Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
2.
Acta Neuropathol ; 140(5): 737-764, 2020 11.
Article in English | MEDLINE | ID: mdl-32642868

ABSTRACT

Impaired neuronal proteostasis is a salient feature of many neurodegenerative diseases, highlighting alterations in the function of the endoplasmic reticulum (ER). We previously reported that targeting the transcription factor XBP1, a key mediator of the ER stress response, delays disease progression and reduces protein aggregation in various models of neurodegeneration. To identify disease modifier genes that may explain the neuroprotective effects of XBP1 deficiency, we performed gene expression profiling of brain cortex and striatum of these animals and uncovered insulin-like growth factor 2 (Igf2) as the major upregulated gene. Here, we studied the impact of IGF2 signaling on protein aggregation in models of Huntington's disease (HD) as proof of concept. Cell culture studies revealed that IGF2 treatment decreases the load of intracellular aggregates of mutant huntingtin and a polyglutamine peptide. These results were validated using induced pluripotent stem cells (iPSC)-derived medium spiny neurons from HD patients and spinocerebellar ataxia cases. The reduction in the levels of mutant huntingtin was associated with a decrease in the half-life of the intracellular protein. The decrease in the levels of abnormal protein aggregation triggered by IGF2 was independent of the activity of autophagy and the proteasome pathways, the two main routes for mutant huntingtin clearance. Conversely, IGF2 signaling enhanced the secretion of soluble mutant huntingtin species through exosomes and microvesicles involving changes in actin dynamics. Administration of IGF2 into the brain of HD mice using gene therapy led to a significant decrease in the levels of mutant huntingtin in three different animal models. Moreover, analysis of human postmortem brain tissue and blood samples from HD patients showed a reduction in IGF2 level. This study identifies IGF2 as a relevant factor deregulated in HD, operating as a disease modifier that buffers the accumulation of abnormal protein species.


Subject(s)
Huntington Disease/metabolism , Huntington Disease/pathology , Insulin-Like Growth Factor II/metabolism , Protein Aggregation, Pathological/metabolism , Animals , Humans , Insulin-Like Growth Factor II/pharmacology , Mice , Mice, Transgenic , Protein Aggregates/drug effects
3.
J Neurosci ; 36(27): 7298-313, 2016 07 06.
Article in English | MEDLINE | ID: mdl-27383602

ABSTRACT

UNLABELLED: Rab35 is a key protein for cargo loading in the recycling endosome. In neuronal immortalized cells, Rab35 promotes neurite differentiation. Here we describe that Rab35 favors axon elongation in rat primary neurons in an activity-dependent manner. In addition, we show that the p53-related protein kinase (PRPK) negatively regulates axonal elongation by reducing Rab35 protein levels through the ubiquitin-proteasome degradation pathway. PRPK-induced Rab35 degradation is regulated by its interaction with microtubule-associated protein 1B (MAP1B), a microtubule stabilizing binding protein essential for axon elongation. Consistently, axon defects found in MAP1B knock-out neurons were reversed by Rab35 overexpression or PRPK inactivation suggesting an epistatic relationship among these proteins. These results define a novel mechanism to support axonal elongation, by which MAP1B prevents PRPK-induced Rab35 degradation. Such a mechanism allows Rab35-mediated axonal elongation and connects the regulation of actin dynamics with membrane trafficking. In addition, our study reveals for the first time that the ubiquitin-proteasome degradation pathway regulates a Rab GTPase. SIGNIFICANCE STATEMENT: Rab35 is required for axonal outgrowth. We define that its protein levels are negatively regulated by p53-related protein kinase (PRPK). We show that microtubule-associated protein 1B (MAP1B) interacts with PRPK, preventing PRPK-dependent Rab35 proteasome degradation. We demonstrate that Rab35 regulates Cdc42 activity in neurons. This is the first evidence showing that a Rab protein is regulated by degradation dependent on the ubiquitin-proteasome system.


Subject(s)
Axons/physiology , Microtubule-Associated Proteins/metabolism , Neurons/cytology , Protein Serine-Threonine Kinases/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Cell Line, Transformed , Cells, Cultured , Chlorocebus aethiops , Embryo, Mammalian , Gene Expression Regulation/genetics , Hippocampus/cytology , Mice , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/metabolism , Oligodeoxyribonucleotides, Antisense/pharmacology , Protein Serine-Threonine Kinases/genetics , Proteolysis/drug effects , RNA, Small Interfering/pharmacology , Rats , cdc42 GTP-Binding Protein/metabolism , rab GTP-Binding Proteins/genetics
4.
J Neurosci ; 36(43): 11107-11119, 2016 10 26.
Article in English | MEDLINE | ID: mdl-27798190

ABSTRACT

Physiological levels of ROS support neurite outgrowth and axonal specification, but the mechanisms by which ROS are able to shape neurons remain unknown. Ca2+, a broad intracellular second messenger, promotes both Rac1 activation and neurite extension. Ca2+ release from the endoplasmic reticulum, mediated by both the IP3R1 and ryanodine receptor (RyR) channels, requires physiological ROS levels that are mainly sustained by the NADPH oxidase (NOX) complex. In this work, we explore the contribution of the link between NOX and RyR-mediated Ca2+ release toward axonal specification of rat hippocampal neurons. Using genetic approaches, we find that NOX activation promotes both axonal development and Rac1 activation through a RyR-mediated mechanism, which in turn activates NOX through Rac1, one of the NOX subunits. Collectively, these data suggest a feedforward mechanism that integrates both NOX activity and RyR-mediated Ca2+ release to support cellular mechanisms involved in axon development. SIGNIFICANCE STATEMENT: High levels of ROS are frequently associated with oxidative stress and disease. In contrast, physiological levels of ROS, mainly sustained by the NADPH oxidase (NOX) complex, promote neuronal development and axonal growth. However, the mechanisms by which ROS shape neurons have not been described. Our work suggests that NOX-derived ROS promote axonal growth by regulating Rac1 activity, a molecular determinant of axonal growth, through a ryanodine receptor (RyR)-mediated Ca2+ release mechanism. In addition, Rac1, one of the NOX subunits, was activated after RyR-mediated Ca2+ release, suggesting a feedforward mechanism between NOX and RyR. Collectively, our data suggest a novel mechanism that is instrumental in sustaining physiological levels of ROS required for axonal growth of hippocampal neurons.


Subject(s)
Axon Guidance/physiology , Calcium Signaling/physiology , Feedback, Physiological/physiology , NADPH Oxidases/metabolism , Neurons/physiology , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Female , Gene Expression Regulation, Developmental/physiology , Hippocampus/physiology , Hippocampus/ultrastructure , Male , Neuronal Plasticity/physiology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
5.
Retrovirology ; 14(1): 54, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29179726

ABSTRACT

BACKGROUND: Stable insertion of the retroviral DNA genome into host chromatin requires the functional association between the intasome (integrase·viral DNA complex) and the nucleosome. The data from the literature suggest that direct protein-protein contacts between integrase and histones may be involved in anchoring the intasome to the nucleosome. Since histone tails are candidates for interactions with the incoming intasomes we have investigated whether they could participate in modulating the nucleosomal integration process. RESULTS: We show here that histone tails are required for an optimal association between HIV-1 integrase (IN) and the nucleosome for efficient integration. We also demonstrate direct interactions between IN and the amino-terminal tail of human histone H4 in vitro. Structure/function studies enabled us to identify amino acids in the carboxy-terminal domain of IN that are important for this interaction. Analysis of the nucleosome-binding properties of catalytically active mutated INs confirmed that their ability to engage the nucleosome for integration in vitro was affected. Pseudovirus particles bearing mutations that affect the IN/H4 association also showed impaired replication capacity due to altered integration and re-targeting of their insertion sites toward dynamic regions of the chromatin with lower nucleosome occupancy. CONCLUSIONS: Collectively, our data support a functional association between HIV-1 IN and histone tails that promotes anchoring of the intasome to nucleosomes and optimal integration into chromatin.


Subject(s)
HIV Integrase/metabolism , HIV-1/metabolism , Histones/metabolism , Nucleosomes/metabolism , Virus Integration , Cell Line, Transformed , Chromatin/virology , DNA, Viral/metabolism , HEK293 Cells , HIV-1/genetics , Histones/chemistry , Host-Parasite Interactions/physiology , Humans , Protein Binding
6.
J Neurosci ; 35(32): 11315-29, 2015 Aug 12.
Article in English | MEDLINE | ID: mdl-26269639

