Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
J Neurosci ; 43(48): 8104-8125, 2023 11 29.
Article in English | MEDLINE | ID: mdl-37816598

ABSTRACT

In the brain, microRNAs (miRNAs) are believed to play a role in orchestrating synaptic plasticity at a higher level by acting as an additional mechanism of translational regulation, alongside the mRNA/polysome system. Despite extensive research, our understanding of the specific contribution of individual miRNA to the function of dopaminergic neurons (DAn) remains limited. By performing a dopaminergic-specific miRNA screening, we have identified miR-218 as a critical regulator of DAn activity in male and female mice. We have found that miR-218 is specifically expressed in mesencephalic DAn and is able to promote dopaminergic differentiation of embryonic stem cells and functional maturation of transdifferentiated induced DA neurons. Midbrain-specific deletion of both genes encoding for miR-218 (referred to as miR-218-1 and mir218-2) affects the expression of a cluster of synaptic-related mRNAs and alters the intrinsic excitability of DAn, as it increases instantaneous frequencies of evoked action potentials, reduces rheobase current, affects the ionic current underlying the action potential after hyperpolarization phase, and reduces dopamine efflux in response to a single electrical stimulus. Our findings provide a comprehensive understanding of the involvement of miR-218 in the dopaminergic system and highlight its role as a modulator of dopaminergic transmission.SIGNIFICANCE STATEMENT In the past decade, several miRNAs have emerged as potential regulators of synapse activity through the modulation of specific gene expression. Among these, we have identified a dopaminergic-specific miRNA, miR-218, which is able to promote dopaminergic differentiation and regulates the translation of an entire cluster of synapse related mRNAs. Deletion of miR-218 has notable effects on dopamine release and alters the intrinsic excitability of dopaminergic neurons, indicating a direct control of dopaminergic activity by miR-218.


Subject(s)
Dopamine , MicroRNAs , Mice , Male , Female , Animals , Dopamine/metabolism , Cell Differentiation , Dopaminergic Neurons/physiology , MicroRNAs/genetics , MicroRNAs/metabolism , Neurotransmitter Agents/metabolism
2.
Int J Mol Sci ; 23(13)2022 Jun 23.
Article in English | MEDLINE | ID: mdl-35805964

ABSTRACT

The development of midbrain dopaminergic (DA) neurons requires a fine temporal and spatial regulation of a very specific gene expression program. Here, we report that during mouse brain development, the microRNA (miR-) 204/211 is present at a high level in a subset of DA precursors expressing the transcription factor Lmx1a, an early determinant for DA-commitment, but not in more mature neurons expressing Th or Pitx3. By combining different in vitro model systems of DA differentiation, we show that the levels of Lmx1a influence the expression of miR-204/211. Using published transcriptomic data, we found a significant enrichment of miR-204/211 target genes in midbrain dopaminergic neurons where Lmx1a was selectively deleted at embryonic stages. We further demonstrated that miR-204/211 controls the timing of the DA differentiation by directly downregulating the expression of Nurr1, a late DA differentiation master gene. Thus, our data indicate the Lmx1a-miR-204/211-Nurr1 axis as a key component in the cascade of events that ultimately lead to mature midbrain dopaminergic neurons differentiation and point to miR-204/211 as the molecular switch regulating the timing of Nurr1 expression.


Subject(s)
Dopaminergic Neurons , LIM-Homeodomain Proteins , MicroRNAs , Nuclear Receptor Subfamily 4, Group A, Member 2 , Animals , Cell Differentiation/physiology , Dopamine/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mesencephalon/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Int J Mol Sci ; 21(11)2020 Jun 03.
Article in English | MEDLINE | ID: mdl-32503161

