Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Front Syst Neurosci ; 16: 941534, 2022.
Article En | MEDLINE | ID: mdl-35910003

Awareness or consciousness in the context of stimulus perception can directly be assessed in well controlled test situations with humans via the persons' reports about their subjective experiences with the stimuli. Since we have no direct access to subjective experiences in animals, their possible awareness or consciousness in stimulus perception tasks has often been inferred from behavior and cognitive abilities previously observed in aware and conscious humans. Here, we analyze published human data primarily on event-related potentials and brain-wave generation during perception and responding to sensory stimuli and extract neural markers (mainly latencies of evoked-potential peaks and of gamma-wave occurrence) indicating that a person became aware or conscious of the perceived stimulus. These neural correlates of consciousness were then applied to sets of corresponding data from various animals including several species of mammals, and one species each of birds, fish, cephalopods, and insects. We found that the neural markers from studies in humans could also successfully be applied to the mammal and bird data suggesting that species in these animal groups can become subjectively aware of and conscious about perceived stimuli. Fish, cephalopod and insect data remained inconclusive. In an evolutionary perspective we have to consider that both awareness of and consciousness about perceived stimuli appear as evolved, attention-dependent options added to the ongoing neural activities of stimulus processing and action generation. Since gamma-wave generation for functional coupling of brain areas in aware/conscious states is energetically highly cost-intensive, it remains to be shown which animal species under which conditions of lifestyle and ecological niche may achieve significant advantages in reproductive fitness by drawing upon these options. Hence, we started our discussion about awareness and consciousness in animals with the question in how far these expressions of brain activity are necessary attributes for perceiving stimuli and responding in an adaptive way.

2.
Front Psychol ; 13: 846159, 2022.
Article En | MEDLINE | ID: mdl-36743633

Near-death experiences (NDEs) including out-of-body experiences (OBEs) have been fascinating phenomena of perception both for affected persons and for communities in science and medicine. Modern progress in the recording of changing brain functions during the time between clinical death and brain death opened the perspective to address and understand the generation of NDEs in brain states of altered consciousness. Changes of consciousness can experimentally be induced in well-controlled clinical or laboratory settings. Reports of the persons having experienced the changes can inform about the similarity of the experiences with those from original NDEs. Thus, we collected neuro-functional models of NDEs including OBEs with experimental backgrounds of drug consumption, epilepsy, brain stimulation, and ischemic stress, and included so far largely unappreciated data from fighter pilot tests under gravitational stress generating cephalic nervous system ischemia. Since we found a large overlap of NDE themes or topics from original NDE reports with those from neuro-functional NDE models, we can state that, collectively, the models offer scientifically appropriate causal explanations for the occurrence of NDEs. The generation of OBEs, one of the NDE themes, can be localized in the temporo-parietal junction (TPJ) of the brain, a multimodal association area. The evaluated literature suggests that NDEs may emerge as hallucination-like phenomena from a brain in altered states of consciousness (ASCs).

3.
PLoS One ; 10(2): e0118024, 2015.
Article En | MEDLINE | ID: mdl-25706271

An in situ hybridization (ISH) study was performed on 2000 murine genes representing around 10% of the protein-coding genes present in the mouse genome using data generated by the EURExpress consortium. This study was carried out in 25 tissues of late gestation embryos (E14.5), with a special emphasis on the developing ear and on five distinct developing sensory organs, including the cochlea, the vestibular receptors, the sensory retina, the olfactory organ, and the vibrissae follicles. The results obtained from an analysis of more than 11,000 micrographs have been integrated in a newly developed knowledgebase, called ImAnno. In addition to managing the multilevel micrograph annotations performed by human experts, ImAnno provides public access to various integrated databases and tools. Thus, it facilitates the analysis of complex ISH gene expression patterns, as well as functional annotation and interaction of gene sets. It also provides direct links to human pathways and diseases. Hierarchical clustering of expression patterns in the 25 tissues revealed three main branches corresponding to tissues with common functions and/or embryonic origins. To illustrate the integrative power of ImAnno, we explored the expression, function and disease traits of the sensory epithelia of the five presumptive sensory organs. The study identified 623 genes (out of 2000) concomitantly expressed in the five embryonic epithelia, among which many (∼12%) were involved in human disorders. Finally, various multilevel interaction networks were characterized, highlighting differential functional enrichments of directly or indirectly interacting genes. These analyses exemplify an under-represention of "sensory" functions in the sensory gene set suggests that E14.5 is a pivotal stage between the developmental stage and the functional phase that will be fully reached only after birth.


Computational Biology/methods , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , In Situ Hybridization/methods , Animals , Choroid Plexus/embryology , Choroid Plexus/metabolism , Databases, Genetic , Ear, Inner/embryology , Ear, Inner/metabolism , Fetal Development/genetics , Gene Ontology , Gene Regulatory Networks , Genomics/methods , Humans , Information Storage and Retrieval/methods , Mice , Olfactory Mucosa/embryology , Olfactory Mucosa/metabolism , Retina/embryology , Retina/metabolism , Sensory Receptor Cells/metabolism , Vibrissae/embryology , Vibrissae/metabolism
4.
Cell Metab ; 20(5): 856-869, 2014 Nov 04.
Article En | MEDLINE | ID: mdl-25200183

Mitochondrial activity is controlled by proteins encoded by both nuclear and mitochondrial DNA. Here, we identify Sirt7 as a crucial regulator of mitochondrial homeostasis. Sirt7 deficiency in mice induces multisystemic mitochondrial dysfunction, which is reflected by increased blood lactate levels, reduced exercise performance, cardiac dysfunction, hepatic microvesicular steatosis, and age-related hearing loss. This link between SIRT7 and mitochondrial function is translatable in humans, where SIRT7 overexpression rescues the mitochondrial functional defect in fibroblasts with a mutation in NDUFSI. These wide-ranging effects of SIRT7 on mitochondrial homeostasis are the consequence of the deacetylation of distinct lysine residues located in the hetero- and homodimerization domains of GABPß1, a master regulator of nuclear-encoded mitochondrial genes. SIRT7-mediated deacetylation of GABPß1 facilitates complex formation with GABPα and the transcriptional activation of the GABPα/GABPß heterotetramer. Altogether, these data suggest that SIRT7 is a dynamic nuclear regulator of mitochondrial function through its impact on GABPß1 function.


GA-Binding Protein Transcription Factor/metabolism , Mitochondria/genetics , Mitochondria/pathology , Sirtuins/genetics , Acetylation , Aging , Amino Acid Sequence , Animals , GA-Binding Protein Transcription Factor/analysis , Gene Knockout Techniques , Humans , Mice , Mice, Knockout , Mitochondria/metabolism , Molecular Sequence Data , Sequence Alignment , Sirtuins/analysis , Sirtuins/metabolism
5.
J Neurosci ; 33(13): 5856-66, 2013 Mar 27.
Article En | MEDLINE | ID: mdl-23536097

The retinaldehyde dehydrogenase 3 (Raldh3) gene encodes a major retinoic acid synthesizing enzyme and is highly expressed in the inner ear during embryogenesis. We found that mice deficient in Raldh3 bear severe impairment in vestibular functions. These mutant mice exhibited spontaneous circling/tilted behaviors and performed poorly in several vestibular-motor function tests. In addition, video-oculography revealed a complete loss of the maculo-ocular reflex and a significant reduction in the horizontal angular vestibulo-ocular reflex, indicating that detection of both linear acceleration and angular rotation were compromised in the mutants. Consistent with these behavioral and functional deficiencies, morphological anomalies, characterized by a smaller vestibular organ with thinner semicircular canals and a significant reduction in the number of otoconia in the saccule and the utricle, were consistently observed in the Raldh3 mutants. The loss of otoconia in the mutants may be attributed, at least in part, to significantly reduced expression of Otop1, which encodes a protein known to be involved in calcium regulation in the otolithic organs. Our data thus reveal a previously unrecognized role of Raldh3 in structural and functional development of the vestibular end organs.


