Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Elife ; 112022 09 09.
Article in English | MEDLINE | ID: mdl-36083801

ABSTRACT

The oncogenic G-protein-coupled receptor (GPCR) Smoothened (SMO) is a key transducer of the hedgehog (HH) morphogen, which plays an essential role in the patterning of epithelial structures. Here, we examine how HH controls SMO subcellular localization and activity in a polarized epithelium using the Drosophila wing imaginal disc as a model. We provide evidence that HH promotes the stabilization of SMO by switching its fate after endocytosis toward recycling. This effect involves the sequential and additive action of protein kinase A, casein kinase I, and the Fused (FU) kinase. Moreover, in the presence of very high levels of HH, the second effect of FU leads to the local enrichment of SMO in the most basal domain of the cell membrane. Together, these results link the morphogenetic effects of HH to the apico-basal distribution of SMO and provide a novel mechanism for the regulation of a GPCR.


Subject(s)
Drosophila Proteins , Hedgehog Proteins , Animals , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Hedgehog Proteins/metabolism , Phosphorylation , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Smoothened Receptor/genetics , Smoothened Receptor/metabolism
2.
Blood Adv ; 6(16): 4691-4704, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35679464

ABSTRACT

Tumor microenvironment exerts a critical role in sustaining homing, retention, and survival of chronic lymphocytic leukemia (CLL) cells in secondary lymphoid organs. Such conditions foster immune surveillance escape and resistance to therapies. The physiological microenvironment is rendered tumor permissive by an interplay of chemokines, chemokine receptors, and adhesion molecules as well as by direct interactions between malignant lymphocytes and stromal cells, T cells, and specialized macrophages referred to as nurselike cells (NLCs). To characterize this complex interplay, we investigated the altered architecture on CLL lymph nodes biopsies and observed a dramatic loss of tissue subcompartments and stromal cell networks as compared with nonmalignant lymph nodes. A supplemental high density of CD68+ cells expressing the homeostatic chemokine CCL21 was randomly distributed. Using an imaging flow cytometry approach, CCL21 mRNA and the corresponding protein were observed in single CD68+ NLCs differentiated in vitro from CLL peripheral blood mononuclear cells. The chemokine was sequestered at the NLC membrane, helping capture of CCR7-high-expressing CLL B cells. Inhibiting the CCL21/CCR7 interaction by blocking antibodies or using therapeutic ibrutinib altered the adhesion of leukemic cells. Our results indicate NLCs as providers of an alternative source of CCL21, taking over the physiological task of follicular reticular cells, whose network is deeply altered in CLL lymph nodes. By retaining malignant B cells, CCL21 provides a protective environment for their niching and survival, thus allowing tumor evasion and resistance to treatment. These findings argue for a specific targeting or reeducation of NLCs as a new immunotherapy strategy for this disease.


Subject(s)
Chemokine CCL21 , Leukemia, Lymphocytic, Chronic, B-Cell , Chemokine CCL21/metabolism , Chemokine CCL21/pharmacology , Chemokines/metabolism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Leukocytes, Mononuclear/metabolism , Lymph Nodes/pathology , Receptors, CCR7/metabolism , Tumor Microenvironment
3.
PLoS Genet ; 16(11): e1009164, 2020 11.
Article in English | MEDLINE | ID: mdl-33175861

ABSTRACT

The chromosome translocations generating PAX3-FOXO1 and PAX7-FOXO1 chimeric proteins are the primary hallmarks of the paediatric fusion-positive alveolar subtype of Rhabdomyosarcoma (FP-RMS). Despite the ability of these transcription factors to remodel chromatin landscapes and promote the expression of tumour driver genes, they only inefficiently promote malignant transformation in vivo. The reason for this is unclear. To address this, we developed an in ovo model to follow the response of spinal cord progenitors to PAX-FOXO1s. Our data demonstrate that PAX-FOXO1s, but not wild-type PAX3 or PAX7, trigger the trans-differentiation of neural cells into FP-RMS-like cells with myogenic characteristics. In parallel, PAX-FOXO1s remodel the neural pseudo-stratified epithelium into a cohesive mesenchyme capable of tissue invasion. Surprisingly, expression of PAX-FOXO1s, similar to wild-type PAX3/7, reduce the levels of CDK-CYCLIN activity and increase the fraction of cells in G1. Introduction of CYCLIN D1 or MYCN overcomes this PAX-FOXO1-mediated cell cycle inhibition and promotes tumour growth. Together, our findings reveal a mechanism that can explain the apparent limited oncogenicity of PAX-FOXO1 fusion transcription factors. They are also consistent with certain clinical reports indicative of a neural origin of FP-RMS.