ABSTRACT

The relatively few dopaminergic neurons in the mammalian brain are mostly located in the midbrain and regulate many important neural functions, including motor integration, cognition, emotive behaviors and reward. Therefore, alteration of their function or degeneration leads to severe neurological and neuropsychiatric diseases. Unraveling the mechanisms of midbrain dopaminergic (mDA) phenotype induction and maturation and elucidating the role of the gene network involved in the development and maintenance of these neurons is of pivotal importance to rescue or substitute these cells in order to restore dopaminergic functions. Recently, in addition to morphogens and transcription factors, microRNAs have been identified as critical players to confer mDA identity. The elucidation of the gene network involved in mDA neuron development and function will be crucial to identify early changes of mDA neurons that occur in pre-symptomatic pathological conditions, such as Parkinson's disease. In addition, it can help to identify targets for new therapies and for cell reprogramming into mDA neurons. In this essay, we review the cascade of transcriptional and posttranscriptional regulation that confers mDA identity and regulates their functions. Additionally, we highlight certain mechanisms that offer important clues to unveil molecular pathogenesis of mDA neuron dysfunction and potential pharmacological targets for the treatment of mDA neuron dysfunction.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/metabolism , Neurodegenerative Diseases/metabolism , Animals , Brain/metabolism , Cell Differentiation , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Developmental , Humans , Mesencephalon/metabolism , Mesencephalon/pathology , MicroRNAs/metabolism , Neurodegenerative Diseases/drug therapy , Neurogenesis/genetics , Parkinson Disease/pathology , Phenotype , Regenerative Medicine , Transcription Factors/metabolism
4.
J Neurochem ; 141(5): 647-661, 2017 06.
Article in English | MEDLINE | ID: mdl-28122114

ABSTRACT

Precise control of dendritic spine density and synapse formation is critical for normal and pathological brain functions. Therefore, signaling pathways influencing dendrite outgrowth and remodeling remain a subject of extensive investigations. Here, we report that prolonged activation of the serotonin 5-HT7 receptor (5-HT7R) with selective agonist LP-211 promotes formation of dendritic spines and facilitates synaptogenesis in postnatal cortical and striatal neurons. Critical role of 5-HT7R in neuronal morphogenesis was confirmed by analysis of neurons isolated from 5-HT7R-deficient mice and by pharmacological inactivation of the receptor. Acute activation of 5-HT7R results in pronounced neurite elongation in postnatal striatal and cortical neurons, thus extending previous data on the morphogenic role of 5-HT7R in embryonic and hippocampal neurons. We also observed decreased number of spines in neurons with either genetically (i.e. 5-HT7R-knock-out) or pharmacologically (i.e. antagonist treatment) blocked 5-HT7R, suggesting that constitutive 5-HT7R activity is critically involved in the spinogenesis. Moreover, cyclin-dependent kinase 5 and small GTPase Cdc42 were identified as important downstream effectors mediating morphogenic effects of 5-HT7R in neurons. Altogether, our data suggest that the 5-HT7R-mediated structural reorganization during the postnatal development might have a crucial role for the development and plasticity of forebrain areas such as cortex and striatum, and thereby can be implicated in regulation of the higher cognitive functions. Read the Editorial Highlight for this article on page 644.


Subject(s)
Cerebral Cortex/cytology , Corpus Striatum/cytology , Dendritic Spines/metabolism , Neurogenesis/genetics , Neurons/cytology , Receptors, Serotonin/metabolism , Synapses/genetics , Animals , Animals, Newborn , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cells, Cultured , Dendritic Spines/drug effects , Diterpenes , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Net/drug effects , Nerve Net/physiology , Neurogenesis/drug effects , Neurons/drug effects , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Receptors, Serotonin/genetics , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Synapses/drug effects , Time Factors
5.
FASEB J ; 27(3): 865-70, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23230282

ABSTRACT

The brain is the most cholesterol-enriched tissue in the body. During brain development, desmosterol, an immediate precursor of cholesterol, transiently accumulates up to 30% of total brain sterols. This massive desmosterol deposition appears to be present in all mammalian species reported so far, including humans, but how it is achieved is not well understood. Here, we propose that desmosterol accumulation in the developing brain may be primarily caused by post-transcriptional repression of 3ß-hydroxysterol 24-reductase (DHCR24) by progesterone. Furthermore, distinct properties of desmosterol may serve to increase the membrane active pool of sterols in the brain: desmosterol cannot be hydroxylated to generate 24S-hydroxycholesterol, a brain derived secretory sterol, desmosterol has a reduced propensity to be esterified as compared to cholesterol, and desmosterol may activate LXR to stimulate astrocyte sterol secretion. This regulated accumulation of desmosterol by progesterone-induced suppression of DHCR24 may facilitate the rapid enrichment and distribution of membrane sterols in the developing brain.