Reflex, Vestibulo-Ocular/drug effects , Reflex, Vestibulo-Ocular/genetics , Tretinoin/pharmacology , Vestibule, Labyrinth/physiopathology , Vitamin A Deficiency/pathology , Aldehyde Dehydrogenase 1 Family , Analysis of Variance , Animals , Behavioral Symptoms/etiology , Behavioral Symptoms/genetics , Embryo, Mammalian , Eye Movements/drug effects , Eye Movements/genetics , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Imaging, Three-Dimensional , Isoenzymes/deficiency , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Motor Activity/drug effects , Motor Activity/genetics , Mutation/genetics , Otolithic Membrane/pathology , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Retinal Dehydrogenase/deficiency , Swimming , Vestibular Function Tests , Vestibule, Labyrinth/ultrastructure , Video Recording , Vitamin A Deficiency/etiology , Walking/physiology
6.
Am J Respir Cell Mol Biol ; 45(1): 81-7, 2011 Jul.
Article En | MEDLINE | ID: mdl-20855652

Cathepsin S (Cat S) is predominantly expressed in antigen-presenting cells and is up-regulated in several preclinical models of antigen-induced inflammation, suggesting a role in the allergic response. Prophylactic dosing of an irreversible Cat S inhibitor has been shown to attenuate pulmonary eosinophilia in mice, supporting the hypothesis that Cat S inhibition before the initiation of airway inflammation is beneficial in airway disease. In addition, Cat S has been shown to play a role in more distal events in the allergic response. To determine where Cat S inhibition may affect the allergic response, we used complementary genetic and pharmacological approaches to investigate the role of Cat S in the early and downstream allergic events in a murine model of antigen-induced lung inflammation. Cat S knockout mice did not develop ovalbumin-induced pulmonary inflammation, consistent with a role for Cat S in the development of the allergic response. Alternatively, wild-type mice were treated with a reversible, highly selective Cat S inhibitor in prophylactic and therapeutic dosing paradigms and assessed for changes in airway inflammation. Although both treatment paradigms resulted in potent Cat S inhibition, only prophylactic Cat S inhibitor dosing blocked lung inflammation, consistent with our findings in Cat S knockout mice. The findings indicate that although Cat S is up-regulated in allergic models, it does not appear to play a significant role in the downstream effector inflammatory phase in this model; however, our results demonstrate that Cat S inhibition in a prophylactic paradigm would ameliorate airway inflammation.


Asthma/prevention & control , Cathepsins/genetics , Cathepsins/pharmacology , Animals , Asthma/genetics , Asthma/metabolism , Cathepsins/biosynthesis , Disease Models, Animal , Drug Evaluation , Humans , Mice , Mice, Knockout , Ovalbumin/adverse effects , Ovalbumin/pharmacology , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/metabolism , Pulmonary Eosinophilia/prevention & control , Up-Regulation/drug effects , Up-Regulation/genetics
7.
Biochim Biophys Acta ; 1792(2): 140-7, 2009 Feb.
Article En | MEDLINE | ID: mdl-19073250

Fibroblast growth factor receptor 3 (FGFR3) is a key regulator of skeletal development and activating mutations in FGFR3 cause skeletal dysplasias, including hypochondroplasia, achondroplasia and thanatophoric dysplasia. The introduction of the Y367C mutation corresponding to the human Y373C thanatophoric dysplasia type I (TDI) mutation into the mouse genome, resulted in dwarfism with a skeletal phenotype remarkably similar to that of human chondrodysplasia. To investigate the role of the activating Fgfr3 Y367C mutation in auditory function, the middle and inner ear of the heterozygous mutant Fgfr3(Y367C/+) mice were examined. The mutant Fgfr3(Y367C/+) mice exhibit fully penetrant deafness with a significantly elevated auditory brainstem response threshold for all frequencies tested. The inner ear defect is mainly associated with an increased number of pillar cells or modified supporting cells in the organ of Corti. Hearing loss in the Fgfr3(Y367C/+) mouse model demonstrates the crucial role of Fgfr3 in the development of the inner ear and provides novel insight on the biological consequences of FGFR3 mutations in chondrodysplasia.


Chondrodysplasia Punctata/metabolism , Chondrodysplasia Punctata/pathology , Hearing Loss/metabolism , Hearing Loss/pathology , Labyrinth Diseases/metabolism , Labyrinth Diseases/pathology , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Animals , Chondrodysplasia Punctata/complications , Chondrodysplasia Punctata/genetics , Disease Models, Animal , Enzyme Activation , Hearing Loss/complications , Heterozygote , Labyrinth Diseases/complications , Labyrinth Diseases/genetics , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Mutation/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Tyrosine/genetics , Tyrosine/metabolism
8.
J Comp Neurol ; 508(6): 879-92, 2008 Jun 20.
Article En | MEDLINE | ID: mdl-18399539

Organs develop through many tissue interactions during embryogenesis, involving numerous signaling cascades and gene products. One of these signaling molecules is retinoic acid (RA), an active vitamin A derivative, which in mammalian embryos is synthesized from maternal retinol by two oxidative reactions involving alcohol/retinol dehydrogenases (ADH/RDHs) and retinaldehyde dehydrogenases (RALDHs), respectively. The activity of RALDHs is known to be crucial for RA synthesis; however, recently a retinol dehydrogenase (RDH10) has been shown to represent a new limiting factor in this synthesis. We investigated the spatiotemporal distribution of Rdh10 gene transcripts by in situ hybridization and quantitative polymerase chain reaction (PCR) during development of the brain and sensory organs. Although Rdh10 relative mRNA levels decline throughout brain development, we show a strong and lasting expression in the meninges and choroid plexuses. Rdh10 expression is also specifically seen in the striatum, a known site of retinoid signaling. In the eye, regional expression is observed both in the prospective pigmented epithelium and neural retina. In the inner ear Rdh10 expression is specific to the endolymphatic system and later the stria vascularis, both organs being involved in endolymph homeostasis. Furthermore, in the peripheral olfactory system and the vibrissae follicles, expression is present from early stages in regions where sensory receptors appear and mesenchymal/epithelial interactions take place. The distribution of Rdh10 transcripts during brain and sensory organ development is consistent with a role of this enzyme in generating region-specific pools of retinaldehyde that will be used by the various RALDHs to refine the patterns of RA synthesis.


Alcohol Oxidoreductases/metabolism , Brain/embryology , Brain/metabolism , Sense Organs/enzymology , Sense Organs/metabolism , Age Factors , Alcohol Oxidoreductases/genetics , Animals , Ear, Inner/embryology , Ear, Inner/metabolism , Embryo, Mammalian , Eye/embryology , Eye/metabolism , Female , Gene Expression Regulation, Developmental/physiology , Mice , Olfactory Pathways/embryology , Olfactory Pathways/metabolism , Pregnancy , RNA, Messenger/metabolism , Vibrissae/embryology , Vibrissae/metabolism
9.
Dev Dyn ; 236(10): 2899-908, 2007 Oct.
Article En | MEDLINE | ID: mdl-17849458

Retinoic acid acts as a signalling molecule regulating many developmental events in vertebrates. As this molecule directly influences gene expression by activating nuclear receptors, its patterns of synthesis have to be tightly regulated, and it is well established that at least three retinaldehyde dehydrogenases (RALDHs) are involved in such tissue-specific synthesis. Whereas embryos from oviparous species can obtain retinaldehyde by metabolizing carotenoids stored in the yolk, placental embryos rely on retinol transferred from the maternal circulation. Here, we show that the gene encoding one of the murine retinol dehydrogenases, Rdh10, is expressed according to complex profiles both during early embryogenesis and organ differentiation. Many of its expression sites correlate with regions of active retinoid signalling and Raldh gene expression, especially with Raldh2 in the early presomitic and somitic mesoderm, retrocardiac and posterior branchial arch region, or later in the pleural mesothelium and kidney cortical region. Rdh10 also shows cell-type and/or regional specificity during development of the palate, teeth, and olfactory system. During limb bud development, it may participate in retinoic acid production in proximal/posterior cells, and eventually in interdigital mesenchyme. These data implicate the retinol to retinaldehyde conversion as the first step in the tissue-specific regulation of retinoic acid synthesis, at least in mammalian embryos.


Alcohol Oxidoreductases/genetics , Embryonic Development , Gene Expression Regulation, Developmental , Organogenesis , Tretinoin/metabolism , Alcohol Oxidoreductases/isolation & purification , Alcohol Oxidoreductases/metabolism , Animals , Mesoderm/metabolism , Mice , Morphogenesis , Retinoids/metabolism , Signal Transduction , Skull/embryology , Somites/metabolism
10.
Behav Brain Res ; 172(2): 307-15, 2006 Sep 25.
Article En | MEDLINE | ID: mdl-16814879

The present study examined the developmental course of the age-related hearing loss and its consequences on the expression of acoustic startle reflex (ASR) and prepulse inhibition (PPI) generated by white-noise bursts in 129S2/SvPas (129) and C57BL/6J (C57) mouse strains and their F(1) hybrids. Auditory brainstem responses (ABR), ASR and PPI were assessed at various time points: 6, 28, 41 and 94 weeks. Both parental strains showed marked ABR threshold shifts with age, with C57 mice having the most pronounced deficits. By contrast, the hybrids displayed only minor hearing loss with age. The time courses of ASR and PPI varied considerably between the mouse strains. From 6 to 41 weeks of age, ASR and PPI elicited by weak stimuli (70-90dB) increased in C57 mice, whereas the startle responses to intense stimuli (95-120dB) declined progressively. In 129 and hybrid mice, PPI levels remained relatively stable during the first year, but a progressive increase of ASR was observed in the hybrids for intense stimuli (95-120dB). When animals reached 94 weeks of age, marked deterioration of ASR was observed in all strains, while deficits in PPI were only seen in 129 and C57 mice. These findings show that the time course and the severity of the hearing loss vary considerably between 129, C57 strains and their hybrids, thus suggesting a marked heterogeneity in the genetic mechanisms underlying deafness in mice. They also demonstrate that the age-related hearing loss may have complex consequences on auditory behavioral performances depending of the severity of the deficits, the genetic background as well as the stimuli parameters.


Aging/physiology , Auditory Threshold/physiology , Hearing Loss , Neural Inhibition/physiology , Presbycusis/physiopathology , Reflex, Startle/physiology , Acoustic Stimulation , Animals , Association Learning/physiology , Hearing Loss/genetics , Hearing Loss/physiopathology , Hybridization, Genetic , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Neural Inhibition/genetics , Presbycusis/genetics , Reflex, Startle/genetics , Species Specificity , Statistics, Nonparametric
11.
J Neurobiol ; 66(7): 687-704, 2006 Jun.
Article En | MEDLINE | ID: mdl-16688766

The inner ear originates from an embryonic ectodermal placode and rapidly develops into a three-dimensional structure (the otocyst) through complex molecular and cellular interactions. Many genes and their products are involved in inner ear induction, organogenesis, and cell differentiation. Retinoic acid (RA) is an endogenous signaling molecule that may play a role during different phases of inner ear development, as shown from pathological observations. To gain insight into the function of RA during inner ear development, we have investigated the spatio-temporal expression patterns of major components of RA signaling pathway, including cellular retinoic acid binding proteins (CRABPs), cellular retinoid binding proteins (CRBPs), retinaldehyde dehydrogenases (RALDHs), catabolic enzymes (CYP26s), and nuclear receptors (RARs). Although the CrbpI, CrabpI, and -II genes are specifically expressed in the inner ear throughout development, loss-of-function studies have revealed that these proteins are dispensable for inner development and function. Several Raldh and Cyp26 gene transcripts are expressed at embryological day (E) 9.0-9.5 in the otocyst and show mainly complementary distributions in the otic epithelium and mesenchyme during following stages. From Western blot, RT-PCR, and in situ hybridization analysis, there is a low expression of Raldhs in the early otocyst at E9, while Cyp26s are strongly expressed. During the following days, there is an up-regulation of Raldhs and a down-regulation for Cyp26s. Specific RA receptor (Rar and Rxr) genes are expressed in the otocyst and during further development of the inner ear. At the otocyst stage, most of the components of the retinoid pathway are present, suggesting that the embryonic inner ear might act as an autocrine system, which is able to synthesize and metabolize RA necessary for its development. We propose a model in which two RA-dependent pathways may control inner ear ontogenesis: one indirect with RA from somitic mesoderm acting to regulate gene expression within the hindbrain neuroepithelium, and another with RA acting directly on the otocyst. Current evidence suggests that RA may regulate several genes involved in mesenchyme-epithelial interactions, thereby controlling inner ear morphogenesis. Our investigations suggest that RA signaling is a critical component not only of embryonic development, but also of postnatal maintenance of the inner ear.


Ear, Inner/embryology , Gene Expression Regulation, Developmental , Models, Biological , Retinoids/physiology , Signal Transduction/physiology , Aldehyde Oxidoreductases/metabolism , Animals , Cytochrome P-450 Enzyme System/metabolism , Humans , Receptors, Retinoic Acid/metabolism , Retinoic Acid 4-Hydroxylase
12.
J Comp Neurol ; 496(5): 643-54, 2006 Jun 10.
Article En | MEDLINE | ID: mdl-16615129

Retinoic acid signaling plays essential roles in morphogenesis and neural development through transcriptional regulation of downstream target genes. It is believed that the balance between the activities of synthesizing and metabolizing enzymes determines the amount of active retinoic acid to which a developing tissue is exposed. In this study, we investigated spatiotemporal expression patterns of four synthesizing enzymes, the retinaldehyde dehydrogenases 1, 2, 3, and 4 (Raldh1, Raldh2, Raldh3, and Raldh4) and two metabolizing enzymes (Cyp26A1 and Cyp26B1) in the embryonic and postnatal mouse inner ear by using quantitative reverse transcriptase polymerase chain reaction (RT-PCR), in situ hybridization, and Western blot analysis. Quantitative RT-PCR analysis and Western blot data revealed that the expression of CYP26s was much higher than that of Raldhs at early embryonic ages but that Cyp26 expression was downregulated during embryonic development. Conversely, the expression levels of Raldh2 and -3 increased during development and were significantly higher than the Cyp26 levels at postnatal day 20. At this age, Raldh3 was expressed predominantly in the cochlea, whereas Raldh2 was present in the vestibular end organ. At early embryonic stages, as observed by in situ hybridization, the synthesizing enzymes were expressed only in the dorsoventral epithelium of the otocyst, whereas the metabolizing enzymes were present mainly in mesenchymal cells surrounding the otic epithelium. At later stages, Raldh2, Raldh3, and Cyp26B1 were confined to the stria vascularis, spiral ganglion, and supporting cells in the cochlear and vestibular epithelia, respectively. The downregulation of Cyp26s and the upregulation of Raldhs after birth during inner ear maturation suggest tissue changes in the sensitivity to retinoic acid concentrations.


Ear, Inner/enzymology , Tretinoin/metabolism , Aldehyde Oxidoreductases/biosynthesis , Animals , Blotting, Western , Cytochrome P-450 Enzyme System/biosynthesis , Ear, Inner/embryology , Immunohistochemistry , In Situ Hybridization , Mice , Oxygenases/biosynthesis , Retinal Dehydrogenase , Retinoic Acid 4-Hydroxylase , Reverse Transcriptase Polymerase Chain Reaction
13.
Addiction ; 100(10): 1546-54, 2005 Oct.
Article En | MEDLINE | ID: mdl-16185216

AIM: To evaluate the effectiveness of the Five-Day Plan (FDP) in helping smokers to stop smoking. DESIGN: Randomized controlled trial comparing intervention and control groups. The primary outcome measure was 12 months continuous abstinence verified by expired air carbon monoxide concentration. Secondary outcome measures were self-reported abstinence at end of treatment, at 3 and 6 months. SETTING: Six towns in France. PARTICIPANTS: 228 smokers, recruited by newspaper and radio advertisement, aged 18 years or over and willing to make an attempt to quit smoking. INTERVENTION: The Intervention group (119 participants) received the FDP, which is a behavioural group-based treatment programme that has been in operation in France since 1965. It involves five consecutive evening behavioural therapy sessions. The Control group (109 participants) received a single session discussing the health effects of smoking. FINDINGS: In the Intervention group, 67 participants (56%) quit smoking at the end of the FDP. After three months this number had been reduced to 30 (25%) and to 19 (16%) by the end of one year. In the Control group these numbers were 14 (13%) and 12 (11%), respectively, after three and 12 months. When considering the rate of cessation without lapse after one year a significant difference was observed with a 13% rate in the Intervention group and 3% in the Control group (P = 0.004). CONCLUSIONS: The FDP may be considered as an aid for smokers who want to quit.


Psychotherapy, Group/methods , Smoking Cessation/methods , Adolescent , Adult , Aged , Counseling/methods , Female , France , Humans , Male , Middle Aged , Smoking Cessation/psychology
14.
Proc Natl Acad Sci U S A ; 102(13): 4789-94, 2005 Mar 29.
Article En | MEDLINE | ID: mdl-15778294

Recent studies demonstrated that stromal cells isolated from adult bone marrow have the competence of differentiating into neuronal cells in vitro and in vivo. However, the capacity of marrow stromal cells or mesenchymal stem cells (MSCs) to differentiate into diverse neuronal cell populations and the identity of molecular factors that confer marrow stromal cells with the competence of a neuronal subtype have yet to be elucidated. Here, we show that Sonic hedgehog (Shh) and retinoic acid (RA), signaling molecules secreted from tissues in the vicinity of peripheral sensory ganglia during embryogenesis, exert synergistic effects on neural-competent MSCs to express a comprehensive set of glutamatergic sensory neuron markers. Application of Shh or RA alone had little or no effect on the expression of these neuronal subtype markers. In addition, incubation of MSCs with embryonic hindbrain/somite/otocyst conditioned medium or prenatal cochlea explants promoted up-regulation of additional sensory neuron markers and process outgrowth. These results identify Shh and RA as sensory competence factors for adult pluripotent cells and establish the importance of interactions between adult pluripotent cells and the host microenvironment in neuronal subtype specification.


Bone Marrow Cells/physiology , Cell Differentiation/drug effects , Neurons/metabolism , Pluripotent Stem Cells/physiology , Trans-Activators/pharmacology , Tretinoin/pharmacology , Animals , Bone Marrow Cells/metabolism , Cell Differentiation/physiology , Colforsin , DNA Primers , Drug Synergism , Fibroblast Growth Factor 2 , Flow Cytometry , Genes, Tumor Suppressor , Hedgehog Proteins , Immunohistochemistry , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Neurons/cytology , Organ of Corti/cytology , Organ of Corti/metabolism , Patched Receptors , Pluripotent Stem Cells/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Reverse Transcriptase Polymerase Chain Reaction , Smoothened Receptor , Trans-Activators/metabolism , Tretinoin/metabolism
15.
Biol Cell ; 97(10): 787-98, 2005 Oct.
Article En | MEDLINE | ID: mdl-15730345

BACKGROUND INFORMATION: The sensing of head movement in mammals depends upon the vestibular endorgan of the inner ear, a complex structure made up of the semicircular canals and otoliths. Due to the similarity between the human and mouse vestibular apparatus, the analysis of mutant mouse is a valuable strategy aiming to identify genes involved in the control of balance and movement. RESULTS: In the course of a genome-wide chemical-mutagenesis programme, we isolated a recessive mutation, named ied (inner ear defect), which induced a severe loss-of-balance. A detailed phenotypic analysis of the mutant mice demonstrates that the balance impairment does not affect the motor activity and can be rescued, in part, by training, despite a complete agenesis of otoconia in the utricule and the saccule of the inner ear. Molecular characterization of the ied mutation revealed a transversion that affects the splicing of the second exon of the Otopetrin1 gene located on mouse chromosome 5. The consequence of such a mutation leads to a disruption of the transcription of the gene. CONCLUSIONS: The identification of the ied knock-down allele strengthens the role of the Otopetrin1 in the sensing of balance. Moreover, the rescue of the ied mutant phenotype in specific behavioural tasks confirmed that other sensory inputs or neural plasticity can compensate, to some extent, for the loss-of-balance. In the future, the ied mutant mice might be helpful to study the genetic control of the compensation strategies developed by organisms to counteract balance defects.


Aging/genetics , Alleles , Chromosome Inversion , Genes, Recessive/genetics , Membrane Proteins/genetics , Postural Balance/physiology , Acoustic Maculae/metabolism , Acoustic Maculae/pathology , Aging/metabolism , Animals , Exons/genetics , Female , Male , Membrane Proteins/metabolism , Mice , Mice, Neurologic Mutants , Mutagenesis , Physical Conditioning, Animal
16.
Gene Expr Patterns ; 4(2): 123-33, 2004 Mar.
Article En | MEDLINE | ID: mdl-15161092

Retinoic acid (RA) plays a pivotal role in patterning and differentiation of the embryonic inner ear. Despite its documented effects during embryonic development, the cellular sites that synthesize or metabolize RA in the inner ear have yet to be determined. Here we describe the distribution of three synthesizing enzymes, retinaldehyde dehydrogenases 1, 2 and 3 (RALDH1, RALDH2 and RALDH3) and two catabolizing enzymes (CYP26A1 and CYP26B1) in the mouse inner ear at embryonic day 18.5 when active cell differentiation is underway. Two detection methods, radioactive and non-radioactive in situ hybridization, were employed to elucidate the tissue distribution and cellular localization of these enzymes, respectively. All of the five enzymes examined, with the exception of CYP26A1, were expressed in both vestibular and cochlear end organs. While expression of the three RALDHs was observed in various cell types, CYP26B1 expression was found only in supporting cells of the vestibular and cochlear end organs. In the cochlea, expression domains of RALDH1-3 and CYP26B1 were complementary to one another. These results reveal specific tissue- and cellular expression patterns of RA synthesizing and catabolizing enzymes in the pre-natal inner ear, and suggest that a precise control of RA concentrations in various cell types of the inner ear is achieved by the balance between RALDHs and CYP26B1 activities.


Ear, Inner/embryology , Enzymes/genetics , Gene Expression , Tretinoin/metabolism , Aldehyde Oxidoreductases/genetics , Aldehyde Oxidoreductases/metabolism , Animals , Cell Differentiation/physiology , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Ear, Inner/metabolism , Enzymes/metabolism , Gene Expression Profiling , In Situ Hybridization , Mice , Retinal Dehydrogenase , Retinoic Acid 4-Hydroxylase
18.
Mech Dev ; 119(2): 213-23, 2002 Dec.
Article En | MEDLINE | ID: mdl-12464434

To define the signal transduction pathway of retinoic acid during inner ear development, we analyzed the expression patterns of transcripts encoding the three retinoic acid receptors (RARalpha, beta, and gamma) and related them to phenotypes resulting from single or compound inactivation of these nuclear receptors. The expression of all three RARs was observed in the developing mouse otocyst as early as embryonic day 10.5 (E10.5)-E12.5 and continued into adulthood. Expression domains of the three RAR receptors, however, were largely non-overlapping: RARalpha was predominantly expressed in the developing sensory epithelium, RARbeta in inner ear mesenchymal tissues and RARgamma in the differentiating otic capsule. In the adult, RARalpha and RARgamma transcripts were found in the organ of Corti and the spiral ganglion, whereas RARbeta transcripts were localized in mesenchyme-derived tissues. RARalpha, beta, and gamma null mutant mice, as well as RARalpha/RARbeta and RARbeta/RARgamma combined null fetuses, did not present any noticeable morphological abnormalities in the inner ear. In contrast, RARalpha/RARgamma null mutants displayed a severe hypoplasia of the otocyst that was already visible at E10.5 without any visible endolymphatic duct. The hypoplastic otocyst in RARalpha/RARgamma null mutants was characterized by impaired chondrocyte differentiation and neural development. After the second week of gestation, these mutant fetuses lacked all of the semi-circular canals and the endolymphatic duct and displayed strong anomalies in the inner ear structures. The morphological deficits were generally more severe in the cochlear portion than in the vestibular portion of the inner ear. Altogether, these results demonstrate that RARalpha and RARgamma play an essential role in the initial differentiation of otic placode derivatives, whereas RARbeta plays a minimal role in this process.


Ear, Inner/embryology , Gene Expression Regulation, Developmental , Receptors, Retinoic Acid/physiology , Animals , Cell Differentiation , Chondrocytes/cytology , Computer Simulation , In Situ Hybridization , Mice , Mice, Knockout , Mice, Mutant Strains , Neurofilament Proteins/metabolism , Neurons/cytology , Protein Structure, Tertiary , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Signal Transduction , Time Factors , Retinoic Acid Receptor gamma
...