Subject(s)
Cell Transdifferentiation/genetics , Cell Transformation, Neoplastic/genetics , Oncogene Proteins, Fusion/metabolism , Paired Box Transcription Factors/metabolism , Rhabdomyosarcoma, Alveolar/genetics , Animals , Biopsy , Chick Embryo , Child , Cyclin D1/genetics , Datasets as Topic , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , N-Myc Proto-Oncogene Protein/genetics , Neoplasm Invasiveness/genetics , Neural Stem Cells/pathology , Neural Tube/cytology , Oncogene Proteins, Fusion/genetics , PAX3 Transcription Factor/genetics , PAX3 Transcription Factor/metabolism , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Paired Box Transcription Factors/genetics , Rhabdomyosarcoma, Alveolar/pathology , S Phase/genetics
4.
J Vis Exp ; (162)2020 08 27.
Article in English | MEDLINE | ID: mdl-32925873

ABSTRACT

Drosophila testes are a powerful model system for studying biological processes including stem cell biology, nuclear architecture, meiosis and sperm development. However, immunolabeling of the whole Drosophila testis is often associated with significant non-uniformity of staining due to antibody penetration. Squashed preparations only partially overcome the problem since it decreases the 3D quality of the analyses. Herein, we describe a whole-mount protocol using NP40 and heptane during fixation together with immunolabeling in liquid media. It preserves the volume suitable for confocal microscopy together with reproducible and reliable labeling. We show different examples of 3D reconstitution of spermatocyte nuclei from confocal sections. The intra- and inter-testes reproducibility allows 3D quantification and comparison of fluorescence between single cells from different genotypes. We used different components of the intranuclear MINT structure (Mad1-containing Intra Nuclear Territory) as well as two components associated with the nuclear pore complex to illustrate this protocol and its applications on the largest cells of the testis, the S4-S5 spermatocytes.


Subject(s)
Drosophila melanogaster/cytology , Imaging, Three-Dimensional , Microscopy, Confocal/methods , Spermatocytes/cytology , Testis/cytology , Animals , Cell Nucleus/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Fluorescent Antibody Technique , Male , RNA Interference , Reproducibility of Results , Spermatocytes/metabolism , TOR Serine-Threonine Kinases/metabolism , Tissue Fixation
5.
Development ; 146(14)2019 07 25.
Article in English | MEDLINE | ID: mdl-31239243

ABSTRACT

Bone morphogenetic proteins (BMPs) are secreted regulators of cell fate in several developing tissues. In the embryonic spinal cord, they control the emergence of the neural crest, roof plate and distinct subsets of dorsal interneurons. Although a gradient of BMP activity has been proposed to determine cell type identity in vivo, whether this is sufficient for pattern formation in vitro is unclear. Here, we demonstrate that exposure to BMP4 initiates distinct spatial dynamics of BMP signalling within the self-emerging epithelia of both mouse and human pluripotent stem cell-derived spinal organoids. The pattern of BMP signalling results in the stereotyped spatial arrangement of dorsal neural tube cell types, and concentration, timing and duration of BMP4 exposure modulate these patterns. Moreover, differences in the duration of competence time-windows between mouse and human account for the species-specific tempo of neural differentiation. Together, this study describes efficient methods for generating patterned subsets of dorsal interneurons in spinal organoids and supports the conclusion that graded BMP activity orchestrates the spatial organization of the dorsal neural tube cellular diversity in mouse and human.


Subject(s)
Bone Morphogenetic Protein 4/physiology , Cell Differentiation/genetics , Organoids/physiology , Smad Proteins/metabolism , Spine/cytology , Animals , Cell Lineage/genetics , Cells, Cultured , Embryo, Mammalian , Gene Expression Regulation, Developmental , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology , Interneurons/cytology , Interneurons/physiology , Mice , Neural Crest/cytology , Neural Crest/physiology , Neural Tube/cytology , Neural Tube/embryology , Neurons/cytology , Neurons/physiology , Organoids/cytology , Signal Transduction/genetics , Smad Proteins/genetics
6.
Cell Mol Life Sci ; 76(24): 4995-5009, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31139847

ABSTRACT

Protein interacting with Amyloid Precursor Protein (APP) tail 1 (PAT1) also called APPBP2 or Ara 67 has different targets such as APP or androgen receptor and is expressed in several tissues. PAT1 is known to be involved in the subcellular trafficking of its targets. We previously observed in primary neurons that PAT1 is poorly associated with APP at the cell surface. Here we show that PAT1 colocalizes with vesicles close to the cell surface labeled with Rab5, Rab4, EEA1 and Rabaptin-5 but not with Rab11 and Rab7. Moreover, PAT1 expression regulates the number of EEA1 and Rab5 vesicles, and endocytosis/recycling of the transferrin receptor. In addition, low levels of PAT1 decrease the size of transferrin-colocalized EEA1 vesicles with time following transferrin uptake. Finally, overexpression of the APP binding domain to PAT1 is sufficient to compromise endocytosis. Altogether, these data suggest that PAT1 is a new actor in transferrin early endocytosis. Whether this new function of PAT1 may have consequences in pathology remains to be determined.


Subject(s)
Amino Acid Transport Systems/genetics , Symporters/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Cell Membrane/genetics , Cell Membrane/metabolism , Endocytosis/genetics , Endosomes/genetics , Endosomes/metabolism , Gene Expression Regulation , Humans , Mice , Neurons/metabolism , Protein Transport , Receptors, Androgen/genetics , Vesicular Transport Proteins/genetics , rab GTP-Binding Proteins/genetics , rab4 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins
7.
Sci Rep ; 8(1): 17121, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30459446

ABSTRACT

The spatial organization of cells depends on coordination between cytoskeletal systems and intracellular organelles. The Arf1 small G protein and its activator GBF1 are important regulators of Golgi organization, maintaining its morphology and function. Here we show that GBF1 and its substrate Arf1 regulate the spatial organization of mitochondria in a microtubule-dependent manner. Miro is a mitochondrial membrane protein that interacts through adaptors with microtubule motor proteins such as cytoplasmic dynein, the major microtubule minus end directed motor. We demonstrate a physical interaction between GBF1 and Miro, and also between the active GTP-bound form of Arf1 and Miro. Inhibition of GBF1, inhibition of Arf1 activation, or overexpression of Miro, caused a collapse of the mitochondrial network towards the centrosome. The change in mitochondrial morphology upon GBF1 inhibition was due to a two-fold increase in the time engaged in retrograde movement compared to control conditions. Electron tomography revealed that GBF1 inhibition also resulted in larger mitochondria with more complex morphology. Miro silencing or drug inhibition of cytoplasmic dynein activity blocked the GBF1-dependent repositioning of mitochondria. Our results show that blocking GBF1 function promotes dynein- and Miro-dependent retrograde mitochondrial transport along microtubules towards the microtubule-organizing center, where they form an interconnected network.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , rho GTP-Binding Proteins/metabolism , ADP-Ribosylation Factor 1/genetics , Brefeldin A/pharmacology , Cells, Cultured , Dyneins/metabolism , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , HeLa Cells , Humans , Microtubules/metabolism , Mitochondria/drug effects , Mitochondrial Proteins/genetics , Mutation , Pyridines/pharmacology , Quinolines/pharmacology , RNA Interference , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , rho GTP-Binding Proteins/genetics
8.
Curr Biol ; 25(19): 2466-78, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26387718

ABSTRACT

In the neocortex, higher-order areas are essential to integrate sensory-motor information and have expanded in size during evolution. How higher-order areas are specified, however, remains largely unknown. Here, we show that the migration and distribution of early-born neurons, the Cajal-Retzius cells (CRs), controls the size of higher-order areas in the mouse somatosensory, auditory, and visual cortex. Using live imaging, genetics, and in silico modeling, we show that subtype-specific differences in the onset, speed, and directionality of CR migration determine their differential invasion of the developing cortical surface. CR migration speed is cell autonomously modulated by vesicle-associated membrane protein 3 (VAMP3), a classically non-neuronal mediator of endosomal recycling. Increasing CR migration speed alters their distribution in the developing cerebral cortex and leads to an expansion of postnatal higher-order areas and congruent rewiring of thalamo-cortical input. Our findings thus identify novel roles for neuronal migration and VAMP3-dependent vesicular trafficking in cortical wiring.


Subject(s)
Cell Movement/physiology , Cerebral Cortex/physiology , Interstitial Cells of Cajal/physiology , Neocortex/physiology , Neurons/metabolism , Animals , Cerebral Cortex/cytology , Interstitial Cells of Cajal/cytology , Mice , Mice, Transgenic , Models, Biological , Neocortex/cytology , Neocortex/metabolism , Vesicle-Associated Membrane Protein 3/metabolism
9.
Nanoscale ; 7(10): 4529-44, 2015 Mar 14.
Article in English | MEDLINE | ID: mdl-25685900

ABSTRACT

Inhalation is the most frequent route of unintentional exposure to nanoparticles (NPs). Our aim was to quantify the translocation of different metallic NPs across human bronchial epithelial cells and to determine the factors influencing this translocation. Calu-3 cells forming a tight epithelial barrier when grown on a porous membrane in a two compartment chamber were exposed to fluorescently labelled NPs to quantify the NP translocation. NP translocation and uptake by cells were also studied by confocal and transmission electron microscopy. Translocation was characterized according to NP size (16, 50, or 100 nm), surface charge (negative or positive SiO2), composition (SiO2 or TiO2), presence of proteins or phospholipids and in an inflammatory context. Our results showed that NPs can translocate through the Calu-3 monolayer whatever their composition (SiO2 or TiO2), but this translocation was increased for the smallest and negatively charged NPs. Translocation was not associated with an alteration of the integrity of the epithelial monolayer, suggesting a transcytosis of the internalized NPs. By modifying the NP corona, the ability of NPs to cross the epithelial barrier differed depending on their intrinsic properties, making positively charged NPs more prone to translocate. NP translocation can be amplified by using agents known to open tight junctions and to allow paracellular passage. NP translocation was also modulated when mimicking an inflammatory context frequently found in the lungs, altering the epithelial integrity and inducing transient tight junction opening. This in vitro evaluation of NP translocation could be extended to other inhaled NPs to predict their biodistribution.


Subject(s)
Bronchi/metabolism , Nanoparticles , Respiratory Mucosa/metabolism , Silicon Dioxide/pharmacokinetics , Titanium/pharmacokinetics , Biological Transport, Active , Cell Line, Tumor , Humans , Silicon Dioxide/pharmacology , Titanium/pharmacology
10.
Neurobiol Aging ; 35(5): 958-68, 2014 May.
Article in English | MEDLINE | ID: mdl-24262202

ABSTRACT

Caspase cleaved amyloid precursor protein (APPcc) and SET are increased and mislocalized in the neuronal cytoplasm in Alzheimer Disease (AD) brains. Translocated SET to the cytoplasm can induce tau hyperphosphorylation. To elucidate the putative relationships between mislocalized APPcc and SET, we studied their level and distribution in the hippocampus of 5 controls, 3 Down syndrome and 10 Alzheimer patients. In Down syndrome and Alzheimer patients, APPcc and SET levels were increased in CA1 and the frequency of both localizations in the neuronal cytoplasm was high in CA1, and low in CA4. As the increase of APPcc is already present at early stages of AD, we overexpressed APPcc in CA1 and the dentate gyrus neurons of adult mice with a lentiviral construct. APPcc overexpression in CA1 and not in the dentate gyrus induced endogenous SET translocation and tau hyperphosphorylation. These data suggest that increase in APPcc in CA1 neurons could be an early event leading to the translocation of SET and the progression of AD through tau hyperphosphorylation.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/metabolism , CA1 Region, Hippocampal/metabolism , Down Syndrome/genetics , Histone Chaperones/metabolism , Transcription Factors/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Animals , CA1 Region, Hippocampal/cytology , Caspases/physiology , Cytoplasm/metabolism , DNA-Binding Proteins , Dentate Gyrus/cytology , Dentate Gyrus/metabolism , Disease Progression , Down Syndrome/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neurons/metabolism , Phosphorylation , tau Proteins/metabolism
11.
J Cell Sci ; 126(Pt 20): 4794-805, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23943872

ABSTRACT

Lipid droplet metabolism and secretory pathway trafficking both require activation of the Arf1 small G protein. The spatiotemporal regulation of Arf1 activation is mediated by guanine nucleotide exchange factors (GEFs) of the GBF and BIG families, but the mechanisms of their localization to multiple sites within cells are poorly understood. Here we show that GBF1 has a lipid-binding domain (HDS1) immediately downstream of the catalytic Sec7 domain, which mediates association with both lipid droplets and Golgi membranes in cells, and with bilayer liposomes and artificial droplets in vitro. An amphipathic helix within HDS1 is necessary and sufficient for lipid binding, both in vitro and in cells. The HDS1 domain of GBF1 is stably associated with lipid droplets in cells, and the catalytic Sec7 domain inhibits this potent lipid-droplet-binding capacity. Additional sequences upstream of the Sec7 domain-HDS1 tandem are required for localization to Golgi membranes. This mechanism provides insight into crosstalk between lipid droplet function and secretory trafficking.


Subject(s)
Golgi Apparatus/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Inclusion Bodies/metabolism , ADP-Ribosylation Factor 1/genetics , ADP-Ribosylation Factor 1/metabolism , Animals , COS Cells , Chlorocebus aethiops , Golgi Apparatus/genetics , Guanine Nucleotide Exchange Factors/genetics , HeLa Cells , Humans , Inclusion Bodies/genetics , Lipid Metabolism , Plasmids , Protein Transport , Secretory Pathway , Transfection
12.
J Mol Biol ; 425(23): 4837-44, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-23810902

ABSTRACT

We analyzed formation of single-stranded DNA (ssDNA) related to SOS induction in nalidixilate (Nal)-treated Escherichia coli, using immunofluorescence microscopy accompanied by computer analysis. We found enhancement of both ssDNA concentrations and cells having ssDNA foci that often localized around cellpoles. Analyzing several mutants deficient in DNA repair or replication, we found, after Nal treatment, that recN, recA, uvrD and dnaB failed to increase ssDNA concentration and that recG and particularly ruvA only partially enhanced it. In Nal-treated recB mutant, despite its failure in SOS induction, ssDNA foci positive cells increased with a slight enhancement of its concentration. These observations suggest the existence of a cellular process that sequesters genotoxic ssDNA as inert form, offering a new concept for SOS suppressor genes action.


Subject(s)
DNA Replication , DNA, Bacterial/metabolism , DNA, Single-Stranded/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Anti-Bacterial Agents/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Escherichia coli/drug effects , Gene Knockout Techniques , Image Processing, Computer-Assisted , Microscopy, Fluorescence , Nalidixic Acid/metabolism , SOS Response, Genetics
13.
Part Fibre Toxicol ; 10: 2, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23388071

ABSTRACT

BACKGROUND: The uptake of nanoparticles (NPs) by cells remains to be better characterized in order to understand the mechanisms of potential NP toxicity as well as for a reliable risk assessment. Real NP uptake is still difficult to evaluate because of the adsorption of NPs on the cellular surface. RESULTS: Here we used two approaches to distinguish adsorbed fluorescently labeled NPs from the internalized ones. The extracellular fluorescence was either quenched by Trypan Blue or the uptake was analyzed using imaging flow cytometry. We used this novel technique to define the inside of the cell to accurately study the uptake of fluorescently labeled (SiO2) and even non fluorescent but light diffracting NPs (TiO2). Time course, dose-dependence as well as the influence of surface charges on the uptake were shown in the pulmonary epithelial cell line NCI-H292. By setting up an integrative approach combining these flow cytometric analyses with confocal microscopy we deciphered the endocytic pathway involved in SiO2 NP uptake. Functional studies using energy depletion, pharmacological inhibitors, siRNA-clathrin heavy chain induced gene silencing and colocalization of NPs with proteins specific for different endocytic vesicles allowed us to determine macropinocytosis as the internalization pathway for SiO2 NPs in NCI-H292 cells. CONCLUSION: The integrative approach we propose here using the innovative imaging flow cytometry combined with confocal microscopy could be used to identify the physico-chemical characteristics of NPs involved in their uptake in view to redesign safe NPs.


Subject(s)
Endocytosis , Epithelial Cells/drug effects , Flow Cytometry/methods , Microscopy, Confocal/methods , Nanoparticles , Silicon Dioxide , Adsorption , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Endocytosis/physiology , Epithelial Cells/metabolism , Fluorescent Dyes/chemistry , Humans , Nanoparticles/chemistry , Nanoparticles/toxicity , Particle Size , Silicon Dioxide/chemistry , Silicon Dioxide/toxicity , Surface Properties , Trypan Blue/chemistry
14.
PLoS Genet ; 8(11): e1003081, 2012.
Article in English | MEDLINE | ID: mdl-23209438

ABSTRACT

Cardiac aging is a complex process, which is influenced by both environmental and genetic factors. Deciphering the mechanisms involved in heart senescence therefore requires identifying the molecular pathways that are affected by age in controlled environmental and genetic conditions. We describe a functional genomic investigation of the genetic control of cardiac senescence in Drosophila. Molecular signatures of heart aging were identified by differential transcriptome analysis followed by a detailed bio-informatic analysis. This approach implicated the JNK/dJun pathway and the transcription factor Vri/dNFIL3 in the transcription regulatory network involved in cardiac senescence and suggested the possible involvement of oxidative stress (OS) in the aging process. To validate these predictions, we developed a new in vivo assay to analyze heart performance in various contexts of adult heart-specific gene overexpression and inactivation. We demonstrate that, as in mammals, OS plays a central role in cardiac senescence, and we show that pharmacological interventions impinging on OS slow heart senescence. These observations strengthen the idea that cardiac aging is controlled by evolutionarily conserved mechanisms, further validating Drosophila as a model to study cardiac senescence. In addition, we demonstrate that Vri, the ortholog of the vertebrate NFIL3/E4B4 transcription factor, is a major genetic regulator of cardiac aging. Vri overexpression leads to major heart dysfunctions, but its loss of function significantly reduces age-related cardiac dysfunctions. Furthermore, we unambiguously show that the JNK/AP1 pathway, the role of which in cardiac aging in mammals is controversial, is activated during cardiac aging and has a detrimental effect on cardiac senescence. This data-driven functional genomic analysis therefore led to the identification of key components of the Gene Regulatory Network of cardiac aging in Drosophila and may prompt to investigate the involvement of their counterparts in the cardiac aging process in mammals.


Subject(s)
Aging , Drosophila Proteins , Drosophila melanogaster , Heart/physiology , MAP Kinase Signaling System/genetics , Transcription Factors , Aging/genetics , Aging/metabolism , Aging/physiology , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Gene Expression Regulation , Humans , Oxidative Stress , Transcription Factors/genetics , Transcription Factors/metabolism
15.
J Neurosci ; 32(26): 9007-22, 2012 Jun 27.
Article in English | MEDLINE | ID: mdl-22745500

ABSTRACT

Vezatin is an integral membrane protein associated with cell-cell adhesion complex and actin cytoskeleton. It is expressed in the developing and mature mammalian brain, but its neuronal function is unknown. Here, we show that Vezatin localizes in spines in mature mouse hippocampal neurons and codistributes with PSD95, a major scaffolding protein of the excitatory postsynaptic density. Forebrain-specific conditional ablation of Vezatin induced anxiety-like behavior and impaired cued fear-conditioning memory response. Vezatin knock-down in cultured hippocampal neurons and Vezatin conditional knock-out in mice led to a significantly increased proportion of stubby spines and a reduced proportion of mature dendritic spines. PSD95 remained tethered to presynaptic terminals in Vezatin-deficient hippocampal neurons, suggesting that the reduced expression of Vezatin does not compromise the maintenance of synaptic connections. Accordingly, neither the amplitude nor the frequency of miniature EPSCs was affected in Vezatin-deficient hippocampal neurons. However, the AMPA/NMDA ratio of evoked EPSCs was reduced, suggesting impaired functional maturation of excitatory synapses. These results suggest a role of Vezatin in dendritic spine morphogenesis and functional synaptic maturation.


Subject(s)
Carrier Proteins/metabolism , Dendritic Spines/physiology , Excitatory Postsynaptic Potentials/physiology , Membrane Proteins/metabolism , Neurogenesis/physiology , Neurons/ultrastructure , Synapses/physiology , Animals , Animals, Newborn , Anxiety/genetics , Avoidance Learning/physiology , Cadherins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cells, Cultured , Conditioning, Psychological/physiology , Electric Stimulation , Embryo, Mammalian , Excitatory Postsynaptic Potentials/genetics , Exploratory Behavior/physiology , Eye Proteins/genetics , Fear/physiology , Gene Expression Regulation/genetics , Glutamate Decarboxylase/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , In Vitro Techniques , Male , Maze Learning/physiology , Membrane Proteins/deficiency , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Microtubule-Associated Proteins/metabolism , N-Methylaspartate/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , RNA, Messenger , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Silver Staining , Statistics, Nonparametric , Synapses/genetics , Synaptosomes/metabolism , Transfection , Vesicle-Associated Membrane Protein 2/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...