Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Trends Biochem Sci ; 47(4): 352-366, 2022 04.
Article in English | MEDLINE | ID: mdl-35184951

ABSTRACT

Benzimidazole 1 (BUB1) and budding uninhibited by benzimidazole 1-related 1 (BUBR1) are multidomain paralogs with key roles in chromosome alignment during mitosis and the spindle assembly checkpoint (SAC), an evolutionarily conserved signaling pathway that monitors errors in chromosome segregation during cell division in eukaryotes. Although BUB1 and BUBR1 share a similar domain organization and short linear interaction motifs (SLiMs), they control distinct aspects of chromosome congression and the SAC. Here we discuss the roles of BUB1 and BUBR1 SLiMs in mitosis and complement this with additional insights gleamed from studying their evolution. We show that BUB1 and BUBR1 SLiMs form highly specific interactions that are carefully orchestrated in space and time and contend that they define BUB1 and BUBR1 as organizing hubs that drive SAC signaling and ensure genome stability.


Subject(s)
Mitosis , Protein Serine-Threonine Kinases , Cell Cycle Proteins/metabolism , Chromosome Segregation , Kinetochores/metabolism , Signal Transduction , Spindle Apparatus/metabolism
2.
Mol Cell Proteomics ; 17(10): 1979-1990, 2018 10.
Article in English | MEDLINE | ID: mdl-30002203

ABSTRACT

Signals from cell surface receptors are often relayed via adaptor proteins. NCK1 and NCK2 are Src-Homology (SH) 2 and 3 domain adaptors that regulate processes requiring a remodeling of the actin cytoskeleton. Evidence from gene inactivation in mouse suggests that NCK1 and NCK2 are functionally redundant, although recent reports support the idea of unique functions for NCK1 and NCK2. We sought to examine this question further by delineating NCK1- and NCK2-specific signaling networks. We used both affinity purification-mass spectrometry and BioID proximity labeling to identify NCK1/2 signaling networks comprised of 98 proteins. Strikingly, we found 30 proteins restricted to NCK1 and 28 proteins specifically associated with NCK2, suggesting differences in their function. We report that Nck2-/-, but not Nck1-/- mouse embryo fibroblasts (MEFs) are multinucleated and display extended protrusions reminiscent of intercellular bridges, which correlate with an extended time spent in cytokinesis as well as a failure of a significant proportion of cells to complete abscission. Our data also show that the midbody of NCK2-deficient cells is not only increased in length, but also altered in composition, as judged by the mislocalization of AURKB, PLK1 and ECT2. Finally, we show that NCK2 function during cytokinesis requires its SH2 domain. Taken together, our data delineate the first high-confidence interactome for NCK1/2 adaptors and highlight several proteins specifically associated with either protein. Thus, contrary to what is generally accepted, we demonstrate that NCK1 and NCK2 are not completely redundant, and shed light on a previously uncharacterized function for the NCK2 adaptor protein in cell division.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytokinesis , Oncogene Proteins/metabolism , Proteomics , Adaptor Proteins, Signal Transducing/chemistry , Animals , HEK293 Cells , HeLa Cells , Humans , Mass Spectrometry , Mice , Oncogene Proteins/chemistry , Protein Interaction Mapping , Protein Transport , Structure-Activity Relationship , src Homology Domains
3.
PLoS Genet ; 11(10): e1005582, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26496431

ABSTRACT

The co-chaperone BAG3, in complex with the heat shock protein HSPB8, plays a role in protein quality control during mechanical strain. It is part of a multichaperone complex that senses damaged cytoskeletal proteins and orchestrates their seclusion and/or degradation by selective autophagy. Here we describe a novel role for the BAG3-HSPB8 complex in mitosis, a process involving profound changes in cell tension homeostasis. BAG3 is hyperphosphorylated at mitotic entry and localizes to centrosomal regions. BAG3 regulates, in an HSPB8-dependent manner, the timely congression of chromosomes to the metaphase plate by influencing the three-dimensional positioning of the mitotic spindle. Depletion of BAG3 caused defects in cell rounding at metaphase and dramatic blebbing of the cortex associated with abnormal spindle rotations. Similar defects were observed upon silencing of the autophagic receptor p62/SQSTM1 that contributes to BAG3-mediated selective autophagy pathway. Mitotic cells depleted of BAG3, HSPB8 or p62/SQSTM1 exhibited disorganized actin-rich retraction fibres, which are proposed to guide spindle orientation. Proper spindle positioning was rescued in BAG3-depleted cells upon addition of the lectin concanavalin A, which restores cortex rigidity. Together, our findings suggest the existence of a so-far unrecognized quality control mechanism involving BAG3, HSPB8 and p62/SQSTM1 for accurate remodelling of actin-based mitotic structures that guide spindle orientation.


Subject(s)
Actin Cytoskeleton/genetics , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Heat-Shock Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Spindle Apparatus/genetics , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Autophagy/genetics , Chromosome Segregation/genetics , Concanavalin A/administration & dosage , HeLa Cells , Heat-Shock Proteins/metabolism , Heat-Shock Response/genetics , Humans , MCF-7 Cells , Mitosis/genetics , Molecular Chaperones , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Proteolysis , RNA, Small Interfering , Sequestosome-1 Protein
4.
J Cell Sci ; 127(Pt 16): 3415-23, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25052095

ABSTRACT

Knl1 (also known as CASC5, UniProt Q8NG31) is an evolutionarily conserved scaffolding protein that is required for proper kinetochore assembly, spindle assembly checkpoint (SAC) function and chromosome congression. A number of recent reports have confirmed the prominence of Knl1 in these processes and provided molecular details and structural features that dictate Knl1 functions in higher organisms. Knl1 recruits SAC components to the kinetochore and is the substrate of certain protein kinases and phosphatases, the interplay of which ensures the exquisite regulation of the aforementioned processes. In this Commentary, we discuss the overall domain organization of Knl1 and the roles of this protein as a versatile docking platform. We present emerging roles of the protein interaction motifs present in Knl1, including the RVSF, SILK, MELT and KI motifs, and their role in the recruitment and regulation of the SAC proteins Bub1, BubR1, Bub3 and Aurora B. Finally, we explore how the regions of low structural complexity that characterize Knl1 are implicated in the cooperative interactions that mediate binding partner recognition and scaffolding activity by Knl1.


Subject(s)
Kinetochores/metabolism , Microtubule-Associated Proteins/metabolism , Animals , Humans , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Protein Binding , Protein Structure, Tertiary
5.
Biochem J ; 457(2): 323-34, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24107129

ABSTRACT

Protein kinase-like domains that lack conserved residues known to catalyse phosphoryl transfer, termed pseudokinases, have emerged as important signalling domains across all kingdoms of life. Although predicted to function principally as catalysis-independent protein-interaction modules, several pseudokinase domains have been attributed unexpected catalytic functions, often amid controversy. We established a thermal-shift assay as a benchmark technique to define the nucleotide-binding properties of kinase-like domains. Unlike in vitro kinase assays, this assay is insensitive to the presence of minor quantities of contaminating kinases that may otherwise lead to incorrect attribution of catalytic functions to pseudokinases. We demonstrated the utility of this method by classifying 31 diverse pseudokinase domains into four groups: devoid of detectable nucleotide or cation binding; cation-independent nucleotide binding; cation binding; and nucleotide binding enhanced by cations. Whereas nine pseudokinases bound ATP in a divalent cation-dependent manner, over half of those examined did not detectably bind nucleotides, illustrating that pseudokinase domains predominantly function as non-catalytic protein-interaction modules within signalling networks and that only a small subset is potentially catalytically active. We propose that henceforth the thermal-shift assay be adopted as the standard technique for establishing the nucleotide-binding and catalytic potential of kinase-like domains.


Subject(s)
Janus Kinase 2/chemistry , Janus Kinase 2/classification , Real-Time Polymerase Chain Reaction/methods , Receptor, ErbB-3/chemistry , Receptor, ErbB-3/classification , Amino Acid Sequence , Animals , Cell Line , Humans , Insecta , Janus Kinase 2/genetics , Molecular Sequence Data , Protein Binding/physiology , Receptor, ErbB-3/genetics
6.
Cell Syst ; 15(6): 544-562.e8, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38861992

ABSTRACT

Most biological processes are regulated by signaling modules that bind to short linear motifs. For protein kinases, substrates may have full or only partial matches to the kinase recognition motif, a property known as "substrate quality." However, it is not clear whether differences in substrate quality represent neutral variation or if they have functional consequences. We examine this question for the kinase CK2, which has many fundamental functions. We show that optimal CK2 sites are phosphorylated at maximal stoichiometries and found in many conditions, whereas minimal substrates are more weakly phosphorylated and have regulatory functions. Optimal CK2 sites tend to be more conserved, and substrate quality is often tuned by selection. For intermediate sites, increases or decreases in substrate quality may be deleterious, as we demonstrate for a CK2 substrate at the kinetochore. The results together suggest a strong role for substrate quality in phosphosite function and evolution. A record of this paper's transparent peer review process is included in the supplemental information.


Subject(s)
Casein Kinase II , Casein Kinase II/metabolism , Phosphorylation , Humans , Substrate Specificity , Kinetochores/metabolism , Evolution, Molecular , Binding Sites
7.
J Biol Chem ; 287(8): 5988-6001, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22187426

ABSTRACT

Kinetochore targeting of the mitotic kinases Bub1, BubR1, and Mps1 has been implicated in efficient execution of their functions in the spindle checkpoint, the self-monitoring system of the eukaryotic cell cycle that ensures chromosome segregation occurs with high fidelity. In all three kinases, kinetochore docking is mediated by the N-terminal region of the protein. Deletions within this region result in checkpoint failure and chromosome segregation defects. Here, we use an interdisciplinary approach that includes biophysical, biochemical, cell biological, and bioinformatics methods to study the N-terminal region of human Mps1. We report the identification of a tandem repeat of the tetratricopeptide repeat (TPR) motif in the N-terminal kinetochore binding region of Mps1, with close homology to the tandem TPR motif of Bub1 and BubR1. Phylogenetic analysis indicates that TPR Mps1 was acquired after the split between deutorostomes and protostomes, as it is distinguishable in chordates and echinoderms. Overexpression of TPR Mps1 resulted in decreased efficiency of both chromosome alignment and mitotic arrest, likely through displacement of endogenous Mps1 from the kinetochore and decreased Mps1 catalytic activity. Taken together, our multidisciplinary strategy provides new insights into the evolution, structural organization, and function of Mps1 N-terminal region.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Computational Biology , M Phase Cell Cycle Checkpoints , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/metabolism , Amino Acid Motifs , Animals , Cattle , Cell Cycle Proteins/genetics , Chromosomes, Human/genetics , Enzyme Stability , Evolution, Molecular , HeLa Cells , Humans , Hydrogen-Ion Concentration , Mice , Models, Molecular , Protein Multimerization , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Protein Transport , Protein-Tyrosine Kinases/genetics , Rats
8.
Mol Cell Proteomics ; 10(1): M110.004457, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20860994

ABSTRACT

Polo-like kinases regulate many aspects of mitotic and meiotic progression from yeast to man. In early mitosis, mammalian Polo-like kinase 1 (Plk1) controls centrosome maturation, spindle assembly, and microtubule attachment to kinetochores. However, despite the essential and diverse functions of Plk1, the full range of Plk1 substrates remains to be explored. To investigate the Plk1-dependent phosphoproteome of the human mitotic spindle, we combined stable isotope labeling by amino acids in cell culture with Plk1 inactivation or depletion followed by spindle isolation and mass spectrometry. Our study identified 358 unique Plk1-dependent phosphorylation sites on spindle proteins, including novel substrates, illustrating the complexity of the Plk1-dependent signaling network. Over 100 sites were validated by in vitro phosphorylation of peptide arrays, resulting in a broadening of the Plk1 consensus motif. Collectively, our data provide a rich source of information on Plk1-dependent phosphorylation, Plk1 docking to substrates, the influence of phosphorylation on protein localization, and the functional interaction between Plk1 and Aurora A on the early mitotic spindle.


Subject(s)
Cell Cycle Proteins/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteome/metabolism , Proto-Oncogene Proteins/metabolism , Spindle Apparatus/enzymology , Amino Acid Motifs , Amino Acid Sequence , Aurora Kinases , Centrosome/enzymology , Consensus Sequence , Enzyme Activation , HeLa Cells , Humans , Kinesins/metabolism , Molecular Sequence Data , Phosphoproteins/chemistry , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Proteome/chemistry , RNA, Small Interfering/metabolism , Reproducibility of Results , Substrate Specificity , Polo-Like Kinase 1
9.
Biochem J ; 448(3): 321-8, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23067341

ABSTRACT

The SAC (spindle assembly checkpoint) is a surveillance system that ensures the timely and accurate transmission of the genetic material to offspring. The process implies kinetochore targeting of the mitotic kinases Bub1 (budding uninhibited by benzamidine 1), BubR1 (Bub1 related) and Mps1 (monopolar spindle 1), which is mediated by the N-terminus of each kinase. In the present study we report the 1.8 Å (1 Å=0.1 nm) crystal structure of the TPR (tetratricopeptide repeat) domain in the N-terminal region of human Mps1. The structure reveals an overall high similarity to the TPR motif of the mitotic checkpoint kinases Bub1 and BubR1, and a number of unique features that include the absence of the binding site for the kinetochore structural component KNL1 (kinetochore-null 1; blinkin), and determinants of dimerization. Moreover, we show that a stretch of amino acids at the very N-terminus of Mps1 is required for dimer formation, and that interfering with dimerization results in mislocalization and misregulation of kinase activity. The results of the present study provide an important insight into the molecular details of the mitotic functions of Mps1 including features that dictate substrate selectivity and kinetochore docking.


Subject(s)
Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/physiology , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/physiology , Heterotrimeric GTP-Binding Proteins/chemistry , Heterotrimeric GTP-Binding Proteins/physiology , M Phase Cell Cycle Checkpoints/physiology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/physiology , Amino Acid Sequence , Crystallography, X-Ray , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Protein Binding/physiology , Protein Structure, Tertiary , Repetitive Sequences, Amino Acid
10.
Nat Genet ; 36(9): 979-83, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15300251

ABSTRACT

The identification of tumor-suppressor genes in solid tumors by classical cancer genetics methods is difficult and slow. We combined nonsense-mediated RNA decay microarrays and array-based comparative genomic hybridization for the genome-wide identification of genes with biallelic inactivation involving nonsense mutations and loss of the wild-type allele. This approach enabled us to identify previously unknown mutations in the receptor tyrosine kinase gene EPHB2. The DU 145 prostate cancer cell line, originating from a brain metastasis, carries a truncating mutation of EPHB2 and a deletion of the remaining allele. Additional frameshift, splice site, missense and nonsense mutations are present in clinical prostate cancer samples. Transfection of DU 145 cells, which lack functional EphB2, with wild-type EPHB2 suppresses clonogenic growth. Taken together with studies indicating that EphB2 may have an essential role in cell migration and maintenance of normal tissue architecture, our findings suggest that mutational inactivation of EPHB2 may be important in the progression and metastasis of prostate cancer.


Subject(s)
Mutation , Prostatic Neoplasms/genetics , Receptor, EphB2/genetics , Cell Line, Tumor , Codon, Nonsense , Emetine/pharmacology , Genes, Tumor Suppressor , Humans , Male , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , RNA Stability , Transfection
11.
J Cell Sci ; 123(Pt 1): 84-94, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20016069

ABSTRACT

The BubR1 checkpoint protein performs multiple functions in mitosis. We have carried out a functional analysis of conserved motifs of human BubR1 (also known as BUB1B) and demonstrate that spindle assembly checkpoint (SAC) and chromosome attachment functions can be uncoupled from each other. Mutation of five proline-directed serine phosphorylation sites, identified in vivo by mass spectrometry, essentially abolishes attachment of chromosomes to the spindle but has no effect on SAC functionality. By contrast, mutation of the two conserved KEN boxes required for SAC function does not impact chromosome congression. Interestingly, the contribution of the two KEN-box motifs is not equal. Cdc20 associates with the N-terminal but not C-terminal KEN box, and mutation of the N-terminal KEN motif results in more severe acceleration of mitotic timing. Moreover, the two KEN motifs are not sufficient for maximal binding of Cdc20 and APC/C, which also requires sequences in the BubR1 C-terminus. Finally, mutation of the GLEBS motif causes loss of Bub3 interaction and mislocalization of BubR1 from the kinetochore; concomitantly, BubR1 phosphorylation as well as SAC activity and chromosome congression are impaired, indicating that the GLEBS motif is strictly required for both major functions of human BubR1.


Subject(s)
Amino Acid Motifs/genetics , Cell Cycle Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Spindle Apparatus/metabolism , CDC2 Protein Kinase/metabolism , Cdc20 Proteins , Cell Cycle Proteins/genetics , Cysteine Proteinase Inhibitors/pharmacology , Genes, cdc/drug effects , HeLa Cells , Humans , Kinetochores/drug effects , Leupeptins/pharmacology , Mitosis/drug effects , Mitosis/genetics , Mutation/genetics , Nocodazole/pharmacology , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Transport/drug effects , Protein Transport/genetics , Spindle Apparatus/drug effects , Spindle Apparatus/genetics , Tubulin Modulators/pharmacology
12.
PLoS One ; 17(7): e0268848, 2022.
Article in English | MEDLINE | ID: mdl-35776709

ABSTRACT

The Rho family Guanine nucleotide exchange factor (GEF) ARHGEF17 (also known as TEM4) is a large protein with only 3 annotated regions: an N-terminal actin-binding domain, a Rho-specific dbl homology (DH)- pleckstrin homology (PH) type GEF domain and a seven bladed ß propeller fold at the C-terminus with unknown function. TEM4 has been implicated in numerous activities that rely on regulation of the cytoskeleton including cell migration, cell-cell junction formation and the spindle assembly checkpoint during mitosis. Here we have assessed the specificity of a TEM4 polyclonal antibody that has been commonly used as a Western blotting and immunocytochemistry probe for TEM4 in mammalian cells. We find that this antibody, in addition to its intended target, cross-reacts with the Nuclear Mitotic Apparatus Protein 1 (NuMA) in Western blotting and immunoprecipitation, and detects NuMA preferentially in immunocytochemistry. This cross-reactivity, with an abundant chromatin- and mitotic spindle-associated factor, is likely to affect the interpretation of experiments that make use of this antibody probe, in particular by immunocytochemistry and immunoprecipitation.


Subject(s)
Antibodies , Cell Cycle Proteins , Rho Guanine Nucleotide Exchange Factors , Spindle Apparatus , Actins/metabolism , Animals , Antibodies/immunology , Cell Cycle Proteins/immunology , Mammals , Microtubules/metabolism , Mitosis , Rho Guanine Nucleotide Exchange Factors/immunology , Spindle Apparatus/metabolism
13.
Methods Enzymol ; 667: 507-534, 2022.
Article in English | MEDLINE | ID: mdl-35525552

ABSTRACT

Budding uninhibited by benzimidazole 1-related protein 1 (BUBR1) is a mitotic checkpoint (better known as the spindle assembly checkpoint) protein that forms part of an inhibitory complex required to delay mitosis when errors occur in the attachment between chromosomes and the mitotic spindle. If these errors remain uncorrected, it could result in unequal distribution of genetic material to each of the nascent daughter cells, leading to potentially disastrous consequences at both the cellular and organismal level. In some higher eukaryotes including vertebrates, BUBR1 has a C-terminal kinase fold that is largely thought to be inactive, whereas in many species this domain has been lost through evolution and the truncated protein is known as mitotic arrest deficient 3 (MAD3). Here we present advice and practical considerations for the design of experiments, their analysis and interpretation to study the functions of the vertebrate BUBR1 during mitosis with emphasis on analysis implicating the pseudokinase domain.


Subject(s)
Kinetochores , M Phase Cell Cycle Checkpoints , Animals , Cell Cycle Proteins/metabolism , HeLa Cells , Humans , Kinetochores/chemistry , Kinetochores/metabolism , Mitosis , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Spindle Apparatus/chemistry , Spindle Apparatus/genetics , Spindle Apparatus/metabolism
14.
Cancers (Basel) ; 14(15)2022 Aug 02.
Article in English | MEDLINE | ID: mdl-35954424

ABSTRACT

Loss of mitotic regulation is commonly observed in cancer and is a major cause of whole-chromosome aneuploidy. The identification of genes that play a role in the proper progression of mitosis can help us to understand the development and evolution of this disease. Here, we generated a list of proteins implicated in mitosis that we used to probe a patient-derived breast cancer (BC) continuum gene-expression dataset generated by our group by human transcriptome analysis of breast lesions of varying aggressiveness (from normal to invasive). We identified cytoskeleton-associated protein 2 (CKAP2) as an important mitotic regulator in invasive BC. The results showed that CKAP2 is overexpressed in invasive BC tumors when compared with normal tissues, and highly expressed in all BC subtypes. Higher expression of CKAP2 is also related to a worse prognosis in overall survival and relapse-free survival in estrogen receptor (ER)-positive and human epidermal growth factor receptor type 2 (HER2)-negative BC patients. Knockdown of CKAP2 in SKBR3 cells impaired cell proliferation and cell migration and reduced aggregate formation in a 3D culture. Our results show the important role of CKAP2 in BC tumorigenesis, and its potential utility as a prognostic marker in BC.

15.
Cell Rep ; 40(1): 111031, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35793621

ABSTRACT

EPH receptors (EPHRs) constitute the largest family among receptor tyrosine kinases in humans. They are mainly involved in short-range cell-cell communication events that regulate cell adhesion, migration, and boundary formation. However, the molecular mechanisms by which EPHRs control these processes are less understood. To address this, we unravel EPHR-associated complexes under native conditions using mass-spectrometry-based BioID proximity labeling. We obtain a composite proximity network from EPHA4, -B2, -B3, and -B4 that comprises 395 proteins, most of which were not previously linked to EPHRs. We examine the contribution of several BioID-identified candidates via loss-of-function in an EPHR-dependent cell-segregation assay. We find that the signaling scaffold PAR-3 is required for cell sorting and that EPHRs directly phosphorylate PAR-3. We also delineate a signaling complex involving the C-terminal SRC kinase (CSK), whose recruitment to PAR-3 is dependent on EPHR signals. Our work describes signaling networks by which EPHRs regulate cellular phenotypes.


Subject(s)
Receptors, Eph Family , Signal Transduction , CSK Tyrosine-Protein Kinase , Cell Communication , Software
16.
Mol Cell Oncol ; 8(2): 1876511, 2021.
Article in English | MEDLINE | ID: mdl-33860079

ABSTRACT

At the metaphase-to-anaphase transition, phosphatase activity feeds back to reverse early mitotic phosphorylation events. Our recent work indicates that the pseudokinase domain of the spindle checkpoint protein BUB1 (Budding Uninhibited by Benzimidazoles 1) mitotic checkpoint serine/threonine kinase B (BUB1B, BUBR1) maintains kinase-phosphatase balance at the outer kinetochore during mitotic exit.

17.
Mol Biol Cell ; 32(9): 880-891, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33085580

ABSTRACT

Accurate chromosome alignment at metaphase facilitates the equal segregation of sister chromatids to each of the nascent daughter cells. Lack of proper metaphase alignment is an indicator of defective chromosome congression and aberrant kinetochore-microtubule attachments which in turn promotes chromosome missegregation and aneuploidy, hallmarks of cancer. Tools to sensitively, accurately, and quantitatively measure chromosome alignment at metaphase will facilitate understanding of the contribution of chromosome segregation errors to the development of aneuploidy. In this work, we have developed and validated a method based on analytical geometry to measure several indicators of chromosome misalignment. We generated semiautomated and flexible ImageJ2/Fiji pipelines to quantify kinetochore misalignment at metaphase plates as well as lagging chromosomes at anaphase. These tools will ultimately allow sensitive and systematic quantitation of these chromosome segregation defects in cells undergoing mitosis.


Subject(s)
Chromosome Segregation/physiology , Image Processing, Computer-Assisted/methods , Metaphase/physiology , Chromatids , HeLa Cells , Humans , Kinetochores/physiology , Microscopy, Fluorescence/methods , Microtubules/physiology , Mitosis/physiology , Models, Theoretical , Spindle Apparatus
18.
Mol Cell Oncol ; 8(6): 1985930, 2021.
Article in English | MEDLINE | ID: mdl-35419475

ABSTRACT

We recently identified Zinc-finger protein 768 (ZNF768) as a novel transcription factor controlling cell fate decision downstream of Rat sarcoma virus (RAS). We showed that ZNF768 depletion impairs cell cycle progression and triggers cellular senescence, while its overexpression allows cells to bypass oncogene-induced senescence. Elevated ZNF768 levels is common in tumors, suggesting that ZNF768 may help to escape cellular senescence, sustain proliferation and promote malignant transformation. Here, we discuss these recent findings and highlight key questions emerging from our work.

19.
Front Immunol ; 12: 650808, 2021.
Article in English | MEDLINE | ID: mdl-34234773

ABSTRACT

The myeloid inhibitory C-type lectin receptor CLEC12A limits neutrophil activation, pro-inflammatory pathways and disease in mouse models of inflammatory arthritis by a molecular mechanism that remains poorly understood. We addressed how CLEC12A-mediated inhibitory signaling counteracts activating signaling by cross-linking CLEC12A in human neutrophils. CLEC12A cross-linking induced its translocation to flotillin-rich membrane domains where its ITIM was phosphorylated in a Src-dependent manner. Phosphoproteomic analysis identified candidate signaling molecules regulated by CLEC12A that include MAPKs, phosphoinositol kinases and members of the JAK-STAT pathway. Stimulating neutrophils with uric acid crystals, the etiological agent of gout, drove the hyperphosphorylation of p38 and Akt. Ultimately, one of the pathways through which CLEC12A regulates uric acid crystal-stimulated release of IL-8 by neutrophils is through a p38/PI3K-Akt signaling pathway. In summary this work defines early molecular events that underpin CLEC12A signaling in human neutrophils to modulate cytokine synthesis. Targeting this pathway could be useful therapeutically to dampen inflammation.


Subject(s)
Lectins, C-Type/immunology , Neutrophil Activation/immunology , Neutrophils/immunology , Phosphatidylinositol 3-Kinases/immunology , Proto-Oncogene Proteins c-akt/immunology , Receptors, Mitogen/immunology , Signal Transduction/immunology , Adult , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , HEK293 Cells , HeLa Cells , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Microscopy, Confocal , Neutrophils/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Mitogen/genetics , Receptors, Mitogen/metabolism , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Nat Commun ; 12(1): 4841, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34404770

ABSTRACT

RAS proteins are GTPases that lie upstream of a signaling network impacting cell fate determination. How cells integrate RAS activity to balance proliferation and cellular senescence is still incompletely characterized. Here, we identify ZNF768 as a phosphoprotein destabilized upon RAS activation. We report that ZNF768 depletion impairs proliferation and induces senescence by modulating the expression of key cell cycle effectors and established p53 targets. ZNF768 levels decrease in response to replicative-, stress- and oncogene-induced senescence. Interestingly, ZNF768 overexpression contributes to bypass RAS-induced senescence by repressing the p53 pathway. Furthermore, we show that ZNF768 interacts with and represses p53 phosphorylation and activity. Cancer genomics and immunohistochemical analyses reveal that ZNF768 is often amplified and/or overexpressed in tumors, suggesting that cells could use ZNF768 to bypass senescence, sustain proliferation and promote malignant transformation. Thus, we identify ZNF768 as a protein linking oncogenic signaling to the control of cell fate decision and proliferation.


Subject(s)
Cellular Senescence/genetics , Genes, ras/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Carcinogenesis , Cell Cycle , Cell Differentiation , Cell Proliferation , Cell Transformation, Neoplastic , DNA Replication , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Genomics , HeLa Cells , Humans , Oncogenes , Phenotype , Phosphoproteins , Phosphorylation , Repression, Psychology , Signal Transduction , ras Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL