Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Front Genet ; 12: 804222, 2021.
Article in English | MEDLINE | ID: mdl-35126468

ABSTRACT

Alcohol Use Disorder (AUD) is one of the most prevalent mental disorders worldwide. Considering the widespread occurrence of AUD, a reliable, cheap, non-invasive biomarker of alcohol consumption is desired by healthcare providers, clinicians, researchers, public health and criminal justice officials. microRNAs could serve as such biomarkers. They are easily detectable in saliva, which can be sampled from individuals in a non-invasive manner. Moreover, microRNAs expression is dynamically regulated by environmental factors, including alcohol. Since excessive alcohol consumption is a hallmark of alcohol abuse, we have profiled microRNA expression in the saliva of chronic, heavy alcohol abusers using microRNA microarrays. We observed significant changes in salivary microRNA expression caused by excessive alcohol consumption. These changes fell into three categories: downregulated microRNAs, upregulated microRNAs, and microRNAs upregulated de novo. Analysis of these combinatorial changes in microRNA expression suggests dysregulation of specific biological pathways leading to impairment of the immune system and development of several types of epithelial cancer. Moreover, some of the altered microRNAs are also modulators of inflammation, suggesting their contribution to pro-inflammatory mechanisms of alcohol actions. Establishment of the cellular source of microRNAs in saliva corroborated these results. We determined that most of the microRNAs in saliva come from two types of cells: leukocytes involved in immune responses and inflammation, and buccal cells, involved in development of epithelial, oral cancers. In summary, we propose that microRNA profiling in saliva can be a useful, non-invasive biomarker allowing the monitoring of alcohol abuse, as well as alcohol-related inflammation and early detection of cancer.

2.
J Neurogenet ; 30(3-4): 195-204, 2016.
Article in English | MEDLINE | ID: mdl-27845601

ABSTRACT

At the core of the changes characteristic of alcoholism are alterations in gene expression in the brain of the addicted individual. These changes are believed to underlie some of the neuroadaptations that promote compulsive drinking. Unfortunately, the mechanisms by which alcohol consumption produces changes in gene expression remain poorly understood. MicroRNAs (miRNAs) have emerged as important regulators of gene expression because they can coordinately modulate the translation efficiency of large sets of specific mRNAs. Here, we investigate the early miRNA responses elicited by an acute sedating dose of alcohol in the Drosophila model organism. In our analysis, we combine the power of next-generation sequencing with Drosophila genetics to identify alcohol-sensitive miRNAs and to functionally test them for a role in modulating alcohol sensitivity. We identified 14 known Drosophila miRNAs, and 13 putative novel miRNAs that respond to an acute sedative exposure to alcohol. Using the GeneSwitch Gal4/UAS system, a subset of these ethanol-responsive miRNAs was functionally tested to determine their individual contribution in modulating ethanol sensitivity. We identified two microRNAs that when overexpressed significantly increased ethanol sensitivity: miR-6 and miR-310. MicroRNA target prediction analysis revealed that the different alcohol-responsive miRNAs target-overlapping sets of mRNAs. Alcoholism is the product of accumulated cellular changes produced by chronic ethanol consumption. Although all of the changes described herein are extremely rapid responses evoked by a single ethanol exposure, understanding the gene expression changes that occur in the first few minutes after ethanol exposure will help us to categorize ethanol responses into those that are near instantaneous and those that are emergent responses produced only by repeated ethanol exposure.


Subject(s)
Alcoholism/genetics , MicroRNAs/drug effects , MicroRNAs/genetics , Transcriptome/drug effects , Animals , Drosophila melanogaster , Ethanol/pharmacology , Female , Gene Expression Profiling
3.
Alcohol ; 49(8): 825-36, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25896098

ABSTRACT

This review summarizes the proceedings of a symposium presented at the "Alcoholism and Stress: A Framework for Future Treatment Strategies" conference held in Volterra, Italy on May 6-9, 2014. The overall goal of the symposium titled "Applying the New Genomics to Alcohol Dependence", chaired by Dr. Adron Harris, was to highlight recent genomic discoveries and applications for profiling alcohol use disorder (AUD). Dr. Sean Farris discussed the gene expression networks related to lifetime consumption of alcohol within human prefrontal cortex. Dr. Andrzej Pietrzykowski presented the effects of alcohol on microRNAs in humans and animal models. Alcohol-induced alterations in the synaptic transcriptome were discussed by Dr. Michael Miles. Dr. Pietro Sanna examined methods to probe the gene regulatory networks that drive excessive alcohol drinking, and Dr. Samir Zakhari served as a panel discussant and summarized the proceedings. Collectively, the presentations emphasized the power of integrating multiple levels of genetics and transcriptomics with convergent biological processes and phenotypic behaviors to determine causal factors of AUD. The combined use of diverse data types demonstrates how unique approaches and applications can help categorize genetic complexities into relevant biological networks using a systems-level model of disease.


Subject(s)
Alcoholism/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation , MicroRNAs/genetics , Transcriptome/genetics , Animals , Gene Expression Profiling , Gene Regulatory Networks , Genomics , Humans , Sequence Analysis, RNA
4.
Gen Comp Endocrinol ; 199: 1-15, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24457251

ABSTRACT

Testosterone plays a key role in preparation of a male domesticated goat (Capra hircus) to breeding season including changes in the urogenital tract of a male goat (buck). microRNAs are important regulators of cellular metabolism, differentiation and function. They are powerful intermediaries of hormonal activity in the body, including the urogenital tract. We investigated seasonal changes in expression of microRNAs in goat buck urine and their potential consequences using next generation sequencing (microRNA-Seq). We determined the location of each microRNA gene in the goat genome. Testosterone was measured by radioimmunoassay and the androgen receptor binding sites (ARBS) in the promoters of the microRNA genes were determined by MatInspector. The overall impact of regulated microRNAs on cellular physiology was assessed by mirPath. We observed high testosterone levels during the breeding season and changes in the expression of forty microRNAs. Nineteen microRNAs were upregulated, while twenty-one were downregulated. We identified several ARBS in the promoters of regulated microRNAs. Notably, the mostly inhibited microRNA, miR-1246, has a unique set of several gene copy variants associated with a cluster of androgen receptor binding sites. Concomitant changes in regulated microRNA expression could promote transcription, proliferation and differentiation of urogenital tract cells. Together, these findings indicate that in a domesticated goat (Capra hircus), there are specific changes in the microRNA expression profile in buck urine during breeding season, which could be attributable to high testosterone levels during breeding, and could help in preparation of the urogenital tract for high metabolic demands of that season.


Subject(s)
Gene Expression Regulation , Goats/genetics , High-Throughput Nucleotide Sequencing/methods , MicroRNAs/genetics , MicroRNAs/urine , Seasons , Animals , Base Sequence , Breeding , Chromosomes, Mammalian/genetics , Cluster Analysis , Computational Biology , Genome , Male , Molecular Sequence Data , Reproduction/genetics , Signal Transduction/genetics
5.
Front Neurosci ; 8: 389, 2014.
Article in English | MEDLINE | ID: mdl-25561905

ABSTRACT

Malfunction of synaptic plasticity in different brain regions, including the amygdala plays a role in impulse control deficits that are characteristics of several psychiatric disorders, such as ADHD, schizophrenia, depression and addiction. Previously, we discovered a locus for impulsivity (Impu1) containing the neuregulin 3 (Nrg3) gene, of which the level of expression determines levels of inhibitory control. MicroRNAs (miRNAs) are potent regulators of gene expression, and have recently emerged as important factors contributing to the development of psychiatric disorders. However, their role in impulsivity, as well as control of Nrg3 expression or malfunction of the amygdala, is not well established. Here, we used the GeneNetwork database of BXD mice to search for correlated traits with impulsivity using an overrepresentation analysis to filter for biologically meaningful traits. We determined that inhibitory control was significantly correlated with expression of miR-190b, -28a, -340, -219a, and -491 in the amygdala, and that the overrepresented correlated traits showed a specific pattern of coregulation with these miRNAs. A bioinformatics analysis identified that miR-190b, by targeting an Nrg3-related network, could affect synaptic plasticity in the amygdala, targeting bot impulsive and compulsive traits. Moreover, miR-28a, -340, -219a, and possibly -491 could act on synaptic function by determining the balance between neuronal outgrowth and differentiation. We propose that these miRNAs are attractive candidates of regulation of amygdala synaptic plasticity, possibly during development but also in maintaining the impulsive phenotype. These results can help us to better understand mechanisms of synaptic dysregulation in psychiatric disorders.

6.
Alcohol Clin Exp Res ; 37(6): 933-40, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23421938

ABSTRACT

BACKGROUND: Voltage-gated calcium channels (VGCCs) in rat neurohypophysial terminals exhibit molecular tolerance to alcohol, including desensitization to the drug and increased current density, after 3 weeks of alcohol drinking. Moreover, after this time, terminals from drinking rats exhibit diminished alcohol inhibition of vasopressin (AVP) release. METHODS: We took advantage of organotypic cultures (explants) of the hypothalamo-neurohypophysial system (HNS) to extend our analysis of molecular tolerance to 2 classes of the VGCC. The isolated HNS explant allows much finer temporal resolution of molecular tolerance than do voluntary drinking paradigms. After exposure of the HNS explant to alcohol, terminals are isolated by mechanical treatment and plated in a dish. Patch clamp recording techniques are used to obtain VGCC currents, and immunohistochemistry is used to determine VGCC distribution. A release assay is used to provide functional readout of AVP release. RESULTS: We show that even a brief, 1-hour exposure to a clinically relevant concentration of alcohol is sufficient to evoke similar changes to those observed after several weeks of exposure. Acute ethanol (EtOH) exposure inhibits high K(+) -induced AVP release from naïve terminals. However, terminals pre-exposed to 20 mM EtOH for 1 hour become tolerant to EtOH, and subsequent exposure has significantly less effect on high K(+) -induced AVP release. Electrophysiological recordings indicate that among different types of VGCCs present in the neuronal terminal, the L-type is the most affected by alcohol. The current density of L-type current is significantly increased (approximately 50%), while its responsiveness to alcohol is significantly diminished (approximately 50%), after brief alcohol exposure. Fluorescent imaging results were consistent with the electrophysiology and suggest that the increased current density of VGCCs after brief exposure is attributable to combined synthesis of 1.2 and 1.3 subtypes of the L-type VGCC and redistribution of channel protein into terminal plasma membrane. CONCLUSIONS: These data indicate that a brief alcohol exposure affects subsequent alcohol sensitivity of VGCCs and neuropeptide release from presynaptic terminals.


Subject(s)
Arginine Vasopressin/metabolism , Calcium Channels, L-Type/drug effects , Central Nervous System Depressants/pharmacology , Drug Tolerance/physiology , Ethanol/pharmacology , Pituitary Gland, Posterior/drug effects , Presynaptic Terminals/drug effects , Animals , Calcium Channels, L-Type/physiology , Electrophysiology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , In Vitro Techniques , Male , Patch-Clamp Techniques , Pituitary Gland, Posterior/physiology , Presynaptic Terminals/physiology , Rats , Rats, Sprague-Dawley
8.
Alcohol Clin Exp Res ; 34(4): 575-87, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20102566

ABSTRACT

Ethanol exerts complex effects on human physiology and health. Ethanol is not only addictive, but it is also a fetal teratogen, an adult neurotoxin, and an etiologic agent in hepatic and cardiovascular disease, inflammation, bone loss, and fracture susceptibility. A large number of genes and signaling mechanisms have been implicated in ethanol's deleterious effects leading to the suggestion that ethanol is a "dirty drug." An important question is, are there cellular "master-switches" that can explain these pleiotropic effects of ethanol? MicroRNAs (miRNAs) have been recently identified as master regulators of the cellular transcriptome and proteome. miRNAs play an increasingly appreciated and crucial role in shaping the differentiation and function of tissues and organs in both health and disease. This critical review discusses new evidence showing that ethanol-sensitive miRNAs are indeed regulatory master-switches. More specifically, miRNAs control the development of tolerance, a crucial component of ethanol addiction. Other drugs of abuse also target some ethanol-sensitive miRNAs suggesting that common biochemical mechanisms underlie addiction. This review also discusses evidence that miRNAs mediate several ethanol pathologies, including disruption of neural stem cell proliferation and differentiation in the exposed fetus, gut leakiness that contributes to endotoxemia and alcoholic liver disease, and possibly also hepatocellular carcinomas and other gastrointestinal cancers. Finally, this review provides a perspective on emerging investigations into potential roles of miRNAs as mediators of ethanol's effects on inflammation and fracture healing, as well as the potential for miRNAs as diagnostic biomarkers and as targets for therapeutic interventions for alcohol-related disorders.


Subject(s)
Alcoholism/genetics , Alcoholism/metabolism , Ethanol/metabolism , Ethanol/toxicity , MicroRNAs/physiology , Alcoholism/pathology , Animals , Cell Death/drug effects , Cell Death/physiology , Humans , Signal Transduction/drug effects , Signal Transduction/physiology
9.
Proc Natl Acad Sci U S A ; 105(45): 17543-8, 2008 Nov 11.
Article in English | MEDLINE | ID: mdl-18981408

ABSTRACT

Tolerance, described as the loss of drug effectiveness over time, is an important component of addiction. The degree of acute behavioral tolerance to alcohol exhibited by a naïve subject can predict the likelihood of alcohol abuse. Thus, the determinants of acute tolerance are important to understand. Calcium- and voltage-gated (BK) potassium channels, consisting of pore forming alpha and modulatory beta subunits, are targets of ethanol (EtOH) action. Here, we examine the role, at the molecular, cellular, and behavioral levels, of the BK beta4 subunit in acute tolerance. Single channel recordings in HEK-293 cells show that, in the absence of beta4, EtOH potentiation of activity exhibits acute tolerance, which is blocked by coexpressing the beta4 subunit. BK channels in acutely isolated medium spiny neurons from WT mice (in which the beta4 subunit is well-represented) exhibit little tolerance. In contrast, neuronal BK channels from beta4 knockout (KO) mice do display acute tolerance. Brain slice recordings showed tolerance to EtOH's effects on spike patterning in KO but not in WT mice. In addition, beta4 KO mice develop rapid tolerance to EtOH's locomotor effects, whereas WT mice do not. Finally, in a restricted access ethanol self-administration assay, beta4 KO mice drink more than their WT counterparts. Taken together, these data indicate that the beta4 subunit controls ethanol tolerance at the molecular, cellular, and behavioral levels, and could determine individual differences in alcohol abuse and alcoholism, as well as represent a therapeutic target for alcoholism.


Subject(s)
Corpus Striatum/metabolism , Ethanol/pharmacology , Feeding Behavior/drug effects , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Locomotion/drug effects , Animals , Cell Line , Drug Tolerance/physiology , Electrophysiology , Humans , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Mice , Mice, Knockout
10.
Neuron ; 59(2): 274-87, 2008 Jul 31.
Article in English | MEDLINE | ID: mdl-18667155

ABSTRACT

Tolerance represents a critical component of addiction. The large-conductance calcium- and voltage-activated potassium channel (BK) is a well-established alcohol target, and an important element in behavioral and molecular alcohol tolerance. We tested whether microRNA, a newly discovered class of gene expression regulators, plays a role in the development of tolerance. We show that in adult mammalian brain, alcohol upregulates microRNA miR-9 and mediates posttranscriptional reorganization in BK mRNA splice variants by miR-9-dependent destabilization of BK mRNAs containing 3'UTRs with a miR-9 Recognition Element (MRE). Different splice variants encode BK isoforms with different alcohol sensitivities. Computational modeling indicates that this miR-9-dependent mechanism contributes to alcohol tolerance. Moreover, this mechanism can be extended to include regulation of additional miR-9 targets relevant to alcohol abuse. Our results describe a mechanism of multiplex regulation of stability of alternatively spliced mRNA by microRNA in drug adaptation and neuronal plasticity.


Subject(s)
Adaptation, Physiological/drug effects , Ethanol/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/metabolism , MicroRNAs/metabolism , Neurons/physiology , Protein Processing, Post-Translational/physiology , RNA Splicing/physiology , RNA Stability/physiology , Adaptation, Physiological/genetics , Animals , Animals, Newborn , Cell Line , Cells, Cultured , Humans , Large-Conductance Calcium-Activated Potassium Channels/genetics , MicroRNAs/genetics , Neurons/drug effects , Protein Processing, Post-Translational/drug effects , Rats , Rats, Sprague-Dawley
11.
Exp Physiol ; 93(7): 908-18, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18403443

ABSTRACT

Large-conductance, calcium-activated potassium (BK(Ca)) channels are regulated by voltage and near-membrane calcium concentrations and are determinants of membrane potential and excitability in airway smooth muscle cells. Since the T helper-2 (Th2) cytokine, interleukin (IL)-4, is an important mediator of airway inflammation, we investigated whether IL-4 rapidly regulated BK(Ca) activity in normal airway smooth muscle cells. On-cell voltage clamp recordings were made on subconfluent, cultured human bronchial smooth muscle cells (HBSMC). Interleukin-4 (50 ng ml(-1)), IL-13 (50 ng ml(-1)) or histamine (10 microm) was added to the bath during the recordings. Immunofluorescence studies with selective antibodies against the alpha and beta1 subunits of BK(Ca) were also performed. Both approaches demonstrated that HBSMC membranes contained large-conductance channels (>200 pS) with both calcium and voltage sensitivity, all of which is characteristic of the BK(Ca) channel. Histamine caused a rapid increase in channel activity, as expected. A new finding was that perfusion with IL-4 stimulated rapid, large increases in BK(Ca) channel activity (77.2 +/- 63.3-fold increase, P < 0.05, n = 18). This large potentiation depended on the presence of external calcium. In contrast, IL-13 (50 ng ml(-1)) had little effect on BK(Ca) channel activity, but inhibited the effect of IL-4. Thus, HBSMC contain functional BK(Ca) channels whose activity is rapidly potentiated by the cytokine, IL-4, but not by IL-13. These findings are consistent with a model in which IL-4 rapidly increases near-membrane calcium concentrations to regulate BK(Ca) activity.


Subject(s)
Interleukin-4/physiology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Muscle, Smooth/physiology , Bronchi/cytology , Bronchi/physiology , Calcium/metabolism , Cells, Cultured , Histamine/physiology , Humans , Interleukin-13/physiology , Membrane Potentials , Muscle, Smooth/cytology , Patch-Clamp Techniques
12.
J Biol Chem ; 283(8): 5090-8, 2008 Feb 22.
Article in English | MEDLINE | ID: mdl-18084004

ABSTRACT

Ethanol tolerance, in which exposure leads to reduced sensitivity, is an important component of alcohol abuse and addiction. The molecular mechanisms underlying this process remain poorly understood. The BKCa channel plays a central role in the behavioral response to ethanol in Caenorhabditis elegans (Davies, A. G., Pierce-Shimomura, J. T., Kim, H., VanHoven, M. K., Thiele, T. R., Bonci, A., Bargmann, C. I., and McIntire, S. L. (2003) Cell 115, 655-666) and Drosophila (Cowmeadow, R. B., Krishnan, H. R., and Atkinson, N. S. (2005) Alcohol. Clin. Exp. Res. 29, 1777-1786) . In neurons, ethanol tolerance in BKCa channels has two components: a reduced number of membrane channels and decreased potentiation of the remaining channels (Pietrzykowski, A. Z., Martin, G. E., Puig, S. I., Knott, T. K., Lemos, J. R., and Treistman, S. N. (2004) J. Neurosci. 24, 8322-8332) . Here, heterologous expression coupled with planar bilayer techniques examines two additional aspects of tolerance in human BKCa channels. 1) Is acute tolerance observed in a single channel protein complex within a lipid environment reduced to only two lipids? 2) Does lipid bilayer composition affect the appearance of acute tolerance? We found that tolerance was observable in BKCa channels in membrane patches pulled from HEK cells and when they are placed into reconstituted 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphatidylethanolamine/1-palmitoyl-2-oleoyl-sn-glycero-3-phosphatidylserine membranes. Furthermore, altering bilayer thickness by incorporating the channel into lipid mixtures of 1,2-dioleoyl-3-phosphatidylethanolamine with phosphatidylcholines of increasing chain length, or with sphingomyelin, strongly affected the sensitivity of the channel, as well as the time course of the acute response. Ethanol sensitivity changed from a strong potentiation in thin bilayers to inhibition in thick sphingomyelin/1,2-dioleoyl-3-phosphatidylethanolamine bilayers. Thus, tolerance can be an intrinsic property of the channel protein-lipid complex, and bilayer thickness plays an important role in shaping the pattern of response to ethanol. As a consequence of these findings the protein-lipid complex should be treated as a unit when studying ethanol action.


Subject(s)
Alcohol-Related Disorders/metabolism , Central Nervous System Depressants/pharmacology , Drug Tolerance/physiology , Ethanol/pharmacology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Membrane Lipids/metabolism , Neurons/metabolism , Alcohol-Related Disorders/genetics , Animals , Behavior, Animal/drug effects , Caenorhabditis elegans , Cell Line , Central Nervous System Depressants/adverse effects , Drug Synergism , Ethanol/adverse effects , Humans , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics
13.
Alcohol Res Health ; 31(4): 298-309, 2008.
Article in English | MEDLINE | ID: mdl-23584007

ABSTRACT

Tolerance is defined as the diminished response to alcohol or other drugs over the course of repeated or prolonged exposure. This mechanism allows physiological processes to achieve stability in a constantly changing environment. The onset of tolerance may occur within minutes, during a single exposure to alcohol (i.e., acute tolerance), or over longer timeframes and with prolonged exposure to alcohol (i.e., rapid or chronic tolerance). Changes in tolerance induced by alcohol may affect several processes at the molecular, cellular, or behavioral level. These effects often are interrelated and may be difficult to separate. This article describes changes at the molecular level that are related to the onset of acute, rapid, or chronic tolerance. It focuses on neuronal membrane-bound channels and the factors that affect their function and production, such as modification of protein synthesis and activity, interaction with the membrane lipid microenvironment, epigenetic effects on cytoplasmic regulation, and gene transcription. Also considered is the genetics of tolerance.


Subject(s)
Alcohol Drinking/genetics , Alcohol Drinking/metabolism , Drug Tolerance/physiology , Ethanol/administration & dosage , Membrane Lipids/genetics , Membrane Proteins/genetics , Animals , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , Humans , Membrane Lipids/metabolism , Membrane Proteins/metabolism
14.
Endocrinology ; 147(5): 2567-74, 2006 May.
Article in English | MEDLINE | ID: mdl-16469804

ABSTRACT

Two well-characterized nongenomic actions of thyroid hormone in cultured brain tissues are: 1) regulation of type 2 iodothyronine 5'deiodinase (D2) activity and 2) regulation of actin polymerization. In particular, the latter is likely to have profound effects on neuronal migration in the developing brain. In this study, we determined whether these nongenomic actions also occurred in vivo during brain development. Neonatal hypothyroidism was induced by propylthiouracil given to pregnant dams beginning on d17 of gestation and continued throughout the neonatal period. On postnatal d 14, rats were injected with either cold or [(125)I]-labeled iodothyronines and killed sequentially after injection. In contrast to reports in the adult rat, all three iodothyronines readily and equally entered developing brain tissues. As expected, cerebrocortical D2 activity was markedly elevated in the hypothyroid brain and both reverse T(3) (rT(3)) and T(4) rapidly decreased D2 to euthyroid levels within 3 h. Furthermore, cerebellar G-actin content in the hypothyroid rat was approximately 5-fold higher than in the euthyroid rat. Again, both rT(3) and T(4) rapidly decreased the G-actin content by approximately 50%, with a reciprocal increase in F-actin content to euthyroid levels without altering total actin. Neither T(3) nor vehicle had any effect on D2 activity in the cortex or G- or F-actin content in the cerebellum. The thyroid hormone-dependent regulation of actin polymerization in the rat brain provides a mechanism by which this morphogenic hormone can influence neuronal migration independent of the need for altered gene transcription. Furthermore, these data suggest a prominent role for rT(3) during brain development.


Subject(s)
Brain/metabolism , Gene Expression Regulation, Developmental , Iodide Peroxidase/metabolism , Thyroid Hormones/metabolism , Actins/metabolism , Animals , Brain/embryology , Cell Movement , Cerebellum/metabolism , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Female , Neurons/metabolism , Rats , Time Factors , Iodothyronine Deiodinase Type II
15.
Alcohol Clin Exp Res ; 30(2): 222-32, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16441271

ABSTRACT

This article presents the proceedings of a symposium entitled "The Tipsy Terminal: Presynaptic Effects of Ethanol" (held at the annual meeting of the Research Society on Alcoholism, in Santa Barbara, CA, June 27, 2005). The objective of this symposium was to focus on a cellular site of ethanol action underrepresented in the alcohol literature, but quickly becoming a "hot" topic. The chairs of the session were Marisa Roberto and George Robert Siggins. Our speakers were chosen on the basis of the diverse electrophysiological and other methods used to discern the effects of acute and chronic ethanol on presynaptic terminals and on the basis of significant insights that their data provide for understanding ethanol actions on neurons in general, as mechanisms underlying problematic behavioral effects of alcohol. The 5 presenters drew from their recent studies examining the effects of acute and chronic ethanol using a range of sophisticated methods from electrophysiological analysis of paired-pulse facilitation and spontaneous and miniature synaptic currents (Drs. Weiner, Valenzuela, Zhu, and Morrisett), to direct recording of ion channel activity and peptide release from acutely isolated synaptic terminals (Dr. Treistman), to direct microscopic observation of vesicular release (Dr. Morrisett). They showed that ethanol administration could both increase and decrease the probability of release of different transmitters from synaptic terminals. The effects of ethanol on synaptic terminals could often be correlated with important behavioral or developmental actions of alcohol. These and other novel findings suggest that future analyses of synaptic effects of ethanol should attempt to ascertain, in multiple brain regions, the role of presynaptic terminals, relevant presynaptic receptors and signal transduction linkages, exocytotic mechanisms, and their involvement in alcohol's behavioral actions. Such studies could lead to new treatment strategies for alcohol intoxication, alcohol abuse, and alcoholism.


Subject(s)
Alcoholic Intoxication/physiopathology , Alcoholism/physiopathology , Brain/drug effects , Receptors, Presynaptic/drug effects , Animals , Brain/physiopathology , Glutamic Acid/metabolism , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neuropeptides/metabolism , Receptors, Presynaptic/physiology , Synaptic Vesicles/drug effects , Synaptic Vesicles/physiology , gamma-Aminobutyric Acid/metabolism
16.
Brain Res Dev Brain Res ; 154(1): 121-35, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15617761

ABSTRACT

The timing of granule cell migration in the developing cerebellum is regulated by thyroid hormone. Granule cell migration depends on the recognition of extracellular neuronal guidance molecule(s), such as laminin, and this, in turn, requires cell surface adhesion molecules (integrins) that are anchored on the cell membrane by the actin cytoskeleton. While many of the actions of thyroid hormone, specifically 3,5,3'-triiodothyronine (T3), are mediated by regulated gene expression, both thyroxine (T4) and 3,3',5'-triiodothyronine (rT3) also exert direct, positive control of the quantity of polymerized actin in cultured astrocytes without affecting gene expression. T4-dependent actin polymerization has been shown to (i) participate in the immobilization of laminin to the cell surface, (ii) help deposit laminin in the molecular layer of the developing cerebellum, and (iii) anchor integrin(s) that recognize laminin present in the extracellular matrix. In this study, we show that both T4 and rT3, but not T3, directly regulate the F-actin content of elongating neurites of cerebellar neurons. T4 and rT3 also promoted extensive granule cell migration from cerebellar explants, as well as, dense cell clustering and extensive neuronal process formation when granule cells were grown on a laminin-coated surface. Both granule cell migration and neuronal process outgrowth were markedly attenuated by the addition of integrin-blocking antibodies or binding peptides, by the absence of thyroid hormone or the presence of T3. These data suggest that the T4-dependent actin polymerization in developing neurons is necessary for these migrating cells to recognize the laminin guidance molecule, thereby providing a novel molecular mechanism for the profound influence of thyroid hormone on brain development that is independent of regulated gene expression.


Subject(s)
Cell Differentiation/physiology , Cell Movement/physiology , Cerebellum/cytology , Neurites/metabolism , Thyroxine/metabolism , Triiodothyronine, Reverse/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Actins/drug effects , Actins/metabolism , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Movement/drug effects , Cells, Cultured , Integrins/antagonists & inhibitors , Integrins/metabolism , Laminin/metabolism , Laminin/pharmacology , Neurites/drug effects , Rats , Thyroid Gland/metabolism , Thyroxine/pharmacology , Triiodothyronine, Reverse/pharmacology
17.
J Neurosci ; 24(38): 8322-32, 2004 Sep 22.
Article in English | MEDLINE | ID: mdl-15385615

ABSTRACT

Tolerance is an important element of drug addiction and provides a model for understanding neuronal plasticity. The hypothalamic-neurohypophysial system (HNS) is an established preparation in which to study the actions of alcohol. Acute application of alcohol to the rat neurohypophysis potentiates large-conductance calcium-sensitive potassium channels (BK), contributing to inhibition of hormone secretion. A cultured HNS explant from adult rat was used to explore the molecular mechanisms of BK tolerance after prolonged alcohol exposure. Ethanol tolerance was intrinsic to the HNS and consisted of: (1) decreased BK potentiation by ethanol, complete within 12 min of exposure, and (2) decreased current density, which was not complete until 24 hr after exposure, indicating that the two components of tolerance represent distinct processes. Single-channel properties were not affected by chronic exposure, suggesting that decreased current density resulted from downregulation of functional channels in the membrane. Indeed, we observed decreased immunolabeling against the BK alpha-subunit on the surface of tolerant terminals. Analysis using confocal microscopy revealed a reduction of BK channel clustering, likely associated with the internalization of the channel.


Subject(s)
Drug Tolerance/physiology , Ethanol/pharmacology , Potassium Channels, Calcium-Activated/drug effects , Potassium Channels, Calcium-Activated/metabolism , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Alcoholism , Animals , Calcium/metabolism , Calcium/pharmacology , Drug Synergism , Hypothalamo-Hypophyseal System/cytology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Immunohistochemistry , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits , Large-Conductance Calcium-Activated Potassium Channels , Male , Neurons/drug effects , Neurons/metabolism , Neurons/ultrastructure , Organ Culture Techniques , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...