Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Betina González is active.

Publication


Featured researches published by Betina González.


PLOS ONE | 2012

Modafinil Abrogates Methamphetamine-Induced Neuroinflammation and Apoptotic Effects in the Mouse Striatum

Mariana Raineri; Betina González; Belén Goitia; Edgar Garcia-Rill; Irina N. Krasnova; Jean Lud Cadet; Francisco J. Urbano; Verónica Bisagno

Methamphetamine is a drug of abuse that can cause neurotoxic damage in humans and animals. Modafinil, a wake-promoting compound approved for the treatment of sleeping disorders, is being prescribed off label for the treatment of methamphetamine dependence. The aim of the present study was to investigate if modafinil could counteract methamphetamine-induced neuroinflammatory processes, which occur in conjunction with degeneration of dopaminergic terminals in the mouse striatum. We evaluated the effect of a toxic methamphetamine binge in female C57BL/6 mice (4×5 mg/kg, i.p., 2 h apart) and modafinil co-administration (2×90 mg/kg, i.p., 1 h before the first and fourth methamphetamine injections) on glial cells (microglia and astroglia). We also evaluated the striatal expression of the pro-apoptotic BAX and anti-apoptotic Bcl-2 proteins, which are known to mediate methamphetamine-induced apoptotic effects. Modafinil by itself did not cause reactive gliosis and counteracted methamphetamine-induced microglial and astroglial activation. Modafinil also counteracted the decrease in tyrosine hydroxylase and dopamine transporter levels and prevented methamphetamine-induced increases in the pro-apoptotic BAX and decreases in the anti-apoptotic Bcl-2 protein expression. Our results indicate that modafinil can interfere with methamphetamine actions and provide protection against dopamine toxicity, cell death, and neuroinflammation in the mouse striatum.


Addiction Biology | 2016

Methamphetamine blunts Ca2+ currents and excitatory synaptic transmission through D1/5 receptor-mediated mechanisms in the mouse medial prefrontal cortex

Betina González; Celeste Rivero-Echeto; Javier A. Muñiz; Jean Lud Cadet; Edgar Garcia-Rill; Francisco J. Urbano; Verónica Bisagno

Psychostimulant addiction is associated with dysfunctions in frontal cortex. Previous data demonstrated that repeated exposure to methamphetamine (METH) can alter prefrontal cortex (PFC)‐dependent functions. Here, we show that withdrawal from repetitive non‐contingent METH administration (7 days, 1 mg/kg) depressed voltage‐dependent calcium currents (ICa) and increased hyperpolarization‐activated cation current (IH) amplitude and the paired‐pulse ratio of evoked excitatory postsynaptic currents (EPSCs) in deep‐layer pyramidal mPFC neurons. Most of these effects were blocked by systemic co‐administration of the D1/D5 receptor antagonist SCH23390 (0.5 and 0.05 mg/kg). In vitro METH (i.e. bath‐applied to slices from naïve‐treated animals) was able to emulate its systemic effects on ICa and evoked EPSCs paired‐pulse ratio. We also provide evidence of altered mRNA expression of (1) voltage‐gated calcium channels P/Q‐type Cacna1a (Cav2.1), N‐type Cacna1b (Cav2.2), T‐type Cav3.1 Cacna1g, Cav3.2 Cacna1h, Cav3.3 Cacna1i and the auxiliary subunit Cacna2d1 (α2δ1); (2) hyperpolarization‐activated cyclic nucleotide‐gated channels Hcn1 and Hcn2; and (3) glutamate receptors subunits AMPA‐type Gria1, NMDA‐type Grin1 and metabotropic Grm1 in the mouse mPFC after repeated METH treatment. Moreover, we show that some of these changes in mRNA expression were sensitive D1/5 receptor blockade. Altogether, these altered mechanisms affecting synaptic physiology and transcriptional regulation may underlie PFC functional alterations that could lead to PFC impairments observed in METH‐addicted individuals.