Subject(s)
Brain Chemistry/physiology , Brain/growth & development , Desmosterol/metabolism , Animals , Brain/cytology , Brain/metabolism , Cell Membrane/metabolism , Liver X Receptors , Mice , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/metabolism , Orphan Nuclear Receptors/metabolism , Oxidoreductases Acting on CH-CH Group Donors/biosynthesis , Progesterone/metabolism
6.
J Neurochem ; 124(2): 159-67, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23134340

ABSTRACT

Research on stem cells has developed as one of the most promising areas of neurobiology. In the beginning of the 1990s, neurogenesis in the adult brain was indisputably accepted, eliciting great research efforts. Neural stem cells in the adult mammalian brain are located in the 'neurogenic' areas of the subventricular and subgranular zones. Nevertheless, many reports indicate that they subsist in other regions of the adult brain. Adult neural stem cells have arisen considerable interest as these studies can be useful to develop new methods to replace damaged neurons and treat severe neurological diseases such as neurodegeneration, stroke or spinal cord lesions. In particular, a promising field is aimed at stimulating or trigger a self-repair system in the diseased brain driven by its own stem cell population. Here, we will revise the latest findings on the characterization of active and quiescent adult neural stem cells in the main regions of neurogenesis and the factors necessary to maintain their active and resting states, stimulate migration and homing in diseased areas, hoping to outline the emerging knowledge for the promotion of regeneration in the brain based on endogenous stem cells.


Subject(s)
Adult Stem Cells/physiology , Brain Diseases/pathology , Brain Diseases/therapy , Neural Stem Cells/physiology , Adult Stem Cells/pathology , Animals , Brain Diseases/physiopathology , Humans , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Nerve Degeneration/therapy , Nerve Regeneration/physiology , Neural Stem Cells/pathology
7.
Exp Cell Res ; 317(4): 464-73, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21093432

ABSTRACT

Krüppel-like factor 7 (KLF7) belongs to the large family of KLF transcription factors, which comprises at least 17 members. Within this family, KLF7 is unique since its expression is strictly restricted within the nervous system during development. We have previously shown that KLF7 is required for neuronal morphogenesis and axon guidance in selected regions of the nervous system, including hippocampus, olfactory bulbs and cortex, as well as in neuronal cell cultures. In the present work, we have furthered our analysis of the role of KLF7 in central nervous system development. By gene expression analysis during brain embryogenesis, we found significant alterations in dopaminergic neurons in Klf7 null mice. In particular, the tyrosine hydroxylase (TH) and dopamine transporter (Dat) transcripts are strongly decreased in the olfactory bulbs and ventral midbrain at birth, compared to wild-type littermates. Interestingly, Klf7-mutant mice show a dramatic reduction of TH-positive neurons in the olfactory bulbs, but no change in GABAergic or midbrain dopaminergic neurons. These observations raise the possibility that a lack of a KLF family member affects dopaminergic neuron development.


Subject(s)
Dopamine , Embryonic Development , Kruppel-Like Transcription Factors/physiology , Neurons/cytology , Olfactory Bulb/growth & development , Animals , Central Nervous System/growth & development , Dopamine Plasma Membrane Transport Proteins/biosynthesis , Dopamine Plasma Membrane Transport Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Olfactory Bulb/cytology , Transcription Factors/physiology , Tyrosine 3-Monooxygenase/biosynthesis , Tyrosine 3-Monooxygenase/genetics
8.
Rev Neurosci ; 33(7): 789-801, 2022 10 26.
Article in English | MEDLINE | ID: mdl-35325516

ABSTRACT

In a million years, under the pressure of natural selection, hominins have acquired the abilities for vocal learning, music, and language. Music is a relevant human activity, highly effective in enhancing sociality, is a universal experience common to all known human cultures, although it varies in rhythmic and melodic complexity. It has been part of human life since the beginning of our history, or almost, and it strengthens the mother-baby relation even within the mother's womb. Music engages multiple cognitive functions, and promotes attention, concentration, imagination, creativity, elicits memories and emotions, and stimulates imagination, and harmony of movement. It changes the chemistry of the brain, by inducing the release of neurotransmitters and hormones (dopamine, serotonin, and oxytocin) and activates the reward and prosocial systems. In addition, music is also used to develop new therapies necessary to alleviate severe illness, especially neurological disorders, and brain injuries.


Subject(s)
Music , Nervous System Diseases , Brain/physiology , Dopamine , Humans , Nervous System Diseases/therapy , Oxytocin , Serotonin
9.
Exp Cell Res ; 316(14): 2365-76, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20580711

ABSTRACT

Previous gene targeting studies in mice have implicated the nuclear protein Krüppel-like factor 7 (KLF7) in nervous system development while cell culture assays have documented its involvement in cell cycle regulation. By employing short hairpin RNA (shRNA)-mediated gene silencing, here we demonstrate that murine Klf7 gene expression is required for in vitro differentiation of neuroectodermal and mesodermal cells. Specifically, we show a correlation of Klf7 silencing with down-regulation of the neuronal marker microtubule-associated protein 2 (Map2) and the nerve growth factor (NGF) tyrosine kinase receptor A (TrkA) using the PC12 neuronal cell line. Similarly, KLF7 inactivation in Klf7-null mice decreases the expression of the neurogenic marker brain lipid-binding protein/fatty acid-binding protein 7 (BLBP/FABP7) in neural stem cells (NSCs). We also report that Klf7 silencing is detrimental to neuronal and cardiomyocytic differentiation of embryonic stem cells (ESCs), in addition to altering the adipogenic and osteogenic potential of mouse embryonic fibroblasts (MEFs). Finally, our results suggest that genes that are key for self-renewal of undifferentiated ESCs repress Klf7 expression in ESCs. Together with previous findings, these results provide evidence that KLF7 has a broad spectrum of regulatory functions, which reflect the discrete cellular and molecular contexts in which this transcription factor operates.


Subject(s)
Cell Differentiation , Gene Expression Regulation , Kruppel-Like Transcription Factors/metabolism , Mesoderm/cytology , Neural Plate/cytology , Neurons/cytology , Animals , Blotting, Western , Cells, Cultured , Embryonic Stem Cells/cytology , Female , Gene Silencing , Homeodomain Proteins/genetics , Immunohistochemistry , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , PC12 Cells , Rats , Reverse Transcriptase Polymerase Chain Reaction
10.
Cells ; 10(4)2021 03 26.
Article in English | MEDLINE | ID: mdl-33810328

ABSTRACT

Dopamine (DA) is a key neurotransmitter involved in multiple physiological functions including motor control, modulation of affective and emotional states, reward mechanisms, reinforcement of behavior, and selected higher cognitive functions. Dysfunction in dopaminergic transmission is recognized as a core alteration in several devastating neurological and psychiatric disorders, including Parkinson's disease (PD), schizophrenia, bipolar disorder, attention deficit hyperactivity disorder (ADHD) and addiction. Here we will discuss the current insights on the role of DA in motor control and reward learning mechanisms and its involvement in the modulation of synaptic dynamics through different pathways. In particular, we will consider the role of DA as neuromodulator of two forms of synaptic plasticity, known as long-term potentiation (LTP) and long-term depression (LTD) in several cortical and subcortical areas. Finally, we will delineate how the effect of DA on dendritic spines places this molecule at the interface between the motor and the cognitive systems. Specifically, we will be focusing on PD, vascular dementia, and schizophrenia.


Subject(s)
Dopamine/metabolism , Movement , Neuronal Plasticity , Neurotransmitter Agents/metabolism , Reward , Animals , Cognition/physiology , Humans
11.
Rev Neurosci ; 2020 Aug 17.
Article in English | MEDLINE | ID: mdl-32924383

ABSTRACT

The environment increased complexity required more neural functions to develop in the hominin brains, and the hominins adapted to the complexity by developing a bigger brain with a greater interconnection between its parts. Thus, complex environments drove the growth of the brain. In about two million years during hominin evolution, the brain increased three folds in size, one of the largest and most complex amongst mammals, relative to body size. The size increase has led to anatomical reorganization and complex neuronal interactions in a relatively small skull. At birth, the human brain is only about 20% of its adult size. That facilitates the passage through the birth canal. Therefore, the human brain, especially cortex, develops postnatally in a rich stimulating environment with continuous brain wiring and rewiring and insertion of billions of new neurons. One of the consequence is that in the newborn brain, neuroplasticity is always turned "on" and it remains active throughout life, which gave humans the ability to adapt to complex and often hostile environments, integrate external experiences, solve problems, elaborate abstract ideas and innovative technologies, store a lot of information. Besides, hominins acquired unique abilities as music, language, and intense social cooperation. Overwhelming ecological, social, and cultural challenges have made the human brain so unique. From these events, as well as the molecular genetic changes that took place in those million years, under the pressure of natural selection, derive the distinctive cognitive abilities that have led us to complex social organizations and made our species successful.

12.
PLoS One ; 15(5): e0233918, 2020.
Article in English | MEDLINE | ID: mdl-32442206

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0030661.].

13.
Behav Brain Res ; 336: 256-260, 2018 01 15.
Article in English | MEDLINE | ID: mdl-28899819

ABSTRACT

Dendritic spines, small protrusions emerging from the dendrites of most excitatory synapses in the mammalian brain, are highly dynamic structures and their shape and number is continuously modulated by memory formation and other adaptive changes of the brain. In this study, using a behavioral paradigm of motor learning, we applied the non-linear analysis of dendritic spines to study spine complexity along dendrites of cortical and subcortical neural systems, such as the basal ganglia, that sustain important motor learning processes. We show that, after learning, the spine organization has greater complexity, as indexed by the maximum Lyapunov exponent (LyE). The positive value of the exponent demonstrates that the system is chaotic, while recurrence plots show that the system is not simply composed by random noise, but displays quasi-periodic behavior. The increase in the maximum LyE and in the system entropy after learning was confirmed by the modification of the reconstructed trajectories in phase-space. Our results suggest that the remodeling of spines, as a result of a chaotic and non-random dynamical process along dendrites, may be a general feature associated with the structural plasticity underlying processes such as long-term memory maintenance. Furthermore, this work indicates that the non-linear method is a very useful tool to allow the detection of subtle stimulus-induced changes in dendritic spine dynamics, giving a key contribution to the study of the relationship between structure and function of spines.


Subject(s)
Dendritic Spines/physiology , Learning/physiology , Animals , Brain/physiology , Dendrites/physiology , Male , Memory, Long-Term/physiology , Mice , Mice, Inbred C57BL , Neuronal Plasticity/physiology , Synapses/physiology
14.
Stem Cell Reports ; 10(4): 1237-1250, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29526736

ABSTRACT

The differentiation of dopaminergic neurons requires concerted action of morphogens and transcription factors acting in a precise and well-defined time window. Very little is known about the potential role of microRNA in these events. By performing a microRNA-mRNA paired microarray screening, we identified miR-34b/c among the most upregulated microRNAs during dopaminergic differentiation. Interestingly, miR-34b/c modulates Wnt1 expression, promotes cell cycle exit, and induces dopaminergic differentiation. When combined with transcription factors ASCL1 and NURR1, miR-34b/c doubled the yield of transdifferentiated fibroblasts into dopaminergic neurons. Induced dopaminergic (iDA) cells synthesize dopamine and show spontaneous electrical activity, reversibly blocked by tetrodotoxin, consistent with the electrophysiological properties featured by brain dopaminergic neurons. Our findings point to a role for miR-34b/c in neuronal commitment and highlight the potential of exploiting its synergy with key transcription factors in enhancing in vitro generation of dopaminergic neurons.


Subject(s)
Cell Differentiation , Dopaminergic Neurons/cytology , Mesencephalon/cytology , MicroRNAs/metabolism , Wnt1 Protein/metabolism , Animals , Base Sequence , Cell Transdifferentiation , Dopaminergic Neurons/metabolism , Fibroblasts/cytology , Gene Expression Regulation , Germ Layers/cytology , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Mice , MicroRNAs/genetics , Neurogenesis/genetics , Transcription Factors/metabolism , Wnt Signaling Pathway
15.
Brain Res ; 1159: 28-39, 2007 Jul 23.
Article in English | MEDLINE | ID: mdl-17574220

ABSTRACT

The glial cell line-derived neurotrophic factor (GDNF) exerts trophic actions on a number of cell types, including mesencephalic dopaminergic (mDA) neurons. Using rat mesencephalic primary cultures enriched in mDA neurons, we show that protracted GDNF stimulation increases their survival and neurite outgrowth. It modulates the expression of genes essential for DA function (tyrosine hydroxylase, TH and dopamine transporter, dat) and of DA high affinity uptake. To identify genes involved in GDNF signaling pathways, we have used DNA microarray on mDA cultures stimulated with GDNF for 3 h. Here we show that GDNF signaling sequentially activates the genes encoding for the transcription factors EGR1 and TIEG. In addition, it increases the expression of cav1, which encodes for the major component of caveolae. GDNF also modulates the expression of the genes encoding for the Calcineurin subunits ppp3R1 and ppp3CB, and inhibits calcium-calmodulin-dependent protein kinase II beta isoform (CaMKIIbeta) gene expression. These proteins are involved in neuronal differentiation and synaptic plasticity. Moreover, GDNF stimulation down regulates the expression of the glycogen synthase kinase 3beta (gsk3beta) gene, involved in neuronal apoptosis. Using inhibitors of specific intracellular signal transduction pathways we show that changes of egr1, tieg, cav1, CaMkIIbeta and gsk3beta genes expression are extracellular-signal regulated kinases 1/2 (ERK)-dependent, while the cAMP-dependent protein kinase (PKA) pathway influences the up-regulation of ppp3R1 and ppp3CB gene expression. These results demonstrate that GDNF stimulation results in the transcriptional modulation of genes involved in neuronal plasticity and survival and in mDA function, mediated in part by ERK and PKA signaling.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/physiology , Mesencephalon/cytology , Neurons/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Dose-Response Relationship, Drug , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Mesencephalon/embryology , Microarray Analysis/methods , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Time Factors , Tritium/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
16.
Brain Res ; 1143: 1-10, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17321503

ABSTRACT

Serotonin (5-HT) is a neurotransmitter involved in a variety of CNS functions during development and in adulthood. 5-HT neurons are also involved in the pathogenesis of a number of psychiatric disorders. FLUOXETINE (FLX), a prototypic antidepressant, is a selective 5-HT uptake inhibitor (SSRI) with a demonstrated clinical efficacy in these disorders. SSRI, in a short-term period, binds 5-HT transporter (SERT) raising 5-HT levels at the synapse. Nevertheless, clinical improvement is observed only after 3-4 weeks of treatment. Recently, it has been shown that antidepressants, besides interfering with neurotransmission, can also display an effect on neural cells' proliferation and differentiation. Therefore it has been proposed that antidepressant may exert their clinical effects also acting on cellular functions other then neurotransmission. Here we show that a mesencephalic neural cell line, mes-c-myc A1 (A1) produces 5-HT and expresses SERT and both peripheral (TPH1) and CNS-specific (TPH2) form of tryptophan hydroxylase, the limiting enzyme in 5-HT biosynthesis. Cyclic AMP-dependent neuronal differentiation of A1 cells modulates the expression of TPHs. FLX, as well as citalopram (CIT), another SSRI inhibitor, modulates expression of serotonergic markers depending on the differentiation status of the cells. Interestingly, long-term but not short-term FLX treatment selectively modulates mRNA levels of TPH2, only in differentiated A1 cells. Finally, FLX and citalopram selectively decrease the proliferation rate of undifferentiated A1 cells, whereas have no effects on NIH-3T3 fibroblasts proliferation. In conclusion, neuronal differentiation of A1 cells not only modulates the expression of serotonergic markers, but appears to affect the response to FLX.


Subject(s)
Cell Differentiation/drug effects , Fluoxetine/pharmacology , Mesencephalon/cytology , Neurons/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin/metabolism , Analysis of Variance , Animals , Cell Count , Cell Line , Citalopram/pharmacology , Dose-Response Relationship, Drug , Embryo, Mammalian , Gene Expression/drug effects , Mice , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Time Factors , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism , Vesicular Monoamine Transport Proteins/genetics , Vesicular Monoamine Transport Proteins/metabolism
17.
Rev Neurosci ; 28(3): 235-245, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28107174

ABSTRACT

Music is a universal language, present in all human societies. It pervades the lives of most human beings and can recall memories and feelings of the past, can exert positive effects on our mood, can be strongly evocative and ignite intense emotions, and can establish or strengthen social bonds. In this review, we summarize the research and recent progress on the origins and neural substrates of human musicality as well as the changes in brain plasticity elicited by listening or performing music. Indeed, music improves performance in a number of cognitive tasks and may have beneficial effects on diseased brains. The emerging picture begins to unravel how and why particular brain circuits are affected by music. Numerous studies show that music affects emotions and mood, as it is strongly associated with the brain's reward system. We can therefore assume that an in-depth study of the relationship between music and the brain may help to shed light on how the mind works and how the emotions arise and may improve the methods of music-based rehabilitation for people with neurological disorders. However, many facets of the mind-music connection still remain to be explored and enlightened.


Subject(s)
Auditory Perception/physiology , Brain/physiology , Emotions/physiology , Music , Reward , Animals , Humans , Mental Recall/physiology , Nervous System Diseases/physiopathology
18.
Neuropsychopharmacology ; 31(9): 1946-56, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16319916

ABSTRACT

Administration of methylphenidate (MPH, Ritalin) to children with attention deficit hyperactivity disorder (ADHD) is an elective therapy, but raises concerns for public health, due to possible persistent neurobehavioral alterations. Wistar adolescent rats (30 to 46 day old) were administered MPH or saline (SAL) for 16 days, and tested for reward-related and motivational-choice behaviors. When tested in adulthood in a drug-free state, MPH-pretreated animals showed increased choice flexibility and economical efficiency, as well as a dissociation between dampened place conditioning and more marked locomotor sensitization induced by cocaine, compared to SAL-pretreated controls. The striatal complex, a core component of the natural reward system, was collected both at the end of the MPH treatment and in adulthood. Genome-wide expression profiling, followed by RT-PCR validation on independent samples, showed that three members of the postsynaptic-density family and five neurotransmitter receptors were upregulated in the adolescent striatum after subchronic MPH administration. Interestingly, only genes for the kainate 2 subunit of ionotropic glutamate receptor (Grik2, also known as KA2) and the 5-hydroxytryptamine (serotonin) receptor 7 (Htr7) (but not GABA(A) subunits and adrenergic receptor alpha1b) were still upregulated in adulthood. cAMP responsive element-binding protein and Homer 1a transcripts were modulated only as a long-term effect. In summary, our data indicate short-term changes in neural plasticity, suggested by modulation of expression of key genes, and functional changes in striatal circuits. These modifications might in turn trigger enduring changes responsible for the adult neurobehavioral profile, that is, altered processing of incentive values and a modified flexibility/habit balance.


Subject(s)
Central Nervous System Stimulants/pharmacology , Conditioning, Operant/drug effects , Gene Expression/drug effects , Methylphenidate/pharmacology , Neostriatum/metabolism , Neuronal Plasticity/drug effects , Reward , Animals , Data Interpretation, Statistical , Female , In Situ Hybridization , Male , Neostriatum/drug effects , Oligonucleotide Array Sequence Analysis , Pregnancy , RNA/biosynthesis , RNA/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/drug effects
19.
Front Hum Neurosci ; 10: 265, 2016.
Article in English | MEDLINE | ID: mdl-27375460

ABSTRACT

Functional magnetic resonance imaging (fMRI) provides a powerful way to visualize brain functions and observe brain activity in response to tasks or thoughts. It allows displaying brain damages that can be quantified and linked to neurobehavioral deficits. fMRI can potentially draw a new cartography of brain functional areas, allow us to understand aspects of brain function evolution or even breach the wall into cognition and consciousness. However, fMRI is not deprived of pitfalls, such as limitation in spatial resolution, poor reproducibility, different time scales of fMRI measurements and neuron action potentials, low statistical values. Thus, caution is needed in the assessment of fMRI results and conclusions. Additional diagnostic techniques based on MRI such as arterial spin labeling (ASL) and the measurement of diffusion tensor imaging (DTI) provide new tools to assess normal brain development or disruption of anatomical networks in diseases. A cutting edge of recent research uses fMRI techniques to establish a "map" of neural connections in the brain, or "connectome". It will help to develop a map of neural connections and thus understand the operation of the network. New applications combining fMRI and real time visualization of one's own brain activity (rtfMRI) could empower individuals to modify brain response and thus could enable researchers or institutions to intervene in the modification of an individual behavior. The latter in particular, as well as the concern about the confidentiality and storage of sensitive information or fMRI and lie detectors forensic use, raises new ethical questions.

20.
Cell Signal ; 16(12): 1425-34, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15381258

ABSTRACT

The glial cell line-derived neurotrophic factor (GDNF) family coreceptor alpha1 (GFRalpha1) is a critical component of the RET receptor kinase signal-transducing complex. The activity of this multicomponent receptor is stimulated by the glial cell line-derived neurotrophic factor (GDNF) and is involved in neuronal cells survival and kidney development. GFRalpha1 pre-mRNA is alternatively spliced and produces two isoforms: GFRalpha1a, which includes the exon 5; and GFRalpha1b, which excludes it. Here we show that the Gfralpha1a isoform is predominantly expressed in neuronal tissues and in PC12 cells differentiated toward a neuronal phenotype. GFRalpha1 splicing is also regulated during kidney development, GFRalpha1a is the minor isoform before birth and then rapidly becomes the major form after birth. We established cell lines expressing either GFRalpha1 isoforms and demonstrated that the GFRalpha1b isoform binds GDNF more efficiently than GFRalpha1a. Consistently, GFRalpha1b promotes a stronger RET phosphorylation than GFRalpha1a. These results indicate that specific inclusion of the GFRalpha1 exon 5 in neuronal tissues or during kidney development may alter the binding properties of GDNF to GFRalpha1, and thus could constitute an additional regulatory mechanism of the RET signaling pathway.


Subject(s)
Alternative Splicing , Gene Expression Regulation, Developmental , Kidney/embryology , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Animals , Base Sequence , Cell Line , Dose-Response Relationship, Drug , Exons , Genome , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Immunoblotting , Ligands , Mitogen-Activated Protein Kinase 3/metabolism , Models, Genetic , Molecular Sequence Data , Nerve Growth Factors/metabolism , Neurons/metabolism , PC12 Cells , Phenotype , Phosphorylation , Protein Binding , Protein Isoforms , Proto-Oncogene Proteins c-ret , RNA/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL