Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Gastroenterology ; 155(3): 799-814.e13, 2018 09.
Article in English | MEDLINE | ID: mdl-29775598

ABSTRACT

BACKGROUND & AIMS: Markers of the epithelial-to-mesenchymal transition (EMT) in gastric tumor tissues are associated with poor patient outcomes. We performed a screen to identify pharmacologic compounds that kill gastric cancer cells with EMT-associated gene expression patterns and investigate their mechanisms. METHODS: We identified 29 gastric cancer cell lines with a gene expression signature previously associated with an EMT subtype, based on data from RNA sequence analyses, and confirmed the mesenchymal phenotypes of 7 lines (Hs746T, SNU1750, MKN1, SK4, SNU484, SNU668, and YCC11), based on invasive activity and protein markers. We screened 1,345 compounds for their ability to kill cells with the EMT signature compared with cell lines without this pattern. We tested the effects of identified compounds in BALB/c nude mice bearing GA077 tumors; mice were given intraperitoneal injections of the compound or vehicle (control) twice daily for 24 days and tumor growth was monitored. Proteins associated with the toxicity of the compounds were overexpressed in MKN1 and SNU484 cells or knocked down in MKN45 and SNU719 using small interfering RNAs. We performed immunohistochemical analyses of 942 gastric cancer tissues and investigated associations between EMT markers and protein expression patterns. RESULTS: The nicotinamide phosphoribosyltransferase inhibitor FK866 killed 6 of 7 gastric cancer cell lines with EMT-associated gene expression signatures but not gastric cancer cells without this signature. The 6 EMT-subtype gastric cell lines expressed significantly low levels of nicotinic acid phosphoribosyltransferase (NAPRT), which makes the cells hypersensitive to nicotinamide phosphoribosyltransferase inhibition. Gastric cell lines that expressed higher levels of NAPRT, regardless of EMT markers, were sensitized to FK866 after knockdown of NAPRT, whereas overexpression of NAPRT in deficient EMT cell lines protected them from FK866-mediated toxicity. Administration of FK866 to nude mice with tumors grown from GA077 cells (human gastric cancer tumors of the EMT subtype) led to tumor regression in 2 weeks; FK866 did not affect tumors grown from MKN45 cells without the EMT expression signature. Loss of NAPRT might promote the EMT, because it stabilizes ß-catenin. We correlated the EMT gene expression signature with lower levels of NAPRT in 942 gastric tumors from patients; we also found lower levels of NAPRT mRNA in colorectal, pancreatic, and lung adenocarcinoma tissues with the EMT gene expression signature. CONCLUSIONS: FK866 selectively kills gastric cancer cells with an EMT gene expression signature by inhibiting nicotinamide phosphoribosyltransferase in cells with NAPRT deficiency. Loss of NAPRT expression, frequently through promoter hypermethylation, is observed in many gastric tumors of the EMT subtype. FK866 might be used to treat patients with tumors of this subtype.


Subject(s)
Acrylamides/pharmacology , Antineoplastic Agents/pharmacology , Cytokines/antagonists & inhibitors , Epithelial-Mesenchymal Transition/drug effects , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Piperidines/pharmacology , Stomach Neoplasms/drug therapy , Animals , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Small Interfering/administration & dosage , Stomach Neoplasms/genetics
2.
J Neurosci ; 35(48): 15934-47, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26631474

ABSTRACT

CNS neurons in adult mammals do not spontaneously regenerate axons after spinal cord injury. Preconditioning peripheral nerve injury allows the dorsal root ganglia (DRG) sensory axons to regenerate beyond the injury site by promoting expression of regeneration-associated genes. We have previously shown that peripheral nerve injury increases the number of macrophages in the DRGs and that the activated macrophages are critical to the enhancement of intrinsic regeneration capacity. The present study identifies a novel chemokine signal mediated by CCL2 that links regenerating neurons with proregenerative macrophage activation. Neutralization of CCL2 abolished the neurite outgrowth activity of conditioned medium obtained from neuron-macrophage cocultures treated with cAMP. The neuron-macrophage interactions that produced outgrowth-promoting conditioned medium required CCL2 in neurons and CCR2/CCR4 in macrophages. The conditioning effects were abolished in CCL2-deficient mice at 3 and 7 d after sciatic nerve injury, but CCL2 was dispensable for the initial growth response and upregulation of GAP-43 at the 1 d time point. Intraganglionic injection of CCL2 mimicked conditioning injury by mobilizing M2-like macrophages. Finally, overexpression of CCL2 in DRGs promoted sensory axon regeneration in a rat spinal cord injury model without harmful side effects. Our data suggest that CCL2-mediated neuron-macrophage interaction plays a critical role for amplification and maintenance of enhanced regenerative capacity by preconditioning peripheral nerve injury. Manipulation of chemokine signaling mediating neuron-macrophage interactions may represent a novel therapeutic approach to promote axon regeneration after CNS injury.


Subject(s)
Chemokine CCL2/metabolism , Macrophages/physiology , Nerve Regeneration/genetics , Neurons/physiology , Peripheral Nerve Injuries/physiopathology , Animals , Cells, Cultured , Chemokine CCL2/genetics , Cholera Toxin/metabolism , Coculture Techniques , Dependovirus/genetics , Disease Models, Animal , Female , Ganglia, Spinal/cytology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Regeneration/physiology , Nerve Tissue Proteins/metabolism , Neurites/physiology , Neurons/cytology , Pain Measurement , Pain Threshold/physiology , Rats , Rats, Sprague-Dawley , Receptors, CCR2/genetics , Receptors, CCR2/metabolism
3.
Biochem Biophys Res Commun ; 471(2): 274-81, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26820531

ABSTRACT

Biomarkers that are identified from a single study often appear to be biologically irrelevant or false positives. Meta-analysis techniques allow integrating data from multiple studies that are related but independent in order to identify biomarkers across multiple conditions. However, existing biomarker meta-analysis methods tend to be sensitive to the dataset being analyzed. Here, we propose a meta-analysis method, iMeta, which integrates t-statistic and fold change ratio for improved robustness. For evaluation of predictive performance of the biomarkers identified by iMeta, we compare our method with other meta-analysis methods. As a result, iMeta outperforms the other methods in terms of sensitivity and specificity, and especially shows robustness to study variance increase; it consistently shows higher classification accuracy on diverse datasets, while the performance of the others is highly affected by the dataset being analyzed. Application of iMeta to 59 drug-induced liver injury studies identified three key biomarker genes: Zwint, Abcc3, and Ppp1r3b. Experimental evaluation using RT-PCR and qRT-PCR shows that their expressional changes in response to drug toxicity are concordant with the result of our method. iMeta is available at http://imeta.kaist.ac.kr/index.html.


Subject(s)
Biomarkers/metabolism , Chemical and Drug Induced Liver Injury/diagnosis , Chemical and Drug Induced Liver Injury/metabolism , Gene Expression Profiling/methods , Meta-Analysis as Topic , Software , Computer Simulation , Data Interpretation, Statistical , Data Mining/methods , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Models, Statistical , Multidrug Resistance-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Protein Phosphatase 1/metabolism , Reproducibility of Results , Sensitivity and Specificity , Systems Integration
4.
Genome Res ; 23(8): 1283-94, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23674306

ABSTRACT

Several studies have sought systematically to identify protein subcellular locations, but an even larger task is to map which of these proteins conditionally relocates in disease (the mislocalizome). Here, we report an integrative computational framework for mapping conditional location and mislocation of proteins on a proteome-wide scale, called a conditional location predictor (CoLP). Using CoLP, we mapped the locations of over 10,000 proteins in normal human brain and in glioma. The prediction showed 0.9 accuracy using 100 location tests of 20 randomly selected proteins. Of the 10,000 proteins, over 150 have a strong likelihood of mislocation under glioma, which is striking considering that few mislocation events have been identified in this disease previously. Using immunofluorescence and Western blotting in both primary cells and tissues, we successfully experimentally confirmed 15 mislocations. The most common type of mislocation occurs between the endoplasmic reticulum and the nucleus; for example, for RNF138, TLX3, and NFRKB. In particular, we found that the gene for the mislocating protein GFRA4 had a nonsynonymous point mutation in exon 2. Moreover, redirection of GFRA4 to its normal location, the plasma membrane, led to marked reductions in phospho-STAT3 and proliferation of glioma cells. This framework has the potential to track changes in protein location in many human diseases.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Proteome/metabolism , Brain/metabolism , Brain Neoplasms/pathology , Cell Proliferation , Disease Progression , Gene Ontology , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Glioma/pathology , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , Humans , Kinesins/metabolism , Molecular Sequence Annotation , Nerve Tissue Proteins/metabolism , Protein Transport , Proto-Oncogene Proteins c-ret/metabolism , STAT3 Transcription Factor/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases , Zebrafish Proteins
5.
BMC Genomics ; 15: 997, 2014 Nov 19.
Article in English | MEDLINE | ID: mdl-25406591

ABSTRACT

BACKGROUND: DNA methylation (DNAm) levels can be used to predict the chronological age of tissues; however, the characteristics of DNAm age signatures in normal and cancer tissues are not well studied using multiple studies. RESULTS: We studied approximately 4000 normal and cancer samples with multiple tissue types from diverse studies, and using linear and nonlinear regression models identified reliable tissue type-invariant DNAm age signatures. A normal signature comprising 127 CpG loci was highly enriched on the X chromosome. Age-hypermethylated loci were enriched for guanine-and-cytosine-rich regions in CpG islands (CGIs), whereas age-hypomethylated loci were enriched for adenine-and-thymine-rich regions in non-CGIs. However, the cancer signature comprised only 26 age-hypomethylated loci, none on the X chromosome, and with no overlap with the normal signature. Genes related to the normal signature were enriched for aging-related gene ontology terms including metabolic processes, immune system processes, and cell proliferation. The related gene products of the normal signature had more than the average number of interacting partners in a protein interaction network and had a tendency not to interact directly with each other. The genomic sequences of the normal signature were well conserved and the age-associated DNAm levels could satisfactorily predict the chronological ages of tissues regardless of tissue type. Interestingly, the age-associated DNAm increases or decreases of the normal signature were aberrantly accelerated in cancer samples. CONCLUSION: These tissue type-invariant DNAm age signatures in normal and cancer can be used to address important questions in developmental biology and cancer research.


Subject(s)
Aging/genetics , DNA Methylation/genetics , Epigenesis, Genetic , Neoplasms/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Chromosomes, Human, X/genetics , CpG Islands , Genome, Human , Humans , Infant , Middle Aged , Neoplasms/pathology
6.
Sci Rep ; 14(1): 7436, 2024 03 28.
Article in English | MEDLINE | ID: mdl-38548901

ABSTRACT

CRISPR/Cas9 technology has effectively targeted cancer-specific oncogenic hotspot mutations or insertion-deletions. However, their limited prevalence in tumors restricts their application. We propose a novel approach targeting passenger single nucleotide variants (SNVs) in haploinsufficient or essential genes to broaden therapeutic options. By disrupting haploinsufficient or essential genes through the cleavage of DNA in the SNV region using CRISPR/Cas9, we achieved the selective elimination of cancer cells without affecting normal cells. We found that, on average, 44.8% of solid cancer patients are eligible for our approach, a substantial increase compared to the 14.4% of patients with CRISPR/Cas9-applicable oncogenic hotspot mutations. Through in vitro and in vivo experiments, we validated our strategy by targeting a passenger mutation in the essential ribosomal gene RRP9 and haploinsufficient gene SMG6. This demonstrates the potential of our strategy to selectively eliminate cancer cells and expand therapeutic opportunities.


Subject(s)
CRISPR-Cas Systems , Neoplasms , Humans , Genes, Essential , Mutation , Nucleotides , Gene Editing , Neoplasms/genetics , Neoplasms/therapy
7.
FEBS Lett ; 581(29): 5640-4, 2007 Dec 11.
Article in English | MEDLINE | ID: mdl-18023281

ABSTRACT

DeltaNp63alpha is exclusively expressed in stem cells and progenitor cells of the stratified epithelia. It promotes cell proliferation by antagonizing p53 and related TAp63/TAp73. Here, we report that specific desumoylation by SUMO protease SuPr-1 provides a fine-tuning mechanism for DeltaNp63alpha repressor activity. We found that disrupting the sumoylation site compromised DeltaNp63alpha repressor activity profoundly against TAp63gamma and TAp73beta-mediated transcription activation, but not to p53-mediated transcription. We further found that SuPr-1 specifically bound to sumoylated DeltaNp63alpha and hydrolyzed SUMO. Consequently, SuPr-1 expression reduced DeltaNp63alpha repressor activity to TAp63gamma and TAp73beta, whereas p53-mediated transactivation was unaffected. Collectively, these data suggest that SuPr-1-mediated DeltaNp63alpha desumoylation elaborately regulates epithelial growth.


Subject(s)
DNA-Binding Proteins/metabolism , Peptide Hydrolases/metabolism , Repressor Proteins/metabolism , SUMO-1 Protein/metabolism , Trans-Activators/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Humans , Lysine/genetics , Lysine/metabolism , Mice , Molecular Sequence Data , Repressor Proteins/genetics , Sequence Alignment , Trans-Activators/genetics , Transcription Factors , Transcription, Genetic , Transfection , Tumor Suppressor Proteins/genetics
8.
Biomed Res Int ; 2017: 1016305, 2017.
Article in English | MEDLINE | ID: mdl-28691013

ABSTRACT

Current multiomics assay platforms facilitate systematic identification of functional entities that are mappable in a biological network, and computational methods that are better able to detect densely connected clusters of signals within a biological network are considered increasingly important. One of the most famous algorithms for detecting network subclusters is Molecular Complex Detection (MCODE). MCODE, however, is limited in simultaneous analyses of multiple, large-scale data sets, since it runs on the Cytoscape platform, which requires extensive computational resources and has limited coding flexibility. In the present study, we implemented the MCODE algorithm in R programming language and developed a related package, which we called MCODER. We found the MCODER package to be particularly useful in analyzing multiple omics data sets simultaneously within the R framework. Thus, we applied MCODER to detect pharmacologically tractable protein-protein interactions selectively elevated in molecular subtypes of ovarian and colorectal tumors. In doing so, we found that a single molecular subtype representing epithelial-mesenchymal transition in both cancer types exhibited enhanced production of the collagen-integrin protein complex. These results suggest that tumors of this molecular subtype could be susceptible to pharmacological inhibition of integrin signaling.


Subject(s)
Antineoplastic Agents/pharmacology , Multiprotein Complexes/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Software , Cluster Analysis , Humans , Time Factors
10.
Sci Rep ; 5: 8580, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25739475

ABSTRACT

Prediction of new disease indications for approved drugs by computational methods has been based largely on the genomics signatures of drugs and diseases. We propose a method for drug repositioning that uses the clinical signatures extracted from over 13 years of electronic medical records from a tertiary hospital, including >9.4 M laboratory tests from >530,000 patients, in addition to diverse genomics signatures. Cross-validation using over 17,000 known drug-disease associations shows this approach outperforms various predictive models based on genomics signatures and a well-known "guilt-by-association" method. Interestingly, the prediction suggests that terbutaline sulfate, which is widely used for asthma, is a promising candidate for amyotrophic lateral sclerosis for which there are few therapeutic options. In vivo tests using zebrafish models found that terbutaline sulfate prevents defects in axons and neuromuscular junction degeneration in a dose-dependent manner. A therapeutic potential of terbutaline sulfate was also observed when axonal and neuromuscular junction degeneration have already occurred in zebrafish model. Cotreatment with a ß2-adrenergic receptor antagonist, butoxamine, suggests that the effect of terbutaline is mediated by activation of ß2-adrenergic receptors.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Drug Repositioning , Electronic Health Records , Medical Informatics/methods , Terbutaline/therapeutic use , Amyotrophic Lateral Sclerosis/genetics , Data Mining , Databases, Factual , Datasets as Topic , Genomics/methods , Humans , Reproducibility of Results , Terbutaline/pharmacology
11.
PLoS One ; 10(9): e0136698, 2015.
Article in English | MEDLINE | ID: mdl-26335687

ABSTRACT

Undesirable toxicity is one of the main reasons for withdrawing drugs from the market or eliminating them as candidates in clinical trials. Although numerous studies have attempted to identify biomarkers capable of predicting pharmacotoxicity, few have attempted to discover robust biomarkers that are coherent across various species and experimental settings. To identify such biomarkers, we conducted meta-analyses of massive gene expression profiles for 6,567 in vivo rat samples and 453 compounds. After applying rigorous feature reduction procedures, our analyses identified 18 genes to be related with toxicity upon comparisons of untreated versus treated and innocuous versus toxic specimens of kidney, liver and heart tissue. We then independently validated these genes in human cell lines. In doing so, we found several of these genes to be coherently regulated in both in vivo rat specimens and in human cell lines. Specifically, mRNA expression of neuronal regeneration-related protein was robustly down-regulated in both liver and kidney cells, while mRNA expression of cathepsin D was commonly up-regulated in liver cells after exposure to toxic concentrations of chemical compounds. Use of these novel toxicity biomarkers may enhance the efficiency of screening for safe lead compounds in early-phase drug development prior to animal testing.


Subject(s)
Biomarkers/analysis , Cathepsin D/analysis , Nerve Tissue Proteins/analysis , Oncogene Proteins/analysis , Toxicogenetics , Animals , Cell Line , Gene Expression , Humans
12.
PLoS One ; 9(2): e88215, 2014.
Article in English | MEDLINE | ID: mdl-24520355

ABSTRACT

Traumatic spinal cord injury (SCI) often leads to debilitating loss of locomotor function. Neuroplasticity of spinal circuitry underlies some functional recovery and therefore represents a therapeutic target to improve locomotor function following SCI. However, the cellular and molecular mechanisms mediating neuroplasticity below the lesion level are not fully understood. The present study performed a gene expression profiling in the rat lumbar spinal cord at 1 and 3 weeks after contusive SCI at T9. Another group of rats received treadmill locomotor training (TMT) until 3 weeks, and gene expression profiles were compared between animals with and without TMT. Microarray analysis showed that many inflammation-related genes were robustly upregulated in the lumbar spinal cord at both 1 and 3 weeks after thoracic injury. Notably, several components involved in an early complement activation pathway were concurrently upregulated. In line with the microarray finding, the number of microglia substantially increased not only in the white matter but also in the gray matter. C3 and complement receptor 3 were intensely expressed in the ventral horn after injury. Furthermore, synaptic puncta near ventral motor neurons were frequently colocalized with microglia after injury, implicating complement activation and microglial cells in synaptic remodeling in the lumbar locomotor circuitry after SCI. Interestingly, TMT did not influence the injury-induced upregulation of inflammation-related genes. Instead, TMT restored pre-injury expression patterns of several genes that were downregulated by injury. Notably, TMT increased the expression of genes involved in neuroplasticity (Arc, Nrcam) and angiogenesis (Adam8, Tie1), suggesting that TMT may improve locomotor function in part by promoting neurovascular remodeling in the lumbar motor circuitry.


Subject(s)
Lumbar Vertebrae/pathology , Motor Activity/genetics , Physical Conditioning, Animal , Spinal Cord Injuries/genetics , Spinal Cord Injuries/physiopathology , Spinal Cord/pathology , Thoracic Injuries/genetics , Animals , Down-Regulation/genetics , Female , Gene Expression Profiling , Inflammation/genetics , Inflammation/pathology , Lumbar Vertebrae/physiopathology , Microglia/metabolism , Microglia/pathology , Oligonucleotide Array Sequence Analysis , Protein Interaction Maps/genetics , Rats , Rats, Sprague-Dawley , Recovery of Function/genetics , Reproducibility of Results , Spinal Cord/physiopathology , Spinal Cord Injuries/pathology , Thoracic Injuries/pathology , Thoracic Injuries/physiopathology , Up-Regulation/genetics
13.
BMC Syst Biol ; 5 Suppl 2: S1, 2011.
Article in English | MEDLINE | ID: mdl-22784566

ABSTRACT

BACKGROUND: Microarray analyses based on differentially expressed genes (DEGs) have been widely used to distinguish samples across different cellular conditions. However, studies based on DEGs have not been able to clearly determine significant differences between samples of pathophysiologically similar HIV-1 stages, e.g., between acute and chronic progressive (or AIDS) or between uninfected and clinically latent stages. We here suggest a novel approach to allow such discrimination based on stage-specific genetic features of HIV-1 infection. Our approach is based on co-expression changes of genes known to interact. The method can identify a genetic signature for a single sample as contrasted with existing protein-protein-based analyses with correlational designs. METHODS: Our approach distinguishes each sample using differentially co-expressed interacting protein pairs (DEPs) based on co-expression scores of individual interacting pairs within a sample. The co-expression score has positive value if two genes in a sample are simultaneously up-regulated or down-regulated. And the score has higher absolute value if expression-changing ratios are similar between the two genes. We compared characteristics of DEPs with that of DEGs by evaluating their usefulness in separation of HIV-1 stage. And we identified DEP-based network-modules and their gene-ontology enrichment to find out the HIV-1 stage-specific gene signature. RESULTS: Based on the DEP approach, we observed clear separation among samples from distinct HIV-1 stages using clustering and principal component analyses. Moreover, the discrimination power of DEPs on the samples (70-100% accuracy) was much higher than that of DEGs (35-45%) using several well-known classifiers. DEP-based network analysis also revealed the HIV-1 stage-specific network modules; the main biological processes were related to "translation," "RNA splicing," "mRNA, RNA, and nucleic acid transport," and "DNA metabolism." Through the HIV-1 stage-related modules, changing stage-specific patterns of protein interactions could be observed. CONCLUSIONS: DEP-based method discriminated the HIV-1 infection stages clearly, and revealed a HIV-1 stage-specific gene signature. The proposed DEP-based method might complement existing DEG-based approaches in various microarray expression analyses.


Subject(s)
Gene Expression Profiling/methods , HIV Infections/genetics , Microarray Analysis/methods , Algorithms , Cluster Analysis , Down-Regulation , HIV Infections/diagnosis , HIV-1/pathogenicity , Humans , RNA Splicing/genetics , Up-Regulation
14.
J Biol Chem ; 284(24): 16501-16512, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19369249

ABSTRACT

During mitosis, establishment of structurally and functionally sound bipolar spindles is necessary for maintaining the fidelity of chromosome segregation. Tumor-associated microtubule-associated protein (TMAP), also known as cytoskeleton-associated protein 2 (CKAP2), is a mitotic spindle-associated protein whose level is frequently up-regulated in various malignancies. Previous reports have suggested that TMAP is a potential regulator of mitotic spindle assembly and dynamics and that it is required for chromosome segregation to occur properly. So far, there have been no reports on how its mitosis-related functions are regulated. Here, we report that TMAP is hyper-phosphorylated at the C terminus specifically during mitosis. At least four different residues (Thr-578, Thr-596, Thr-622, and Ser-627) were responsible for the mitosis-specific phosphorylation of TMAP. Among these, Thr-622 was specifically phosphorylated by Cdk1-cyclin B1 both in vitro and in vivo. Interestingly, compared with the wild type, a phosphorylation-deficient mutant form of TMAP, in which Thr-622 had been replaced with an alanine (T622A), induced a significant increase in the frequency of metaphase cells with abnormal bipolar spindles, which often displayed disorganized, asymmetrical, or narrow and elongated morphologies. Formation of these abnormal bipolar spindles subsequently resulted in misalignment of metaphase chromosomes and ultimately caused a delay in the entry into anaphase. Moreover, such defects resulting from the T622A mutation were associated with a decrease in the rate of protein turnover at spindle microtubules. These findings suggest that Cdk1-cyclin B1-mediated phosphorylation of TMAP is important for and contributes to proper regulation of microtubule dynamics and establishment of functional bipolar spindles during mitosis.


Subject(s)
CDC2 Protein Kinase/metabolism , Cyclin B/metabolism , Cytoskeletal Proteins/metabolism , Neoplasms/metabolism , Spindle Apparatus/metabolism , CDC2 Protein Kinase/genetics , Cyclin B/genetics , Cyclin B1 , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Cytoskeleton/metabolism , HeLa Cells , Humans , Kidney/cytology , Mitosis/physiology , Mutagenesis , Neoplasms/pathology , Phosphorylation/physiology , Protein Structure, Tertiary , RNA, Small Interfering , Threonine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL