Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 223(2)2024 02 05.
Article in English | MEDLINE | ID: mdl-38059900

ABSTRACT

Subcellular location and activation of Tank Binding Kinase 1 (TBK1) govern precise progression through mitosis. Either loss of activated TBK1 or its sequestration from the centrosomes causes errors in mitosis and growth defects. Yet, what regulates its recruitment and activation on the centrosomes is unknown. We identified that NAK-associated protein 1 (NAP1) is essential for mitosis, binding to and activating TBK1, which both localize to centrosomes. Loss of NAP1 causes several mitotic and cytokinetic defects due to inactivation of TBK1. Our quantitative phosphoproteomics identified numerous TBK1 substrates that are not only confined to the centrosomes but are also associated with microtubules. Substrate motifs analysis indicates that TBK1 acts upstream of other essential cell cycle kinases like Aurora and PAK kinases. We also identified NAP1 as a TBK1 substrate phosphorylating NAP1 at S318 to promote its degradation by the ubiquitin proteasomal system. These data uncover an important distinct function for the NAP1-TBK1 complex during cell division.


Subject(s)
Adaptor Proteins, Signal Transducing , Cytokinesis , Mitosis , Protein Serine-Threonine Kinases , Humans , Cell Cycle , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
3.
J Cell Biol ; 220(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33464298

ABSTRACT

Genome-wide CRISPR screens have transformed our ability to systematically interrogate human gene function, but are currently limited to a subset of cellular phenotypes. We report a novel pooled screening approach for a wider range of cellular and subtle subcellular phenotypes. Machine learning and convolutional neural network models are trained on the subcellular phenotype to be queried. Genome-wide screening then utilizes cells stably expressing dCas9-KRAB (CRISPRi), photoactivatable fluorescent protein (PA-mCherry), and a lentiviral guide RNA (gRNA) pool. Cells are screened by using microscopy and classified by artificial intelligence (AI) algorithms, which precisely identify the genetically altered phenotype. Cells with the phenotype of interest are photoactivated and isolated via flow cytometry, and the gRNAs are identified by sequencing. A proof-of-concept screen accurately identified PINK1 as essential for Parkin recruitment to mitochondria. A genome-wide screen identified factors mediating TFEB relocation from the nucleus to the cytosol upon prolonged starvation. Twenty-one of the 64 hits called by the neural network model were independently validated, revealing new effectors of TFEB subcellular localization. This approach, AI-photoswitchable screening (AI-PS), offers a novel screening platform capable of classifying a broad range of mammalian subcellular morphologies, an approach largely unattainable with current methodologies at genome-wide scale.


Subject(s)
CRISPR-Cas Systems/genetics , Genetic Testing , Genome , Imaging, Three-Dimensional , Artificial Intelligence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cyclic AMP Response Element-Binding Protein A/metabolism , Deep Learning , Green Fluorescent Proteins , HEK293 Cells , Humans , Models, Biological , Neural Networks, Computer , Phenotype , Reproducibility of Results , Single-Cell Analysis , Support Vector Machine , Ubiquitin-Protein Ligases/metabolism , RNA, Guide, CRISPR-Cas Systems
4.
Mol Cell ; 80(5): 779-795.e10, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33207181

ABSTRACT

Protein aggregates disrupt cellular homeostasis, causing toxicity linked to neurodegeneration. Selective autophagic elimination of aggregates is critical to protein quality control, but how aggregates are selectively targeted for degradation is unclear. We compared the requirements for autophagy receptor proteins: OPTN, NBR1, p62, NDP52, and TAX1BP1 in clearance of proteotoxic aggregates. Endogenous TAX1BP1 is recruited to and required for the clearance of stress-induced aggregates, whereas ectopic expression of TAX1BP1 increases clearance through autophagy, promoting viability of human induced pluripotent stem cell-derived neurons. In contrast, TAX1BP1 depletion sensitizes cells to several forms of aggregate-induced proteotoxicity. Furthermore, TAX1BP1 is more specifically expressed in the brain compared to other autophagy receptor proteins. In vivo, loss of TAX1BP1 results in accumulation of high molecular weight ubiquitin conjugates and premature lipofuscin accumulation in brains of young TAX1BP1 knockout mice. TAX1BP1 mediates clearance of a broad range of cytotoxic proteins indicating therapeutic potential in neurodegenerative diseases.


Subject(s)
Apoptosis Regulatory Proteins/deficiency , Autophagy , Brain/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Neoplasm Proteins/deficiency , Neurodegenerative Diseases/metabolism , Protein Aggregation, Pathological/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Brain/pathology , Female , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lipofuscin/genetics , Lipofuscin/metabolism , Male , Mice , Mice, Knockout , Neoplasm Proteins/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Rats , Rats, Sprague-Dawley , Ubiquitin/genetics , Ubiquitin/metabolism
5.
J Mol Biol ; 432(8): 2510-2524, 2020 04 03.
Article in English | MEDLINE | ID: mdl-31689437

ABSTRACT

Selective autophagy of mitochondria, or mitophagy, refers to the specific removal and degradation of damaged or surplus mitochondria via targeting to the lysosome for destruction. Disruptions in this homeostatic process may contribute to disease. The identification of diverse mitophagic pathways and how selectivity for each of these pathways is conferred is just beginning to be understood. The removal of both damaged and healthy mitochondria under disease and physiological conditions is controlled by either ubiquitin-dependent or receptor-dependent mechanisms. In this review, we will discuss the known types of mitophagy observed in mammals, recent findings related to PINK1/Parkin-mediated mitophagy (which is the most well-studied form of mitophagy), the implications of defective mitophagy to neurodegenerative processes, and unanswered questions inspiring future research that would enhance our understanding of mitochondrial quality control.


Subject(s)
Mitochondria/pathology , Mitophagy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , Humans , Mitochondria/metabolism , Signal Transduction , Ubiquitinated Proteins/metabolism , Ubiquitination
6.
Cell Rep ; 29(1): 225-235.e5, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31577952

ABSTRACT

PINK1 and Parkin are established mediators of mitophagy, the selective removal of damaged mitochondria by autophagy. PINK1 and Parkin have been proposed to act as tumor suppressors, as loss-of-function mutations are correlated with enhanced tumorigenesis. However, it is unclear how PINK1 and Parkin act in coordination during mitophagy to influence the cell cycle. Here we show that PINK1 and Parkin genetically interact with proteins involved in cell cycle regulation, and loss of PINK1 and Parkin accelerates cell growth. PINK1- and Parkin-mediated activation of TBK1 at the mitochondria during mitophagy leads to a block in mitosis due to the sequestration of TBK1 from its physiological role at centrosomes during mitosis. Our study supports a diverse role for the far-reaching, regulatory effects of mitochondrial quality control in cellular homeostasis and demonstrates that the PINK1/Parkin pathway genetically interacts with the cell cycle, providing a framework for understanding the molecular basis linking PINK1 and Parkin to mitosis.


Subject(s)
Cell Cycle/genetics , Mitochondria/genetics , Mitosis/genetics , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Autophagy/genetics , Cell Line, Tumor , Cell Proliferation/genetics , HCT116 Cells , HEK293 Cells , HeLa Cells , Homeostasis/genetics , Humans , Mitophagy/genetics
7.
Cell Stress Chaperones ; 24(3): 481-492, 2019 05.
Article in English | MEDLINE | ID: mdl-31073902

ABSTRACT

The first Autumn School on Proteostasis was held at the Mediterranean Institute for Life Sciences (MedILS) in Split, Croatia, from November 12th-16th, 2018, bringing together 44 graduate students and postdoctoral fellows and 22 principal investigators from around the world. This meeting was geared towards providing students with an in-depth understanding of the field of proteostasis, with the aim of broadening their perspectives of the field. Session topics covered multiple aspects of cellular and organismal proteostasis, including fundamental principles, responses to heat shock, aging and disease, and protein folding, misfolding, and degradation. The structure of the meeting and the restricted number of participants afforded the students and postdocs the opportunity to interact with principal investigators to discuss not only their latest research, but also their career prospects and progression in a close, supportive environment.


Subject(s)
Aging/pathology , Molecular Chaperones/metabolism , Proteostasis Deficiencies , Proteostasis/physiology , Animals , Heat-Shock Response/physiology , Homeostasis , Humans , Protein Folding , Proteolysis
9.
Sci Signal ; 11(527)2018 04 24.
Article in English | MEDLINE | ID: mdl-29692360

ABSTRACT

In this issue of Science Signaling, Lu et al reveal a role for Parkin-mediated mitophagy in beige-to-white adipocyte transition. In the absence of Parkin, mitochondria in thermogenic beige adipocytes are preserved even after the elimination of cold mimetic stimuli-in contrast to their typical elimination during the white transition.


Subject(s)
Adipocytes, Beige , Adipose Tissue, White , Mitophagy , Thermogenesis , Ubiquitin-Protein Ligases
10.
Elife ; 72018 01 23.
Article in English | MEDLINE | ID: mdl-29360040

ABSTRACT

Damaged mitochondria are selectively eliminated by mitophagy. Parkin and PINK1, gene products mutated in familial Parkinson's disease, play essential roles in mitophagy through ubiquitination of mitochondria. Cargo ubiquitination by E3 ubiquitin ligase Parkin is important to trigger selective autophagy. Although autophagy receptors recruit LC3-labeled autophagic membranes onto damaged mitochondria, how other essential autophagy units such as ATG9A-integrated vesicles are recruited remains unclear. Here, using mammalian cultured cells, we demonstrate that RABGEF1, the upstream factor of the endosomal Rab GTPase cascade, is recruited to damaged mitochondria via ubiquitin binding downstream of Parkin. RABGEF1 directs the downstream Rab proteins, RAB5 and RAB7A, to damaged mitochondria, whose associations are further regulated by mitochondrial Rab-GAPs. Furthermore, depletion of RAB7A inhibited ATG9A vesicle assembly and subsequent encapsulation of the mitochondria by autophagic membranes. These results strongly suggest that endosomal Rab cycles on damaged mitochondria are a crucial regulator of mitophagy through assembling ATG9A vesicles.


Subject(s)
Endosomes/enzymology , Guanine Nucleotide Exchange Factors/metabolism , Mitophagy , Ubiquitin-Protein Ligases/metabolism , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/metabolism , Autophagy-Related Proteins/metabolism , HeLa Cells , Humans , Membrane Proteins/metabolism , Protein Interaction Maps , Vesicular Transport Proteins/metabolism , rab7 GTP-Binding Proteins
12.
mBio ; 7(4)2016 07 19.
Article in English | MEDLINE | ID: mdl-27435465

ABSTRACT

UNLABELLED: Coxiella burnetii replicates in an acidified lysosome-derived vacuole. Biogenesis of the Coxiella-containing vacuole (CCV) requires bacterial effector proteins delivered into host cells by the Dot/Icm secretion system. Genetic and cell biological analysis revealed that an effector protein called Cig2 promotes constitutive fusion of autophagosomes with the CCV to maintain this compartment in an autolysosomal stage of maturation. This distinguishes the CCV from other pathogen-containing vacuoles that are targeted by the host autophagy pathway, which typically confers host resistance to infection by delivering the pathogen to a toxic lysosomal environment. By maintaining the CCV in an autolysosomal stage of maturation, Cig2 enabled CCV homotypic fusion and enhanced bacterial virulence in the Galleria mellonella (wax moth) model of infection by a mechanism that decreases host tolerance. Thus, C. burnetii residence in an autolysosomal organelle alters host tolerance of infection, which indicates that Cig2-dependent manipulation of a lysosome-derived vacuole influences the host response to infection. IMPORTANCE: Coxiella burnetii is an obligate, intracellular bacterial pathogen that replicates inside a unique, lysosome-like compartment called the Coxiella-containing vacuole (CCV). Over 130 bacterial effector proteins are delivered into the host cell cytosol by the C. burnetii Dot/Icm type IV secretion system. Although the Dot/Icm system is essential for pathogenesis, the functions of most effectors remain unknown. Here we show that the effector protein Cig2 is essential for converting the CCV to an organelle that is similar to the autolysosome. Cig2 function promotes constitutive fusion between the CCV and autophagosomes generated by selective autophagy. Cig2-directed biogenesis of an autolysosomal vacuole is essential for the unique fusogenic properties of the CCV and for virulence in an animal model of disease. This work highlights how bacterial subversion of the host autophagy pathway can influence the cell biological properties of the CCV and influence the host response to infection.


Subject(s)
Autophagosomes/metabolism , Bacterial Proteins/metabolism , Coxiella burnetii/pathogenicity , Host-Pathogen Interactions , Vacuoles/metabolism , Vacuoles/microbiology , Animals , Disease Models, Animal , Disease Resistance , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/microbiology , Lepidoptera
13.
Nature ; 524(7565): 309-314, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26266977

ABSTRACT

Protein aggregates and damaged organelles are tagged with ubiquitin chains to trigger selective autophagy. To initiate mitophagy, the ubiquitin kinase PINK1 phosphorylates ubiquitin to activate the ubiquitin ligase parkin, which builds ubiquitin chains on mitochondrial outer membrane proteins, where they act to recruit autophagy receptors. Using genome editing to knockout five autophagy receptors in HeLa cells, here we show that two receptors previously linked to xenophagy, NDP52 and optineurin, are the primary receptors for PINK1- and parkin-mediated mitophagy. PINK1 recruits NDP52 and optineurin, but not p62, to mitochondria to activate mitophagy directly, independently of parkin. Once recruited to mitochondria, NDP52 and optineurin recruit the autophagy factors ULK1, DFCP1 and WIPI1 to focal spots proximal to mitochondria, revealing a function for these autophagy receptors upstream of LC3. This supports a new model in which PINK1-generated phospho-ubiquitin serves as the autophagy signal on mitochondria, and parkin then acts to amplify this signal. This work also suggests direct and broader roles for ubiquitin phosphorylation in other autophagy pathways.


Subject(s)
Autophagy/physiology , Mitophagy/physiology , Nuclear Proteins/metabolism , Protein Kinases/metabolism , Transcription Factor TFIIIA/metabolism , Autophagy-Related Protein-1 Homolog , Autophagy-Related Proteins , Carrier Proteins/metabolism , Cell Cycle Proteins , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Models, Biological , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
14.
Mol Cell ; 56(3): 360-375, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25284222

ABSTRACT

Phosphorylation is often used to promote protein ubiquitylation, yet we rarely understand quantitatively how ligase activation and ubiquitin (UB) chain assembly are integrated with phosphoregulation. Here we employ quantitative proteomics and live-cell imaging to dissect individual steps in the PINK1 kinase-PARKIN UB ligase mitochondrial control pathway disrupted in Parkinson's disease. PINK1 plays a dual role by phosphorylating PARKIN on its UB-like domain and poly-UB chains on mitochondria. PARKIN activation by PINK1 produces canonical and noncanonical UB chains on mitochondria, and PARKIN-dependent chain assembly is required for accumulation of poly-phospho-UB (poly-p-UB) on mitochondria. In vitro, PINK1 directly activates PARKIN's ability to assemble canonical and noncanonical UB chains and promotes association of PARKIN with both p-UB and poly-p-UB. Our data reveal a feedforward mechanism that explains how PINK1 phosphorylation of both PARKIN and poly-UB chains synthesized by PARKIN drives a program of PARKIN recruitment and mitochondrial ubiquitylation in response to mitochondrial damage.


Subject(s)
Mitochondria/enzymology , Polyubiquitin/biosynthesis , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Feedback, Physiological , HeLa Cells , Humans , Membrane Potential, Mitochondrial , Mutation, Missense , Parkinson Disease/enzymology , Phosphorylation , Protein Kinases/metabolism , Protein Multimerization , Protein Transport , Proteomics , Ubiquitin-Protein Ligases/genetics
15.
J Cell Biol ; 205(2): 143-53, 2014 Apr 28.
Article in English | MEDLINE | ID: mdl-24751536

ABSTRACT

PINK1 kinase activates the E3 ubiquitin ligase Parkin to induce selective autophagy of damaged mitochondria. However, it has been unclear how PINK1 activates and recruits Parkin to mitochondria. Although PINK1 phosphorylates Parkin, other PINK1 substrates appear to activate Parkin, as the mutation of all serine and threonine residues conserved between Drosophila and human, including Parkin S65, did not wholly impair Parkin translocation to mitochondria. Using mass spectrometry, we discovered that endogenous PINK1 phosphorylated ubiquitin at serine 65, homologous to the site phosphorylated by PINK1 in Parkin's ubiquitin-like domain. Recombinant TcPINK1 directly phosphorylated ubiquitin and phospho-ubiquitin activated Parkin E3 ubiquitin ligase activity in cell-free assays. In cells, the phosphomimetic ubiquitin mutant S65D bound and activated Parkin. Furthermore, expression of ubiquitin S65A, a mutant that cannot be phosphorylated by PINK1, inhibited Parkin translocation to damaged mitochondria. These results explain a feed-forward mechanism of PINK1-mediated initiation of Parkin E3 ligase activity.


Subject(s)
Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Amino Acid Substitution , Animals , Cell Line , Drosophila melanogaster , Enzyme Activation/physiology , Humans , Mutation, Missense , Phosphorylation/physiology , Protein Kinases/genetics , Protein Structure, Tertiary , Ubiquitin/genetics , Ubiquitin-Protein Ligases/genetics
16.
Nature ; 496(7445): 372-6, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23503661

ABSTRACT

The PARKIN ubiquitin ligase (also known as PARK2) and its regulatory kinase PINK1 (also known as PARK6), often mutated in familial early-onset Parkinson's disease, have central roles in mitochondrial homeostasis and mitophagy. Whereas PARKIN is recruited to the mitochondrial outer membrane (MOM) upon depolarization via PINK1 action and can ubiquitylate porin, mitofusin and Miro proteins on the MOM, the full repertoire of PARKIN substrates--the PARKIN-dependent ubiquitylome--remains poorly defined. Here we use quantitative diGly capture proteomics (diGly) to elucidate the ubiquitylation site specificity and topology of PARKIN-dependent target modification in response to mitochondrial depolarization. Hundreds of dynamically regulated ubiquitylation sites in dozens of proteins were identified, with strong enrichment for MOM proteins, indicating that PARKIN dramatically alters the ubiquitylation status of the mitochondrial proteome. Using complementary interaction proteomics, we found depolarization-dependent PARKIN association with numerous MOM targets, autophagy receptors, and the proteasome. Mutation of the PARKIN active site residue C431, which has been found mutated in Parkinson's disease patients, largely disrupts these associations. Structural and topological analysis revealed extensive conservation of PARKIN-dependent ubiquitylation sites on cytoplasmic domains in vertebrate and Drosophila melanogaster MOM proteins. These studies provide a resource for understanding how the PINK1-PARKIN pathway re-sculpts the proteome to support mitochondrial homeostasis.


Subject(s)
Membrane Potential, Mitochondrial , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Proteome/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Animals , Cell Line , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Humans , Mice , Mitochondria/chemistry , Protein Kinases/metabolism , Proteomics
17.
Dev Cell ; 18(2): 167-8, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20159586

ABSTRACT

The TRF1 subunit of the shelterin complex controls telomere length by regulating telomerase access to chromosome ends. Work from Zeng et al. (2010) now reveals in atomic detail how a battle between the SCF(FBX4) ubiquitin ligase and the shelterin component TIN2 controls TRF1 abundance and telomere length homeostasis.


Subject(s)
Telomere/genetics , Telomere/metabolism , Telomeric Repeat Binding Protein 1/metabolism , Humans , Models, Biological , SKP Cullin F-Box Protein Ligases/metabolism , Shelterin Complex , Telomere-Binding Proteins/metabolism
18.
Mol Cell ; 19(5): 667-78, 2005 Sep 02.
Article in English | MEDLINE | ID: mdl-16137622

ABSTRACT

DNA methylation is interpreted by a family of methyl-CpG binding domain (MBD) proteins that repress transcription through recruitment of corepressors that modify chromatin. To compare in vivo binding of MeCP2 and MBD2, we analyzed immunoprecipitated chromatin from primary human cells. Genomic sites occupied by the two MBD proteins were mutually exclusive. As MeCP2 was unable to colonize sites vacated by depletion of MBD2, we tested the hypothesis that methyl-CpG alone is insufficient to direct MeCP2 binding. In vitro selection for MeCP2 bound DNA-enriched fragments containing A/T bases ([A/T] > or = 4) adjacent to methyl-CpG. [A/T] > or = 4 was found to be essential for high-affinity binding at selected sites and at known MeCP2 target regions in the Bdnf and Dlx6 genes. MBD2 binding, however, did not require an A/T run. The unexpected restriction of MeCP2 to a defined subset of methyl-CpG sites will facilitate identification of genomic targets that are relevant to Rett Syndrome.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , DNA/metabolism , Repressor Proteins/metabolism , Adenine/chemistry , Base Sequence , Brain-Derived Neurotrophic Factor/genetics , Cell Line , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/genetics , CpG Islands/genetics , DNA Methylation , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Humans , Methyl-CpG-Binding Protein 2 , Mutation , Protein Binding , Repressor Proteins/genetics , Thymine/chemistry
19.
Cancer Genet Cytogenet ; 161(1): 63-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16080959

ABSTRACT

Cell lines derived from tumors contain numerous chromosomal aberrations and are the focus of study in tumor evolution. The ovarian teratocarcinoma cell line PA-1 demonstrates a single chromosomal aberration: a reciprocal t(15;20)(p11.2;q11.2). A complete molecular genetic analysis was undertaken to characterize this cell line. The PA-1 cell line was studied with fluorescence in situ hybridization (FISH), spectral karyotyping (SKY), bacterial artificial chromosome (BAC) microarray, and Western blotting. Amplification of 20q is frequently implicated in both breast and ovarian cancer; this region contains a number of oncogenes including MDM2, ZNF217, and the ovarian tumor marker WFDC2 (alias HE4). FISH revealed gene amplification of AIB1 (now known as NCOA3) but not STK15 (now known as AURKA). Immunoblot analysis demonstrated 3.6-fold overexpression of the AIB1 protein product, but no elevation of the STK15. BAC cancer gene microarray analysis showed gene amplification of > or =1.20 for five oncogenes. The presence of a consistent single change in PA-1, the t(15;20)(p11.2;q11.2), suggests that the aberration is significant with respect to the transformation status of the cell line. This translocation appears to cause overexpression of AIB1 (and perhaps other proteins), which may provide an immortalizing effect on this cell line.


Subject(s)
Chromosome Aberrations , Chromosomes, Artificial, Bacterial/genetics , Ovarian Neoplasms/genetics , Teratocarcinoma/genetics , Translocation, Genetic/genetics , Blotting, Western , Chromosome Banding , Chromosomes, Human, Pair 20/genetics , Chromosomes, Human, Pair 8/genetics , Female , Humans , In Situ Hybridization, Fluorescence , Microarray Analysis , Oncogenes/physiology , Ovarian Neoplasms/pathology , Spectral Karyotyping , Teratocarcinoma/pathology , Tumor Cells, Cultured
20.
J Mol Diagn ; 7(2): 183-6, 2005 May.
Article in English | MEDLINE | ID: mdl-15858141

ABSTRACT

Chronic granulomatous disease (CGD) is a rare inherited disorder in which antimicrobial activity of phagocytes is impaired due to the lack of reactive oxygen species, or oxidative burst, produced by NADPH oxidase. The X-linked form of CGD, representing approximately 70% of all cases, is caused by mutations in the cytochrome b beta subunit (CYBB) gene, which maps to chromosome Xp21.1. CYBB encodes the gp91-phox protein, a necessary component in the NADPH oxidase pathway. A wide variety of mutations have been identified in X-linked CGD patients, all of which lead to deletion of the functional protein and no oxidative burst activity. The mutations vary from single nucleotide substitutions to deletions of the entire gene. In this article, we report a mutation detection method for probands of female relatives at risk for carrier status of large deletions of the CYBB gene. Through fluorescent in situ hybridization of metaphase chromosomes, we were able to consistently distinguish carriers from noncarriers using polymerase chain reaction-derived, labeled DNA specific for exons 2 to 13 of the CYBB region at Xp21.1.


Subject(s)
Gene Deletion , Genetic Carrier Screening/methods , Granulomatous Disease, Chronic/genetics , In Situ Hybridization, Fluorescence/methods , Membrane Glycoproteins/genetics , NADPH Oxidases/genetics , Exons , Female , Heterozygote , Humans , Male , NADPH Oxidase 2 , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...