ABSTRACT

Acquisition of neuronal polarity is a complex process involving cellular and molecular events. The second messenger cAMP is involved in axonal specification through activation of protein kinase A. However, an alternative cAMP-dependent mechanism involves the exchange protein directly activated by cAMP (EPAC), which also responds to physiological changes in cAMP concentration, promoting activation of the small Rap GTPases. Here, we present evidence that EPAC signaling contributes to axon specification and elongation. In primary rat hippocampal neurons, EPAC isoforms were expressed differentially during axon specification. Furthermore, 8-pCPT, an EPAC pharmacological activator, and genetic manipulations of EPAC in neurons induced supernumerary axons indicative of Rap1b activation. Moreover, 8-pCPT-treated neurons expressed ankyrin G and other markers of mature axons such as synaptophysin and axonal accumulation of vGLUT1. In contrast, pharmacological inhibition of EPAC delayed neuronal polarity. Genetic manipulations to inactivate EPAC1 using either shRNA or neurons derived from EPAC1 knock-out (KO) mice led to axon elongation and polarization defects. Interestingly, multiaxonic neurons generated by 8-pCPT treatments in wild-type neurons were not found in EPAC1 KO mice neurons. Altogether, these results propose that EPAC signaling is an alternative and complementary mechanism for cAMP-dependent axon determination. SIGNIFICANCE STATEMENT: This study identifies the guanine exchange factor responsible for Rap1b activation during neuronal polarization and provides an alternate explanation for cAMP-dependent acquisition of neuronal polarity.


Subject(s)
Cell Polarity/physiology , Guanine Nucleotide Exchange Factors/metabolism , Hippocampus/metabolism , Neurons/metabolism , rap GTP-Binding Proteins/metabolism , Animals , Axons/metabolism , Hippocampus/cytology , Mice , Neurons/cytology , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
7.
EMBO J ; 31(10): 2322-35, 2012 May 16.
Article in English | MEDLINE | ID: mdl-22510886

ABSTRACT

Adaptation to endoplasmic reticulum (ER) stress depends on the activation of the unfolded protein response (UPR) stress sensor inositol-requiring enzyme 1α (IRE1α), which functions as an endoribonuclease that splices the mRNA of the transcription factor XBP-1 (X-box-binding protein-1). Through a global proteomic approach we identified the BCL-2 family member PUMA as a novel IRE1α interactor. Immun oprecipitation experiments confirmed this interaction and further detected the association of IRE1α with BIM, another BH3-only protein. BIM and PUMA double-knockout cells failed to maintain sustained XBP-1 mRNA splicing after prolonged ER stress, resulting in early inactivation. Mutation in the BH3 domain of BIM abrogated the physical interaction with IRE1α, inhibiting its effects on XBP-1 mRNA splicing. Unexpectedly, this regulation required BCL-2 and was antagonized by BAD or the BH3 domain mimetic ABT-737. The modulation of IRE1α RNAse activity by BH3-only proteins was recapitulated in a cell-free system suggesting a direct regulation. Moreover, BH3-only proteins controlled XBP-1 mRNA splicing in vivo and affected the ER stress-regulated secretion of antibodies by primary B cells. We conclude that a subset of BCL-2 family members participates in a new UPR-regulatory network, thus assuming apoptosis-unrelated functions.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Endoribonucleases/metabolism , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Unfolded Protein Response , Animals , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Gene Knockout Techniques , Immunoprecipitation , Membrane Proteins/genetics , Mice , Protein Binding , Protein Interaction Mapping , Proteome/analysis , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics
8.
Pflugers Arch ; 466(11): 2113-26, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24566975

ABSTRACT

Microtubule-associated protein B is a cytoskeleton protein consisting of heavy and light (LC) chains that play important roles in the regulation of neuronal morphogenesis and function. LC1 is also well known to interact with diverse ionotropic receptors at postsynapse. Much less is known, however, regarding the role of LC1 at presynaptic level where voltage-gated N-type Ca(2+) channels couple membrane depolarization to neurotransmitter release. Here, we investigated whether LC1 interacts with the N-type channels. Co-localization analysis revealed spatial proximity of the two proteins in hippocampal neurons. The interaction between LC1 and the N-type channel was demonstrated using co-immunoprecipitation experiments and in vitro pull-down assays. Detailed biochemical analysis suggested that the interaction occurs through the N-terminal of LC1 and the C-terminal of the pore-forming CaVα1 subunit of the channels. Patch-clamp studies in HEK-293 cells revealed a significant decrease in N-type currents upon LC1 expression, without apparent changes in kinetics. Recordings performed in the presence of MG132 prevented the actions of LC1 suggesting enhanced channel proteasomal degradation. Interestingly, using the yeast two-hybrid system and immunoprecipitation assays in HEK-293 cells, we revealed an interaction between LC1 and the ubiquitin-conjugating enzyme UBE2L3. Furthermore, we found that the LC1/UBE2L3 complex could interact with the N-type channels, suggesting that LC1 may act as a scaffold protein to increase UBE2L3-mediated channel ubiquitination. Together these results revealed a novel functional coupling between LC1 and the N-type channels.


Subject(s)
Calcium Channels, N-Type/metabolism , Cell Membrane/metabolism , Microtubule-Associated Proteins/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination/physiology , Cells, Cultured , HEK293 Cells , Hippocampus/metabolism , Humans , Immunoprecipitation/methods , Neurons/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Two-Hybrid System Techniques , Ubiquitin/metabolism
9.
BMC Struct Biol ; 13: 14, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23844665

ABSTRACT

BACKGROUND: Retroviral integrases (INs) catalyze the integration of viral DNA in the chromosomal DNA of the infected cell. This reaction requires the multimerization of IN to coordinate a nucleophilic attack of the 3' ends of viral DNA at two staggered phosphodiester bonds on the recipient DNA. Several models indicate that a tetramer of IN would be required for two-end concerted integration. Complementation assays have shown that the N-terminal domain (NTD) of integrase is essential for concerted integration, contributing to the formation of a multimer through protein-protein interaction. The isolated NTD of Mo-MLV integrase behave as a dimer in solution however the structure of the dimer in solution is not known. RESULTS: In this work, crosslinking and mass spectrometry were used to identify regions involved in the dimerization of the isolated Mo-MLV NTD. The distances between the crosslinked lysines within the monomer are in agreement with the structure of the NTD monomer found in 3NNQ. The intermolecular crosslinked peptides corresponding to Lys 20-Lys 31, Lys 24-Lys 24 and Lys 68-Lys 88 were identified. The 3D coordinates of 3NNQ were used to derive a theoretical structure of the NTD dimer with the suite 3D-Dock, based on shape and electrostatics complementarity, and filtered with the distance restraints determined in the crosslinking experiments. CONCLUSIONS: The crosslinking results are consistent with the monomeric structure of NTD in 3NNQ, but for the dimer, in our model both polypeptides are oriented in parallel with each other and the contacting areas between the monomers would involve the interactions between helices 1 and helices 3 and 4.


Subject(s)
Integrases/chemistry , Moloney murine leukemia virus/enzymology , Viral Proteins/chemistry , Amino Acid Sequence , Animals , Chromatography, High Pressure Liquid , Dimerization , Integrases/metabolism , Mass Spectrometry , Mice , Molecular Docking Simulation , Molecular Sequence Data , Peptides/analysis , Protein Structure, Tertiary , Sequence Alignment , Static Electricity , Viral Proteins/metabolism
10.
Nat Cell Biol ; 20(8): 942-953, 2018 08.
Article in English | MEDLINE | ID: mdl-30013108

ABSTRACT

Maintenance of endoplasmic reticulum (ER) proteostasis is controlled by a signalling network known as the unfolded protein response (UPR). Here, we identified filamin A as a major binding partner of the ER stress transducer IRE1α. Filamin A is an actin crosslinking factor involved in cytoskeleton remodelling. We show that IRE1α controls actin cytoskeleton dynamics and affects cell migration upstream of filamin A. The regulation of cytoskeleton dynamics by IRE1α is independent of its canonical role as a UPR mediator, serving instead as a scaffold that recruits and regulates filamin A. Targeting IRE1α expression in mice affected normal brain development, generating a phenotype resembling periventricular heterotopia, a disease linked to the loss of function of filamin A. IRE1α also modulated cell movement and cytoskeleton dynamics in fly and zebrafish models. This study unveils an unanticipated biological function of IRE1α in cell migration, whereby filamin A operates as an interphase between the UPR and the actin cytoskeleton.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Movement , Endoribonucleases/metabolism , Fibroblasts/metabolism , Filamins/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Endoribonucleases/deficiency , Endoribonucleases/genetics , Evolution, Molecular , Female , Filamins/genetics , HEK293 Cells , Humans , Kinetics , Male , Mice , Mice, Knockout , Neurons/pathology , Periventricular Nodular Heterotopia/genetics , Periventricular Nodular Heterotopia/metabolism , Periventricular Nodular Heterotopia/pathology , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Unfolded Protein Response , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
Sci Rep ; 6: 30069, 2016 07 18.
Article in English | MEDLINE | ID: mdl-27425640

ABSTRACT

Microtubule-associated protein 1B (MAP1B) is expressed predominantly during the early stages of development of the nervous system, where it regulates processes such as axonal guidance and elongation. Nevertheless, MAP1B expression in the brain persists in adult stages, where it participates in the regulation of the structure and physiology of dendritic spines in glutamatergic synapses. Moreover, MAP1B expression is also found in presynaptic synaptosomal preparations. In this work, we describe a presynaptic phenotype in mature neurons derived from MAP1B knockout (MAP1B KO) mice. Mature neurons express MAP1B, and its deficiency does not alter the expression levels of a subgroup of other synaptic proteins. MAP1B KO neurons display a decrease in the density of presynaptic and postsynaptic terminals, which involves a reduction in the density of synaptic contacts, and an increased proportion of orphan presynaptic terminals. Accordingly, MAP1B KO neurons present altered synaptic vesicle fusion events, as shown by FM4-64 release assay, and a decrease in the density of both synaptic vesicles and dense core vesicles at presynaptic terminals. Finally, an increased proportion of excitatory immature symmetrical synaptic contacts in MAP1B KO neurons was detected. Altogether these results suggest a novel role for MAP1B in presynaptic structure and physiology regulation in vitro.


Subject(s)
Dendritic Spines/physiology , Hippocampus/metabolism , Microtubule-Associated Proteins/genetics , Neurons/cytology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Astrocytes/cytology , Cells, Cultured , Coculture Techniques , Dendritic Spines/metabolism , Excitatory Amino Acids/metabolism , Hippocampus/cytology , Hippocampus/embryology , Mice , Mice, Knockout , Pyridinium Compounds , Quaternary Ammonium Compounds , RNA Interference , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Secretory Vesicles/metabolism , Synapses/metabolism , Synaptic Vesicles/metabolism
14.
PLoS One ; 7(12): e53123, 2012.
Article in English | MEDLINE | ID: mdl-23300879

ABSTRACT

Microtubule-associated protein 1B (MAP1B) is a neuronal protein involved in the stabilization of microtubules both in the axon and somatodendritic compartments. Acute, genetic inactivation of MAP1B leads to delayed axonal outgrowth, most likely due to changes in the post-translational modification of tubulin subunits, which enhances microtubule polymerization. Furthermore, MAP1B deficiency is accompanied by abnormal actin microfilament polymerization and dramatic changes in the activity of small GTPases controlling the actin cytoskeleton. In this work, we showed that MAP1B interacts with a guanine exchange factor, termed Tiam1, which specifically activates Rac1. These proteins co-segregated in neurons, and interact in both heterologous expression systems and primary neurons. We dissected the molecular domains involved in the MAP1B-Tiam1 interaction, and demonstrated that pleckstrin homology (PH) domains in Tiam1 are responsible for MAP1B binding. Interestingly, only the light chain 1 (LC1) of MAP1B was able to interact with Tiam1. Moreover, it was able to increase the activity of the small GTPase, Rac1. These results suggest that the interaction between Tiam1 and MAP1B, is produced by the binding of LC1 with PH domains in Tiam1. The formation of such a complex impacts on the activation levels of Rac1 confirming a novel function of MAP1B related with the control of small GTPases. These results also support the idea of cross-talk between cytoskeleton compartments inside neuronal cells.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Neurons/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cytoskeleton/genetics , Cytoskeleton/metabolism , Guanine Nucleotide Exchange Factors/genetics , Hippocampus/cytology , Hippocampus/metabolism , Microtubule-Associated Proteins/genetics , Microtubules/genetics , Microtubules/metabolism , Neoplasm Proteins/genetics , Neurons/cytology , Rats , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , rac1 GTP-Binding Protein/genetics
15.
J Alzheimers Dis ; 29(1): 63-77, 2012.
Article in English | MEDLINE | ID: mdl-22204905

ABSTRACT

The neuronal cytoskeleton regulates numerous processes that occur in normal homeostasis. Under pathological conditions such as those of Alzheimer's disease (AD), major alterations in cytoskeleton organization have been observed and changes in both microtubules and actin filaments have been reported. Many neurodegenerative consequences of AD are linked to the production and accumulation of amyloid peptides (Aß) and their oligomers, produced from the internal cleavage of the amyloid-ß protein precursor. We previously reported that fibrillar Aß1-42 (fAß) treatment of hippocampal neurons induced an increase in Rac1 and Cdc42 activities linking fAß effects with changes in actin dynamics. Here we show fAß-induces increased activity of PAK1 and cyclin-dependent kinase 5, and that p21-activated kinase (PAK1) activation targets the LIMK1-cofilin signaling pathway. Increased cofilin dephosphorylation under conditions of enhanced LIM-Kinase 1 (LIMK1) activity suggests that fAß co-stimulates bifurcating pathways impacting cofilin phosphorylation. Overexpression of slingshot (SSH) prevents the augment of F-actin induced by fAß after 24 h, suggesting that fAß-induced changes in actin assembly involve both LIMK1 and SSH. These results suggest that fAb may alter the PAK1/LIMK1/cofilin axis and therefore actin organization in AD.


Subject(s)
Actin Depolymerizing Factors/metabolism , Actins/physiology , Amyloid beta-Peptides/physiology , Amyloid/physiology , Neuropeptides/physiology , Peptide Fragments/physiology , Phosphoprotein Phosphatases/physiology , cdc42 GTP-Binding Protein/physiology , rac GTP-Binding Proteins/physiology , Animals , Cells, Cultured , Mice , Mice, Transgenic , Phosphorylation , Rats , Rats, Sprague-Dawley , rac1 GTP-Binding Protein
16.
Cytoskeleton (Hoboken) ; 69(7): 464-85, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22605667

ABSTRACT

The highly dynamic remodeling and cross talk of the microtubule and actin cytoskeleton support neuronal morphogenesis. Small RhoGTPases family members have emerged as crucial regulators of cytoskeletal dynamics. In this review we will comprehensively analyze findings that support the participation of RhoA, Rac, Cdc42, and TC10 in different neuronal morphogenetic events ranging from migration to synaptic plasticity. We will specifically address the contribution of these GTPases to support neuronal polarity and axonal elongation.


Subject(s)
Cell Polarity , Monomeric GTP-Binding Proteins/metabolism , Morphogenesis , Neurons/cytology , Neurons/enzymology , Animals , Humans , Models, Biological
18.
Biol Res ; 39(4): 661-8, 2006.
Article in English | MEDLINE | ID: mdl-17657347

ABSTRACT

Salmonella entérica serovar Typhimurium cells expressing the cadA gene of Geobacillus stearothermophilus LV exhibit a hypersensitive phenotype to cadmium chloride. Deletion of the ORF STM3576 from the Salmonella genome resulted in cadmium, lead and zinc sensitivity, confirming that this ORF is a homologue of the zntA gene. The observed sensitivity was reverted upon expression of the G. stearothermophilus LV cadA gene. These results indicate that the cadA gene product is involved in Cd, Pb and Zn resistance as a classical P-type ATPase and strongly suggest that the observed hypersensitive phenotype to these metals can be related to the function of the host .zntA gene product.


Subject(s)
Adenosine Triphosphatases/genetics , Geobacillus stearothermophilus/genetics , Salmonella typhimurium/drug effects , Cadmium/metabolism , Cadmium/toxicity , Drug Resistance, Bacterial/genetics , Gene Expression Regulation, Bacterial , Lead/metabolism , Lead/toxicity , Mutation , Phenotype , Salmonella typhimurium/enzymology , Salmonella typhimurium/genetics , Zinc/metabolism , Zinc/toxicity
19.
Biol. Res ; 39(4): 661-668, 2006. ilus
Article in English | LILACS | ID: lil-456601

ABSTRACT

Salmonella entérica serovar Typhimurium cells expressing the cadA gene of Geobacillus stearothermophilus LV exhibit a hypersensitive phenotype to cadmium chloride. Deletion of the ORF STM3576 from the Salmonella genome resulted in cadmium, lead and zinc sensitivity, confirming that this ORF is a homologue of the zntA gene. The observed sensitivity was reverted upon expression of the G. stearothermophilus LV cadA gene. These results indicate that the cadA gene product is involved in Cd, Pb and Zn resistance as a classical P-type ATPase and strongly suggest that the observed hypersensitive phenotype to these metals can be related to the function of the host ·zntA gene product.


Subject(s)
Adenosine Triphosphatases/genetics , Geobacillus stearothermophilus/genetics , Salmonella typhimurium/drug effects , Cadmium/pharmacology , Drug Resistance, Bacterial/genetics , Gene Expression Regulation, Bacterial , Lead/pharmacology , Mutation , Phenotype , Salmonella typhimurium/enzymology , Salmonella typhimurium/genetics , Zinc/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL