZ cells as they exited the cell cycle, and TGFbeta1 increased the number p21-positive cells exiting the cell cycle. Collectively, these data show the following: (1) TGFbeta1 promotes cell cycle exit, (2) p21 upregulation is correlated with cell cycle exit, and (3) TGFbeta1-induced cell cycle exit is mediated by p21.
Collapse
Affiliation(s)
- Julie A Siegenthaler
- Department of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
| | | |
Collapse
|
24
|
Miller MW. Ruminations of a Jersey boy: Ferdinand under the cork tree. 2005 Henry L. Rosett Award. Alcohol Clin Exp Res 2006; 30:180-4. [PMID: 16441266 DOI: 10.1111/j.1530-0277.2006.00021.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Michael W Miller
- Department of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, New York 13210, USA.
| |
Collapse
|
25
|
Nguyen MP, Bittner GD, Fishman HM. Critical interval of somal calcium transient after neurite transection determines B 104 cell survival. J Neurosci Res 2005; 81:805-16. [PMID: 16049977 PMCID: PMC1237108 DOI: 10.1002/jnr.20606] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Nerve cells may survive or die after axonal or dendritic transection. After neurite transection near (<50 mum) the cell body of Fura-2-loaded B 104 neuroblastoma (rat brain-derived) cells, the somal calcium concentration (SCC) undergoes a three-phase transient change: a rapid (0-0.15-min post-transection [PT]) rise phase, followed by an early (0.15--1.5-min PT) rapid decay phase, and succeeded by a late (1.5-60-min PT) slower decay phase that restores SCC to preinjury levels. The SCC in a critical interval (1.5-12.5 min PT) of the third transient phase correlates with cell fate, i.e., most transected cells that exclude dye (restore a barrier) and die have a significantly higher (P<0.005) SCC in this critical interval than do transected cells that exclude dye and survive at 24-hr PT. Loading BAPTA (chelation of somal Ca(2+)) before, but not after, the critical interval increases the percentage of cells that survive compared to that of cells transected without BAPTA loading. Furthermore, most transected cells that die despite successful barrier restoration exhibit characteristics consistent with apoptosis initiated during the critical interval of the SCC, including caspase activation and plasmalemmal phosphatidylserine translocation. These data suggest that decreased cell survival for injuries near the soma is due to Ca(2+)-initiated apoptosis during the critical interval of the third phase of the SCC transient. (c) 2005 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Michael P. Nguyen
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555-0641
| | - George D. Bittner
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555-0641
- Neurobiology Section, School of Biological Sciences, Institute for Neuroscience, and College of Pharmacy, The University of Texas at Austin, Austin, TX 78712
| | - Harvey M. Fishman
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555-0641
- * Correspondence to: Harvey M. Fishman, Tel: 409-772-2975, Fax: 409-772-6442,
| |
Collapse
|
26
|
Siegenthaler JA, Miller MW. Ethanol disrupts cell cycle regulation in developing rat cortex interaction with transforming growth factor beta1. J Neurochem 2005; 95:902-12. [PMID: 16190877 DOI: 10.1111/j.1471-4159.2005.03461.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ethanol is a potent teratogenic agent that disrupts several aspects of neuronogenesis, including the proliferation rate of cortical precursors. With regard to corticogenesis, possible targets of ethanol toxicity include soluble factors, like transforming growth factor beta1 (TGFbeta1), that regulate cortical growth and cell cycle proteins that control the kinetics of the cell cycle. The effect of ethanol on normal cell proliferation and TGFbeta1-regulated cell proliferation in the developing cortex was assessed using an organotypic slice culture model. Ethanol elongated the cell cycle, possibly through a decrease in the expression of G1 cell cycle protein cyclin D1. Further, ethanol exposure antagonized the anti-proliferative action of TGFbeta1 and blocked TGFbeta1-dependent increases in cell cycle inhibitor p21. Collectively, this evidence suggests that disruption of appropriate cell cycle protein expression and inhibition of TGFbeta1 activity are potential mechanisms underlying the effect of ethanol on cortical development.
Collapse
Affiliation(s)
- Julie A Siegenthaler
- Department of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, New York 13210, USA
| | | |
Collapse
|
27
|
Vallés SL, Blanco AM, Pascual M, Guerri C. Chronic ethanol treatment enhances inflammatory mediators and cell death in the brain and in astrocytes. Brain Pathol 2005; 14:365-71. [PMID: 15605983 PMCID: PMC8095743 DOI: 10.1111/j.1750-3639.2004.tb00079.x] [Citation(s) in RCA: 195] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Inflammatory processes and cytokine expression have been implicated in the pathogenesis of several neurodegenerative disorders. Chronic ethanol intake induces brain damage, although the mechanisms involved in this effect are not well understood. We tested the hypothesis that activation of glial cells by ethanol would induce stimulation of signaling pathways and inflammatory mediators in brain, and would cause neurotoxicity. We used cerebral cortex from control and chronic ethanol-fed rats, which received ethanol-liquid diet for 5 months and cultured of astrocytes exposed to 75 mM ethanol for 7 days. Our results demonstrate that chronic ethanol treatment up-regulates iNOS, COX-2 and IL-1beta in rat cerebral cortex and in cultured astrocytes. Under both experimental conditions, up-regulation of these inflammatory mediators and IL-1RI concomitantly occurs with the stimulation of IRAK and MAP kinases, including ERK1/2, p-38 and JNK, which trigger the downstream activation of oxidant-sensitive transcription factors NF-KB and AP-1. These effects were associated with an increased in both caspase-3 and apoptosis in ethanol-fed rats and in astrocytes exposed to ethanol. In conclusion, chronic ethanol treatment stimulates glial cells, up-regulating the production and the expression of inflammatory mediators in the brain, and activating signalling pathways and transcription factors involved in inflammatory damage and cell death.
Collapse
Affiliation(s)
- Soraya L. Vallés
- Instituto de Investigaciones Citológicas (FVIB), Valencia, Spain
| | - Ana M. Blanco
- Instituto de Investigaciones Citológicas (FVIB), Valencia, Spain
| | - María Pascual
- Instituto de Investigaciones Citológicas (FVIB), Valencia, Spain
| | - Consuelo Guerri
- Instituto de Investigaciones Citológicas (FVIB), Valencia, Spain
| |
Collapse
|
28
|
Abstract
Chronic ethanol abuse is associated with liver injury, neurotoxicity, hypertension, cardiomyopathy, modulation of immune responses and increased risk for cancer, whereas moderate alcohol consumption exerts protective effect on coronary heart disease. However, the signal transduction mechanisms underlying these processes are not well understood. Emerging evidences highlight a central role for mitogen activated protein kinase (MAPK) family in several of these effects of ethanol. MAPK signaling cascade plays an essential role in the initiation of cellular processes such as proliferation, differentiation, development, apoptosis, stress and inflammatory responses. Modulation of MAPK signaling pathway by ethanol is distinctive, depending on the cell type; acute or chronic; normal or transformed cell phenotype and on the type of agonist stimulating the MAPK. Acute exposure to ethanol results in modest activation of p42/44 MAPK in hepatocytes, astrocytes, and vascular smooth muscle cells. Acute ethanol exposure also results in potentiation or prolonged activation of p42/44MAPK in an agonist selective manner. Acute ethanol treatment also inhibits serum stimulated p42/44 MAPK activation and DNA synthesis in vascular smooth muscle cells. Chronic ethanol treatment causes decreased activation of p42/44 MAPK and inhibition of growth factor stimulated p42/44 MAPK activation and these effects of ethanol are correlated to suppression of DNA synthesis, impaired synaptic plasticity and neurotoxicity. In contrast, chronic ethanol treatment causes potentiation of endotoxin stimulated p42/44 MAPK and p38 MAPK signaling in Kupffer cells leading to increased synthesis of tumor necrosis factor. Acute exposure to ethanol activates pro-apoptotic JNK pathway and anti-apoptotic p42/44 MAPK pathway. Apoptosis caused by chronic ethanol treatment may be due to ethanol potentiation of TNF induced activation of p38 MAPK. Ethanol induced activation of MAPK signaling is also involved in collagen expression in stellate cells. Ethanol did not potentiate serum stimulated or Gi-protein dependent activation of p42/44 MAPK in normal hepatocytes but did so in embryonic liver cells and transformed hepatocytes leading to enhanced DNA synthesis. Ethanol has a 'triangular effect' on MAPK that involve direct effects of ethanol, its metabolically derived mediators and oxidative stress. Acetaldehyde, phosphatidylethanol, fatty acid ethyl ester and oxidative stress, mediate some of the effects seen after ethanol alone whereas ethanol modulation of agonist stimulated MAPK signaling appears to be mediated by phosphatidylethanol. Nuclear MAPKs are also affected by ethanol. Ethanol modulation of nuclear p42/44 MAPK occurs by both nuclear translocation of p42/44 MAPK and its activation in the nucleus. Of interest is the observation that ethanol caused selective acetylation of Lys 9 of histone 3 in the hepatocyte nucleus. It is plausible that ethanol modulation of cross talk between phosphorylation and acetylations of histone may regulate chromatin remodeling. Taken together, these recent developments place MAPK in a pivotal position in relation to cellular actions of ethanol. Furthermore, they offer promising insights into the specificity of ethanol effects and pharmacological modulation of MAPK signaling. Such molecular signaling approaches have the potential to provide mechanism-based therapy for the management of deleterious effects of ethanol or for exploiting its beneficial effects.
Collapse
Affiliation(s)
- Annayya R Aroor
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, USA.
| | | |
Collapse
|
29
|
Mooney SM, Miller MW. Ethanol-induced neuronal death in organotypic cultures of rat cerebral cortex. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2003; 147:135-41. [PMID: 14741758 DOI: 10.1016/j.devbrainres.2003.08.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ethanol can affect normal development of the cerebral cortex, e.g., it can disrupt cell migration and exacerbate cell death. In vitro studies using primary cultures or cell lines provide further evidence that cell migration and death are altered by ethanol exposure. Organotypic cultures are more complex than primary cell cultures, and maintain some normal connectivity, thus providing a "more in vivo-like" model of brain development. We predict that exposing organotypic cultures of fetal rat cerebral cortex to ethanol results in changes similar to those described in vivo. Organotypic cultures of brains from 16-day-old fetuses were exposed to ethanol (0, 200, 400 or 800 mg/dl) for 72 h. Stereological methods were used to assess the frequency of viable and dying cells. Dying cells were identified as having DNA with polyadenylated tails or as having condensed chromatin. A small amount of cell death was evident in the marginal zone (MZ) and cortical plate (CP) of control cultures. The MZ, normally a cell body-poor layer, was enriched with somata following exposure to 400 mg/dl ethanol. Ethanol-induced cell death in the MZ; the amount of cell death was doubled following exposure to 800 mg/dl ethanol. The CP was more sensitive than the MZ; cell death increased following treatment with 400 mg/dl ethanol. Thus, organotypic cultures show that ethanol disrupts neuronal migration and increases cell death in the developing cerebral cortex. The effects of ethanol were site-specific and concentration-dependent. These changes are similar to those described in vivo.
Collapse
Affiliation(s)
- Sandra M Mooney
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse NY 13210, USA
| | | |
Collapse
|
30
|
Miller MW. Balance of cell proliferation and death among dynamic populations: a mathematical model. ACTA ACUST UNITED AC 2003; 57:172-82. [PMID: 14556283 DOI: 10.1002/neu.10265] [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] [Indexed: 12/26/2022]
Abstract
Developmental changes in cell numbers represent the dynamic balance between cell proliferation and death. One obstacle to assessing this balance is an inability to quantify the total amount of cell death, i.e., with a positive indicator such as terminal dUTP nick end labeling (TUNEL) or caspase activity. A novel mathematical model is described wherein data on daily cell growth (the change in cell number) and cell cycle kinetics can be used to determine the total amount of cell death. Two sets of data from previously published studies were tested in this model; primary cultured cortical neurons and B104 neuroblastoma cells. These two preparations have contrasting features: neuronal cultures are heterogeneous and have relatively few cells that are actively cycling (i.e., the growth fraction for these cells is low), whereas B104 cells are relatively homogeneous cultures in which the growth fraction is high. In primary cortical cultures, there was a balance in cell production and death. Treatment with a potent anti-mitogen, ethanol (400 mg/dl), affected this balance principally by reducing cell production, although the rate of cell death was also increased. In untreated B104 cells, there was eight-fold more cell production than cell death. Growth factors such as platelet-derived growth factor BB doubled cell production. Ethanol reduced cell production by >60%, and it eliminated growth factor-mediated cell production. All of these changes occurred in the absence of an effect on the amount of cell death. Thus, the model is ideal for predicting the effects of an epigenetic factor (e.g., a growth factor, toxin, or pharmacological agent) on cell development and can be useful in determining the consequences of a genetic manipulation as well.
