Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Int J Mol Sci ; 22(1)2020 Dec 29.
Article in English | MEDLINE | ID: mdl-33383772

ABSTRACT

The mitochondrial translocator protein (TSPO) has been shown to bind cholesterol with high affinity and is involved in mediating its availability for steroidogenesis. We recently reported that targeted Tspo gene deletion in MA-10 mouse tumor Leydig cells resulted in reduced cAMP-stimulated steroid formation and significant reduction in the mitochondrial membrane potential (ΔΨm) compared to control cells. We hypothesized that ΔΨm reduction in the absence of TSPO probably reflects the dysregulation and/or maintenance failure of some basic mitochondrial function(s). To explore the consequences of TSPO depletion via CRISPR-Cas9-mediated deletion (indel) mutation in MA-10 cells, we assessed the transcriptome changes in TSPO-mutant versus wild-type (Wt) cells using RNA-seq. Gene expression profiles were validated using real-time PCR. We report herein that there are significant changes in nuclear gene expression in Tspo mutant versus Wt cells. The identified transcriptome changes were mapped to several signaling pathways including the regulation of membrane potential, calcium signaling, extracellular matrix, and phagocytosis. This is a retrograde signaling pathway from the mitochondria to the nucleus and is probably the result of changes in expression of several transcription factors, including key members of the NF-κB pathway. In conclusion, TSPO regulates nuclear gene expression through intracellular signaling. This is the first evidence of a compensatory response to the loss of TSPO with transcriptome changes at the cellular level.


Subject(s)
Leydig Cells/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Receptors, GABA/deficiency , Sertoli-Leydig Cell Tumor/etiology , Sertoli-Leydig Cell Tumor/metabolism , Signal Transduction , Animals , CRISPR-Cas Systems , Cell Line, Tumor , Chromosome Mapping , Extracellular Matrix/metabolism , Gene Editing , Gene Expression Profiling , Gene Expression Regulation , INDEL Mutation , Male , Mice , NF-kappa B/metabolism , Sertoli-Leydig Cell Tumor/pathology , Transcriptome
2.
Biochem J ; 475(1): 75-85, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29127254

ABSTRACT

The translocator protein (TSPO) has been proposed to act as a key component in a complex important for mitochondrial cholesterol importation, which is the rate-limiting step in steroid hormone synthesis. However, TSPO function in steroidogenesis has recently been challenged by the development of TSPO knockout (TSPO-KO) mice, as they exhibit normal baseline gonadal testosterone and adrenal corticosteroid production. Here, we demonstrate that despite normal androgen levels in young male TSPO-KO mice, TSPO deficiency alters steroidogenic flux and results in reduced total steroidogenic output. Specific reductions in the levels of progesterone and corticosterone as well as age-dependent androgen deficiency were observed in both young and aged male TSPO-KO mice. Collectively, these findings indicate that while TSPO is not critical for achieving baseline testicular and adrenal steroidogenesis, either indirect effects of TSPO on steroidogenic processes, or compensatory mechanisms and functional redundancy, lead to subtle steroidogenic abnormalities which become exacerbated with aging.


Subject(s)
Adrenal Glands/metabolism , Aging/genetics , Gene Expression Regulation, Developmental , Receptors, GABA/genetics , Testis/metabolism , Adrenal Glands/growth & development , Aging/metabolism , Aldosterone/biosynthesis , Androgens/biosynthesis , Animals , Corticosterone/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Progesterone/biosynthesis , Receptors, GABA/deficiency , Testis/growth & development
3.
Int J Mol Sci ; 20(13)2019 Jul 09.
Article in English | MEDLINE | ID: mdl-31323920

ABSTRACT

The 18 kDa translocator protein (TSPO) is an evolutionary conserved cholesterol binding protein localized in the outer mitochondrial membrane. It has been implicated in the regulation of various cellular processes including oxidative stress, proliferation, apoptosis, and steroid hormone biosynthesis. Since the expression of TSPO in activated microglia is upregulated in various neuroinflammatory and neurodegenerative disorders, we set out to examine the role of TSPO in an immortalized human microglia C20 cell line. To this end, we performed a dual approach and used (i) lentiviral shRNA silencing to reduce TSPO expression, and (ii) the CRISPR/Cas9 technology to generate complete TSPO knockout microglia cell lines. Functional characterization of control and TSPO knockdown as well as knockout cells, revealed only low de novo steroidogenesis in C20 cells, which was not dependent on the level of TSPO expression or influenced by the treatment with TSPO-specific ligands. In contrast to TSPO knockdown C20 cells, which did not show altered mitochondrial function, the TSPO deficient knockout cells displayed a significantly decreased mitochondrial membrane potential and cytosolic Ca2+ levels, as well as reduced respiratory function. Performing the rescue experiment by lentiviral overexpression of TSPO in knockout cells, increased oxygen consumption and restored respiratory function. Our study provides further evidence for a significant role of TSPO in cellular and mitochondrial metabolism and demonstrates that different phenotypes of mitochondrial function are dependent on the level of TSPO expression.


Subject(s)
CRISPR-Cas Systems/physiology , Microglia/metabolism , Receptors, GABA/metabolism , CRISPR-Cas Systems/genetics , Calcium/metabolism , Cell Line , Cells, Cultured , Humans , Membrane Potential, Mitochondrial/physiology , Oxidative Phosphorylation , Receptors, GABA/deficiency , Receptors, GABA/genetics , Steroids/metabolism
4.
Nature ; 468(7321): 305-9, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-21048709

ABSTRACT

Stroke is a leading cause of disability, but no pharmacological therapy is currently available for promoting recovery. The brain region adjacent to stroke damage-the peri-infarct zone-is critical for rehabilitation, as it shows heightened neuroplasticity, allowing sensorimotor functions to re-map from damaged areas. Thus, understanding the neuronal properties constraining this plasticity is important for the development of new treatments. Here we show that after a stroke in mice, tonic neuronal inhibition is increased in the peri-infarct zone. This increased tonic inhibition is mediated by extrasynaptic GABA(A) receptors and is caused by an impairment in GABA (γ-aminobutyric acid) transporter (GAT-3/GAT-4) function. To counteract the heightened inhibition, we administered in vivo a benzodiazepine inverse agonist specific for α5-subunit-containing extrasynaptic GABA(A) receptors at a delay after stroke. This treatment produced an early and sustained recovery of motor function. Genetically lowering the number of α5- or δ-subunit-containing GABA(A) receptors responsible for tonic inhibition also proved beneficial for recovery after stroke, consistent with the therapeutic potential of diminishing extrasynaptic GABA(A) receptor function. Together, our results identify new pharmacological targets and provide the rationale for a novel strategy to promote recovery after stroke and possibly other brain injuries.


Subject(s)
Motor Cortex/physiology , Motor Cortex/physiopathology , Recovery of Function/physiology , Stroke/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Benzodiazepines/pharmacology , Cerebral Infarction/metabolism , Cerebral Infarction/pathology , Cerebral Infarction/physiopathology , Disease Models, Animal , Drug Inverse Agonism , GABA Antagonists/pharmacology , GABA Plasma Membrane Transport Proteins/metabolism , Imidazoles/pharmacology , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Motor Cortex/metabolism , Motor Cortex/pathology , Neuronal Plasticity/physiology , Receptors, GABA/deficiency , Receptors, GABA/genetics , Receptors, GABA/metabolism , Stroke/drug therapy , Stroke/pathology , Synapses/metabolism , Time Factors
5.
J Biol Chem ; 289(20): 13769-81, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24692541

ABSTRACT

Translocator protein of 18 kDa (TSPO) is a highly conserved, ubiquitous protein localized in the outer mitochondrial membrane, where it is thought to play a key role in the mitochondrial transport of cholesterol, a key step in the generation of steroid hormones. However, it was first characterized as the peripheral benzodiazepine receptor because it appears to be responsible for high affinity binding of a number of benzodiazepines to non-neuronal tissues. Ensuing studies have employed natural and synthetic ligands to assess the role of TSPO function in a number of natural and pathological circumstances. Largely through the use of these compounds and biochemical associations, TSPO has been proposed to play a role in the mitochondrial permeability transition pore (PTP), which has been associated with cell death in many human pathological conditions. Here, we critically assess the role of TSPO in the function of the PTP through the generation of mice in which the Tspo gene has been conditionally eliminated. Our results show that 1) TSPO plays no role in the regulation or structure of the PTP, 2) endogenous and synthetic ligands of TSPO do not regulate PTP activity through TSPO, 3) outer mitochondrial membrane regulation of PTP activity occurs though a mechanism that does not require TSPO, and 4) hearts lacking TSPO are as sensitive to ischemia-reperfusion injury as hearts from control mice. These results call into question a wide variety of studies implicating TSPO in a number of pathological processes through its actions on the PTP.


Subject(s)
Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/metabolism , Receptors, GABA/metabolism , Animals , Female , Gene Deletion , Liver/cytology , Liver/metabolism , Male , Mice , Mitochondrial Permeability Transition Pore , Myocardium/cytology , Myocardium/metabolism , Permeability , Receptors, GABA/deficiency , Receptors, GABA/genetics
6.
J Neurochem ; 131(5): 566-72, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25113583

ABSTRACT

Serotonin (5-HT)2C receptors play a role in psychoaffective disorders and often contribute to the antidepressant and anxiolytic effects of psychotropic drugs. During stress, activation of these receptors exerts a negative feedback on 5-HT release, probably by increasing the activity of GABAergic interneurons. However, to date, the GABA receptor types that mediate the 5-HT2C receptor-induced feedback inhibition are still unknown. To address this question, we assessed the inhibition of 5-HT turnover by a 5-HT2C receptor agonist (RO 60-0175) at the hippocampal level and under conditions of stress, after pharmacological or genetic inactivation of either GABA-A or GABA-B receptors in mice. Neither the GABA-B receptor antagonist phaclofen nor the specific genetic ablation of either GABA-B1a or GABA-B1b subunits altered the inhibitory effect of RO 60-0175, although 5-HT turnover was markedly decreased in GABA-B1a knock-out mice in both basal and stress conditions. In contrast, the 5-HT2C receptor-mediated inhibition of 5-HT turnover was reduced by the GABA-A receptor antagonist bicuculline. However, a significant effect of 5-HT2C receptor activation persisted in mutant mice deficient in the α3 subunit of GABA-A receptors. It can be inferred that non-α3 subunit-containing GABA-A receptors, but not GABA-B receptors, mediate the 5-HT2C -induced inhibition of stress-induced increase in hippocampal 5-HT turnover in mice.


Subject(s)
GABA Agents/pharmacology , Receptor, Serotonin, 5-HT2C/metabolism , Receptors, GABA/genetics , Stress, Psychological/genetics , Stress, Psychological/metabolism , Animals , Disease Models, Animal , Ethylamines/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hippocampus/drug effects , Hippocampus/metabolism , Hydroxyindoleacetic Acid/metabolism , Indoles/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, GABA/deficiency , Serotonin/metabolism , Stress, Psychological/drug therapy , Stress, Psychological/pathology
7.
Mol Psychiatry ; 16(4): 383-406, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21079608

ABSTRACT

Increasing evidence points to an association between major depressive disorders (MDDs) and diverse types of GABAergic deficits. In this review, we summarize clinical and preclinical evidence supporting a central and causal role of GABAergic deficits in the etiology of depressive disorders. Studies of depressed patients indicate that MDDs are accompanied by reduced brain concentration of the inhibitory neurotransmitter γ-aminobutyric acid (GABA) and by alterations in the subunit composition of the principal receptors (GABA(A) receptors) mediating GABAergic inhibition. In addition, there is abundant evidence that suggests that GABA has a prominent role in the brain control of stress, the most important vulnerability factor in mood disorders. Furthermore, preclinical evidence suggests that currently used antidepressant drugs (ADs) designed to alter monoaminergic transmission and nonpharmacological therapies may ultimately act to counteract GABAergic deficits. In particular, GABAergic transmission has an important role in the control of hippocampal neurogenesis and neural maturation, which are now established as cellular substrates of most if not all antidepressant therapies. Finally, comparatively modest deficits in GABAergic transmission in GABA(A) receptor-deficient mice are sufficient to cause behavioral, cognitive, neuroanatomical and neuroendocrine phenotypes, as well as AD response characteristics expected of an animal model of MDD. The GABAergic hypothesis of MDD suggests that alterations in GABAergic transmission represent fundamentally important aspects of the etiological sequelae of MDDs that are reversed by monoaminergic AD action.


Subject(s)
Depressive Disorder, Major/genetics , Genetic Predisposition to Disease , gamma-Aminobutyric Acid/deficiency , gamma-Aminobutyric Acid/genetics , Animals , Antidepressive Agents/therapeutic use , Brain/drug effects , Brain/metabolism , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/pathology , Disease Models, Animal , Gene Expression Regulation/genetics , Humans , Mice , Mutation/genetics , Receptors, GABA/deficiency , Receptors, GABA/genetics , Risk Factors
8.
Arch Ital Biol ; 149(4): 454-66, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22205591

ABSTRACT

REM sleep triggers a potent suppression of postural muscle tone - i.e., REM atonia. However, motor control during REM sleep is paradoxical because overall brain activity is maximal, but motor output is minimal. The skeletal motor system remains quiescent during REM sleep because somatic motoneurons are powerfully inactivated. Determining the mechanisms triggering loss of motoneuron function during REM sleep is important because breakdown in REM sleep motor control underlies sleep disorders such as REM sleep behavior disorder (RBD) and cataplexy/narcolepsy. For example, RBD is characterized by dramatic REM motor activation resulting in dream enactment and subsequent patient injury. In contrast, cataplexy a pathognomonic symptom of narcolepsy - is caused by the involuntary onset of REM-like atonia during wakefulness. This review highlights recent work from my laboratory that examines how motoneuron function is lost during normal REM sleep and it also identifies potential biochemical mechanisms underlying abnormal motor control in both RBD and cataplexy. First, I show that both GABAB and GABAA/glycine mediated inhibition of motoneurons is required for generating REM atonia. Next, I show that impaired GABA and glycine neurotransmission triggers the cardinal features of RBD in a transgenic mouse model. Last, I show that loss of an excitatory noradrenergic drive onto motoneurons is, at least in part, responsible for the loss of postural muscle tone during cataplexy in narcoleptic mice. Together, this research indicates that multiple transmitters systems are responsible for regulating postural muscle tone during REM sleep, RBD and cataplexy.


Subject(s)
Motor Neurons/physiology , Muscle Tonus/physiology , Narcolepsy/pathology , REM Sleep Behavior Disorder/pathology , Sleep, REM/physiology , Trigeminal Nuclei/cytology , Action Potentials/drug effects , Action Potentials/genetics , Animals , Humans , Mice , Mice, Transgenic , Narcolepsy/genetics , REM Sleep Behavior Disorder/genetics , Receptors, GABA/deficiency , Receptors, Glycine/deficiency , Trigeminal Nerve/cytology
9.
Biochemistry ; 49(23): 4766-78, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20469933

ABSTRACT

Translocator protein TSPO is an 18 kDa protein implicated in numerous cell functions and is highly expressed in secretory and glandular tissues, especially in steroidogenic cells. TSPO expression is altered in pathological conditions such as certain cancers and neurological diseases. In search of the factors regulating Tspo expression, we recently showed that high levels of TSPO in steroidogenic cells may be due to high constitutive expression of protein kinase Cepsilon (PKCepsilon), while phorbol 12-myristate 13-acetate (PMA) activation of PKCepsilon drives inducible TSPO expression in nonsteroidogenic cells, likely through activator protein 1 (AP1). In this study, we aimed to identify the signal transduction pathway through which PKCepsilon regulates Tspo gene expression. The MEK1/2 specific inhibitor U0126, but not NFkappaB inhibitors, reduced basal Tspo promoter activity in TSPO-rich steroidogenic cells (MA-10 Leydig), as well as basal and PMA-induced Tspo promoter levels in TSPO-poor nonsteroidogenic cells (NIH-3T3 fibroblasts). AP1 and signal transducer and activation of transcription 3 (STAT3) have binding sites in the Tspo promoter and are downstream targets of PKCepsilon and MAPK (Raf-1-ERK1/2) pathways. PKCepsilon overexpression induced STAT3 phosphorylation in NIH-3T3 cells, while PKCepsilon knockdown reduced STAT3 and c-Jun phosphorylation in Leydig cells. MEK1/2, ERK2, c-Jun, and STAT3 knockdown reduced Tspo mRNA and protein levels in Leydig cells. Additionally, Raf-1 reduced Tspo mRNA levels in the same cells. MEK1/2, c-Jun, and STAT3 knockdown also reduced basal as well as PMA-induced Tspo mRNA levels in NIH-3T3 cells. Together, these results demonstrate that PKCepsilon regulates Tspo gene expression through a MAPK (Raf-1-MEK1/2-ERK1/2) signal transduction pathway, acting at least in part through c-Jun and STAT3 transcription factors.


Subject(s)
Gene Expression Regulation, Enzymologic , MAP Kinase Signaling System/genetics , Protein Kinase C-epsilon/physiology , Proto-Oncogene Proteins c-jun/metabolism , Receptors, GABA/biosynthesis , Receptors, GABA/genetics , STAT3 Transcription Factor/metabolism , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Gene Targeting/methods , MAP Kinase Kinase 1/physiology , MAP Kinase Kinase 2/physiology , Mice , NIH 3T3 Cells , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-jun/physiology , Proto-Oncogene Proteins c-raf/physiology , Receptors, GABA/deficiency , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/physiology
10.
Mol Pharmacol ; 74(2): 443-53, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18492798

ABSTRACT

As a promiscuous xenobiotic sensor, the constitutive androstane receptor (CAR; NR1I3) regulates the expression of multiple drug-metabolizing enzymes and transporters in liver. The constitutively activated nature of CAR in the cell-based transfection assays has hindered its use as a predictor of metabolism-based drug-drug interactions. Here, we have identified 1-(2-chlorophenylmethylpropyl)-3-isoquinoline-carboxamide (PK11195), a typical peripheral benzodiazepine receptor (PBR) ligand, as a selective and potent inhibitor of human (h) CAR. In cell-based transfection assays, PK11195 inhibited the constitutive activity of hCAR more than 80% at the concentration of 10 microM, and the PK11195-inhibited activity was efficiently reactivated by the direct CAR activator, 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde-O-(3,4-dichlorobenzyl) oxime, but not by the indirect hCAR activator, phenobarbital. Mammalian two-hybrid and GST pull-down assays showed that PK11195 repressed the interactions of hCAR with the coactivators steroid receptor coactivator-1 and glucocorticoid receptor-interacting protein 1 to inhibit hCAR activity. The inhibition by PK11195 specifically occurred to the hCAR: PK1195 strongly activated human pregnane X receptor (PXR), whereas it did not alter the activity of the mouse CAR and mouse PXR. In addition, PBR played no role in the PK11195 inhibition of hCAR because the inhibition fully occurred in the HeLa cells in which the PBR was knocked down by small interfering RNA. In the Car(-/-) mouse liver, PK11195 translocated enhanced yellow fluorescent protein-hCAR into the nucleus. These results are consistent with the conclusion that PK11195 is a novel hCAR-specific antagonist that represses the CAR-coactivator interactions to inhibit the receptor activity inside the nucleus. Thus, PK11195 can be used as a chemical tool for studying the molecular basis of CAR function.


Subject(s)
Isoquinolines/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, GABA-A/metabolism , Receptors, GABA/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Animals , COS Cells , Cell Line, Tumor , Cells, Cultured , Chlorocebus aethiops , Constitutive Androstane Receptor , HeLa Cells , Humans , Isoquinolines/chemistry , Isoquinolines/pharmacology , Ligands , Liver/drug effects , Liver/enzymology , Liver/metabolism , Mice , Mice, Mutant Strains , Receptors, GABA/deficiency , Receptors, GABA/genetics
11.
Neuropharmacology ; 136(Pt A): 106-116, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29106983

ABSTRACT

Following the discovery of GABAB receptors by Norman Bowery and colleagues, cloning and biochemical efforts revealed that GABAB receptors assemble multi-subunit complexes composed of principal and auxiliary subunits. The principal receptor subunits GABAB1a, GABAB1b and GABAB2 form two heterodimeric GABAB(1a,2) and GABAB(1b,2) receptors that can associate with tetramers of auxiliary KCTD (K+ channel tetramerization domain) subunits. Experiments with subunit knock-out mice revealed that GABAB(1b,2) receptors activate slow inhibitory postsynaptic currents (sIPSCs) while GABAB(1a,2) receptors function as heteroreceptors and inhibit glutamate release. Both GABAB(1a,2) and GABAB(1b,2) receptors can serve as autoreceptors and inhibit GABA release. Auxiliary KCTD subunits regulate the duration of sIPSCs and scaffold effector channels at the receptor. GABAB receptors are well known to contribute to thalamic spindle oscillations. Spindles are generated through alternating burst-firing in reciprocally connected glutamatergic thalamocortical relay (TCR) and GABAergic thalamic reticular nucleus (TRN) neurons. The available data implicate postsynaptic GABAB receptors in TCR cells in the regulation of spindle frequency. We now used electrical or optogenetic activation of thalamic spindles and pharmacological experiments in acute slices of knock-out mice to study the impact of GABAB(1a,2) and GABAB(1b,2) receptors on spindle oscillations. We found that selectively GABAB(1a,2) heteroreceptors at TCR to TRN cell synapses regulate oscillation strength, while GABAB(1b,2) receptors control oscillation frequency. The auxiliary subunit KCTD16 influences both oscillation strength and frequency, supporting that KCTD16 regulates network activity through GABAB(1a,2) and GABAB(1b,2) receptors. This article is part of the "Special Issue Dedicated to Norman G. Bowery".


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, GABA-B/metabolism , Thalamus/metabolism , Animals , Glutamic Acid/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mice, Knockout , Nerve Tissue Proteins/genetics , Neurons/metabolism , Receptors, GABA/deficiency , Receptors, GABA/genetics , Receptors, GABA-B/genetics , Synapses/metabolism , Tissue Culture Techniques
12.
Sci Rep ; 8(1): 16213, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30385779

ABSTRACT

Heart failure (HF) is characterized by abnormal mitochondrial calcium (Ca2+) handling, energy failure and impaired mitophagy resulting in contractile dysfunction and myocyte death. We have previously shown that the 18-kDa mitochondrial translocator protein of the outer mitochondrial membrane (TSPO) can modulate mitochondrial Ca2+ uptake. Experiments were designed to test the role of the TSPO in a murine pressure-overload model of HF induced by transverse aortic constriction (TAC). Conditional, cardiac-specific TSPO knockout (KO) mice were generated using the Cre-loxP system. TSPO-KO and wild-type (WT) mice underwent TAC for 8 weeks. TAC-induced HF significantly increased TSPO expression in WT mice, associated with a marked reduction in systolic function, mitochondrial Ca2+ uptake, complex I activity and energetics. In contrast, TSPO-KO mice undergoing TAC had preserved ejection fraction, and exhibited fewer clinical signs of HF and fibrosis. Mitochondrial Ca2+ uptake and energetics were restored in TSPO KO mice, associated with decreased ROS, improved complex I activity and preserved mitophagy. Thus, HF increases TSPO expression, while preventing this increase limits the progression of HF, preserves ATP production and decreases oxidative stress, thereby preventing metabolic failure. These findings suggest that pharmacological interventions directed at TSPO may provide novel therapeutics to prevent or treat HF.


Subject(s)
Blood Pressure , Heart Failure/etiology , Heart Failure/physiopathology , Receptors, GABA/deficiency , Animals , Biomarkers , Calcium/metabolism , Cardiomegaly/etiology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Disease Models, Animal , Gene Expression Regulation , Heart Failure/pathology , Heart Function Tests , Mice , Mice, Knockout , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Ventricular Remodeling
13.
DNA Cell Biol ; 36(2): 103-108, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28004979

ABSTRACT

The mitochondrial membrane 18 kDa translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor, is constitutively expressed in most organs, most abundantly in hormonal tissue and cells of mononuclear phagocyte lineage, while in the brain, TSPO expression is induced in the wake of injury, inflammation, and neurodegeneration. Increased TSPO expression is also prominent in several cancerous tissues where it appears to correlate with the degree of malignancy. Currently, TSPO is thus actively investigated as a generic biomarker for disease activity and a therapeutic target for a wide range of diseases. In this study, we report a Jurkat human T cell leukemia cell line that has only trace expression of TSPO mRNA. Through the use of bisulphite genomic sequencing, we show that the Jurkat TSPO promoter is highly methylated except for CpG sites that are adjacent to the transcription start site. Control measurements in HEK-293, HeLa, and U87-MG cells with high TSPO mRNA expression showed low levels of TSPO promoter methylation. Demethylation with 5-aza-2'-deoxycytidine (5-aza-dC) caused a dose-dependent increase in TSPO mRNA with a corresponding demethylation of the TSPO promoter in Jurkat cells. Treating HeLa and U87-MG cells with 5-aza-dC caused no change in the level of TSPO mRNA. These observations confirm the epigenetic regulation of TSPO and suggest it to be a more common mechanism by which the differential expression of TSPO in various cell types and in health and disease may be explained.


Subject(s)
Epigenesis, Genetic , Gene Silencing , Leukemia, T-Cell/pathology , Receptors, GABA/deficiency , Receptors, GABA/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Base Sequence , Cell Line, Tumor , DNA Methylation/drug effects , DNA Methylation/genetics , Decitabine , Epigenesis, Genetic/drug effects , Gene Silencing/drug effects , Humans , Promoter Regions, Genetic/genetics
14.
Schizophr Res ; 171(1-3): 125-30, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26796540

ABSTRACT

BACKGROUND: Although altered gamma-aminobutyric acid (GABA) neurotransmission has been implicated in the pathophysiology of schizophrenia, it is unclear whether the influence of GABA on working memory processes is confounded by nicotine use in this population. It is therefore crucial to evaluate working memory and its underlying mechanisms in non-smokers with schizophrenia to eliminate the confounding effects of nicotine on behavior and neurophysiology. METHODS: In this cross-sectional study, working memory was assessed using the verbal N-back task, while GABAergic function was assessed through motor cortical inhibition using single and paired-pulse transcranial magnetic stimulation (TMS) to the left primary motor cortex in 11 non-smokers with schizophrenia and 13 non-smoker healthy subjects. RESULTS: Similar to previously published studies, working memory performance was significantly impaired in the 3-back condition in patients with schizophrenia compared to healthy subjects (p=0.036). In addition, GABAA receptor function was significantly reduced in schizophrenia as assessed by short interval cortical inhibition (SICI) (p=0.005). A positive correlation was found between GABAA inhibition and working memory performance on the 3-back task (r(23)=0.55, p=0.006), suggesting that greater GABAA inhibition is associated with better performance on tasks of working memory. CONCLUSIONS: Our findings highlight the role of GABAA receptor dysfunction in working memory and the pathophysiology of schizophrenia, and may represent a selective characteristic of schizophrenia. Moreover, these findings suggest a potential therapeutic role for targeting GABA receptor activity to improve cognition and quality of life in patients with schizophrenia.


Subject(s)
Memory Disorders/etiology , Memory, Short-Term/physiology , Neural Inhibition/physiology , Receptors, GABA/deficiency , Schizophrenia/complications , Adolescent , Adult , Analysis of Variance , Cross-Sectional Studies , Electromyography , Evoked Potentials, Motor/physiology , Female , Humans , Male , Middle Aged , Neuropsychological Tests , Psychiatric Status Rating Scales , Reaction Time/physiology , Transcranial Magnetic Stimulation , Young Adult
15.
J Neurosci ; 22(10): 4163-74, 2002 May 15.
Article in English | MEDLINE | ID: mdl-12019334

ABSTRACT

Inhibition is crucial for normal function in the nervous system. In the CNS, inhibition is mediated primarily by the amino acid GABA via activation of two ionotropic GABA receptors, GABA(A) and GABA(C). GABA(A) receptor composition and function have been well characterized, whereas much less is known about native GABA(C) receptors. Differences in molecular composition, anatomical distributions, and physiological properties strongly suggest that GABA(A) receptors and GABA(C) receptors have distinct functional roles in the CNS. To determine the functional role of GABA(C) receptors, we eliminated their expression in mice using a knock-out strategy. Although native rodent GABA(C) receptors are composed of rho1 and rho2 subunits, we show that after rho1 subunit expression was selectively eliminated there was no GABA(C) receptor expression. We assessed GABA(C) receptor function in the retina because GABA(C) receptors are highly expressed on the axon terminals of rod bipolar cells and because this site modulates the visual signal to amacrine and ganglion cells. In GABA(C)rho1 null mice, GABA-evoked responses, normally mediated by GABA(C) receptors, were eliminated, and signaling from rod bipolar cells to third order cells was altered. These data demonstrate that elimination of the GABA(C)rho1 subunit, via gene targeting, results in the absence of GABA(C) receptors in the retina and selective alterations in normal visual processing.


Subject(s)
Protein Subunits , Receptors, GABA/deficiency , Receptors, GABA/metabolism , Retina/metabolism , Vision, Ocular/physiology , Animals , Dark Adaptation/physiology , Electroretinography/drug effects , GABA Antagonists/pharmacology , Gene Targeting , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Inhibition/physiology , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques , Photic Stimulation/methods , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Receptors, GABA/genetics , Recombination, Genetic , Retina/cytology , Retina/drug effects , Retinal Rod Photoreceptor Cells/physiology , Sequence Deletion/genetics , Stem Cells , Stimulation, Chemical , Vision, Ocular/genetics , gamma-Aminobutyric Acid/pharmacology
16.
PLoS One ; 6(12): e28750, 2011.
Article in English | MEDLINE | ID: mdl-22205965

ABSTRACT

Esters of phthalic acid (phthalates) are largely used in industrial plastics, medical devices, and pharmaceutical formulations. They are easily released from plastics into the environment and can be found in measurable levels in human fluids. Phthalates are agonists for peroxisome proliferator-activated receptors (PPARs), through which they regulate translocator protein (TSPO; 18 kDa) transcription in a tissue-specific manner. TSPO is a drug- and cholesterol-binding protein involved in mitochondrial respiration, steroid formation, and cell proliferation. TSPO has been shown to increase during differentiation and decrease during maturation in mouse adipocytes. The purpose of this study was to establish the effect of mono-(2-ethylhexyl) phthalate (MEHP) on the differentiation of human SW 872 preadipocyte cells, and examine the role of TSPO in the process. After 4 days of treatment with 10 µM MEHP, we observed changes in the transcription of acetyl-CoA carboxylase alpha, adenosine triphosphate citrate lyase, glucose transporters 1 and 4, and the S100 calcium binding protein B, all of which are markers of preadipocyte differentiation. These observed gene expression changes coincided with a decrease in cellular proliferation without affecting cellular triglyceride content. Taken together, these data suggest that MEHP exerts a differentiating effect on human preadipocytes. Interestingly, MEHP was able to temporarily increase TSPO mRNA levels through the PPAR-α and ß/δ pathways. These results suggest that TSPO can be considered an important player in the differentiation process itself, or alternatively a factor whose presence is essential for adipocyte development.


Subject(s)
Cell Differentiation/drug effects , Diethylhexyl Phthalate/analogs & derivatives , Endocrine Disruptors/pharmacology , Liposarcoma/pathology , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Animals , Biomarkers/metabolism , Bromodeoxyuridine/metabolism , Cell Line, Tumor , Diethylhexyl Phthalate/pharmacology , Gene Knockdown Techniques , Humans , Mice , Peroxisome Proliferator-Activated Receptors/genetics , Protein Kinase C-epsilon/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, GABA/deficiency , Receptors, GABA/genetics , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic/drug effects
17.
J Neurophysiol ; 101(6): 2974-83, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19321639

ABSTRACT

GABA(C) receptors (GABA(C)Rs) are widely expressed in the mammalian subcortical visual system, particularly in the retina and superior colliculus (SC). GABA(C)Rs are composed of specific rho1-3 subunits the expression of which varies among visual structures. Thus rho1 subunits are most abundant in retina, and their loss eliminates GABA(C)R expression and function. In the SC, rho2 subunit expression may be equal to or stronger than rho1 subunit expression; however, results across studies vary considerably. To more directly assess the expression of GABA(C)R subunits, we characterized inhibition in the SC of wild-type (WT) and GABA(C) rho1 Null mice that lack expression of GABA(C) rho1 subunits. We used whole cell patch-clamp recordings and evaluated GABA(C)R-mediated modulation of electrically evoked post synaptic currents using either agonists or antagonists in WT mice. In GABA(C) rho1 Null stratum griseum superficiale (SGS) cells, inhibitory postsynaptic currents were shorter in duration and their excitatory postsynaptic currents (EPSCs) were longer, indicating that a slow GABA(C)R-mediated inhibitory component was reduced in each case. In contrast to retina, GABA(C)R-mediated currents in the SC were altered but not eliminated in GABA(C) rho1 Null mice. In the majority of SC cells in GABA(C) rho1 Null mice, GABA(C)R activation could still be induced to alter EPSC peak amplitudes in putative interneurons and in many projection neurons. These results, compared with previously published data, indicate a fundamental difference between retina and SC in the control of GABA(C)R expression and subunit composition.


Subject(s)
Neural Inhibition/physiology , Protein Subunits/deficiency , Receptors, GABA/physiology , Superior Colliculi/cytology , Animals , Drug Interactions , Electric Stimulation/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Female , GABA Agonists/pharmacology , GABA Antagonists/pharmacology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Neural Inhibition/drug effects , Neural Inhibition/genetics , Patch-Clamp Techniques , Receptors, GABA/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL