Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
SLAS Discov ; 25(6): 591-604, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32425084

ABSTRACT

Kidney fibrosis presents a hallmark of chronic kidney disease. With ever-increasing patient numbers and limited treatment options available, novel strategies for therapeutic intervention in kidney disease are warranted. Fibrosis commonly results from a wound healing response to repeated or chronic tissue damage, irrespective of the underlying etiology, and can occur in virtually any solid organ or tissue. In order to identify targets relevant for kidney fibrosis, we aimed to employ CRISPR screening in primary human kidney fibroblasts. We demonstrate that CRISPR technology can be applied in primary kidney fibroblasts and can furthermore be used to conduct arrayed CRISPR screening using a high-content imaging readout in a whole genome-wide manner. Hits coming out of this screen were validated using orthogonal approaches and present starting points for validation of novel targets relevant to kidney disease.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Fibrosis/genetics , Kidney Diseases/genetics , Kidney/drug effects , Fibroblasts/metabolism , Fibrosis/drug therapy , Fibrosis/pathology , Genome, Human/genetics , High-Throughput Nucleotide Sequencing/trends , Humans , Kidney/pathology , Kidney Diseases/drug therapy , Kidney Diseases/pathology , Molecular Targeted Therapy/trends
2.
Nat Commun ; 10(1): 1547, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30948712

ABSTRACT

The Hippo pathway maintains tissue homeostasis by negatively regulating the oncogenic transcriptional co-activators YAP and TAZ. Though functional inactivation of the Hippo pathway is common in tumors, mutations in core pathway components are rare. Thus, understanding how tumor cells inactivate Hippo signaling remains a key unresolved question. Here, we identify the kinase STK25 as an activator of Hippo signaling. We demonstrate that loss of STK25 promotes YAP/TAZ activation and enhanced cellular proliferation, even under normally growth-suppressive conditions both in vitro and in vivo. Notably, STK25 activates LATS by promoting LATS activation loop phosphorylation independent of a preceding phosphorylation event at the hydrophobic motif, which represents a form of Hippo activation distinct from other kinase activators of LATS. STK25 is significantly focally deleted across a wide spectrum of human cancers, suggesting STK25 loss may represent a common mechanism by which tumor cells functionally impair the Hippo tumor suppressor pathway.


Subject(s)
Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Cell Line , Cell Proliferation , Genes, Tumor Suppressor , Hippo Signaling Pathway , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
3.
Sci Signal ; 8(397): ra100, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26443704

ABSTRACT

The serine and threonine kinase MST1 is the mammalian homolog of Hippo. MST1 is a critical mediator of the migration, adhesion, and survival of T cells; however, these functions of MST1 are independent of signaling by its typical effectors, the kinase LATS and the transcriptional coactivator YAP. The kinase NDR1, a member of the same family of kinases as LATS, functions as a tumor suppressor by preventing T cell lymphomagenesis, which suggests that it may play a role in T cell homeostasis. We generated and characterized mice with a T cell-specific double knockout of Ndr1 and Ndr2 (Ndr DKO). Compared with control mice, Ndr DKO mice exhibited a substantial reduction in the number of naïve T cells in their secondary lymphoid organs. Mature single-positive thymocytes accumulated in the thymus in Ndr DKO mice. We also found that NDRs acted downstream of MST1 to mediate the egress of mature thymocytes from the thymus, as well as the interstitial migration of naïve T cells within popliteal lymph nodes. Together, our findings indicate that the kinases NDR1 and NDR2 function as downstream effectors of MST1 to mediate thymocyte egress and T cell migration.


Subject(s)
Lymphocytes/cytology , Lymphopenia/enzymology , Protein Serine-Threonine Kinases/physiology , Thymocytes/cytology , Thymus Gland/pathology , Transendothelial and Transepithelial Migration/physiology , Actins/physiology , Animals , Apoptosis , Cell Movement , Chemotaxis , Cytoskeleton/ultrastructure , Lymphocyte Count , Lymphoid Tissue/pathology , Lymphopenia/pathology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , T-Lymphocyte Subsets/cytology , rho GTP-Binding Proteins/physiology
4.
Nature ; 522(7555): 179-84, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-26017310

ABSTRACT

Genome sequencing has uncovered a new mutational phenomenon in cancer and congenital disorders called chromothripsis. Chromothripsis is characterized by extensive genomic rearrangements and an oscillating pattern of DNA copy number levels, all curiously restricted to one or a few chromosomes. The mechanism for chromothripsis is unknown, but we previously proposed that it could occur through the physical isolation of chromosomes in aberrant nuclear structures called micronuclei. Here, using a combination of live cell imaging and single-cell genome sequencing, we demonstrate that micronucleus formation can indeed generate a spectrum of genomic rearrangements, some of which recapitulate all known features of chromothripsis. These events are restricted to the mis-segregated chromosome and occur within one cell division. We demonstrate that the mechanism for chromothripsis can involve the fragmentation and subsequent reassembly of a single chromatid from a micronucleus. Collectively, these experiments establish a new mutational process of which chromothripsis is one extreme outcome.


Subject(s)
Chromosome Breakage , DNA Damage , Micronuclei, Chromosome-Defective , Cell Line , Cell Survival , Chromosome Segregation/genetics , DNA Copy Number Variations/genetics , Gene Rearrangement/genetics , Genomic Instability/genetics , Humans , Mutation/genetics , Neoplasms/genetics , S Phase/genetics , Single-Cell Analysis
5.
Nat Commun ; 6: 6822, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25879913

ABSTRACT

Artifacts introduced in whole-genome amplification (WGA) make it difficult to derive accurate genomic information from single-cell genomes and require different analytical strategies from bulk genome analysis. Here, we describe statistical methods to quantitatively assess the amplification bias resulting from whole-genome amplification of single-cell genomic DNA. Analysis of single-cell DNA libraries generated by different technologies revealed universal features of the genome coverage bias predominantly generated at the amplicon level (1-10 kb). The magnitude of coverage bias can be accurately calibrated from low-pass sequencing (∼0.1 × ) to predict the depth-of-coverage yield of single-cell DNA libraries sequenced at arbitrary depths. We further provide a benchmark comparison of single-cell libraries generated by multi-strand displacement amplification (MDA) and multiple annealing and looping-based amplification cycles (MALBAC). Finally, we develop statistical models to calibrate allelic bias in single-cell whole-genome amplification and demonstrate a census-based strategy for efficient and accurate variant detection from low-input biopsy samples.


Subject(s)
Alleles , Genome, Human , Genomics/methods , Single-Cell Analysis/methods , Statistics as Topic , Bias , Calibration , Cell Line , Gene Library , Humans , Nucleic Acid Amplification Techniques , Polymerase Chain Reaction , Sequence Analysis, DNA
6.
Cell ; 158(4): 833-848, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25126788

ABSTRACT

Genetically unstable tetraploid cells can promote tumorigenesis. Recent estimates suggest that ∼37% of human tumors have undergone a genome-doubling event during their development. This potentially oncogenic effect of tetraploidy is countered by a p53-dependent barrier to proliferation. However, the cellular defects and corresponding signaling pathways that trigger growth suppression in tetraploid cells are not known. Here, we combine RNAi screening and in vitro evolution approaches to demonstrate that cytokinesis failure activates the Hippo tumor suppressor pathway in cultured cells, as well as in naturally occurring tetraploid cells in vivo. Induction of the Hippo pathway is triggered in part by extra centrosomes, which alter small G protein signaling and activate LATS2 kinase. LATS2 in turn stabilizes p53 and inhibits the transcriptional regulators YAP and TAZ. These findings define an important tumor suppression mechanism and uncover adaptive mechanisms potentially available to nascent tumor cells that bypass this inhibitory regulation.


Subject(s)
Cytokinesis , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Cell Line, Tumor , Centrosome/metabolism , Epithelial Cells/metabolism , Hepatocytes/metabolism , Hippo Signaling Pathway , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Tetraploidy , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
7.
Cell Cycle ; 10(12): 1897-904, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21593588

ABSTRACT

The mammalian genome encodes four members of the NDR/LATS kinase family: NDR1 (STK38), NDR2 (STK38L), LATS1 and LATS2, which are highly conserved from yeast to man. Members of the NDR/LATS kinase family have been implicated in a variety of biological processes ranging from cell division and morphology to apoptosis and tumor suppression. In mammals, LATS1/2 function as central parts of the HIPPO tumor suppressor pathway by restricting the activity of the YAP/TAZ proto-oncogenes. Recent evidence suggested that NDR1/2 are also part of an extended HIPPO tumor suppressor pathway. Apart from functions in apoptosis signaling and tumor suppression, NDR1/2 have been implicated in controlling centrosome duplication and mitotic chromosome alignment downstream of the HIPPO kinase homologs MST1 and MST2. Significantly, we also reported recently that NDR1/2 are controlling G 1/S transition downstream of a third MST family member MST3. Intriguingly, this newly described MST3-NDR1/2 axis promotes G 1 progression by stabilizing c-myc and preventing p21 accumulation, indicating a potential pro-tumorigenic role for NDR kinases. Here, we discuss these novel cell cycle functions of NDR kinases in a broader context and elaborate on possible explanations for the opposing functions of NDR kinases in normal and tumor biology.


Subject(s)
Cell Cycle , Cyclin-Dependent Kinase Inhibitor p21/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-myc/physiology , Tumor Suppressor Proteins/physiology , Animals , Humans , Protein Stability
8.
Mol Cell Biol ; 31(7): 1382-95, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21262772

ABSTRACT

The G(1) phase of the cell cycle is an important integrator of internal and external cues, allowing a cell to decide whether to proliferate, differentiate, or die. Multiple protein kinases, among them the cyclin-dependent kinases (Cdks), control G(1)-phase progression and S-phase entry. With the regulation of apoptosis, centrosome duplication, and mitotic chromosome alignment downstream of the HIPPO pathway components MST1 and MST2, mammalian NDR kinases have been implicated to function in cell cycle-dependent processes. Although they are well characterized in terms of biochemical regulation and upstream signaling pathways, signaling mechanisms downstream of mammalian NDR kinases remain largely unknown. We identify here a role for human NDR in regulating the G(1)/S transition. In G(1) phase, NDR kinases are activated by a third MST kinase (MST3). Significantly, interfering with NDR and MST3 kinase expression results in G(1) arrest and subsequent proliferation defects. Furthermore, we describe the first downstream signaling mechanisms by which NDR kinases regulate cell cycle progression. Our findings suggest that NDR kinases control protein stability of the cyclin-Cdk inhibitor protein p21 by direct phosphorylation. These findings establish a novel MST3-NDR-p21 axis as an important regulator of G(1)/S progression of mammalian cells.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , G1 Phase , Protein Serine-Threonine Kinases/metabolism , S Phase , Cell Cycle Proteins/metabolism , Cell Line , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/chemistry , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Enzyme Activation , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Gene Knockdown Techniques , Humans , Phosphorylation , Phosphoserine/metabolism , Protein Binding , Protein Conformation , Protein Stability , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
9.
Mol Cell Biol ; 30(18): 4507-20, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20624913

ABSTRACT

MOB proteins are integral components of signaling pathways controlling important cellular processes, such as mitotic exit, centrosome duplication, apoptosis, and cell proliferation in eukaryotes. The human MOB protein family consists of six distinct members (human MOB1A [hMOB1A], -1B, -2, -3A, -3B, and -3C), with hMOB1A/B the best studied due to their putative tumor-suppressive functions through the regulation of NDR/LATS kinases. The roles of the other MOB proteins are less well defined. Accordingly, we characterized all six human MOB proteins in the context of NDR/LATS binding and their abilities to activate NDR/LATS kinases. hMOB3A/B/C proteins neither bind nor activate any of the four human NDR/LATS kinases. We found that both hMOB2 and hMOB1A bound to the N-terminal region of NDR1. However, our data suggest that the binding modes differ significantly. Our work revealed that hMOB2 competes with hMOB1A for NDR binding. hMOB2, in contrast to hMOB1A/B, is bound to unphosphorylated NDR. Moreover, RNA interference (RNAi) depletion of hMOB2 resulted in increased NDR kinase activity. Consistent with these findings, hMOB2 overexpression interfered with the functional roles of NDR in death receptor signaling and centrosome overduplication. In summary, our data indicate that hMOB2 is a negative regulator of human NDR kinases in biochemical and biological settings.


Subject(s)
Nerve Tissue Proteins/metabolism , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Humans , Nerve Tissue Proteins/genetics , Protein Binding , Protein Isoforms/genetics , Protein Serine-Threonine Kinases/genetics , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology
10.
Sci Signal ; 3(126): ra47, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20551432

ABSTRACT

Defective apoptosis contributes to the development of various human malignancies. The kinases nuclear Dbf2-related 1 (NDR1) and NDR2 mediate apoptosis downstream of the tumor suppressor proteins RASSF1A (Ras association domain family member 1A) and MST1 (mammalian Ste20-like kinase 1). To further analyze the role of NDR1 in apoptosis, we generated NDR1-deficient mice. Although NDR1 is activated by both intrinsic and extrinsic proapoptotic stimuli, which indicates a role for NDR1 in regulating apoptosis, NDR1-deficient T cells underwent apoptosis in a manner similar to that of wild-type cells in response to different proapoptotic stimuli. Analysis of the abundances of NDR1 and NDR2 proteins revealed that loss of NDR1 was functionally compensated for by an increase in the abundance of NDR2 protein. Despite this compensation, NDR1(-/-) and NDR1(+/-) mice were more prone to the development of T cell lymphomas than were wild-type mice. Tumor development in mice and humans was accompanied by a decrease in the overall amounts of NDR proteins in T cell lymphoma samples. Thus, reduction in the abundance of NDR1 triggered a decrease in the total amount of both isoforms. Together, our data suggest that a reduction in the abundances of the NDR proteins results in defective responses to proapoptotic stimuli, thereby facilitating the development of tumors.


Subject(s)
Lymphoma, T-Cell/etiology , Protein Serine-Threonine Kinases/physiology , Animals , Apoptosis , Humans , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/deficiency
11.
Curr Biol ; 19(20): 1692-702, 2009 Nov 03.
Article in English | MEDLINE | ID: mdl-19836237

ABSTRACT

BACKGROUND: Human MST/hSAV/LATS/hMOB tumor suppressor cascades are regulators of cell death and proliferation; however, little is known about other functions of MST/hMOB signaling. Mob1p, one of two MOB proteins in yeast, appears to play a role in spindle pole body duplication (the equivalent of mammalian centrosome duplication). We therefore investigated the role of human MOB proteins in centrosome duplication. We also addressed the regulation of human centrosome duplication by mammalian serine/threonine Ste20-like (MST) kinases, considering that MOB proteins can function together with Ste20-like kinases in eukaryotes. RESULTS: By studying the six human MOB proteins and five MST kinases, we found that MST1/hMOB1 signaling controls centrosome duplication. Overexpression of hMOB1 caused centrosome overduplication, whereas RNAi depletion of hMOB1 or MST1 impaired centriole duplication. Significantly, we delineated an hMOB1/MST1/NDR1 signaling pathway regulating centrosome duplication. More specifically, analysis of shRNA-resistant hMOB1 and NDR1 mutants revealed that a functional NDR/hMOB1 complex is critical for MST1 to phosphorylate NDR on the hydrophobic motif that in turn is required for human centrosome duplication. Furthermore, shRNA-resistant MST1 variants revealed that MST1 kinase activity is crucial for centrosome duplication whereas MST1 binding to the hSAV and RASSF1A tumor suppressor proteins is dispensable. Finally, by studying the PLK4/HsSAS-6/CP110 centriole assembly machinery, we also observed that normal daughter centriole formation depends on intact MST1/hMOB1/NDR signaling, although HsSAS-6 centriolar localization is not affected. CONCLUSIONS: Our observations propose a novel pathway in control of human centriole duplication after recruitment of HsSAS-6 to centrioles.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Centrosome/metabolism , Hepatocyte Growth Factor/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/physiology , Tumor Suppressor Proteins/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , COS Cells , Cell Line , Centrioles/metabolism , Chlorocebus aethiops , HeLa Cells , Hepatocyte Growth Factor/metabolism , Humans , Microtubules/metabolism , Phosphorylation , Proto-Oncogene Proteins/metabolism , RNA Interference , Signal Transduction , Tumor Suppressor Proteins/metabolism
12.
Blood ; 113(15): 3568-76, 2009 Apr 09.
Article in English | MEDLINE | ID: mdl-19204327

ABSTRACT

The mechanism of cell transformation by Fms-like tyrosine kinase 3 (FLT3) in acute myeloid leukemia (AML) is incompletely understood. The most prevalent activated mutant FLT3 ITD exhibits an altered signaling quality, including strong activation of the STAT5 transcription factor. FLT3 ITD has also been found partially retained as a high-mannose precursor in an intracellular compartment. To analyze the role of intracellular retention of FLT3 for transformation, we have generated FLT3 versions that are anchored in the perinuclear endoplasmic reticulum (ER) by appending an ER retention sequence containing a RRR (R3) motif. ER retention of R3, but not of corresponding A3 FLT3 versions, is shown by biochemical, fluorescence-activated cell sorting, and immunocytochemical analyses. ER anchoring reduced global autophosphorylation and diminished constitutive activation of ERK1/2 and AKT of the constitutively active FLT3 versions. ER anchoring was, however, associated with elevated signaling to STAT3. Transforming activity of the FLT3 D835Y mutant was suppressed by ER anchoring. In contrast, ER-anchored FLT3 ITD retained STAT5-activating capacity and was transforming in vitro and in vivo. The findings highlight another aspect of the different signaling quality of FLT3 ITD: It can transform cells from an intracellular location.


Subject(s)
Endoplasmic Reticulum/metabolism , Leukemia, Myeloid, Acute/metabolism , Signal Transduction/physiology , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism , Animals , Cell Division/physiology , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Gene Duplication , Humans , Kidney/cytology , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Inbred C3H , Mutagenesis , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism
13.
Curr Biol ; 18(23): 1889-95, 2008 Dec 09.
Article in English | MEDLINE | ID: mdl-19062280

ABSTRACT

Human NDR1 and 2 (NDR1/2) are serine-threonine protein kinases in a subgroup of the AGC kinase family. The mechanisms of physiological NDR1/2 activation and their function remain largely unknown. Here we report that Fas and TNF-alpha receptor stimulation activates human NDR1/2 by promoting phosphorylation at the hydrophobic motif (Thr444/442). Moreover, NDR1/2 are essential for Fas receptor-induced apoptosis as shown by the fact that NDR knockdown significantly reduced cell death whereas overexpression of the NDR1 kinase further potentiated apoptosis. Activation of NDR1/2 by death receptor stimulation is mediated by the tumor suppressor RASSF1A. Furthermore, RASSF1A-induced apoptosis largely depends on the presence of NDR1/2. Fas receptor stimulation promoted direct phosphorylation and activation of NDR1/2 by the mammalian STE20-like kinase 1 (MST1), a downstream effector of RASSF1A. Concurrently, the NDR1/2 coactivator MOB1 induced MST1-NDR-MOB1 complex formation, which is crucial for MST1-induced NDR1/2 phosphorylation upon induction of apoptosis. Our findings identify NDR1/2 as novel proapoptotic kinases and key members of the RASSF1A/MST1 signaling cascade.


Subject(s)
Enzyme Activation , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , fas Receptor/metabolism , Apoptosis , Cell Line , Gene Deletion , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Tumor Suppressor Proteins/genetics , fas Receptor/genetics
14.
Cell Signal ; 20(8): 1564-77, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18555663

ABSTRACT

The nuclear Dbf2-related protein kinases 1 and 2 (NDR1/2) are closely-related AGC family kinases that are strongly conserved through evolution. In mammals, they are activated inter alia by phosphorylation of an hydrophobic domain threonine-residue [NDR1(Thr-444)/NDR2(Thr-442)] by an extrinsic protein kinase followed by autophosphorylation of a catalytic domain serine-residue [NDR1(Ser-281)/NDR2(Ser-282)]. We examined NDR1/2 expression and regulation in primary cultures of neonatal rat cardiac myocytes and in perfused adult rat hearts. In myocytes, transcripts for NDR2, but not NDR1, were induced by the hypertrophic agonist, endothelin-1. NDR1(Thr-444) and NDR2(Thr-442) were rapidly phosphorylated (maximal in 15-30 min) in myocytes exposed to some phosphoprotein Ser-/Thr-phosphatase 1/2 inhibitors (calyculin A, okadaic acid) and, to a lesser extent, by hyperosmotic shock, low concentrations of H(2)O(2), or chelerythrine. In myocytes adenovirally-transduced to express FLAG-NDR2 (which exhibited a mainly-cytoplasmic localisation), the same agents increased FLAG-NDR2 activity as assessed by in vitro protein kinase assays, indicative of FLAG-NDR2(Ser-282/Thr-442) phosphorylation. Calyculin A-induced phosphorylation of NDR1(Thr-444)/NDR2(Thr-442) and activation of FLAG-NDR2 were inhibited by staurosporine, but not by other protein kinase inhibitors tested. In ex vivo rat hearts, NDR1(Thr-444)/NDR2(Thr-442) were phosphorylated in response to ischaemia-reperfusion or calyculin A. From a pathological viewpoint, we conclude that activities of NDR1 and NDR2 are responsive to cytotoxic stresses in heart preparations and this may represent a previously-unidentified response to myocardial ischaemia in vivo.


Subject(s)
Enzyme Inhibitors/pharmacology , Myocardium/enzymology , Myocytes, Cardiac/enzymology , Phosphoprotein Phosphatases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Cloning, Molecular , Enzyme Activation , Female , Marine Toxins , Myocardial Reperfusion Injury/enzymology , Osmotic Pressure , Oxazoles/pharmacology , Oxidative Stress , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Rats , Threonine/metabolism
15.
Biochim Biophys Acta ; 1784(1): 3-15, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17881309

ABSTRACT

The NDR (nuclear Dbf2-related) family of kinases is highly conserved from yeast to human, and has been classified as a subgroup of the AGC group of protein kinases based on the sequence of the catalytic domain. Like all other members of the AGC class of protein kinases, NDR kinases require the phosphorylation of conserved Ser/Thr residues for activation. Importantly, NDR family members have two unique stretches of primary sequence: an N-terminal regulatory (NTR) domain and an insert of several residues between subdomains VII and VIII of the kinase domain. The kinase domain insert functions as an auto-inhibitory sequence (AIS), while binding of the co-activator MOB (Mps-one binder) proteins to the NTR domain releases NDR kinases from inhibition of autophosphorylation. However, despite such advances in our understanding of the molecular activation mechanism(s) and physiological functions of NDR kinases in yeast and invertebrates, most biological NDR substrates still remain to be identified. Nevertheless, by showing that the centrosomal subpopulation of human NDR1/2 is required for proper centrosome duplication, the first biological role of human NDR1/2 kinases has been defined recently. How far NDR-driven centrosome overduplication could actually contribute to cellular transformation will also be discussed.


Subject(s)
Centrosome/physiology , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle/physiology , Humans , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...