Collapse
Affiliation(s)
- Michael W Miller
- Department of Neuroscience and Physiology, State University of New York-Upstate Medical University, 750 East Adams Street, Syracuse, New York 13210, USA.
| |
Collapse
|
31
|
|
32
|
Smith TL, Navratilova E. The effect of ethanol exposure on mitogen-activated protein kinase activity and expression in cultured rat astrocytes. Neurosci Lett 2003; 341:91-4. [PMID: 12686373 DOI: 10.1016/s0304-3940(03)00179-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The effects of ethanol exposures on mitogen-activated protein kinase (MAPK) activity were determined in confluent astrocyte monolayers prepared from neonatal rat cerebral cortex. Acute 30 min exposure to 50 mM ethanol had no significant effect on MAPK activity. However, chronic exposure to ethanol for 4 days elicited a concentration-dependent increase in the basal level of this enzyme activity with no parallel increase in its protein expression. In addition, the magnitude of MAPK activation by epidermal growth factor, basic fibroblast growth factor and platelet-derived growth factor was significantly increased above corresponding control values in cells chronically exposed to ethanol. Immunolabeling experiments indicated that the protein expression of receptors for these growth factors was unaffected by ethanol treatment. Our results suggest that even after chronic ethanol treatment, MAPK phosphorylation and, hence, activation remains elevated.
Collapse
Affiliation(s)
- Thomas L Smith
- Research Health Care Group (0-151), Southern AZ VA Health Care System, Tucson, AZ, USA.
| | | |
Collapse
|
33
|
Effects of Ethanol and Transforming Growth Factor ?? (TGF??) on Neuronal Proliferation and nCAM Expression. Alcohol Clin Exp Res 2002. [DOI: 10.1097/00000374-200208000-00022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
34
|
Miller MW, Luo J. Effects of Ethanol and Transforming Growth Factor beta (TGFbeta) on Neuronal Proliferation and nCAM Expression. Alcohol Clin Exp Res 2002. [DOI: 10.1111/j.1530-0277.2002.tb02668.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
35
|
Effects of Ethanol and Basic Fibroblast Growth Factor on the Transforming Growth Factor ??1 Regulated Proliferation of Cortical Astrocytes and C6 Astrocytoma Cells. Alcohol Clin Exp Res 2002. [DOI: 10.1097/00000374-200205000-00011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
36
|
Miller MW, Luo J. Effects of Ethanol and Basic Fibroblast Growth Factor on the Transforming Growth Factor beta1 Regulated Proliferation of Cortical Astrocytes and C6 Astrocytoma Cells. Alcohol Clin Exp Res 2002. [DOI: 10.1111/j.1530-0277.2002.tb02589.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
37
|
Yagle K, Lu H, Guizzetti M, Möller T, Costa LG. Activation of mitogen-activated protein kinase by muscarinic receptors in astroglial cells: role in DNA synthesis and effect of ethanol. Glia 2001; 35:111-20. [PMID: 11460267 DOI: 10.1002/glia.1076] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Mitogen-activated protein kinase (MAPK) can be phosphorylated by mitogens binding to G-protein-coupled receptors and is considered a major pathway involved in cell proliferation. In this study, we report on the activation of MAPK by muscarinic acetylcholine receptors in astroglial cells, namely the 1321N1 human astrocytoma cell line, primary rat cortical astrocytes, and fetal human astrocytes. Carbachol caused a rapid and transient phorphorylation of MAPK (ERK1/2) in all cell types, with an increase in MAPK activity, without changing the levels of MAPK proteins. Human astrocytoma cells were used to characterize the effect of carbachol on MAPK. Experiments with M2- and M3-receptor subtype-selective antagonists, and with pertussis toxin, indicated that the M3 subtype is responsible for activating MAPK in glial cells. Pretreatment of cells with the protein kinase C (PKC) inhibitor bisindolylmaleimide I, or downregulation of PKC by 24-h treatment with the phorbol ester TPA inhibited carbachol-induced MAPK activation. Additional experiments with PKC alpha- or PKC epsilon-specific compounds indicated that the epsilon isozyme of PKC is primarily involved in MAPK activation by carbachol. Chelation of calcium also inhibited MAPK activation by carbachol. Two MEK (MAPK kinase) inhibitors inhibited carbachol-induced DNA synthesis but only at concentrations that exceeded those sufficient to block carbachol-induced MAPK activation. Ethanol (< or =200 mM) had no effect on MAPK when present alone and did not affect carbachol-induced MAPK activation under various experimental conditions, although it inhibits carbachol-induced DNA synthesis at low concentrations (10-100 mM). These results suggest that activation of MAPK by carbachol may be necessary but not sufficient for its mitogenic effect in astroglial cells, and that does not represent a target for ethanol-induced inhibition of DNA synthesis elicited by muscarinic receptors.
Collapse
Affiliation(s)
- K Yagle
- Department of Environmental Health, University of Washington, Seattle, Washington 98105, USA
| | | | | | | | | |
Collapse
|
38
|
Seiler AE, Ross BN, Rubin R. Inhibition of insulin-like growth factor-1 receptor and IRS-2 signaling by ethanol in SH-SY5Y neuroblastoma cells. J Neurochem 2001; 76:573-81. [PMID: 11208920 DOI: 10.1046/j.1471-4159.2001.00025.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The effect of ethanol on insulin-like growth factor-1 (IGF-I)-mediated signal transduction and functional activation in neuronal cells was examined. In human SH-SY5Y neuroblastoma cells, ethanol inhibited tyrosine autophosphorylation of the IGF-I receptor. This corresponded to the inhibition of IGF-I-induced phosphorylation of p42/p44 mitogen-activated/extracellular signal-regulated protein kinase (MAPK) by ethanol. Insulin-related substrate-2 (IRS-2) and focal adhesion kinase phosphorylation were reduced in the presence of ethanol, which corresponded to the prevention of lamellipodia formation (30 min). By contrast, ethanol had no effect on Shc phosphorylation when measured up to 1 h, and did not affect the association of Grb-2 with Shc. Neurite formation at 24 h was similarly unaffected by ethanol. The data indicate that the IGF-I receptor is a target for ethanol in SH-SY5Y cells However, there is diversity in the sensitivity of signaling elements within the IGF-I receptor tyrosine kinase signaling cascades to ethanol, which can be related to the inhibition of specific functional events in neuronal activation.
Collapse
Affiliation(s)
- A E Seiler
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia 19107, USA
| | | | | |
Collapse
|
39
|
Platelet-derived growth factor-mediated signal transduction underlying astrocyte proliferation: site of ethanol action. J Neurosci 1999. [PMID: 10559409 DOI: 10.1523/jneurosci.19-22-10014.1999] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Platelet-derived growth factor (PDGF) is a critical regulator of cell proliferation. Because ethanol inhibits cell proliferation in vivo and in vitro, we hypothesize that ethanol-induced inhibition results from differential interference with signal transduction pathways activated by PDGF. Cultured cortical astrocytes were used to examine the effects of ethanol on PDGF-mediated signal transduction, on the expression of two PDGF monomers (A- and B-chains), and on the expression of two PDGF receptor subunits (PDGFalphar and PDGFbetar). PDGF-B chain homodimer (PDGF-BB), and to a lesser extent PDGF-A chain homodimer (PDGF-AA), stimulated the proliferation of astrocytes raised in a serum-free medium. Ethanol attenuated these actions in a concentration-dependent manner. Ethanol inhibited both PDGF-AA- and PDGF-BB-mediated phosphorylation of PDGFalphar, but it had little effect on PDGFbetar autophosphorylation. Likewise, ethanol abolished the association of PDGFalphar to Ras GTPase-activating protein (Ras-GAP), but it did not affect the binding of Ras-GAP to PDGFbetar. PDGF stimulated the activities of mitogen-activated protein kinase (MAPK) in protein kinase C (PKC) independent and dependent manners. Ethanol inhibited the PKC-independent, acute activation of MAPK; however, it stimulated the PKC-dependent, sustained activation of MAPK. The expression of neither ligand was altered by exposure to ethanol for 3 d. Moreover, such treatment specifically upregulated PDGFalphar expression in a concentration-dependent manner. It did not, however, affect the binding affinity of either receptor. Thus, the signal transduction pathways initiated by PDGF-AA and PDGF-BB were differentially affected by ethanol. This differential vulnerability resulted from the preferential effects of ethanol on PDGFalphar autophosphorylation. Hence, ethanol-induced alterations are transduced through specific receptors of mitogenic growth factors.
Collapse
|