Molecular Neurobiology | 2018

Methamphetamine Induces TET1- and TET3-Dependent DNA Hydroxymethylation of Crh and Avp Genes in the Rat Nucleus Accumbens

Subramaniam Jayanthi; Betina González; Michael T. McCoy; Bruce Ladenheim; Verónica Bisagno; Jean Lud Cadet

Methamphetamine (METH) addiction is a biopsychosocial disorder that is accompanied by multiple relapses even after prolonged abstinence, suggesting the possibilities of long-lasting maladaptive epigenetic changes in the brain. Here, we show that METH administration produced time-dependent increases in the expression of corticotropin-releasing hormone (Crh/Crf), arginine vasopressin (Avp), and cocaine- and amphetamine-regulated transcript prepropeptide (Cartpt) mRNAs in the rat nucleus accumbens (NAc). Chromatin immunoprecipitation (ChIP) assays revealed that METH increased the abundance of phosphorylated CREB (pCREB) at the promoter of Cartpt but not at Avp or Crh DNA sequences. In contrast, METH produced DNA hypomethylation at sites near the Crh transcription start site (TSS) and at intragenic Avp sequences. METH also increased DNA hydroxymethylation at the Crh TSS and at intragenic Avp sites. In addition, METH increased the protein expression of ten-eleven-translocation enzymes that catalyze DNA hydroxymethylation. Importantly, METH increased TET1 binding at the Crh promoter and increased TET3 binding at Avp intragenic regions. We further tested the role of TET enzymes in METH-induced changes in gene expression by using the TET inhibitor, 1,5-isoquinolinediol (IQD), and found that IQD blocked METH-induced increases in Crh and Avp mRNA expression. Together, these results indicate that METH produced changes in neuropeptide transcription by both activation of the cAMP/CREB pathway and stimulation of TET-dependent DNA hydroxymethylation. These results provide molecular evidence for epigenetic controls of METH-induced changes in the expression of neuropeptides.


PLOS ONE | 2015

Psychostimulant-Induced Testicular Toxicity in Mice: Evidence of Cocaine and Caffeine Effects on the Local Dopaminergic System

Candela Rocío González; Betina González; María E. Matzkin; Javier A. Muñiz; Jean Lud Cadet; Edgar Garcia-Rill; Francisco J. Urbano; Alfredo Daniel Vitullo; Verónica Bisagno

Several organ systems can be affected by psychostimulant toxicity. However, there is not sufficient evidence about the impact of psychostimulant intake on testicular physiology and catecholaminergic systems. The aim of the present study was to further explore potential toxic consequences of chronic exposure to cocaine, caffeine, and their combination on testicular physiology. Mice were injected with a 13-day chronic binge regimen of caffeine (3x5mg/kg), cocaine (3×10mg/kg), or combined administration. Mice treated with cocaine alone or combined with caffeine showed reduced volume of the seminiferous tubule associated to a reduction in the number of spermatogonia. Cocaine-only and combined treatments induced increased lipid peroxidation evaluated by TBARS assay and decreased glutathione peroxidase mRNA expression. Importantly, caffeine-cocaine combination potentiated the cocaine-induced germ cell loss, and induced pro-apoptotic BAX protein expression and diminished adenosine receptor A1 mRNA levels. We analyzed markers of dopaminergic function in the testis and detected the presence of tyrosine hydroxylase (TH) in the cytoplasm of androgen-producing Leydig cells, but also in meiotic germs cells within seminiferous tubules. Moreover, using transgenic BAC-Drd1a-tdTomato and D2R-eGFP mice, we report for the first time the presence of dopamine receptors (DRs) D1 and D2 in testicular mouse Leydig cells. Interestingly, the presence of DRD1 was also detected in the spermatogonia nearest the basal lamina of the seminiferous tubules, which did not show TH staining. We observed that psychostimulants induced downregulation of DRs mRNA expression and upregulation of TH protein expression in the testis. These findings suggest a potential role of the local dopaminergic system in psychostimulant-induced testicular pathology.


Progress in Neuro-psychopharmacology & Biological Psychiatry | 2017

Repeated methamphetamine and modafinil induce differential cognitive effects and specific histone acetylation and DNA methylation profiles in the mouse medial prefrontal cortex.

Betina González; Subramaniam Jayanthi; Natalia Gomez; Oscar V. Torres; Máximo H. Sosa; Alejandra Bernardi; Francisco J. Urbano; Edgar Garcia-Rill; Jean-Lud Cadet; Verónica Bisagno

&NA; Methamphetamine (METH) and modafinil are psychostimulants with different long‐term cognitive profiles: METH is addictive and leads to cognitive decline, whereas modafinil has little abuse liability and is a cognitive enhancer. Increasing evidence implicates epigenetic mechanisms of gene regulation behind the lasting changes that drugs of abuse and other psychotropic compounds induce in the brain, like the control of gene expression by histones 3 and 4 tails acetylation (H3ac and H4ac) and DNA cytosine methylation (5‐mC). Mice were treated with a seven‐day repeated METH, modafinil or vehicle protocol and evaluated in the novel object recognition (NOR) test or sacrificed 4 days after last injection for molecular assays. We evaluated total H3ac, H4ac and 5‐mC levels in the medial prefrontal cortex (mPFC), H3ac and H4ac promotor enrichment (ChIP) and mRNA expression (RT‐PCR) of neurotransmitter systems involved in arousal, wakefulness and cognitive control, like dopaminergic (Drd1 and Drd2), &agr;‐adrenergic (Adra1a and Adra1b), orexinergic (Hcrtr1 and Hcrtr2), histaminergic (Hrh1 and Hrh3) and glutamatergic (AMPA Gria1 and NMDA Grin1) receptors. Repeated METH and modafinil treatment elicited different cognitive outcomes in the NOR test, where modafinil‐treated mice performed as controls and METH‐treated mice showed impaired recognition memory. METH‐treated mice also showed i) decreased levels of total H3ac and H4ac, and increased levels of 5‐mC, ii) decreased H3ac enrichment at promoters of Drd2, Hcrtr1/2, Hrh1 and Grin1, and increased H4ac enrichment at Drd1, Hrh1 and Grin1, iii) increased mRNA of Drd1a, Grin1 and Gria1. Modafinil‐treated mice shared none of these effects and showed increased H3ac enrichment and mRNA expression at Adra1b. Modafinil and METH showed similar effects linked to decreased H3ac in Hrh3, increased H4ac in Hcrtr1, and decreased mRNA expression of Hcrtr2. The specific METH‐induced epigenetic and transcriptional changes described here may be related to the long‐term cognitive decline effects of the drug and its detrimental effects on mPFC function. The lack of similar epigenetic effects of chronic modafinil administration supports this notion. HighlightsRepeated METH administration impairs object recognition memory whereas repeated modafinil does not.Only METH decreased total H3ac and H4ac and increased total DNA methylation in the mPFC.Only METH altered H3ac and H4ac at specific DA, orexin, histamine and glutamate receptors promoters and/or mRNA in mPFC.Only modafinil increased promoter H3ac and mRNA levels of alpha(1B)adrenoreceptor in mPFC.METH‐specific effects may be related to mPFC malfunction and long‐term cognitive decline.


Frontiers in Behavioral Neuroscience | 2017

Cocaine and Caffeine Effects on the Conditioned Place Preference Test: Concomitant Changes on Early Genes within the Mouse Prefrontal Cortex and Nucleus Accumbens

Javier A. Muñiz; José Pedro Prieto; Betina González; Máximo H. Sosa; Jean Lud Cadet; Cecilia Scorza; Francisco J. Urbano; Verónica Bisagno

Caffeine is the worlds most popular psychostimulant and is frequently used as an active adulterant in many illicit drugs including cocaine. Previous studies have shown that caffeine can potentiate the stimulant effects of cocaine and cocaine-induced drug seeking behavior. However, little is known about the effects of this drug combination on reward-related learning, a key process in the maintenance of addiction and vulnerability to relapse. The goal of the present study was thus to determine caffeine and cocaine combined effects on the Conditioned Place Preference (CPP) test and to determine potential differential mRNA expression in the Nucleus Accumbens (NAc) and medial prefrontal cortex (mPFC) of immediate-early genes (IEGs) as well as dopamine and adenosine receptor subunits. Mice were treated with caffeine (5 mg/kg, CAF), cocaine (10 mg/kg, COC), or their combination (caffeine 5 mg/kg + cocaine 10 mg/kg, CAF-COC) and trained in the CPP test or treated with repeated injections inside the home cage. NAc and mPFC tissues were dissected immediately after the CPP test, after a single conditioning session or following psychostimulant injection in the home cage for mRNA expression analysis. CAF-COC induced a marked change of preference to the drug conditioned side of the CPP and a significant increase in locomotion compared to COC. Gene expression analysis after CPP test revealed specific up-regulation in the CAF-COC group of Drd1a, cFos, and FosB in the NAc, and cFos, Egr1, and Npas4 in the mPFC. Importantly, none of these changes were observed when animals received same treatments in their home cage. With a single conditioning session, we found similar effects in both CAF and CAF-COC groups: increased Drd1a and decreased cFos in the NAc, and increased expression of Drd1a and Drd2, in the mPFC. Interestingly, we found that cFos and Npas4 gene expression were increased only in the mPFC of the CAF-COC. Our study provides evidence that caffeine acting as an adulterant could potentiate reward-associated memories elicited by cocaine. This is associated with specific changes in IEGs expression that were observed almost exclusively in mice that received the combination of both psychostimulants in the context of CPP memory encoding and retrieval. Our results highlight the potential relevance of caffeine in the maintenance of cocaine addiction which might be mediated by modifying neural plasticity mechanisms that strengthen learning of the association between drug and environment.


Progress in Neuro-psychopharmacology & Biological Psychiatry | 2019

The effects of single-dose injections of modafinil and methamphetamine on epigenetic and functional markers in the mouse medial prefrontal cortex: potential role of dopamine receptors

Betina González; Oscar V. Torres; Subramaniam Jayanthi; Natalia Gomez; Máximo H. Sosa; Alejandra Bernardi; Francisco J. Urbano; Edgar Garcia-Rill; Jean-Lud Cadet; Verónica Bisagno

Abstract METH use causes neuroadaptations that negatively impact the prefrontal cortex (PFC) leading to addiction and associated cognitive decline in animals and humans. In contrast, modafinil enhances cognition by increasing PFC function. Accumulated evidence indicates that psychostimulant drugs, including modafinil and METH, regulate gene expression via epigenetic modifications. In this study, we measured the effects of single‐dose injections of modafinil and METH on the protein levels of acetylated histone H3 (H3ac) and H4ac, deacetylases HDAC1 and HDAC2, and of the NMDA subunit GluN1 in the medial PFC (mPFC) of mice euthanized 1 h after drug administration. To test if dopamine (DA) receptors (DRs) participate in the biochemical effects of the two drugs, we injected the D1Rs antagonist, SCH23390, or the D2Rs antagonist, raclopride, 30 min before administration of METH and modafinil. We evaluated each drug effect on glutamate synaptic transmission in D1R‐expressing layer V pyramidal neurons. We also measured the enrichment of H3ac and H4ac at the promoters of several genes including DA, NE, orexin, histamine, and glutamate receptors, and their mRNA expression, since they are responsive to chronic modafinil and METH treatment. Acute modafinil and METH injections caused similar effects on total histone acetylation, increasing H3ac and decreasing H4ac, and they also increased HDAC1, HDAC2 and GluN1 protein levels in the mouse mPFC. In addition, the effects of the drugs were prevented by pre‐treatment with D1Rs and D2Rs antagonists. Specifically, the changes in H4ac, HDAC2, and GluN1 were responsive to SCH23390, whereas those of H3ac and GluN1 were responsive to raclopride. Whole‐cell patch clamp in transgenic BAC‐Drd1a‐tdTomato mice showed that METH, but not modafinil, induced paired‐pulse facilitation of EPSCs, suggesting reduced presynaptic probability of glutamate release onto layer V pyramidal neurons. Analysis of histone 3/4 enrichment at specific promoters revealed: i) distinct effects of the drugs on histone 3 acetylation, with modafinil increasing H3ac at Drd1 and Adra1b promoters, but METH increasing H3ac at Adra1a; ii) distinct effects on histone 4 acetylation enrichment, with modafinil increasing H4ac at the Drd2 promoter and decreasing it at Hrh1, but METH increasing H4ac at Drd1; iii) comparable effects of both psychostimulants, increasing H3ac at Drd2, Hcrtr1, and Hrh1 promoters, decreasing H3ac at Hrh3, increasing H4ac at Hcrtr1, and decreasing H4ac at Hcrtr2, Hrh3, and Grin1 promoters. Interestingly, only METH altered mRNA levels of genes with altered histone acetylation status, inducing increased expression of Drd1a, Adra1a, Hcrtr1, and Hrh1, and decreasing Grin1. Our study suggests that although acute METH and modafinil can both increase DA neurotransmission in the mPFC, there are similar and contrasting epigenetic and transcriptional consequences that may account for their divergent clinical effects. HighlightsBoth Modafinil and METH increased H3ac and decreased H4ac in the mPFC.Histone 3 and 4 acetylation appear to be regulated by different DRs activation.H3ac status was restored by D2R antagonist but H4ac was sensitive to D1R antagonist.Only METH increased glutamate PPR in D1R‐expressing layer V pyramidal neurons.Modafinil and METH altered H3/H4ac at specific receptors promoters.


Sleep Science | 2015

Pedunculopontine arousal system physiology-Effects of psychostimulant abuse.

Francisco J. Urbano; Verónica Bisagno; Betina González; María Celeste Rivero-Echeto; Javier A. Muñiz; Brennon Luster; Stasia D’Onofrio; Susan Mahaffey; Edgar Garcia-Rill

This review describes the interactions between the pedunculopontine nucleus (PPN), the ventral tegmental area (VTA), and the thalamocortical system. Experiments using modulators of cholinergic receptors in the PPN clarified its role on psychostimulant-induced locomotion. PPN activation was found to be involved in the animal’s voluntary search for psychostimulants. Every PPN neuron is known to generate gamma band oscillations. Voltage-gated calcium channels are key elements in the generation and maintenance of gamma band activity of PPN neurons. Calcium channels are also key elements mediating psychostimulant-induced alterations in the thalamic targets of PPN output. Thus, the PPN is a key substrate for maintaining arousal and REM sleep, but also in modulating psychostimulant self-administration.


Neuropharmacology | 2014

Modafinil improves methamphetamine-induced object recognition deficits and restores prefrontal cortex ERK signaling in mice.

Betina González; Mariana Raineri; Jean Lud Cadet; Edgar Garcia-Rill; Francisco J. Urbano; Verónica Bisagno


Pharmacological Research | 2016

Cognitive enhancers versus addictive psychostimulants: The good and bad side of dopamine on prefrontal cortical circuits

Verónica Bisagno; Betina González; Francisco J. Urbano

Collaboration


Dive into the Betina González's collaboration.

Top Co-Authors

Avatar

Verónica Bisagno

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Edgar Garcia-Rill

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Jean Lud Cadet

National Institute on Drug Abuse

View shared research outputs
Top Co-Authors

Avatar

Javier A. Muñiz

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Candela Rocío González

Instituto de Biología y Medicina Experimental

View shared research outputs
Top Co-Authors

Avatar

Máximo H. Sosa

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Ricardo S. Calandra

Instituto de Biología y Medicina Experimental

View shared research outputs
Top Co-Authors

Avatar

Mariana Raineri

National Scientific and Technical Research Council

View shared research outputs
Top Co-Authors

Avatar

Subramaniam Jayanthi

National Institute on Drug Abuse

View shared research outputs
Researchain Logo
Decentralizing Knowledge