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










Publication year range
1.
Stem Cells ; 36(6): 943-950, 2018 06.
Article in English | MEDLINE | ID: mdl-29430853

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) are necessary for life-long blood production and replenishment of the hematopoietic system during stress. We recently reported that nuclear factor I/X (Nfix) promotes HSPC survival post-transplant. Here, we report that ectopic expression of Nfix in primary mouse HSPCs extends their ex vivo culture from about 20 to 40 days. HSPCs overexpressing Nfix display hypersensitivity to supportive cytokines and reduced apoptosis when subjected to cytokine deprivation relative to controls. Ectopic Nfix resulted in elevated levels of c-Mpl transcripts and cell surface protein on primary murine HSPCs as well as increased phosphorylation of STAT5, which is known to be activated down-stream of c-MPL. Blocking c-MPL signaling by removal of thrombopoietin or addition of a c-MPL neutralizing antibody negated the antiapoptotic effect of Nfix overexpression on cultured HSPCs. Furthermore, NFIX was capable of binding to and transcriptionally activating a proximal c-Mpl promoter fragment. In sum, these data suggest that NFIX-mediated upregulation of c-Mpl transcription can protect primitive hematopoietic cells from stress ex vivo. Stem Cells 2018;36:943-950.


Subject(s)
Hematopoietic Stem Cells/metabolism , NFI Transcription Factors/metabolism , Receptors, Thrombopoietin/metabolism , Animals , Humans , Mice , Signal Transduction
3.
J Exp Med ; 213(3): 433-49, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26880577

ABSTRACT

Understanding the molecular regulation of hematopoietic stem and progenitor cell (HSPC) engraftment is paramount to improving transplant outcomes. To discover novel regulators of HSPC repopulation, we transplanted >1,300 mice with shRNA-transduced HSPCs within 24 h of isolation and transduction to focus on detecting genes regulating repopulation. We identified 17 regulators of HSPC repopulation: Arhgef5, Armcx1, Cadps2, Crispld1, Emcn, Foxa3, Fstl1, Glis2, Gprasp2, Gpr56, Myct1, Nbea, P2ry14, Smarca2, Sox4, Stat4, and Zfp251. Knockdown of each of these genes yielded a loss of function, except in the cases of Armcx1 and Gprasp2, whose loss enhanced hematopoietic stem cell (HSC) repopulation. The discovery of multiple genes regulating vesicular trafficking, cell surface receptor turnover, and secretion of extracellular matrix components suggests active cross talk between HSCs and the niche and that HSCs may actively condition the niche to promote engraftment. We validated that Foxa3 is required for HSC repopulating activity, as Foxa3(-/-) HSC fails to repopulate ablated hosts efficiently, implicating for the first time Foxa genes as regulators of HSPCs. We further show that Foxa3 likely regulates the HSC response to hematologic stress. Each gene discovered here offers a window into the novel processes that regulate stable HSPC engraftment into an ablated host.


Subject(s)
Genetic Association Studies , Hematopoietic Stem Cells/cytology , Amino Acid Motifs , Animals , Cell Proliferation , Cytoprotection , Enhancer Elements, Genetic/genetics , Genetic Testing , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Hepatocyte Nuclear Factor 3-gamma/metabolism , Mice, Inbred C57BL , Protein Binding , Reproducibility of Results , Signal Transduction , Stress, Physiological
4.
Blood ; 122(17): 2987-96, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24041575

ABSTRACT

Hematopoietic stem cells are both necessary and sufficient to sustain the complete blood system of vertebrates. Here we show that Nfix, a member of the nuclear factor I (Nfi) family of transcription factors, is highly expressed by hematopoietic stem and progenitor cells (HSPCs) of murine adult bone marrow. Although short hairpin RNA-mediated knockdown of Nfix expression in Lineage(-)Sca-1(+)c-Kit(+) HSPCs had no effect on in vitro cell growth or viability, Nfix-depleted HSPCs displayed a significant loss of colony-forming potential, as well as short- and long-term in vivo hematopoietic repopulating activity. Analysis of recipient mice at 4 to 20 days posttransplant revealed that Nfix-depleted HSPCs are established in the bone marrow, but fail to persist due to increased apoptotic cell death. Gene expression profiling of Nfix-depleted HSPCs reveals that loss of Nfix expression in HSPCs is concomitant with a decrease in the expression of multiple genes known to be important for HSPCs survival, such as Erg, Mecom, and Mpl. These data reveal that Nfix is a novel regulator of HSPCs survival posttransplantation and establish a role for Nfi genes in the regulation of this cellular compartment.


Subject(s)
Adult Stem Cells/metabolism , Bone Marrow Cells/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , NFI Transcription Factors/genetics , Adult Stem Cells/cytology , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Apoptosis , Bone Marrow Cells/cytology , Cell Survival , Gene Expression Profiling , Gene Expression Regulation , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , NFI Transcription Factors/deficiency , NFI Transcription Factors/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Thrombopoietin/genetics , Receptors, Thrombopoietin/metabolism , Signal Transduction , Transcription Factors , Transcriptional Regulator ERG
5.
Mol Biol Cell ; 22(23): 4588-601, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21998205

ABSTRACT

Septin family proteins oligomerize through guanosine 5'-triphosphate-binding domains into core heteromers, which in turn polymerize at the cleavage furrow of dividing fungal and animal cells. Septin assemblies during the interphase of animal cells remain poorly defined and are the topic of this report. In this study, we developed protocols for visualization of authentic higher-order assemblies using tagged septins to effectively replace the endogenous gene product within septin core heteromers in human cells. Our analysis revealed that septins assemble into microtubule-supported, disk-like structures at the plasma membrane. In the absence of cell substrate adhesion, this is the predominant higher-order arrangement in interphase cells and each of the seven to eight septin family members expressed by the two analyzed cell types appears equally represented. However, studies of myeloid and lymphoid cell model systems revealed cell type-specific alterations of higher-order septin arrangements in response to substrate adhesion. Live-cell observations suggested that all higher-order septin assemblies are mutually exclusive with plasma membrane regions undergoing remodeling. The combined data point to a mechanism by which densely arranged cortical microtubules, which are typical for nonadhered spherical cells, support plasma membrane-bound, disk-like septin assemblies.


Subject(s)
Cell Membrane/metabolism , Microtubules/metabolism , Septins/metabolism , Cell Adhesion , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cell Membrane/ultrastructure , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Jurkat Cells , K562 Cells , Leukemia, Erythroblastic, Acute/pathology , Permeability , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Septins/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/ultrastructure
6.
Exp Cell Res ; 316(12): 2017-26, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20399773

ABSTRACT

Op18/stathmin (Op18) is a microtubule-destabilizing protein that is phosphorylation-inactivated during mitosis and its normal function is to govern tubulin subunit partitioning during interphase. Human tumors frequently overexpress Op18 and a tumor-associated Q18-->E mutation has been identified that confers hyperactivity, destabilizes spindle microtubules, and causes mitotic aberrancies, polyploidization, and chromosome loss in K562 leukemia cells. Here we determined whether wild-type and mutant Op18 have the potential to cause chromosomal instability by some means other than interference with spindle assembly, and thereby bypassing the spindle assembly checkpoint. Our approach was based on Op18 derivatives with distinct temporal order of activity during mitosis, conferred either by differential phosphorylation inactivation or by anaphase-specific degradation through fusion with the destruction box of cyclin B1. We present evidence that excessive Op18 activity generates chromosomal instability through interference occurring subsequent to the metaphase-to-anaphase transition, which reduces the fidelity of chromosome segregation to spindle poles during anaphase. Similar to uncorrected merotelic attachment, this mechanism evades detection by the spindle assembly checkpoint and thus provides an additional route to chromosomal instability.


Subject(s)
Chromosomal Instability/genetics , Spindle Apparatus/metabolism , Stathmin/genetics , Stathmin/metabolism , Up-Regulation , Anaphase , Humans , Jurkat Cells , K562 Cells , Metaphase , Spindle Apparatus/genetics , Transfection
7.
Cell Mol Life Sci ; 66(20): 3263-76, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19585080

ABSTRACT

The microtubule-system organizes the cytoplasm during interphase and segregates condensed chromosomes during mitosis. Four unrelated conserved proteins, XMAP215/Dis1/TOGp, MCAK, MAP4 and Op18/stathmin, have all been implicated as predominant regulators of tubulin monomer-polymer partitioning in animal cells. However, while studies employing the Xenopus egg extract model system indicate that the partitioning is largely governed by the counteractive activities of XMAP215 and MCAK, studies of human cell lines indicate that MAP4 and Op18 are the predominant regulators of the interphase microtubule-array. Here, we review functional interplay of these proteins during interphase and mitosis in various cell model systems. We also review the evidence that MAP4 and Op18 have interphase-specific, counteractive and phosphorylation-inactivated activities that govern tubulin subunit partitioning in many mammalian cell types. Finally, we discuss evidence indicating that partitioning regulation by MAP4 and Op18 may be of significance to establish cell polarity.


Subject(s)
Cell Polarity , Mammals/metabolism , Tubulin/metabolism , Animals , Cell Line , Humans , Interphase/physiology , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/physiology , Microtubules/metabolism , Models, Biological , Protein Processing, Post-Translational , Stathmin/metabolism , Stathmin/physiology , Tubulin/physiology
8.
Infect Immun ; 77(3): 1144-54, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19124604

ABSTRACT

The Sm16/SmSLP/SmSPO-1 (Sm16) protein is secreted by the parasite Schistosoma mansoni during skin penetration and has been ascribed immunosuppressive activities. Here we describe the strategy behind the design of a modified Sm16 protein with a decreased aggregation propensity, thus facilitating the expression and purification of an Sm16 protein that is soluble in physiological buffers. The Stokes radii and sedimentation coefficients of recombinant and native proteins indicate that Sm16 is an approximately nine-subunit oligomer. Analysis of truncated Sm16 derivatives showed that both oligomerization and binding to the plasma membrane of human cells depend on multiple C-terminal regions. For analysis of immunomodulatory activities, Sm16 was expressed in Pichia pastoris to facilitate the preparation of a pyrogen/endotoxin-free purified protein. Recombinant Sm16 was found to have no effect on T-lymphocyte activation, cell proliferation, or the basal level of cytokine production by whole human blood or monocytic cells. However, Sm16 exerts potent inhibition of the cytokine response to the Toll-like receptor (TLR) ligands lipopolysaccharide (LPS) and poly(I:C) while being less efficient at inhibiting the response to the TLR ligand peptidoglycan or a synthetic lipopeptide. Since Sm16 specifically inhibits the degradation of the IRAK1 signaling protein in LPS-stimulated monocytes, our findings indicate that inhibition is exerted proximal to the TLR complex.


Subject(s)
Helminth Proteins/biosynthesis , Helminth Proteins/immunology , Recombinant Proteins/biosynthesis , Schistosoma mansoni/immunology , Signal Transduction/immunology , Toll-Like Receptors/immunology , Animals , Cytokines/biosynthesis , Cytokines/immunology , Flow Cytometry , Helminth Proteins/chemistry , Humans , Immunoblotting , Lymphocyte Activation/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology
9.
Mol Biol Cell ; 19(7): 2897-906, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18434595

ABSTRACT

Op18/stathmin (Op18), a conserved microtubule-depolymerizing and tubulin heterodimer-binding protein, is a major interphase regulator of tubulin monomer-polymer partitioning in diverse cell types in which Op18 is abundant. Here, we addressed the question of whether the microtubule regulatory function of Op18 includes regulation of tubulin heterodimer synthesis. We used two human cell model systems, K562 and Jurkat, combined with strategies for regulatable overexpression or depletion of Op18. Although Op18 depletion caused extensive overpolymerization and increased microtubule content in both cell types, we did not detect any alteration in polymer stability. Interestingly, however, we found that Op18 mediates positive regulation of tubulin heterodimer content in Jurkat cells, which was not observed in K562 cells. By analysis of cells treated with microtubule-poisoning drugs, we found that Jurkat cells regulate tubulin mRNA levels by a posttranscriptional mechanism similarly to normal primary cells, whereas this mechanism is nonfunctional in K562 cells. We present evidence that Op18 mediates posttranscriptional regulation of tubulin mRNA in Jurkat cells through the same basic autoregulatory mechanism as microtubule-poisoning drugs. This, combined with potent regulation of tubulin monomer-polymer partitioning, enables Op18 to exert global regulation of the microtubule system.


Subject(s)
Gene Expression Regulation , Interphase , Microtubule Proteins/metabolism , Microtubules/metabolism , Stathmin/metabolism , Colchicine/pharmacology , Dimerization , Humans , Jurkat Cells , K562 Cells , Phenotype , Polymers/chemistry , RNA Processing, Post-Transcriptional , Reverse Transcriptase Polymerase Chain Reaction , Tubulin/chemistry
10.
Exp Cell Res ; 314(6): 1367-77, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18262179

ABSTRACT

Op18/stathmin (Op18) is a phosphorylation-regulated and differentially expressed microtubule-destabilizing protein in animal cells. Op18 regulates tubulin monomer-polymer partitioning of the interphase microtubule system and forms complexes with tubulin heterodimers. Recent reports have shown that specific tubulin-folding cofactors and related proteins may disrupt tubulin heterodimers. We therefore investigated whether Op18 protects unpolymerized tubulin from such disruptive activities. Our approach was based on inducible overexpression of two tubulin-disrupting proteins, namely TBCE, which is required for tubulin biogenesis, and E-like, which has been proposed to regulate tubulin turnover and microtubule stability. Expression of either of these proteins was found to cause a rapid degradation of both alpha-tubulin and beta-tubulin subunits of unpolymerized, but not polymeric, tubulin heterodimers. We found that depletion of Op18 by means of RNA interference increased the susceptibility of tubulin to TBCE or E-like mediated disruption, while overexpressed Op18 exerted a tubulin-protective effect. Tubulin protection was shown to depend on Op18 levels, binding affinity, and the partitioning between tubulin monomers and polymers. Hence, the present study reveals that Op18 at physiologically relevant levels functions to preserve the integrity of tubulin heterodimers, which may serve to regulate tubulin turnover rates.


Subject(s)
Leukemia/metabolism , Leukemia/pathology , Microtubule-Associated Proteins/metabolism , Molecular Chaperones/metabolism , Stathmin/metabolism , Dimerization , Gene Expression Regulation, Leukemic , Humans , K562 Cells , Kinesins/metabolism , Leukemia/genetics , Membrane Proteins/metabolism , Protein Binding , Protein Processing, Post-Translational , Stathmin/genetics , Tubulin/metabolism
11.
Environ Microbiol ; 10(5): 1320-34, 2008 May.
Article in English | MEDLINE | ID: mdl-18279347

ABSTRACT

Comparatively little is known about directed motility of environmental bacteria to common aromatic pollutants. Here, by expressing different parts of a (methyl)phenol-degradative pathway and the use of specific mutants, we show that taxis of Pseudomonas putida towards (methyl)phenols is dictated by its ability to catabolize the aromatic compound. Thus, in contrast to previously described chemoreceptor-mediated chemotaxis mechanisms towards benzoate, naphthalene and toluene, taxis in response to (methyl)phenols is mediated by metabolism-dependent behaviour. Here we show that P. putida differentially expresses three Aer-like receptors that are all polar-localized through interactions with CheA, and that inactivation of the most abundant Aer2 protein significantly decreases taxis towards phenolics. In addition, the participation of a sensory signal transduction protein composed of a PAS, a GGDEF and an EAL domain in motility towards these compounds is demonstrated. The results are discussed in the context of the versatility of metabolism-dependent coupling and the necessity for P. putida to integrate diverse metabolic signals from its native heterogeneous soil and water environments.


Subject(s)
Bacterial Proteins/metabolism , Cell Polarity , Chemotaxis , Gene Expression Regulation, Bacterial , Phenols , Pseudomonas/metabolism , Signal Transduction , Amino Acid Sequence , Bacterial Proteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Methyl-Accepting Chemotaxis Proteins , Molecular Sequence Data , Mutation , Phenols/chemistry , Phenols/metabolism , Plasmids/genetics , Pseudomonas/genetics , Pseudomonas/physiology
12.
Mol Biochem Parasitol ; 156(2): 225-34, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17913257

ABSTRACT

Sm16/SmSLP/SPO-1 (Sm16) has been identified as a developmentally regulated protein that is released from specific glands of the Schistosoma mansoni parasite during skin penetration. Sm16 has been ascribed both anti-inflammatory activities and a functional similarity with the conserved cytosolic tubulin-binding protein stathmin/Op18. Here we used a cell line to confirm signal peptide-dependent secretion and to define the secreted form of Sm16 for production in E. coli. We present evidence from both in vitro experiments and studies on transfected human cells that refute any functional similarity with stathmin/Op18. Instead of an Op18-like activity, we found that targeting of Sm16 to the cytosol of human cells, which was achieved by ectopic expression of Sm16 lacking the signal peptide, results in a caspase-dependent apoptotic response. Interestingly, by analysis of recombinant preparations we found that the secreted form of Sm16 is a lipid bilayer-binding protein that efficiently binds to the surface of diverse cell types by a polyanion-independent mechanism, which results in uptake by endocytosis. While the significance of the pro-apoptotic activity exerted by cytosolic Sm16 remains unclear, the present findings on cell-surface-binding properties of Sm16 seems likely to be of functional relevance during skin penetration of the parasite.


Subject(s)
Helminth Proteins/metabolism , Lipid Bilayers/metabolism , Microtubules/metabolism , Schistosoma mansoni/metabolism , Animals , Cell Line , Endocytosis , Escherichia coli/genetics , Humans , Protein Binding , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
13.
Mol Biol Cell ; 18(5): 1909-17, 2007 May.
Article in English | MEDLINE | ID: mdl-17344472

ABSTRACT

The microtubule cytoskeleton is differentially regulated by a diverse array of proteins during interphase and mitosis. Op18/stathmin (Op18) and microtubule-associated protein (MAP)4 have been ascribed opposite general microtubule-directed activities, namely, microtubule destabilization and stabilization, respectively, both of which can be inhibited by phosphorylation. Here, using three human cell models, we depleted cells of Op18 and/or MAP4 by expression of interfering hairpin RNAs and we analyzed the resulting phenotypes. We found that the endogenous levels of Op18 and MAP4 have opposite and counteractive activities that largely govern the partitioning of tubulin dimers in the microtubule array at interphase. Op18 and MAP4 were also found to be the downstream targets of Ca(2+)- and calmodulin-dependent protein kinase IV and PAR-1/MARK2 kinase, respectively, that control the demonstrated counteractive phosphorylation-mediated regulation of tubulin dimer partitioning. Furthermore, to address mechanisms regulating microtubule polymerization in response to cell signals, we developed a system for inducible gene product replacement. This approach revealed that site-specific phosphorylation of Op18 is both necessary and sufficient for polymerization of microtubules in response to the multifaceted signaling event of stimulation of the T cell antigen receptor complex, which activates several signal transduction pathways.


Subject(s)
Interphase/physiology , Tubulin/chemistry , Tubulin/metabolism , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinase Type 4 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , DNA/genetics , Dimerization , Humans , Jurkat Cells , K562 Cells , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Quaternary , Signal Transduction , Stathmin/antagonists & inhibitors , Stathmin/genetics , Stathmin/metabolism , Transfection
14.
Mol Biol Cell ; 17(7): 2921-30, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16624860

ABSTRACT

Op18/stathmin (Op18) is a phosphorylation-regulated microtubule destabilizer that is frequently overexpressed in tumors. The importance of Op18 in malignancy was recently suggested by identification of a somatic Q18-->E mutation of Op18 in an adenocarcinoma. We addressed the functional consequences of aberrant Op18 expression in leukemias by analyzing the cell cycle of K562 cells either depleted of Op18 by expression of interfering hairpin RNA or induced to express wild-type or Q18E substituted Op18. We show here that although Op18 depletion increases microtubule density during interphase, the density of mitotic spindles is essentially unaltered and cells divide normally. This is consistent with phosphorylation-inactivation of Op18 during mitosis. Overexpression of wild-type Op18 results in aneugenic activities, manifest as aberrant mitosis, polyploidization, and chromosome loss. One particularly significant finding was that the aneugenic activity of Op18 was dramatically increased by the Q18-->E mutation. The hyperactivity of mutant Op18 is apparent in its unphosphorylated state, and this mutation also suppresses phosphorylation-inactivation of the microtubule-destabilizing activity of Op18 without any apparent effect on its phosphorylation status. Thus, although Op18 is dispensable for mitosis, the hyperactive Q18-->E mutant, or overexpressed wild-type Op18, exerts aneugenic effects that are likely to contribute to chromosomal instability in tumors.


Subject(s)
Chromosomal Instability , Leukemia/genetics , Spindle Apparatus/metabolism , Stathmin/genetics , Stathmin/metabolism , Aneugens/metabolism , Cell Cycle/genetics , Humans , Leukemia/metabolism , Leukemia/pathology , Microtubules/metabolism , Mitosis/genetics , Mutation , Phosphorylation , RNA Interference , Stathmin/antagonists & inhibitors , Tubulin/metabolism
15.
EMBO J ; 24(6): 1256-66, 2005 Mar 23.
Article in English | MEDLINE | ID: mdl-15775983

ABSTRACT

MCAK, a member of the kinesin-13 family, is a microtubule (MT) depolymerase that is necessary to ensure proper kinetochore MT attachment during spindle formation. Regulation of MCAK activity and localization is controlled in part by Aurora B kinase at the centromere. Here we analyzed human cells depleted of the ubiquitous Ca(2+)/calmodulin-dependent protein kinase IIgamma isoform (CaMKIIgamma) by RNA interference and found that CaMKIIgamma was necessary to suppress MCAK depolymerase activity in vivo. A functional overlap with TOGp, a MT regulator known to counteract MCAK, was suggested by similar CaMKIIgamma- and TOGp-depletion phenotypes, namely disorganized multipolar spindles. A replicating vector system, which permits inducible overexpression in cells that simultaneously synthesize interfering short hairpin RNAs, was used to dissect the functional interplay between CaMKIIgamma, TOGp, and MCAK. Our results revealed two distinct but functionally overlapping mechanisms for negative regulation of the cytosolic/centrosomal pool of MCAK. These two mechanisms, involving CaMKIIgamma and TOGp, respectively, are both essential for spindle bipolarity in a normal physiological context, but not in MCAK-depleted cells.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Kinesins/metabolism , Spindle Apparatus/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Division/genetics , Cell Division/physiology , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/physiology , Nucleic Acid Conformation , RNA Interference , RNA, Small Interfering/genetics
16.
EMBO J ; 23(3): 627-37, 2004 Feb 11.
Article in English | MEDLINE | ID: mdl-14749730

ABSTRACT

XMAP215/TOGp family members and KinI kinesins are conserved microtubule (MT)-regulatory proteins, and have been viewed as possessing prominent antagonistic stabilizing/destabilizing activities that must be balanced. Here, interdependencies between TOGp and the KinI kinesin MCAK were analyzed in human leukemia cells. A system was established that permits inducible overexpression in homogeneous cell populations that simultaneously synthesize interfering short hairpin RNAs. We present evidence that the functional interplay of TOGp and MCAK proteins is manifested as three distinct phenotypes during the cell cycle. The first involves a role for TOGp in protecting spindle MTs from MCAK activity at the centrosome, which appears essential to prevent the formation of disorganized multipolar spindles. The second phenotype involves TOGp-dependent counteraction of excessive MCAK activity during mitosis, which recapitulates the previously established plus-end specific counteractive activities in vitro. The third involves an unexpected destabilization of the interphase MTs by overexpressed TOGp, a phenotype that requires endogenous MCAK. We hypothesize that TOGp-dependent prevention of MCAK-mediated spindle disorganization, as evidenced by depletion experiments, reflects a primary physiological role for TOGp in human somatic cells.


Subject(s)
Interphase/physiology , Kinesins/metabolism , Microtubule-Associated Proteins/metabolism , Mitosis/physiology , Spindle Apparatus/metabolism , Humans , K562 Cells
17.
Mol Biol Cell ; 14(9): 3716-29, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12972559

ABSTRACT

The Op18/stathmin family of microtubule regulators includes the ubiquitous cytosolic Op18/stathmin (Op18) and the neuronal, primarily Golgi-associated proteins SCG10 and RB3, which all form ternary complexes with two head-to-tail-aligned tubulin heterodimers. To understand the physiological significance of previously observed differences in ternary complex stability, we have fused each of the heterodimer-binding regions of these three proteins with the CD2 cell surface protein to generate confined plasma membrane localization of the resulting CD2 chimeras. Herein, we show that, in contrast to constitutively active CD2-Op18-tetraA, both the CD2-SCG10 and CD2-RB3 chimeras sequestered tubulin at the plasma membrane, which results in >35% reduction of cytosolic tubulin heterodimer levels and consequent delayed formation of mitotic spindles. However, all three CD2 chimeras, including the tubulin sequestration-incompetent CD2-Op18-tetraA, destabilize interphase microtubules. Given that microtubules are in extensive contact with the plasma membrane during interphase, but not during mitosis, these findings indicate that Op18-like proteins have the potential to destabilize microtubules by both sequestration and direct interaction with microtubules. However, the differences in tubulin binding observed in cells also indicate conceptual differences between the functions of low-abundance neural family members, which will accumulate tubulin at specific cellular compartments, and the abundant cytosolic Op18 protein, which will not.


Subject(s)
Microtubule Proteins , Microtubules/metabolism , Nerve Growth Factors/metabolism , Phosphoproteins/metabolism , Tubulin/metabolism , CD2 Antigens , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Membrane/physiology , Dimerization , Fluorescent Antibody Technique , Humans , Interphase/physiology , K562 Cells , Membrane Proteins , Microtubules/physiology , Mitosis/physiology , Nerve Growth Factors/genetics , Nerve Growth Factors/physiology , Phosphoproteins/genetics , Phosphoproteins/physiology , Protein Binding , Protein Structure, Secondary/genetics , Protein Structure, Secondary/physiology , Recombinant Proteins , Spindle Apparatus/metabolism , Spindle Apparatus/physiology , Stathmin , Surface Plasmon Resonance , Tubulin/physiology
18.
J Cell Sci ; 116(Pt 18): 3701-11, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12890753

ABSTRACT

The microtubule-associated protein 4 (MAP4) has recently been shown to counteract destabilization of interphase microtubules caused by catastrophe promotion but not by tubulin sequestering. To address how MAP4 discriminates between destabilization of microtubules by these two mechanisms, we have evaluated the combined phenotypes of MAP4 coexpressed with Op18/stathmin family member derivatives with either catastrophe-promoting or sequestering activities. This approach relies on the finding that overexpression of MAP4 alone stabilizes microtubules during all phases of the cell cycle in human leukemia cells, and causes a potent mitotic block and a dramatic, previously unobserved, phenotype characterized by large monoastral spindles. Coexpression of either catastrophe-promoting or tubulin-sequestration-specific Op18 derivatives was found to modulate the activity of ectopic MAP4 during mitosis, but with differential functional outcome. Interestingly, the tubulin-sequestering derivative suppressed the monoastral mitotic phenotype of MAP4 (i.e. coexpression facilitated the formation of functional spindles). To evaluate whether this phenotypic suppression could be explained by tubulin-sequestration-dependent modulation of MAP4 activity, a plasma-membrane-targeted, tubulin-sequestering chimera was constructed to decrease the cytosolic free tubulin concentration substantially. This chimera likewise suppressed the monoastral phenotype caused by overexpression of MAP4, suggesting a direct downregulation of MAP4 activity by reduced free tubulin concentrations.


Subject(s)
Interphase/physiology , Microtubule Proteins , Microtubule-Associated Proteins/biosynthesis , Microtubules/metabolism , Spindle Apparatus/metabolism , Tubulin/metabolism , Flow Cytometry , Humans , Jurkat Cells , K562 Cells , Microscopy, Fluorescence , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Recombinant Fusion Proteins/metabolism , Stathmin , Tumor Cells, Cultured
19.
J Cell Sci ; 116(Pt 1): 197-205, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12456729

ABSTRACT

Op18 is the prototypical member of a family of phosphorylation-responsive regulators of microtubule (MT) dynamics. Previous dissection of Op18 has suggested that it has a functional dichotomy in which an intact N-terminus is required for catastrophe promotion (i.e. transition from growing to shrinking MTs), whereas an intact C-terminus is required for efficient ternary Op18-tubulin complex formation and the resultant tubulin-sequestering activity. Here we have expressed and functionally analyzed the properties of the N-terminus of Op18. The data show that the N-terminal 57 residues are sufficient for low-affinity tubulin interactions, as shown by inhibition of basal GTP hydrolysis of soluble heterodimers. In addition, high concentrations of the Op18 N-terminal portion increased the catastrophe rate during MT assembly in vitro. Overexpression of the N-terminus in a human cell line results in MT destabilization in interphase and phosphorylation-modulated accumulation of metaphase-arrested cells with dense short MTs. These results demonstrate that the N-terminus of Op18 has autonomous activity. Evidently, this activity is enhanced by the increase in tubulin affinity that is provided by the extended alpha-helical portion of native Op18.


Subject(s)
Eukaryotic Cells/metabolism , Microtubule Proteins , Microtubules/metabolism , Phosphoproteins/metabolism , Amino Acid Sequence/physiology , Cell Cycle/physiology , Dimerization , Eukaryotic Cells/cytology , Gene Expression/physiology , Guanosine Triphosphate/metabolism , Humans , Hydrolysis , Interphase/physiology , Metaphase/physiology , Mitosis/physiology , Phosphorylation , Protein Structure, Secondary/physiology , Protein Structure, Tertiary/physiology , Stathmin , Tumor Cells, Cultured
20.
FEBS Lett ; 533(1-3): 35-41, 2003 Jan 02.
Article in English | MEDLINE | ID: mdl-12505155

ABSTRACT

Internalisation of the human pathogen Yersinia pseudotuberculosis via interaction of bacterial invasin with host beta1 integrins depends on the actin cytoskeleton and involves Src family kinases, focal adhesion kinase, p130Crk-associated substrate, proline-rich tyrosine kinase 2, Rac, Arp 2/3 complex and WASP family members. We show here that Rho GTPases are regulated by the microtubule system during bacterial uptake. Interfering with microtubule organisation using nocodazole or paclitaxel suppressed uptake by HeLa cells. The nocodazole effect on microtubule depolymerisation was partially inhibited through overexpression of Rac, Cdc42, RhoG or RhoA and completely prevented by expression of Vav2. This suggests that microtubules influence Rho GTPases during invasin-mediated phagocytosis and in the absence of functional microtubules Vav2 can mimic their effect on one, or more, of the Rho family GTPases. Lastly, overexpression of p50 dynamitin partially inhibited bacterial uptake and this effect was also blocked by co-expression of Vav2, thus further implicating this guanine nucleotide exchange factor in activating Rho GTPases for internalisation during loss of microtubule function.


Subject(s)
Yersinia pseudotuberculosis/pathogenicity , rho GTP-Binding Proteins/metabolism , Biological Transport, Active/drug effects , Dynactin Complex , Dyneins/metabolism , GTP Phosphohydrolases/metabolism , HeLa Cells , Humans , Microtubule-Associated Proteins/metabolism , Microtubules/drug effects , Microtubules/metabolism , Nocodazole/pharmacology , Oncogene Proteins/metabolism , Paclitaxel/pharmacology , Proto-Oncogene Proteins c-vav , Recombinant Fusion Proteins/metabolism , cdc42 GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...