Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Oncotarget ; 11(14): 1290-1291, 2020 Apr 07.
Article in English | MEDLINE | ID: mdl-32292578

ABSTRACT

[This corrects the article DOI: 10.18632/oncotarget.15343.].

2.
Oncotarget ; 8(13): 20813-20824, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28209915

ABSTRACT

We identified target proteins modified by phenanthrenes that cause exclusive eradication of human cancer cells. The cytotoxic activity of the phenanthrenes in a variety of human cancer cells is attributed by these findings to post translational modifications of NuMA and kinesins HSET/kifC1 and kif18A. Their activity prevented the binding of NuMA to α-tubulin and kinesins in human cancer cells, and caused aberrant spindles. The most efficient cytotoxic activity of the phenanthridine PJ34, caused significantly smaller aberrant spindles with disrupted spindle poles and scattered extra-centrosomes and chromosomes. Concomitantly, PJ34 induced tumor growth arrest of human malignant tumors developed in athymic nude mice, indicating the relevance of its activity for cancer therapy.


Subject(s)
Biomarkers, Tumor/metabolism , Mitosis/physiology , Neoplasms/pathology , Protein Processing, Post-Translational/drug effects , Spindle Apparatus/pathology , Animals , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Cell Cycle Proteins , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kinesins/genetics , Kinesins/metabolism , Mice , Mitosis/drug effects , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Phenanthrenes/pharmacology , Spindle Apparatus/drug effects , Spindle Apparatus/genetics , Spindle Apparatus/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
J Vis Exp ; (78): e50568, 2013 Aug 21.
Article in English | MEDLINE | ID: mdl-23995751

ABSTRACT

Phenanthrene derivatives acting as potent PARP1 inhibitors prevented the bi-focal clustering of supernumerary centrosomes in multi-centrosomal human cancer cells in mitosis. The phenanthridine PJ-34 was the most potent molecule. Declustering of extra-centrosomes causes mitotic failure and cell death in multi-centrosomal cells. Most solid human cancers have high occurrence of extra-centrosomes. The activity of PJ-34 was documented in real-time by confocal imaging of live human breast cancer MDA-MB-231 cells transfected with vectors encoding for fluorescent γ-tubulin, which is highly abundant in the centrosomes and for fluorescent histone H2b present in the chromosomes. Aberrant chromosomes arrangements and de-clustered γ-tubulin foci representing declustered centrosomes were detected in the transfected MDA-MB-231 cells after treatment with PJ-34. Un-clustered extra-centrosomes in the two spindle poles preceded their cell death. These results linked for the first time the recently detected exclusive cytotoxic activity of PJ-34 in human cancer cells with extra-centrosomes de-clustering in mitosis, and mitotic failure leading to cell death. According to previous findings observed by confocal imaging of fixed cells, PJ-34 exclusively eradicated cancer cells with multi-centrosomes without impairing normal cells undergoing mitosis with two centrosomes and bi-focal spindles. This cytotoxic activity of PJ-34 was not shared by other potent PARP1 inhibitors, and was observed in PARP1 deficient MEF harboring extracentrosomes, suggesting its independency of PARP1 inhibition. Live confocal imaging offered a useful tool for identifying new molecules eradicating cells during mitosis.


Subject(s)
Breast Neoplasms/pathology , Microscopy, Confocal/methods , Mitosis/physiology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Centrosome/pathology , Female , Humans , Mice , Mitosis/drug effects , Phenanthrenes/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Spindle Apparatus/pathology
4.
Mech Ageing Dev ; 134(10): 496-505, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23583690

ABSTRACT

The cerebellum is exquisitely sensitive to deficiencies in the cellular response to specific DNA lesions. Genetic disorders caused by such deficiencies involve relentless, progressive cerebellar atrophy with striking loss of Purkinje and granule neurons. The reason for the extreme sensitivity of these cells to defective response to certain DNA lesions is unclear. This is particularly true for ataxia-telangiectasia (A-T) - a genomic instability syndrome whose major symptom is cerebellar atrophy. It is important to understand whether the DNA damage response in the cerebellum, particularly in Purkinje neurons, has special characteristics that stem from the unique features of these cells. Murine cerebellar organotypic cultures provide a valuable experimental system for this purpose since they retain the tissue organization for several weeks in culture and appear to provide the delicate Purkinje neurons with a physiological environment close to that in vivo. We have optimized this system and are using it to examine the Atm-mediated DNA damage response (DDR) in the cerebellum, with special emphasis on Purkinje cells. Our results to date, which indicate special chromatin organization in Purkinje cells that affects certain pathways of the DDR, demonstrate the usefulness of cerebellar organotypic cultures for addressing the above questions.


Subject(s)
Chromatin/metabolism , DNA Damage , Genomic Instability , Purkinje Cells/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Chromatin/genetics , Chromatin/pathology , Chromobox Protein Homolog 5 , Humans , Mice , Mice, Knockout , Purkinje Cells/pathology , Tissue Culture Techniques
5.
ACS Med Chem Lett ; 4(3): 323-8, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-24900668

ABSTRACT

The cytotoxic activities and subcellular localizations of clinically used and synthetic analogues of the anthracycline family of chemotherapeutic agents were studied. The structures of the anthracycline derivatives affected their cytotoxicity and the time required for these compounds to exert cytotoxic effects on tumor cells. Fluorescent DNA intercalator displacement experiments demonstrated that there was no correlation between the DNA intercalation properties and the cytotoxicity of the studied anthracycline derivatives. Confocal microscopy experiments indicated that structural differences led to differences in subcellular localization. All studied anthracycline derivatives were observed in lysosomes, suggesting that this organelle, which is involved in several processes leading to malignancy, may contain previously unidentified molecular targets for these antitumor agents.

6.
ACS Appl Mater Interfaces ; 4(7): 3542-9, 2012 Jul 25.
Article in English | MEDLINE | ID: mdl-22724437

ABSTRACT

The use of artificial, prepatterned neuronal networks in vitro is a promising approach for studying the development and dynamics of small neural systems in order to understand the basic functionality of neurons and later on of the brain. The present work presents a high fidelity and robust procedure for controlling neuronal growth on substrates such as silicon wafers and glass, enabling us to obtain mature and durable neural networks of individual cells at designed geometries. It offers several advantages compared to other related techniques that have been reported in recent years mainly because of its high yield and reproducibility. The procedure is based on surface chemistry that allows the formation of functional, tailormade neural architectures with a micrometer high-resolution partition, that has the ability to promote or repel cells attachment. The main achievements of this work are deemed to be the creation of a large scale neuronal network at low density down to individual cells, that develop intact typical neurites and synapses without any glia-supportive cells straight from the plating stage and with a relatively long term survival rate, up to 4 weeks. An important application of this method is its use on 3D nanopillars and 3D nanowire-device arrays, enabling not only the cell bodies, but also their neurites to be positioned directly on electrical devices and grow with registration to the recording elements underneath.


Subject(s)
Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Nanowires/chemistry , Nerve Net/cytology , Neurons/cytology , Tissue Scaffolds , Animals , Cell Adhesion , Cell Communication , Cell Survival , Cells, Cultured , Embryo, Mammalian , Materials Testing , Rats , Rats, Sprague-Dawley , Silicon/chemistry , Surface Properties , Synapses/physiology , Tissue Scaffolds/chemistry
7.
BMC Cancer ; 11: 412, 2011 Sep 26.
Article in English | MEDLINE | ID: mdl-21943092

ABSTRACT

BACKGROUND: Cells of most human cancers have supernumerary centrosomes. To enable an accurate chromosome segregation and cell division, these cells developed a yet unresolved molecular mechanism, clustering their extra centrosomes at two poles, thereby mimicking mitosis in normal cells. Failure of this bipolar centrosome clustering causes multipolar spindle structures and aberrant chromosomes segregation that prevent normal cell division and lead to 'mitotic catastrophe cell death'. METHODS: We used cell biology and biochemical methods, including flow cytometry, immunocytochemistry and live confocal imaging. RESULTS: We identified a phenanthrene derived PARP inhibitor, known for its activity in neuroprotection under stress conditions, which exclusively eradicated multi-centrosomal human cancer cells (mammary, colon, lung, pancreas, ovarian) while acting as extra-centrosomes de-clustering agent in mitosis. Normal human proliferating cells (endothelial, epithelial and mesenchymal cells) were not impaired. Despite acting as PARP inhibitor, the cytotoxic activity of this molecule in cancer cells was not attributed to PARP inhibition alone. CONCLUSION: We identified a water soluble phenanthridine that exclusively targets the unique dependence of most human cancer cells on their supernumerary centrosomes bi-polar clustering for their survival. This paves the way for a new selective cancer-targeting therapy, efficient in a wide range of human cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Centrosome/drug effects , Enzyme Inhibitors/pharmacology , Neoplasms/enzymology , Neoplasms/genetics , Phenanthrenes/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chromosome Segregation/drug effects , Humans , Mitosis/drug effects , Mitosis/genetics , Spindle Apparatus/drug effects
8.
Biochem J ; 438(2): 337-47, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21635224

ABSTRACT

The nuclear protein PARP-1 [poly(ADP-ribose) polymerase-1] is activated in cardiomyocytes exposed to hypoxia causing DNA breaks. Unlike this stress-induced PARP-1 activation, our results provide evidence for Ca(2+)-induced PARP-1 activation in contracting newborn cardiomyocytes treated with growth factors and hormones that increased their contraction rate, induced intracellular Ca(2+) mobilization and its rhythmical and transient translocation into the nucleus. Furthermore, activated PARP-1 up-regulated the activity of phosphorylated ERK (extracellular-signal-regulated kinase) in the nucleus, promoting expression of the Elk1 target gene c-fos. Up-regulation of the transcription factor c-Fos/GATA-4 promoted ANF (atrial natriuretic factor) expression. Given that expression of ANF is known to be implicated in morphological changes, growth and development of cardiomyocytes, these results outline a PARP-1-dependent signal transduction mechanism that links contraction rate and Ca(2+) mobilization with the expression of genes underlying morphological changes in cardiomyocytes.


Subject(s)
Atrial Natriuretic Factor/metabolism , Calcium/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Poly(ADP-ribose) Polymerases/metabolism , Angiotensin II/pharmacology , Animals , Animals, Newborn , Atrial Natriuretic Factor/genetics , Calcium/metabolism , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , GATA4 Transcription Factor/metabolism , Gene Expression Regulation/drug effects , Intracellular Space/drug effects , Intracellular Space/metabolism , Myocardial Contraction/drug effects , Phosphorylation/drug effects , Poly (ADP-Ribose) Polymerase-1 , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Ribose/metabolism
9.
DNA Repair (Amst) ; 6(1): 128-34, 2007 Jan 04.
Article in English | MEDLINE | ID: mdl-17178256

ABSTRACT

Ataxia-telangiectasia (A-T) is a multi-system genomic instability syndrome that is caused by loss or inactivation of the ATM protein kinase. ATM is largely nuclear in proliferating cells, and activates an extensive network of pathways in response to double strand breaks (DSBs) in the DNA by phosphorylating key proteins in these pathways. The prominent symptom of A-T is neuronal degeneration, making the elucidation of ATM's functions in neurons essential to understanding the disease. It has been suggested that ATM is cytoplasmic in neurons and functions in processes that are not associated with the DNA damage response. Recently we showed that in human neuron-like cells obtained by in vitro differentiation of neuroblastomas, ATM was largely nuclear and mediated the DSB response as in proliferating cells. We have now extended these studies to two additional model systems: neurons derived from human embryonic stem cells, and cortical neurons derived from neural stem cells. The results substantiate the notion that ATM is nuclear in human neurons and mediates the DSB response, the same as it does in proliferating cells. We present here unique and powerful model systems to further study the ATM-mediated network in neurons.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/physiology , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Nucleus/metabolism , DNA Repair , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Humans , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
10.
Mol Cell Biol ; 26(18): 6819-31, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16943424

ABSTRACT

The p53 tumor suppressor plays a major role in maintaining genomic stability. Its activation and stabilization in response to double strand breaks (DSBs) in DNA are regulated primarily by the ATM protein kinase. ATM mediates several posttranslational modifications on p53 itself, as well as phosphorylation of p53's essential inhibitors, Hdm2 and Hdmx. Recently we showed that ATM- and Hdm2-dependent ubiquitination and subsequent degradation of Hdmx following DSB induction are mediated by phosphorylation of Hdmx on S403, S367, and S342, with S403 being targeted directly by ATM. Here we show that S367 phosphorylation is mediated by the Chk2 protein kinase, a downstream kinase of ATM. This phosphorylation, which is important for subsequent Hdmx ubiquitination and degradation, creates a binding site for 14-3-3 proteins which controls nuclear accumulation of Hdmx following DSBs. Phosphorylation of S342 also contributed to optimal 14-3-3 interaction and nuclear accumulation of Hdmx, but phosphorylation of S403 did not. Our data indicate that binding of a 14-3-3 dimer and subsequent nuclear accumulation are essential steps toward degradation of p53's inhibitor, Hdmx, in response to DNA damage. These results demonstrate a sophisticated control by ATM of a target protein, Hdmx, which itself is one of several ATM targets in the ATM-p53 axis of the DNA damage response.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage/genetics , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , 14-3-3 Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Nucleus/metabolism , Cells, Cultured , Checkpoint Kinase 2 , Humans , Mice , Models, Biological , Phosphorylation , Phosphoserine/metabolism , Protein Binding , Protein Processing, Post-Translational
11.
J Cell Biol ; 174(2): 195-206, 2006 Jul 17.
Article in English | MEDLINE | ID: mdl-16847100

ABSTRACT

Mitotic cell death (MCD) is a prominent but poorly defined form of death that stems from aberrant mitosis. One of the early steps in MCD is premature mitosis and uneven chromatin condensation (UCC). The mechanism underlying this phenomenon is currently unknown. In this study, we show that DNA damage in cells with a compromised p53-mediated G2/M checkpoint triggers the unscheduled activation of cyclin-dependent kinase 1 (Cdk1), activation and chromatin loading of the condensin I complex, and UCC followed by the appearance of multimicronucleated cells, which is evidence of MCD. We demonstrate that these processes engage some of the players of normal mitotic chromatin packaging but not those that drive the apoptotic chromatin condensation. Our findings establish a link between the induction of DNA damage and mitotic abnormalities (UCC) through the unscheduled activation of Cdk1 and recruitment of condensin I. These results demonstrate a clear distinction between the mechanisms that drive MCD-associated and apoptosis-related chromatin condensation and provide mechanistic insights and new readouts for a major cell death process in treated tumors.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromatin/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Mitosis , Multiprotein Complexes/metabolism , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Cell Death , Cell Nucleolus/metabolism , Chromatin/genetics , Chromosomal Proteins, Non-Histone/metabolism , Enzyme Activation , G2 Phase/genetics , HCT116 Cells , HeLa Cells , Humans , Models, Genetic , Nuclear Proteins/metabolism , Protein Transport , Tumor Cells, Cultured , Cohesins
12.
J Biol Chem ; 281(25): 17482-17491, 2006 Jun 23.
Article in English | MEDLINE | ID: mdl-16627474

ABSTRACT

The protein kinase ATM (ataxia-telangiectasia mutated) activates the cellular response to double strand breaks (DSBs), a highly cytotoxic DNA lesion. ATM is activated by DSBs and in turn phosphorylates key players in numerous damage response pathways. ATM is missing or inactivated in the autosomal recessive disorder ataxia-telangiectasia (A-T), which is characterized by neuronal degeneration, immunodeficiency, genomic instability, radiation sensitivity, and cancer predisposition. The predominant symptom of A-T is a progressive loss of movement coordination due to ongoing degeneration of the cerebellar cortex and peripheral neuropathy. A major deficiency in understanding A-T is the lack of information on the role of ATM in neurons. It is unclear whether the ATM-mediated DSB response operates in these cells similarly to proliferating cells. Furthermore, ATM was reported to be cytoplasmic in neurons and suggested to function in these cells in capacities other than the DNA damage response. Recently we obtained genetic molecular evidence that the neuronal degeneration in A-T does result from defective DNA damage response. We therefore undertook to investigate this response in a model system of human neuron-like cells (NLCs) obtained by neuronal differentiation in culture. ATM was largely nuclear in NLCs, and their ATM-mediated responses to DSBs were similar to those of proliferating cells. Knocking down ATM did not interfere with neuronal differentiation but abolished ATM-mediated damage responses in NLCs. We concluded that nuclear ATM mediates the DSB response in NLCs similarly to in proliferating cells. Attempts to understand the neurodegeneration in A-T should be directed to investigating the DSB response in the nervous system.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , DNA Damage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/chemistry , Cell Differentiation , Cell Line , Cell Line, Tumor , Cell Proliferation , DNA Repair , DNA-Binding Proteins/chemistry , Genetic Vectors , HeLa Cells , Humans , Neurodegenerative Diseases/metabolism , Protein Serine-Threonine Kinases/chemistry , Tumor Suppressor Proteins/chemistry
13.
J Infect Dis ; 189(11): 1965-73, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15143461

ABSTRACT

Aspergillus fumigatus is an opportunistic pathogenic fungus that predominantly infects the respiratory system. Penetration of the lung alveolar epithelium is a key step in the infectious process. The cytoskeleton of alveolar epithelial cells forms the cellular basis for the formation of a physical barrier between the cells and their surroundings. This study focused on the distinct effects of A. fumigatus on the actin cytoskeleton of A549 lung pneumocytes. Of the 3 major classes of cytoskeletal fibers--actin microfilaments, microtubules, and intermediate filaments--only the actin cytoskeleton was found to undergo major structural changes in response to infection, including loss of actin stress fibers, formation of actin aggregates, disruption of focal adhesion sites, and cell blebbing. These changes could be specifically blocked in wild-type strains of A. fumigatus by the addition of antipain, a serine and cysteine protease inhibitor, and were not induced by an alkaline serine protease-deficient strain of A. fumigatus. Antipain also reduced, by approximately 50%, fungal-induced A549 cell detachment from the plates and reduction in viability. Our findings suggest that A. fumigatus breaches the alveolar epithelial cell barrier by secreting proteases that act together to disorganize the actin cytoskeleton and destroy cell attachment to the substrate by disrupting focal adhesions.


Subject(s)
Actins/metabolism , Aspergillosis/microbiology , Aspergillus fumigatus/enzymology , Cytoskeleton/metabolism , Endopeptidases/metabolism , Antipain/pharmacology , Aspergillus fumigatus/physiology , Benzenesulfonates/chemistry , Cell Adhesion/physiology , Cell Line, Tumor , Formazans/chemistry , Humans , Immunohistochemistry , Lung/microbiology , Lung/ultrastructure , Microscopy, Confocal , Microtubules , Protease Inhibitors/pharmacology , Vinculin/physiology
14.
J Biol Chem ; 279(27): 28531-8, 2004 Jul 02.
Article in English | MEDLINE | ID: mdl-15123709

ABSTRACT

An octapeptide was previously described that protects neurons against a wide variety of insults directly and indirectly as a result of interactions (at femtomolar concentrations) with supporting glial cells. The current study set out to identify the octapeptide binding molecules so as to understand the high affinity mechanisms of cellular protection. Studies utilizing affinity chromatography of brain extracts identified tubulin, the brain major protein, as the octapeptide-binding ligand. Dot blot analysis with pure tubulin and the biotinylated octapeptide verified this finding. When added to cerebral cortical astrocytes, the octapeptide (10(-15)-10(-10) m) induced a rapid microtubule reorganization into distinct microtubular structures that were stained by monoclonal tubulin antibodies and visualized by confocal microscopy. Fluorescein-labeled octapeptide induced a similar change and was detected in the intracellular milieu, even when cells were incubated at 4 degrees C or at low pH. In a cell-free system, the octapeptide stimulated tubulin assembly into microtubules. Furthermore, treatment of astrocytes with zinc chloride resulted in microtubule disassembly and cell death that was protected by the octapeptide. In conclusion, the results suggest that the octapeptide crosses the plasma membrane and interacts directly with tubulin, the microtubule subunit, to induce microtubule reorganization and improved survival. Because microtubules are the key component of the neuronal and glial cytoskeleton that regulates cell division, differentiation, and protection, this finding may explain the breadth and efficiency of the cellular protective capacities of the octapeptide.


Subject(s)
Astrocytes/metabolism , Peptides/pharmacology , Tubulin/chemistry , Zinc/chemistry , Amino Acid Sequence , Animals , Biotinylation , Brain/metabolism , Cattle , Cell Death , Cell Differentiation , Cell Division , Cell Membrane/metabolism , Cell-Free System , Cells, Cultured , Cerebral Cortex/cytology , Chromatography, Affinity , Fluorescein/pharmacology , Fluorescent Dyes/pharmacology , Hydrogen-Ion Concentration , Ligands , Microscopy, Confocal , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Molecular Sequence Data , Neuroglia/metabolism , Neurons/metabolism , Peptides/chemistry , Protein Binding , Protein Transport , Rats , Sequence Homology, Amino Acid , Temperature , Time Factors , Zinc/pharmacology
15.
EMBO J ; 22(20): 5612-21, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14532133

ABSTRACT

The ATM protein kinase is a primary activator of the cellular response to DNA double-strand breaks (DSBs). In response to DSBs, ATM is activated and phosphorylates key players in various branches of the DNA damage response network. ATM deficiency causes the genetic disorder ataxia-telangiectasia (A-T), characterized by cerebellar degeneration, immunodeficiency, radiation sensitivity, chromosomal instability and cancer predisposition. The MRN complex, whose core contains the Mre11, Rad50 and Nbs1 proteins, is involved in the initial processing of DSBs. Hypomorphic mutations in the NBS1 and MRE11 genes lead to two other genomic instability disorders: the Nijmegen breakage syndrome (NBS) and A-T like disease (A-TLD), respectively. The order in which ATM and MRN act in the early phase of the DSB response is unclear. Here we show that functional MRN is required for ATM activation, and consequently for timely activation of ATM-mediated pathways. Collectively, these and previous results assign to components of the MRN complex roles upstream and downstream of ATM in the DNA damage response pathway and explain the clinical resemblance between A-T and A-TLD.


Subject(s)
Ataxia Telangiectasia/genetics , Cell Cycle Proteins/genetics , DNA Damage , DNA-Binding Proteins/genetics , Gene Expression Regulation , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Saccharomyces cerevisiae Proteins/genetics , Ataxia Telangiectasia/pathology , Ataxia Telangiectasia Mutated Proteins , Cell Line , Cell Survival , DNA-Binding Proteins/deficiency , Humans , MRE11 Homologue Protein , Nuclear Proteins/deficiency , Phosphorylation , Reference Values , Tumor Suppressor Proteins
16.
Nephrol Dial Transplant ; 18(8): 1493-504, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12897086

ABSTRACT

BACKGROUND: Hepatocyte growth factor/scatter factor (HGF/SF) binds to its tyrosine kinase receptor, Met, thereby stimulating diverse cellular responses. The multifunctional docking site in the C-terminal domain mediates the signal of phosphorylated Met receptors to multiple transducers. The tyrosine at position 1356 of the Met docking site is crucial for cell motility and morphogenesis. METHODS: We examined the in situ distribution patterns of the Tyr1356-phosphorylated form of Met with a novel monoclonal antibody following renal injury and renal hypertrophy in rats. Sections of the kidney following either sham operation, transient ischaemia of one kidney or unilateral nephrectomy were analysed using indirect immunofluorescence staining and confocal laser scanning microscopy analysis of total Met protein levels and Tyr1356-phosphorylated Met (Met and pMet, respectively). RESULTS: At 6 h post-treatment, pMet increases in ischaemic kidneys compared with sham-operated kidneys, and these changes become substantial after 48 h in both medulla and cortex of ischaemic kidneys (P < 0.001). We also show significant up-regulation of Met predominantly in the medulla of ischaemic kidneys, 48 h following injury (P < 0.009). Inter-estingly, the stimulus for hypertrophy in the remnant kidney after uninephrectomy and the contra-lateral kidney during ischaemia is not accom-panied by significant up-regulation of Met or pMet staining compared with sham operation at both time points. CONCLUSIONS: We demonstrate in this work, for the first time, in situ detection of tyrosine kinase growth factor receptor docking site activation during pathological processes in the kidney. Using this methodology, we show a significant increase in Met docking site activity in both renal medulla and cortex solely following stimulation by ischaemia and repair.


Subject(s)
Hepatocyte Growth Factor/metabolism , Kidney Cortex/pathology , Kidney Medulla/pathology , Proto-Oncogene Proteins c-met/metabolism , Animals , Antibodies, Monoclonal , Binding Sites , Hyperplasia , Hypertrophy , Kidney Cortex/physiopathology , Kidney Medulla/physiopathology , Male , Microscopy, Confocal , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Up-Regulation/physiology
17.
Methods Cell Sci ; 24(4): 169-80, 2002.
Article in English | MEDLINE | ID: mdl-12843706

ABSTRACT

Spermatogenesis consists of spermatogonial proliferation, meiosis and spermatid differentiation. Laser scanning confocal microscopy (LSCM) may be used as an advanced analytical tool to follow spermatogenesis inside the seminiferous tubules without performing histological sections. For this purpose, separated seminiferous tubules are fixed in 0.5% paraformaldehyde, stained for DNA with propidium iodide and analyzed by LSCM. By producing longitudinal optical sections in the layer of spermatogonia, spermatocytes and spermatids, stage-specific changes in their structure may be followed within the tubules by LSCM. Longitudinal z-sections may be obtained to produce three-dimensional images of the seminiferous tubules. In addition, different proteins may be followed during spermatogenesis in a stage specific manner within the tubule by incubation of the fixed seminiferous tubules with appropriate antibodies. As an example of the spermatogenesis studies using described LSCM techniques, detailed examination of spermatogonia, spermatocytes and spermatids during golden hamster spermatogenesis is presented. LSCM analysis of c-kit and SC3 protein expression at different stages of hamster spermatogenesis is demonstrated.


Subject(s)
Microscopy, Confocal/methods , Seminiferous Tubules/cytology , Spermatogenesis/physiology , Animals , Antibodies , Coloring Agents , Cricetinae , DNA/metabolism , Immunohistochemistry/methods , Male , Mesocricetus , Microscopy, Confocal/instrumentation , Models, Biological , Proto-Oncogene Proteins c-kit/metabolism , Seminiferous Tubules/physiology , Spermatids/cytology , Spermatids/physiology , Spermatocytes/cytology , Spermatocytes/physiology , Spermatogonia/cytology , Spermatogonia/physiology , Tissue Fixation/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...