Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
Protoplasma ; 254(3): 1187-1199, 2017 May.
Article in English | MEDLINE | ID: mdl-28074286

ABSTRACT

The microtubule cytoskeleton is critically important for spatio-temporal organization of eukaryotic cells. The nucleation of new microtubules is typically restricted to microtubule organizing centers (MTOCs) and requires γ-tubulin that assembles into multisubunit complexes of various sizes. γ-Tubulin ring complexes (TuRCs) are efficient microtubule nucleators and are associated with large number of targeting, activating and modulating proteins. γ-Tubulin-dependent nucleation of microtubules occurs both from canonical MTOCs, such as spindle pole bodies and centrosomes, and additional sites such as Golgi apparatus, nuclear envelope, plasma membrane-associated sites, chromatin and surface of pre-existing microtubules. Despite many advances in structure of γ-tubulin complexes and characterization of γTuRC interacting factors, regulatory mechanisms of microtubule nucleation are not fully understood. Here, we review recent work on the factors and regulatory mechanisms that are involved in centrosomal and non-centrosomal microtubule nucleation.


Subject(s)
Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Spindle Pole Bodies/metabolism , Tubulin/metabolism , Centrosome/metabolism , Golgi Apparatus/metabolism , Humans , Multiprotein Complexes/metabolism , Nuclear Envelope/metabolism
3.
Biochim Biophys Acta ; 1863(6 Pt A): 1282-97, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27012601

ABSTRACT

Microtubule nucleation from γ-tubulin complexes, located at the centrosome, is an essential step in the formation of the microtubule cytoskeleton. However, the signaling mechanisms that regulate microtubule nucleation in interphase cells are largely unknown. In this study, we report that γ-tubulin is in complexes containing G protein-coupled receptor kinase-interacting protein 1 (GIT1), p21-activated kinase interacting exchange factor (ßPIX), and p21 protein (Cdc42/Rac)-activated kinase 1 (PAK1) in various cell lines. Immunofluorescence microscopy revealed association of GIT1, ßPIX and activated PAK1 with centrosomes. Microtubule regrowth experiments showed that depletion of ßPIX stimulated microtubule nucleation, while depletion of GIT1 or PAK1 resulted in decreased nucleation in the interphase cells. These data were confirmed for GIT1 and ßPIX by phenotypic rescue experiments, and counting of new microtubules emanating from centrosomes during the microtubule regrowth. The importance of PAK1 for microtubule nucleation was corroborated by the inhibition of its kinase activity with IPA-3 inhibitor. GIT1 with PAK1 thus represent positive regulators, and ßPIX is a negative regulator of microtubule nucleation from the interphase centrosomes. The regulatory roles of GIT1, ßPIX and PAK1 in microtubule nucleation correlated with recruitment of γ-tubulin to the centrosome. Furthermore, in vitro kinase assays showed that GIT1 and ßPIX, but not γ-tubulin, serve as substrates for PAK1. Finally, direct interaction of γ-tubulin with the C-terminal domain of ßPIX and the N-terminal domain of GIT1, which targets this protein to the centrosome, was determined by pull-down experiments. We propose that GIT1/ßPIX signaling proteins with PAK1 kinase represent a novel regulatory mechanism of microtubule nucleation in interphase cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Microtubules/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , p21-Activated Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Cycle Proteins/genetics , Cell Line, Transformed , Cell Line, Tumor , Centrosome/metabolism , HEK293 Cells , Humans , Immunoblotting , Microscopy, Fluorescence , Phosphorylation , Protein Binding , Rho Guanine Nucleotide Exchange Factors/genetics , Signal Transduction , Tubulin/metabolism , p21-Activated Kinases/genetics
4.
J Immunol ; 194(9): 4099-111, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25821222

ABSTRACT

Ag-mediated activation of mast cells initiates signaling events leading to Ca(2+) response, release of allergic mediators from cytoplasmic granules, and synthesis of cytokines and chemokines. Although microtubule rearrangement during activation has been described, the molecular mechanisms that control their remodeling are largely unknown. Microtubule nucleation is mediated by complexes that are formed by γ-tubulin and γ-tubulin complex proteins. In this study, we report that, in bone marrow-derived mast cells (BMMCs), γ-tubulin interacts with p21-activated kinase interacting exchange factor ß (ßPIX) and G protein-coupled receptor kinase-interacting protein (GIT)1. Microtubule regrowth experiments showed that the depletion of ßPIX in BMMCs stimulated microtubule nucleation, whereas depletion of GIT1 led to the inhibition of nucleation compared with control cells. Phenotypic rescue experiments confirmed that ßPIX and GIT1 represent negative and positive regulators of microtubule nucleation in BMMCs, respectively. Live-cell imaging disclosed that both proteins are associated with centrosomes. Immunoprecipitation and pull-down experiments revealed that an enhanced level of free cytosolic Ca(2+) affects γ-tubulin properties and stimulates the association of GIT1 and γ-tubulin complex proteins with γ-tubulin. Microtubule nucleation also was affected by Ca(2+) level. Moreover, in activated BMMCs, γ-tubulin formed complexes with tyrosine-phosphorylated GIT1. Further experiments showed that GIT1 and ßPIX are involved in the regulation of such important physiological processes as Ag-induced chemotaxis and degranulation. Our study provides for the first time, to our knowledge, a possible mechanism for the concerted action of tyrosine kinases, GIT1/ßPIX proteins, and Ca(2+) in the propagation of signals leading to the regulation of microtubule nucleation in activated mast cells.


Subject(s)
Bone Marrow Cells/cytology , Calcium/metabolism , Cell Cycle Proteins/metabolism , GTPase-Activating Proteins/metabolism , Mast Cells/cytology , Microtubules/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Animals , Mice , Mice, Inbred BALB C
5.
Brain ; 138(Pt 2): 336-55, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25567323

ABSTRACT

Severe progressive neurological paediatric disease mucopolysaccharidosis III type C is caused by mutations in the HGSNAT gene leading to deficiency of acetyl-CoA: α-glucosaminide N-acetyltransferase involved in the lysosomal catabolism of heparan sulphate. To understand the pathophysiology of the disease we generated a mouse model of mucopolysaccharidosis III type C by germline inactivation of the Hgsnat gene. At 6-8 months mice showed hyperactivity, and reduced anxiety. Cognitive memory decline was detected at 10 months and at 12-13 months mice showed signs of unbalanced hesitant walk and urinary retention. Lysosomal accumulation of heparan sulphate was observed in hepatocytes, splenic sinus endothelium, cerebral microglia, liver Kupffer cells, fibroblasts and pericytes. Starting from 5 months, brain neurons showed enlarged, structurally abnormal mitochondria, impaired mitochondrial energy metabolism, and storage of densely packed autofluorescent material, gangliosides, lysozyme, phosphorylated tau, and amyloid-ß. Taken together, our data demonstrate for the first time that deficiency of acetyl-CoA: α-glucosaminide N-acetyltransferase causes lysosomal accumulation of heparan sulphate in microglial cells followed by their activation and cytokine release. They also show mitochondrial dysfunction in the neurons and neuronal loss explaining why mucopolysaccharidosis III type C manifests primarily as a neurodegenerative disease.


Subject(s)
Mitochondrial Diseases/pathology , Mucopolysaccharidosis III/pathology , Neuritis/pathology , Neurodegenerative Diseases/pathology , Acetyltransferases/deficiency , Acetyltransferases/genetics , Animals , Behavior, Animal , Energy Metabolism/physiology , Gangliosides/metabolism , Glycosaminoglycans/metabolism , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Mitochondrial Diseases/etiology , Mucopolysaccharidosis III/complications , Mucopolysaccharidosis III/psychology , Neuritis/etiology , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/psychology , Neurologic Examination , Proteostasis Deficiencies/pathology
6.
Methods Mol Biol ; 1192: 161-76, 2014.
Article in English | MEDLINE | ID: mdl-25149491

ABSTRACT

A better understanding of the molecular mechanisms leading to mast cell migration and chemotaxis is the long-term goal in mast cell research and is essential for comprehension of mast cell function in health and disease. Various techniques have been developed in recent decades for in vitro and in vivo assessment of mast cell motility and chemotaxis. In this chapter three microscopy assays facilitating real-time quantification of mast cell chemotaxis and migration are described, focusing on individual cell tracking and data analysis.


Subject(s)
Cell Migration Assays/methods , Cell Movement , Mast Cells/cytology , Mast Cells/physiology , Microscopy/methods , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Movement/drug effects , Cell Tracking/methods , Cells, Cultured , Chemotactic Factors/pharmacology , Chemotaxis , Humans , Mast Cells/drug effects
7.
J Immunol Methods ; 395(1-2): 63-70, 2013 Sep 30.
Article in English | MEDLINE | ID: mdl-23851142

ABSTRACT

Microtubules formed by αß-tubulin dimers represent cellular structures that are indispensable for the maintenance of cell morphology and for cell motility generation. Microtubules in intact cells are in highly regulated equilibrium with cellular pools of soluble tubulin dimers. Sensitive, reproducible and rapid assays are necessary to monitor tubulin changes in cytosolic pools after treatment with anti-mitotic drugs, during the cell cycle or activation and differentiation events. Here we describe new assays for α-tubulin quantification. The assays are based on sandwich ELISA, and the signal is amplified with biotinyl-tyramide or immuno-PCR. Matching monoclonal antibody pair recognizes phylogenetically highly conserved epitopes localized outside the C-terminal isotype-defining region. This makes it possible to detect α-tubulin isotypes in different cell types of various species. Biotinyl-tyramide amplification and immuno-PCR amplification enable detection of tubulin at concentrations 2.5ng/ml and 0.086ng/ml, respectively. Immuno-PCR detection shows enhanced sensitivity and wider dynamic range when compared to ELISA with biotinyl-tyramide detection. Our results on taxol-treated and activated bone marrow-derived mast cells demonstrate, that the assays allow sensitive quantification of tubulin in complex biological fluids.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Tubulin/analysis , Animals , Antibodies, Monoclonal , Biotin/analogs & derivatives , Cell Line , Enzyme-Linked Immunosorbent Assay/statistics & numerical data , Epitope Mapping , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Paclitaxel/pharmacology , Polymerase Chain Reaction/methods , Protein Isoforms/analysis , Protein Isoforms/genetics , Protein Isoforms/immunology , Thapsigargin/pharmacology , Tubulin/genetics , Tubulin/immunology , Tyramine/analogs & derivatives
8.
PLoS One ; 7(7): e40949, 2012.
Article in English | MEDLINE | ID: mdl-22859959

ABSTRACT

A glycolytic profile unifies a group of pheochromocytomas and paragangliomas (PHEOs/PGLs) with distinct underlying gene defects, including von Hippel-Lindau (VHL) and succinate dehydrogenase B (SDHB) mutations. Nevertheless, their tumor aggressiveness is distinct: PHEOs/PGLs metastasize rarely in VHL-, but frequently in SDHB-patients. To date, the molecular mechanisms causing the more aggressive phenotype in SDHB-PHEOs/PGLs remain largely unknown. Recently, however, an excellent model to study aggressive PHEOs (mouse tumor tissue (MTT) cells) has been developed from mouse PHEO cells (MPC). We employed this model for a proteomics based approach to identify changes characteristic for tumor aggressiveness, which we then explored in a homogeneous set of human SDHB- and VHL-PHEOs/PGLs. The increase of glucose transporter 1 in VHL, and of hexokinase 2 in VHL and SDHB, confirmed their glycolytic profile. In agreement with the cell model and in support of decoupling of glycolysis, the Krebs cycle and oxidative phosphorylation (OXPHOS), SDHB tumors showed increased lactate dehydrogenase levels. In SDHB-PGLs OXPHOS complex activity was increased at complex III and, as expected, decreased at complex II. Moreover, protein and mRNA expression of all tested OXPHOS-related genes were higher in SDHB- than in VHL-derived tumors. Although there was no direct evidence for increased reactive oxygen species production, elevated superoxide dismutase 2 expression may reflect elevated oxidative stress in SDHB-derived PHEOs/PGLs. For the first time, we show that despite dysfunction in complex II and evidence for a glycolytic phenotype, the Warburg effect does not seem to fully apply to SDHB-PHEOs/PGLs with respect to decreased OXPHOS. In addition, we present evidence for increased LDHA and SOD2 expression in SDHB-PHEOs/PGLs, proteins that have been proposed as promising therapeutic targets in other cancers. This study provides new insight into pathogenic mechanisms in aggressive human PHEOs/PGLs, which may lead to identifying new diagnostic and prognostic markers in the near future.


Subject(s)
Adrenal Gland Neoplasms/pathology , Paraganglioma/pathology , Pheochromocytoma/pathology , Adrenal Gland Neoplasms/metabolism , Adrenal Medulla/metabolism , Animals , Cell Line, Tumor , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex II/genetics , Electron Transport Complex II/metabolism , Electron Transport Complex III/genetics , Electron Transport Complex III/metabolism , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Gene Expression , Glycolysis , Humans , L-Lactate Dehydrogenase/metabolism , Mitochondria/metabolism , Oxidative Phosphorylation , Oxygen Consumption , Paraganglioma/metabolism , Pheochromocytoma/metabolism , Proteome/metabolism , Reactive Oxygen Species/metabolism
9.
Dalton Trans ; 40(22): 5916-20, 2011 Jun 14.
Article in English | MEDLINE | ID: mdl-21512698

ABSTRACT

Reactions between the methylated arenes ArMe(n) [where ArMe(n) = C(6)Me(n)H((6-n)), and n = 1-6] and FeCl(2) in heptane at 90 °C in the presence of anhydrous AlCl(3) give, for the arenes with n = 1-5, extensive isomerisations and disproportionations involving the methyl groups on the arene rings, and the formation of mixtures of [Fe(ArMe(n))(2)](2+) dications that defy separation into pure species. GC-MS studies of AlCl(3)/mesitylene and AlCl(3)/durene reactions in the absence of FeCl(2) (90 °C, 2 h) allow quantitative assessments of the rearrangements, and the EINS mechanism (electrophile-induced nucleophilic substitution) is applied to rationalise the phenomena. By contrast, ArMe(n) / FeCl(2) /AlCl(3) reactions in heptane for 24-36 h at room-temperature proceed with no rearrangements, allowing the synthesis of the complete series of pure [Fe(ArMen)](2+) cations in yields of 48-71%. The pure compounds are characterised by (1)H NMR spectroscopy and electrospray-ionization mass-spectrometry (ESI-MS), and the structures of [Fe(m-xylene)(2)][PF(6)](2) and [Fe(durene)(2)][PF(6)](2) are established by single-crystal X-ray diffraction analyses.

10.
J Immunol ; 186(2): 913-23, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21160048

ABSTRACT

Activation of mast cells by aggregation of the high-affinity IgE receptors (FcεRI) initiates signaling events leading to the release of inflammatory and allergic mediators stored in cytoplasmic granules. A key role in this process play changes in concentrations of intracellular Ca(2+) controlled by store-operated Ca(2+) entry (SOCE). Although microtubules are also involved in the process leading to degranulation, the molecular mechanisms that control microtubule rearrangement during activation are largely unknown. In this study, we report that activation of bone marrow-derived mast cells (BMMCs) induced by FcεRI aggregation or treatment with pervanadate or thapsigargin results in generation of protrusions containing microtubules (microtubule protrusions). Formation of these protrusions depended on the influx of extracellular Ca(2+). Changes in cytosolic Ca(2+)concentration also affected microtubule plus-end dynamics detected by microtubule plus-end tracking protein EB1. Experiments with knockdown or reexpression of STIM1, the key regulator of SOCE, confirmed the important role of STIM1 in the formation of microtubule protrusions. Although STIM1 in activated cells formed puncta associated with microtubules in protrusions, relocation of STIM1 to a close proximity of cell membrane was independent of growing microtubules. In accordance with the inhibition of Ag-induced Ca(2+) response and decreased formation of microtubule protrusions in BMMCs with reduced STIM1, the cells also exhibited impaired chemotactic response to Ag. We propose that rearrangement of microtubules in activated mast cells depends on STIM1-induced SOCE, and that Ca(2+) plays an important role in the formation of microtubule protrusions in BMMCs.


Subject(s)
Mast Cells/immunology , Mast Cells/metabolism , Membrane Proteins/physiology , Microtubules/immunology , Microtubules/metabolism , Neoplasm Proteins/physiology , Amino Acid Sequence , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Calcium Signaling/immunology , Cell Communication/immunology , Cells, Cultured , HEK293 Cells , Humans , Mast Cells/cytology , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Molecular Sequence Data , Neoplasm Proteins/metabolism , Stromal Interaction Molecule 1
SELECTION OF CITATIONS
SEARCH DETAIL
...