Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Hum Mol Genet ; 28(15): 2573-2588, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31009951

RESUMO

Mutations in subunits of the cilia-specific cytoplasmic dynein-2 (CD2) complex cause short-rib thoracic dystrophy syndromes (SRTDs), characterized by impaired bone growth and life-threatening perinatal respiratory complications. Different SRTD mutations result in varying disease severities. It remains unresolved whether this reflects the extent of retained hypomorphic protein functions or relative importance of the affected subunits for the activity of the CD2 holoenzyme. To define the contribution of the LC8-type dynein light chain subunit to the CD2 complex, we have generated Dynll1-deficient mouse strains, including the first-ever conditional knockout (KO) mutant for any CD2 subunit. Germline Dynll1 KO mice exhibit a severe ciliopathy-like phenotype similar to mice lacking another CD2 subunit, Dync2li1. Limb mesoderm-specific loss of Dynll1 results in severe bone shortening similar to human SRTD patients. Mechanistically, loss of Dynll1 leads to a partial depletion of other SRTD-related CD2 subunits, severely impaired retrograde intra-flagellar transport, significant thickening of primary cilia and cilia signaling defects. Interestingly, phenotypes of Dynll1-deficient mice are very similar to entirely cilia-deficient Kif3a/Ift88-null mice, except that they never present with polydactyly and retain relatively higher signaling outputs in parts of the hedgehog pathway. Compared to complete loss of Dynll1, maintaining very low DYNLL1 levels in mice lacking the Dynll1-transcription factor ASCIZ (ATMIN) results in significantly attenuated phenotypes and improved CD2 protein levels. The results suggest that primary cilia can maintain some functionality in the absence of intact CD2 complexes and provide a viable animal model for the analysis of the underlying bone development defects of SRTDs.


Assuntos
Doenças do Desenvolvimento Ósseo/metabolismo , Cílios/metabolismo , Ciliopatias/metabolismo , Dineínas do Citoplasma/genética , Osteogênese , Animais , Doenças do Desenvolvimento Ósseo/genética , Doenças do Desenvolvimento Ósseo/fisiopatologia , Células Cultivadas , Cílios/fisiologia , Ciliopatias/genética , Ciliopatias/fisiopatologia , Dineínas do Citoplasma/metabolismo , Dineínas do Citoplasma/fisiologia , Extremidades/patologia , Extremidades/fisiopatologia , Proteínas Hedgehog/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Transdução de Sinais , Fatores de Transcrição/metabolismo
2.
Nucleic Acids Res ; 47(12): 6236-6249, 2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-30982887

RESUMO

The tumor suppressor protein 53BP1 plays key roles in response to DNA double-strand breaks (DSBs) by serving as a master scaffold at the damaged chromatin. Current evidence indicates that 53BP1 assembles a cohort of DNA damage response (DDR) factors to distinctly execute its repertoire of DSB responses, including checkpoint activation and non-homologous end joining (NHEJ) repair. Here, we have uncovered LC8 (a.k.a. DYNLL1) as an important 53BP1 effector. We found that LC8 accumulates at laser-induced DNA damage tracks in a 53BP1-dependent manner and requires the canonical H2AX-MDC1-RNF8-RNF168 signal transduction cascade. Accordingly, genetic inactivation of LC8 or its interaction with 53BP1 resulted in checkpoint defects. Importantly, loss of LC8 alleviated the hypersensitivity of BRCA1-depleted cells to ionizing radiation and PARP inhibition, highlighting the 53BP1-LC8 module in counteracting BRCA1-dependent functions in the DDR. Together, these data establish LC8 as an important mediator of a subset of 53BP1-dependent DSB responses.


Assuntos
Dineínas do Citoplasma/fisiologia , Quebras de DNA de Cadeia Dupla , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo , Proteína BRCA1/genética , Linhagem Celular , Cromatina/metabolismo , Dineínas do Citoplasma/química , Dineínas do Citoplasma/metabolismo , Reparo do DNA , Humanos , Inibidores de Poli(ADP-Ribose) Polimerases , Radiação Ionizante
3.
Mol Biol Cell ; 29(2): 180-190, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29142075

RESUMO

Teleosts and amphibians exhibit retinomotor movements, morphological changes in photoreceptors regulated by light and circadian rhythms. Cone myoid elongation occurs during dark adaptation, leading to the positioning of the cone outer segment closer to the retinal pigment epithelium. Although it has been shown that microtubules are essential for cone myoid elongation, the underlying mechanism has not been established. In this work, we generated a transgenic line of zebrafish expressing a photoconvertible form of α-tubulin (tdEOS-tubulin) specifically in cone photoreceptors. Using superresolution structured illumination microscopy in conjunction with both pharmacological and genetic manipulation, we show that cytoplasmic dynein-1, which localizes to the junction between the ellipsoid and myoid, functions to shuttle microtubules from the ellipsoid into the myoid during the course of myoid elongation. We propose a novel model by which stationary complexes of cytoplasmic dynein-1 are responsible for the shuttling of microtubules between the ellipsoid and myoid is the underlying force for the morphological change of myoid elongation.


Assuntos
Dineínas do Citoplasma/fisiologia , Microtúbulos/fisiologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Peixe-Zebra/anatomia & histologia , Animais , Animais Geneticamente Modificados , Adaptação à Escuridão , Microtúbulos/ultraestrutura , Nocodazol/farmacologia , Epitélio Pigmentado Ocular , Células Fotorreceptoras Retinianas Cones/ultraestrutura , Moduladores de Tubulina/farmacologia
4.
Mol Biol Cell ; 28(19): 2543-2554, 2017 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-28720664

RESUMO

The efficient transport of cargoes within axons and dendrites is critical for neuronal function. Although we have a basic understanding of axonal transport, much less is known about transport in dendrites. We used an optogenetic approach to recruit motor proteins to cargo in real time within axons or dendrites in hippocampal neurons. Kinesin-1, a robust axonal motor, moves cargo less efficiently in dendrites. In contrast, cytoplasmic dynein efficiently navigates both axons and dendrites; in both compartments, dynamic microtubule plus ends enhance dynein-dependent transport. To test the predictions of the optogenetic assay, we examined the contribution of dynein to the motility of an endogenous dendritic cargo and found that dynein inhibition eliminates the retrograde bias of BDNF/TrkB trafficking. However, inhibition of microtubule dynamics has no effect on BDNF/TrkB motility, suggesting that dendritic kinesin motors may cooperate with dynein to drive the transport of signaling endosomes into the soma. Collectively our data highlight compartment-specific differences in kinesin activity that likely reflect specialized tuning for localized cytoskeletal determinants, whereas dynein activity is less compartment specific but is more responsive to changes in microtubule dynamics.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Dineínas do Citoplasma/fisiologia , Dendritos/fisiologia , Endossomos/fisiologia , Receptor trkB/metabolismo , Animais , Transporte Axonal , Axônios/metabolismo , Células Cultivadas , Dineínas do Citoplasma/metabolismo , Citoesqueleto/metabolismo , Citoesqueleto/fisiologia , Dendritos/metabolismo , Endossomos/metabolismo , Hipocampo/metabolismo , Cinesinas/metabolismo , Microtúbulos/metabolismo , Miosinas/metabolismo , Neurônios/metabolismo , Transporte Proteico , Ratos , Transdução de Sinais
5.
J Virol ; 91(10)2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28250122

RESUMO

How murine leukemia virus (MLV) travels from the cell membrane to the nucleus and the mechanism for nuclear entry of MLV DNA in dividing cells still remain unclear. It seems likely that the MLV preintegration complex (PIC) interacts with cellular proteins to perform these tasks. We recently published that the microtubule motor cytoplasmic dynein complex and its regulator proteins interact with the MLV PIC at early times of infection, suggesting a functional interaction between the incoming viral particles, the dynein complex, and dynein regulators. To better understand the role of the dynein complex in MLV infection, we performed short hairpin RNA (shRNA) screening of the dynein light chains on MLV infection. We found that silencing of a specific light chain of the cytoplasmic dynein complex, DYNLRB2, reduced the efficiency of infection by MLV reporter viruses without affecting HIV-1 infection. Furthermore, the overexpression of DYNLRB2 increased infection by MLV. We conclude that the DYNLRB2 light chain of the cytoplasmic dynein complex is an important and specific piece of the host machinery needed for MLV infection.IMPORTANCE Retroviruses must reach the chromatin of their host to integrate their viral DNA, but first they must get into the nucleus. The cytoplasm is a crowded environment in which simple diffusion is slow, and thus viruses utilize retrograde transport along the microtubule network, mediated by the dynein complex. Different viruses use different components of this multisubunit complex. We have found that murine leukemia virus (MLV) associates functionally and specifically with the dynein light chain DYNLRB2, which is required for infection. Our study provides more insight into the molecular requirements for retrograde transport of the MLV preintegration complex and demonstrates, for the first time, a role for DYNLRB2 in viral infection.


Assuntos
Dineínas do Citoplasma/genética , Dineínas do Citoplasma/fisiologia , Interações Hospedeiro-Patógeno , Vírus da Leucemia Murina/fisiologia , Animais , Transporte Biológico , Linhagem Celular , Núcleo Celular/virologia , Células HEK293 , HIV-1/fisiologia , Interações Hospedeiro-Patógeno/genética , Humanos , Camundongos , Microtúbulos/virologia , Células NIH 3T3
6.
Bioessays ; 38(6): 514-25, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27143631

RESUMO

Cytoplasmic dynein is the major minus-end-directed motor protein in eukaryotes, and has functions ranging from organelle and vesicle transport to spindle positioning and orientation. The mode of regulation of dynein in the cell remains elusive, but a tantalising possibility is that dynein is maintained in an inhibited, non-motile state until bound to cargo. In vivo, stable attachment of dynein to the cell membrane via anchor proteins enables dynein to produce force by pulling on microtubules and serves to organise the nuclear material. Anchor proteins of dynein assume diverse structures and functions and differ in their interaction with the membrane. In yeast, the anchor protein has come to the fore as one of the key mediators of dynein activity. In other systems, much is yet to be discovered about the anchors, but future work in this area will prove invaluable in understanding dynein regulation in the cell.


Assuntos
Dineínas do Citoplasma/metabolismo , Eucariotos/metabolismo , Animais , Transporte Biológico , Membrana Celular/metabolismo , Dineínas do Citoplasma/fisiologia , Humanos , Microtúbulos/metabolismo , Fuso Acromático/metabolismo
7.
Mol Biol Cell ; 25(5): 669-78, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24403603

RESUMO

The timely delivery of membranous organelles and macromolecules to specific locations within the majority of eukaryotic cells depends on microtubule-based transport. Here we describe a screening method to identify mutations that have a critical effect on intracellular transport and its regulation using mutagenesis, multicolor-fluorescence microscopy, and multiplex genome sequencing. This screen exploits the filamentous fungus Aspergillus nidulans, which has many of the advantages of yeast molecular genetics but uses long-range microtubule-based transport in a manner more similar to metazoan cells. Using this method, we identified seven mutants that represent novel alleles of components of the intracellular transport machinery: specifically, kinesin-1, cytoplasmic dynein, and the dynein regulators Lis1 and dynactin. The two dynein mutations identified in our screen map to dynein's AAA+ catalytic core. Single-molecule studies reveal that both mutations reduce dynein's velocity in vitro. In vivo these mutants severely impair the distribution and velocity of endosomes, a known dynein cargo. In contrast, another dynein cargo, the nucleus, is positioned normally in these mutants. These results reveal that different dynein functions have distinct stringencies for motor performance.


Assuntos
Aspergillus nidulans/metabolismo , Transporte Biológico , Dineínas/metabolismo , Organelas/metabolismo , Aspergillus nidulans/genética , Aspergillus nidulans/ultraestrutura , Dineínas do Citoplasma/genética , Dineínas do Citoplasma/fisiologia , Complexo Dinactina , Dineínas/genética , Proteínas Fúngicas/genética , Proteínas Fúngicas/fisiologia , Hidroliases/genética , Cinesinas/genética , Cinesinas/fisiologia , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/fisiologia , Microtúbulos/metabolismo , Organelas/ultraestrutura , Peroxissomos , Temperatura
8.
J Cell Biol ; 200(4): 385-95, 2013 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-23401002

RESUMO

During meiosis, telomeres cluster and promote homologous chromosome pairing. Telomere clustering requires the interaction of telomeres with the nuclear membrane proteins SUN (Sad1/UNC-84) and KASH (Klarsicht/ANC-1/Syne homology). The mechanism by which telomeres gather remains elusive. In this paper, we show that telomere clustering in fission yeast depends on microtubules and the microtubule motors, cytoplasmic dynein, and kinesins. Furthermore, the γ-tubulin complex (γ-TuC) is recruited to SUN- and KASH-localized telomeres to form a novel microtubule-organizing center that we termed the "telocentrosome." Telocentrosome formation depends on the γ-TuC regulator Mto1 and on the KASH protein Kms1, and depletion of either Mto1 or Kms1 caused severe telomere clustering defects. In addition, the dynein light chain (DLC) contributes to telocentrosome formation, and simultaneous depletion of DLC and dynein also caused severe clustering defects. Thus, the telocentrosome is essential for telomere clustering. We propose that telomere-localized SUN and KASH induce telocentrosome formation and that subsequent microtubule motor-dependent aggregation of telocentrosomes via the telocentrosome-nucleated microtubules causes telomere clustering.


Assuntos
Dineínas do Citoplasma/fisiologia , Meiose/genética , Centro Organizador dos Microtúbulos/metabolismo , Schizosaccharomyces/metabolismo , Telômero/metabolismo , Dineínas do Citoplasma/genética , Deleção de Genes , Centro Organizador dos Microtúbulos/ultraestrutura , Microtúbulos/metabolismo , Microtúbulos/fisiologia , Microtúbulos/ultraestrutura , Modelos Biológicos , Schizosaccharomyces/genética , Schizosaccharomyces/ultraestrutura , Proteínas de Schizosaccharomyces pombe/metabolismo , Proteínas de Schizosaccharomyces pombe/fisiologia , Telômero/ultraestrutura , Proteínas de Ligação a Telômeros/metabolismo , Proteínas de Ligação a Telômeros/fisiologia
9.
J Immunol ; 190(3): 1312-8, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23293355

RESUMO

NF-κB is one of the key transcription factors activated by receptor activator of NF-κB ligand (RANKL) during osteoclast differentiation. The 8-kDa dynein L chain (LC8) was previously identified as a novel NF-κB regulator. However, its physiological role as an NF-κB inhibitor remains elusive. In this study, we showed the inhibitory role of LC8 in RANKL-induced osteoclastogenesis and signaling pathways and its protective role in osteolytic animal models. LC8 suppressed RANKL-induced osteoclast differentiation, actin ring formation, and osteoclastic bone resorption. LC8 inhibited RANKL-induced phosphorylation and subsequent degradation of IκBα, the expression of c-Fos, and the consequent activation of NFATc1, which is a pivotal determinant of osteoclastogenesis. LC8 also inhibited RANKL-induced activation of JNK and ERK. LC8-transgenic mice exhibited a mild osteopetrotic phenotype. Moreover, LC8 inhibited inflammation-induced bone erosion and protected against ovariectomy-induced bone loss in mice. Thus, our results suggest that LC8 inhibits osteoclast differentiation by regulating NF-κB and MAPK pathways and provide the molecular basis of a new strategy for treating osteoporosis and other bone diseases.


Assuntos
Reabsorção Óssea/prevenção & controle , Dineínas do Citoplasma/fisiologia , Osteoclastos/patologia , Osteólise/prevenção & controle , Ligante RANK/antagonistas & inibidores , Transdução de Sinais/fisiologia , Actinas/análise , Animais , Diferenciação Celular , Dineínas do Citoplasma/genética , Dineínas do Citoplasma/toxicidade , Avaliação Pré-Clínica de Medicamentos , Ativação Enzimática , Regulação da Expressão Gênica/fisiologia , Genes fos , Humanos , Proteínas I-kappa B/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/biossíntese , Fatores de Transcrição NFATC/genética , Osteólise/fisiopatologia , Osteopetrose/genética , Osteoporose Pós-Menopausa/fisiopatologia , Osteoporose Pós-Menopausa/prevenção & controle , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-fos/biossíntese , Proteínas Recombinantes de Fusão/fisiologia , Proteínas Recombinantes de Fusão/toxicidade
10.
Biochem Biophys Res Commun ; 428(3): 333-8, 2012 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-23079622

RESUMO

km23-1 was originally identified as a TGFß receptor-interacting protein that plays an important role in TGFß signaling. Moreover, km23-1 is actually part of an ancient superfamily of NTPase-regulatory proteins, widely represented in archaea and bacteria. To further elucidate the function of km23-1, we identified novel protein interacting partners for km23-1 by using tandem affinity purification (TAP) and tandem mass spectrometry (MS). Here we show that km23-1 interacted with a class of proteins involved in actin-based cell motility and modulation of the actin cytoskeleton. We further showed that km23-1 modulates the formation of a highly organized stress fiber network. More significantly, we demonstrated that knockdown (KD) of km23-1 decreased RhoA activation in Mv1Lu epithelial cells. Finally, our results demonstrated for the first time that depletion of km23-1 inhibited cell migration of human colon carcinoma cells (HCCCs) in wound-healing assays. Overall, our findings demonstrate that km23-1 regulates RhoA and motility-associated actin modulating proteins, suggesting that km23-1 may represent a novel target for anti-metastatic therapy.


Assuntos
Actinas/biossíntese , Movimento Celular , Dineínas do Citoplasma/fisiologia , Metástase Neoplásica/patologia , Proteína rhoA de Ligação ao GTP/biossíntese , Dineínas do Citoplasma/genética , Células HCT116 , Células HEK293 , Humanos , Cicatrização
11.
Assay Drug Dev Technol ; 10(5): 432-56, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22830992

RESUMO

Rapid ligand-induced trafficking of glucocorticoid nuclear hormone receptor (GR) from the cytoplasm to the nucleus is an extensively studied model for intracellular retrograde cargo transport employed in constructive morphogenesis and many other cellular functions. Unfortunately, potent and selective small-molecule disruptors of this process are lacking, which has restricted pharmacological investigations. We describe here the development and validation of a 384-well high-content screening (HCS) assay to identify inhibitors of the rapid ligand-induced retrograde translocation of cytoplasmic glucocorticoid nuclear hormone receptor green fluorescent fusion protein (GR-GFP) into the nuclei of 3617.4 mouse mammary adenocarcinoma cells. We selected 3617.4 cells, because they express GR-GFP under the control of a tetracycline (Tet)-repressible promoter and are exceptionally amenable to image acquisition and analysis procedures. Initially, we investigated the time-dependent expression of GR-GFP in 3617.4 cells under Tet-on and Tet-off control to determine the optimal conditions to measure dexamethasone (Dex)-induced GR-GFP nuclear translocation on the ArrayScan-VTI automated imaging platform. We then miniaturized the assay into a 384-well format and validated the performance of the GR-GFP nuclear translocation HCS assay in our 3-day assay signal window and dimethylsulfoxide validation tests. The molecular chaperone heat shock protein 90 (Hsp90) plays an essential role in the regulation of GR steroid binding affinity and ligand-induced retrograde trafficking to the nucleus. We verified that the GR-GFP HCS assay captured the concentration-dependent inhibition of GR-GFP nuclear translocation by 17-AAG, a benzoquinone ansamycin that selectively blocks the binding and hydrolysis of ATP by Hsp90. We screened the 1280 compound library of pharmacologically active compounds set in the Dex-induced GR-GFP nuclear translocation assay and used the multi-parameter HCS data to eliminate cytotoxic compounds and fluorescent outliers. We identified five qualified hits that inhibited the rapid retrograde trafficking of GR-GFP in a concentration-dependent manner: Bay 11-7085, 4-phenyl-3-furoxancarbonitrile, parthenolide, apomorphine, and 6-nitroso-1,2-benzopyrone. The data presented here demonstrate that the GR-GFP HCS assay provides an effective phenotypic screen and support the proposition that screening a larger library of diversity compounds will yield novel small-molecule probes that will enable the further exploration of intracellular retrograde transport of cargo along microtubules, a process which is essential to the morphogenesis and function of all cells.


Assuntos
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Dineínas do Citoplasma/antagonistas & inibidores , Descoberta de Drogas/métodos , Ensaios de Triagem em Larga Escala/métodos , Receptores de Glucocorticoides/metabolismo , Animais , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Citoplasma/efeitos dos fármacos , Dineínas do Citoplasma/fisiologia , Dexametasona/química , Dexametasona/metabolismo , Dexametasona/farmacologia , Camundongos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia
12.
J Biol Chem ; 287(31): 26453-63, 2012 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-22637579

RESUMO

We have previously elucidated the signaling events that are required for TGFß1 autoinduction (Yue, J., and Mulder, K. M. (2000) J. Biol. Chem. 275, 30765-30773). Further, we have reported that the TGFß receptor (TßR)-interacting protein km23-1 plays an important role in TGFß signal transduction (Jin, Q., Ding, W., and Mulder, K. M. (2007) J. Biol. Chem. 282, 19122-19132). Here we examined the role of km23-1 in TGFß1 autoinduction in TGFß-sensitive epithelial cells. siRNA blockade of km23-1 reduced TGFß1 mRNA expression, as well as DNA binding and transcriptional activation of the relevant activator protein-1 site in the human TGFß1 promoter. Further, knockdown of km23-1 inhibited TGFß-mediated activation of ERK and JNK, phosphorylation of c-Jun, and transactivation of the c-Jun promoter. Sucrose gradient analyses indicate that km23-1 was present in lipid rafts together with Ras and TßRII after TGFß treatment. Immunoprecipitation/blot analyses revealed the formation of a TGFß-inducible complex between Ras and km23-1 in vivo within minutes of TGFß addition. Moreover, we demonstrate for the first time that km23-1 is required for Ras activation by TGFß. Our results indicate that km23-1 is required for TGFß1 autoinduction through Smad2-independent Ras/ERK/JNK pathways. More importantly, our findings demonstrate that km23-1 functions as a critical adaptor coupling TßR activation to activation of Ras effector pathways downstream.


Assuntos
Dineínas do Citoplasma/fisiologia , Ativação Transcricional , Fator de Crescimento Transformador beta1/metabolismo , Proteínas ras/metabolismo , Sítios de Ligação , Linhagem Celular , Dineínas do Citoplasma/genética , Dineínas do Citoplasma/metabolismo , Ativação Enzimática , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Sistema de Sinalização das MAP Quinases , Regiões Promotoras Genéticas , Ligação Proteica , Interferência de RNA , Proteína Smad2/metabolismo , Fator de Transcrição AP-1/metabolismo , Fator de Crescimento Transformador beta1/genética
13.
Assay Drug Dev Technol ; 10(1): 46-60, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21919741

RESUMO

Agonist-induced glucocorticoid receptor [GR] transport from the cytoplasm to the nucleus was used as a model to identify dynein-mediated cargo transport inhibitors. Cell-based screening of the library of pharmacologically active compound (LOPAC)-1280 collection identified several small molecules that stalled the agonist-induced transport of GR-green fluorescent protein (GFP) in a concentration-dependent manner. Fluorescent images of microtubule organization, nuclear DNA staining, expression of GR-GFP, and its subcellular distribution were inspected and quantified by image analysis to evaluate the impact of compounds on cell morphology, toxicity, and GR transport. Given the complexity of the multi-protein complex involved in dynein-mediated cargo transport and the variety of potential mechanisms for interruption of that process, we therefore developed and validated a panel of biochemical assays to investigate some of the more likely intracellular target(s) of the GR transport inhibitors. Although the apomorphine enantiomers exhibited the most potency toward the ATPase activities of cytoplasmic dynein, myosin, and the heat-shock proteins (HSPs), their apparent lack of specificity made them unattractive for further study in our quest. Other molecules appeared to be nonspecific inhibitors that targeted reactive cysteines of proteins. Ideally, specific retrograde transport inhibitors would either target dynein itself or one of the other important proteins associated with the transport process. Although the hits from the cell-based screen of the LOPAC-1280 collection did not exhibit this desired profile, this screening platform provided a promising phenotypic system for the discovery of dynein/HSP modulators.


Assuntos
Núcleo Celular/metabolismo , Dineínas do Citoplasma/fisiologia , Avaliação Pré-Clínica de Medicamentos/métodos , Receptores de Glucocorticoides/antagonistas & inibidores , Receptores de Glucocorticoides/metabolismo , Animais , Apomorfina/metabolismo , Apomorfina/farmacologia , Bovinos , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Transporte Proteico/fisiologia
14.
J Cell Sci ; 124(Pt 24): 4267-85, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22193958

RESUMO

In several migratory cells, the microtubule-organizing center (MTOC) is repositioned between the leading edge and nucleus, creating a polarized morphology. Although our understanding of polarization has progressed as a result of various scratch-wound and cell migration studies, variations in culture conditions required for such assays have prevented a unified understanding of the intricacies of MTOC and nucleus positioning that result in cell polarization. Here, we employ a new SMRT (for sparse, monolayer, round, triangular) analysis that uses a universal coordinate system based on cell centroid to examine the pathways regulating MTOC and nuclear positions in cells plated in a variety of conditions. We find that MTOC and nucleus positioning are crucially and independently affected by cell shape and confluence; MTOC off-centering correlates with the polarization of single cells; acto-myosin contractility and microtubule dynamics are required for single-cell polarization; and end binding protein 1 and light intermediate chain 1, but not Par3 and light intermediate chain 2, are required for single-cell polarization and directional cell motility. Using various cellular geometries and conditions, we implement a systematic and reproducible approach to identify regulators of MTOC and nucleus positioning that depend on extracellular guidance cues.


Assuntos
Núcleo Celular/fisiologia , Polaridade Celular , Dineínas do Citoplasma/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Centro Organizador dos Microtúbulos/fisiologia , Actinas/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Moléculas de Adesão Celular/fisiologia , Proteínas de Ciclo Celular , Movimento Celular , Núcleo Celular/ultraestrutura , Forma Celular , Células Cultivadas , Dineínas do Citoplasma/antagonistas & inibidores , Laminas/fisiologia , Camundongos , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Centro Organizador dos Microtúbulos/ultraestrutura , Microtúbulos/fisiologia , Miosina Tipo II/fisiologia
15.
FEBS J ; 278(17): 2964-79, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21777385

RESUMO

Dynein, which is a minus-end-directed microtubule motor, is crucial to a range of cellular processes. The mass of its motor domain is about 10 times that of kinesin, the other microtubule motor. Its large size and the difficulty of expressing and purifying mutants have hampered progress in dynein research. Recently, however, electron microscopy, X-ray crystallography and single-molecule nanometry have shed light on several key unsolved questions concerning how the dynein molecule is organized, what conformational changes in the molecule accompany ATP hydrolysis, and whether two or three motor domains are coordinated in the movements of dynein. This minireview describes our current knowledge of the molecular organization and the force-generating mechanism of dynein, with emphasis on findings from electron microscopy and single-molecule nanometry.


Assuntos
Dineínas/fisiologia , Microtúbulos/metabolismo , Subunidades Proteicas/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Dineínas do Axonema/química , Dineínas do Axonema/fisiologia , Transporte Biológico , Dineínas do Citoplasma/química , Dineínas do Citoplasma/fisiologia , Dineínas/química , Humanos , Microtúbulos/química , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Subunidades Proteicas/química
16.
FEBS J ; 278(17): 2980-96, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21777386

RESUMO

The LC8 family members of dynein light chains (DYNLL1 and DYNLL2 in vertebrates) are highly conserved ubiquitous eukaryotic homodimer proteins that interact, besides dynein and myosin 5a motor proteins, with a large (and still incomplete) number of proteins involved in diverse biological functions. Despite an earlier suggestion that LC8 light chains function as cargo adapters of the above molecular motors, they are now recognized as regulatory hub proteins that interact with short linear motifs located in intrinsically disordered protein segments. The most prominent LC8 function is to promote dimerization of their binding partners that are often scaffold proteins of various complexes, including the intermediate chains of the dynein motor complex. Structural and functional aspects of this intriguing hub protein will be highlighted in this minireview.


Assuntos
Dineínas do Citoplasma/fisiologia , Citoesqueleto/metabolismo , Subunidades Proteicas/fisiologia , Animais , Transporte Biológico , Dineínas do Citoplasma/química , Dineínas/metabolismo , Humanos , Domínios e Motivos de Interação entre Proteínas , Subunidades Proteicas/química
17.
J Cell Physiol ; 226(10): 2666-74, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21792925

RESUMO

In living cells, a fluctuating torque is exerted on the nuclear surface but the origin of the torque is unclear. In this study, we found that the nuclear rotation angle is directionally persistent on a time scale of tens of minutes, but rotationally diffusive on longer time scales. Rotation required the activity of the microtubule motor dynein. We formulated a model based on microtubules undergoing dynamic instability, with tensional forces between a stationary centrosome and the nuclear surface mediated by dynein. Model simulations suggest that the persistence in rotation angle is due to the transient asymmetric configuration of microtubules exerting a net torque in one direction until the configuration is again randomized by dynamic instability. The model predicts that the rotational magnitude must depend on the distance between the nucleus and the centrosome. To test this prediction, rotation was quantified in patterned cells in which the cell's centrosome was close to the projected nuclear centroid. Consistent with the prediction, the angular displacement was found to decrease in these cells relative to unpatterned cells. This work provides the first mechanistic explanation for how nuclear dynein interactions with discrete microtubules emanating from a stationary centrosome cause rotational torque on the nucleus.


Assuntos
Núcleo Celular/fisiologia , Simulação por Computador , Dineínas do Citoplasma/fisiologia , Microtúbulos/fisiologia , Modelos Biológicos , Animais , Fenômenos Biomecânicos/fisiologia , Centrossomo/fisiologia , Corantes Fluorescentes/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Células NIH 3T3 , Rotação , Torque
18.
J Cell Biol ; 193(7): 1229-44, 2011 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-21690306

RESUMO

In animals, the female meiotic spindle is positioned at the egg cortex in a perpendicular orientation to facilitate the disposal of half of the chromosomes into a polar body. In Caenorhabditis elegans, the metaphase spindle lies parallel to the cortex, dynein is dispersed on the spindle, and the dynein activators ASPM-1 and LIN-5 are concentrated at spindle poles. Anaphase-promoting complex (APC) activation results in dynein accumulation at spindle poles and dynein-dependent rotation of one spindle pole to the cortex, resulting in perpendicular orientation. To test whether the APC initiates spindle rotation through cyclin B-CDK-1 inactivation, separase activation, or degradation of an unknown dynein inhibitor, CDK-1 was inhibited with purvalanol A in metaphase-I-arrested, APC-depleted embryos. CDK-1 inhibition resulted in the accumulation of dynein at spindle poles and dynein-dependent spindle rotation without chromosome separation. These results suggest that CDK-1 blocks rotation by inhibiting dynein association with microtubules and with LIN-5-ASPM-1 at meiotic spindle poles and that the APC promotes spindle rotation by inhibiting CDK-1.


Assuntos
Proteína Quinase CDC2/fisiologia , Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/genética , Dineínas/fisiologia , Meiose/fisiologia , Fuso Acromático/metabolismo , Ciclossomo-Complexo Promotor de Anáfase , Animais , Proteína Quinase CDC2/antagonistas & inibidores , Caenorhabditis elegans/citologia , Caenorhabditis elegans/ultraestrutura , Proteínas de Caenorhabditis elegans/antagonistas & inibidores , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclina B/fisiologia , Dineínas do Citoplasma/metabolismo , Dineínas do Citoplasma/fisiologia , Embrião não Mamífero/citologia , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Inibidores Enzimáticos/farmacologia , Microtúbulos/metabolismo , Purinas/farmacologia , Fuso Acromático/ultraestrutura , Complexos Ubiquitina-Proteína Ligase/fisiologia
19.
Cell Adh Migr ; 5(1): 42-7, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20935494

RESUMO

During vertebrate brain development, migration of neurons from the germinal zones to their final laminar positions is essential to establish functional neural circuits. Whereas key insights into neuronal migration initially came from landmark studies identifying the genes mutated in human cortical malformations, cell biology has recently greatly advanced our understanding of how cytoskeletal proteins and molecular motors drive the morphogenic cell movements that build the developing brain. This Commentary & View reviews recent studies examining the role of the molecular motors during neuronal migration and critically examines current models of acto-myosin function in the two-step neuronal migration cycle. Given the apparent emerging diversity of neuronal sub-type cytoskeletal organizations, we propose that two approaches must be taken to resolve differences between the current migration models: the mechanisms of radial and tangential migration must be compared and the loci of tension generation, migration substrates, and sites of adhesion dynamics must be precisely examined in an integrated manner.


Assuntos
Adesão Celular/fisiologia , Movimento Celular/fisiologia , Neuroglia/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Actinas/fisiologia , Dineínas do Citoplasma/fisiologia , Humanos , Miosina Tipo II/fisiologia , Neuroglia/citologia , Neurônios/metabolismo
20.
Nat Neurosci ; 13(12): 1463-71, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21037580

RESUMO

Radial glial progenitor cells exhibit bidirectional cell cycle-dependent nuclear oscillations. The purpose and underlying mechanism of this unusual 'interkinetic nuclear migration' are poorly understood. We investigated the basis for this behavior by live imaging of nuclei, centrosomes and microtubules in embryonic rat brain slices, coupled with the use of RNA interference (RNAi) and the myosin inhibitor blebbistatin. We found that nuclei migrated independent of centrosomes and unidirectionally away from or toward the ventricular surface along microtubules, which were uniformly oriented from the ventricular surface to the pial surface of the brain. RNAi directed against cytoplasmic dynein specifically inhibited nuclear movement toward the apical surface. An RNAi screen of kinesin genes identified Kif1a, a member of the kinesin-3 family, as the motor for basally directed nuclear movement. These observations provide direct evidence that kinesins are involved in nuclear migration and neurogenesis and suggest that a cell cycle-dependent switch between distinct microtubule motors drives interkinetic nuclear migration.


Assuntos
Movimento Celular/fisiologia , Núcleo Celular/fisiologia , Dineínas do Citoplasma/fisiologia , Células-Tronco Embrionárias/fisiologia , Cinesinas/fisiologia , Células-Tronco Neurais/fisiologia , Animais , Ciclo Celular/fisiologia , Células-Tronco Embrionárias/citologia , Cinese/fisiologia , Microtúbulos/fisiologia , Células-Tronco Neurais/citologia , Células PC12 , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA