Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
EMBO Rep ; 24(11): e57264, 2023 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-37702953

RESUMO

Cytoplasmic microtubules are tubular polymers that can harbor small proteins or filaments inside their lumen. The identities of these objects and mechanisms for their accumulation have not been conclusively established. Here, we used cryogenic electron tomography of Drosophila S2 cell protrusions and found filaments inside the microtubule lumen, which resemble those reported recently in human HAP1 cells. The frequency of these filaments increased upon inhibition of the sarco/endoplasmic reticulum Ca2+ ATPase with the small molecule drug thapsigargin. Subtomogram averaging showed that the luminal filaments adopt a helical structure reminiscent of cofilin-bound actin (cofilactin). Consistent with this, we observed cofilin dephosphorylation, an activating modification, in cells under the same conditions that increased luminal filament occurrence. Furthermore, RNA interference knock-down of cofilin reduced the frequency of luminal filaments with cofilactin morphology. These results suggest that cofilin activation stimulates its accumulation on actin filaments inside the microtubule lumen.


Assuntos
Citoesqueleto de Actina , Citoesqueleto , Humanos , Citoesqueleto/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Microtúbulos/metabolismo
2.
EMBO J ; 33(2): 96-8, 2014 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-24421323

RESUMO

Proteins residing at the plus and minus ends of microtubules have been thought not to communicate with each other, but recent findings on bona fide nucleation factors also regulating microtubule dynamics have challenged this notion. New work by Bouissou et al (2014) in The EMBO Journal now reveals that interplay between the nucleation factor γ­TuRC and the plus­end tracking protein EB1 controls mitotic spindle positioning by affecting the stability and dynamics of astral microtubules.


Assuntos
Proteínas Associadas aos Microtúbulos/fisiologia , Microtúbulos/metabolismo , Fuso Acromático/fisiologia , Tubulina (Proteína)/fisiologia , Animais , Humanos
3.
J Cell Sci ; 129(1): 121-34, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26585311

RESUMO

Coordination between different cytoskeletal systems is crucial for many cell biological functions, including cell migration and mitosis, and also plays an important role during tissue morphogenesis. Proteins of the class of cytoskeletal crosslinkers, or cytolinkers, have the ability to interact with more than one cytoskeletal system at a time and are prime candidates to mediate any coordination. One such class comprises the Gas2-like proteins, combining a conserved calponin-homology-type actin-binding domain and a Gas2 domain predicted to bind microtubules (MTs). This domain combination is also found in spectraplakins, huge cytolinkers that play important roles in many tissues in both invertebrates and vertebrates. Here, we dissect the ability of the single Drosophila Gas2-like protein Pigs to interact with both actin and MT cytoskeletons, both in vitro and in vivo, and illustrate complex regulatory interactions that determine the localisation of Pigs to and its effects on the cytoskeleton.


Assuntos
Polaridade Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Microtúbulos/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Motivos de Aminoácidos , Animais , Células Cultivadas , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Mutação/genética , Ligação Proteica , Estrutura Terciária de Proteína
4.
J Cell Sci ; 129(17): 3282-94, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27422099

RESUMO

Axonal growth and targeting are fundamental to the organization of the nervous system, and require active engagement of the cytoskeleton. Polymerization and stabilization of axonal microtubules is central to axonal growth and maturation of neuronal connectivity. Studies have suggested that members of the tubulin polymerization promoting protein (TPPP, also known as P25α) family are involved in cellular process extension. However, no in vivo knockout data exists regarding its role in axonal growth during development. Here, we report the characterization of Ringmaker (Ringer; CG45057), the only Drosophila homolog of long p25α proteins. Immunohistochemical analyses indicate that Ringer expression is dynamically regulated in the embryonic central nervous system (CNS). ringer-null mutants show cell misplacement, and errors in axonal extension and targeting. Ultrastructural examination of ringer mutants revealed defective microtubule morphology and organization. Primary neuronal cultures of ringer mutants exhibit defective axonal extension, and Ringer expression in cells induced microtubule stabilization and bundling into rings. In vitro assays showed that Ringer directly affects tubulin, and promotes microtubule bundling and polymerization. Together, our studies uncover an essential function of Ringer in axonal extension and targeting through proper microtubule organization.


Assuntos
Axônios/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Drosophila melanogaster/metabolismo , Desenvolvimento Embrionário , Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Alelos , Sequência de Aminoácidos , Animais , Proteínas de Drosophila/química , Drosophila melanogaster/ultraestrutura , Embrião não Mamífero/anormalidades , Embrião não Mamífero/metabolismo , Loci Gênicos , Microtúbulos/ultraestrutura , Mutação/genética , Proteínas do Tecido Nervoso/química , Polimerização
5.
Dev Biol ; 394(1): 6-14, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25127992

RESUMO

Epithelia form the building blocks of many tissue and organ types. Epithelial cells often form a contiguous 2-dimensional sheet that is held together by strong adhesions. The mechanical properties conferred by these adhesions allow the cells to undergo dramatic three-dimensional morphogenetic movements while maintaining cell-cell contacts during embryogenesis and post-embryonic development. The Drosophila Folded gastrulation pathway triggers epithelial cell shape changes that drive gastrulation and tissue folding and is one of the most extensively studied examples of epithelial morphogenesis. This pathway has yielded key insights into the signaling mechanisms and cellular machinery involved in epithelial remodeling. In this review, we discuss principles of morphogenesis and signaling that have been discovered through genetic and cell biological examination of this pathway. We also consider various regulatory mechanisms and the system׳s relevance to mammalian development. We propose future directions that will continue to broaden our knowledge of morphogenesis across taxa.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Drosophila/crescimento & desenvolvimento , Gastrulação , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Adesão Celular , Movimento Celular , Proteínas de Drosophila/genética , Células Epiteliais , Transdução de Sinais , Proteínas rho de Ligação ao GTP/metabolismo
6.
Mol Pharmacol ; 85(4): 586-97, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24435554

RESUMO

The G12/13 class of heterotrimeric G proteins, comprising the α-subunits Gα12 and Gα13, regulates multiple aspects of cellular behavior, including proliferation and cytoskeletal rearrangements. Although guanine nucleotide exchange factors for the monomeric G protein Rho (RhoGEFs) are well characterized as effectors of this G protein class, a variety of other downstream targets has been reported. To identify Gα12 determinants that mediate specific protein interactions, we used a structural and evolutionary comparison between the G12/13, Gs, Gi, and Gq classes to identify "class-distinctive" residues in Gα12 and Gα13. Mutation of these residues in Gα12 to their deduced ancestral forms revealed a subset necessary for activation of serum response element (SRE)-mediated transcription, a G12/13-stimulated pathway implicated in cell proliferative signaling. Unexpectedly, this subset of Gα12 mutants showed impaired binding to heat-shock protein 90 (Hsp90) while retaining binding to RhoGEFs. Corresponding mutants of Gα13 exhibited robust SRE activation, suggesting a Gα12-specific mechanism, and inhibition of Hsp90 by geldanamycin or small interfering RNA-mediated lowering of Hsp90 levels resulted in greater downregulation of Gα12 than Gα13 signaling in SRE activation experiments. Furthermore, the Drosophila G12/13 homolog Concertina was unable to signal to SRE in mammalian cells, and Gα12:Concertina chimeras revealed Gα12-specific determinants of SRE activation within the switch regions and a C-terminal region. These findings identify Gα12 determinants of SRE activation, implicate Gα12:Hsp90 interaction in this signaling mechanism, and illuminate structural features that arose during evolution of Gα12 and Gα13 to allow bifurcated mechanisms of signaling to a common cell proliferative pathway.


Assuntos
Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Elemento de Resposta Sérica , Animais , Linhagem Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Células HEK293 , Humanos , Mutação , Filogenia , Ligação Proteica , Transdução de Sinais , Ativação Transcricional , Proteínas rho de Ligação ao GTP/metabolismo
7.
bioRxiv ; 2023 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-37034688

RESUMO

Cytoplasmic microtubules are tubular polymers that can harbor small proteins or filaments inside their lumen. The identity of these objects and what causes their accumulation has not been conclusively established. Here, we used cryogenic electron tomography (cryoET) of Drosophila S2 cell protrusions and found filaments inside the microtubule lumen, which resemble those reported recently in human HAP1 cells. The frequency of these filaments increased upon inhibition of the sarco/endoplasmic reticulum Ca 2+ ATPase (SERCA) with the small-molecule drug thapsigargin. Subtomogram averaging showed that the luminal filaments adopt a helical structure reminiscent of cofilin-bound actin (cofilactin). Consistent with this, cofilin was activated in cells under the same conditions that increased luminal filament occurrence. Furthermore, RNAi knock-down of cofilin reduced the frequency of luminal filaments with cofilactin morphology. These results suggest that cofilin activation stimulates its accumulation on actin filaments inside the microtubule lumen.

8.
Blood ; 116(16): 3108-17, 2010 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-20664058

RESUMO

Regulated vascular endothelial growth factor (VEGF) signaling is required for proper angiogenesis, and excess VEGF signaling results in aberrantly formed vessels that do not function properly. Tumor endothelial cells have excess centrosomes and are aneuploid, properties that probably contribute to the morphologic and functional abnormalities of tumor vessels. We hypothesized that endothelial cell centrosome number is regulated by signaling via angiogenic factors, such as VEGF. We found that endothelial cells in developing vessels exposed to elevated VEGF signaling display centrosome overduplication. Signaling from VEGF, through either MEK/ERK or AKT to cyclin E/Cdk2, is amplified in association with centrosome overduplication, and blockade of relevant pathway components rescued the centrosome overduplication defect. Endothelial cells exposed to elevated FGF also had excess centrosomes, suggesting that multiple angiogenic factors regulate centrosome number. Endothelial cells with excess centrosomes survived and formed aberrant spindles at mitosis. Developing vessels exposed to elevated VEGF signaling also exhibited increased aneuploidy of endothelial cells, which is associated with cellular dysfunction. These results provide the first link between VEGF signaling and regulation of the centrosome duplication cycle, and suggest that endothelial cell centrosome overduplication contributes to aberrant angiogenesis in developing vessel networks exposed to excess angiogenic factors.


Assuntos
Indutores da Angiogênese/metabolismo , Vasos Sanguíneos/crescimento & desenvolvimento , Centrossomo/metabolismo , Células Endoteliais/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Aneuploidia , Animais , Vasos Sanguíneos/metabolismo , Linhagem Celular , Proliferação de Células , Células Cultivadas , Ciclina E/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Células Endoteliais/citologia , Humanos , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Saco Vitelino/citologia
9.
Nat Cell Biol ; 7(3): 235-45, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15723056

RESUMO

Regulation of microtubule polymerization and depolymerization is required for proper cell development. Here, we report that two proteins of the Drosophila melanogaster kinesin-13 family, KLP10A and KLP59C, cooperate to drive microtubule depolymerization in interphase cells. Analyses of microtubule dynamics in S2 cells depleted of these proteins indicate that both proteins stimulate depolymerization, but alter distinct parameters of dynamic instability; KLP10A stimulates catastrophe (a switch from growth to shrinkage) whereas KLP59C suppresses rescue (a switch from shrinkage to growth). Moreover, immunofluorescence and live analyses of cells expressing tagged kinesins reveal that KLP10A and KLP59C target to polymerizing and depolymerizing microtubule plus ends, respectively. Our data also suggest that KLP10A is deposited on microtubules by the plus-end tracking protein, EB1. Our findings support a model in which these two members of the kinesin-13 family divide the labour of microtubule depolymerization.


Assuntos
Interfase , Cinesinas/fisiologia , Microtúbulos/ultraestrutura , Animais , Western Blotting , Linhagem Celular , Drosophila , Drosophila melanogaster , Glutationa Transferase/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Cinesinas/química , Cinesinas/metabolismo , Microscopia de Fluorescência , Modelos Biológicos , Polímeros/química , Estrutura Terciária de Proteína , Interferência de RNA , RNA de Cadeia Dupla/química , Fatores de Tempo
10.
J Cell Biol ; 176(5): 641-51, 2007 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-17325206

RESUMO

Dynactin links cytoplasmic dynein and other motors to cargo and is involved in organizing radial microtubule arrays. The largest subunit of dynactin, p150(glued), binds the dynein intermediate chain and has an N-terminal microtubule-binding domain. To examine the role of microtubule binding by p150(glued), we replaced the wild-type p150(glued) in Drosophila melanogaster S2 cells with mutant DeltaN-p150 lacking residues 1-200, which is unable to bind microtubules. Cells treated with cytochalasin D were used for analysis of cargo movement along microtubules. Strikingly, although the movement of both membranous organelles and messenger ribonucleoprotein complexes by dynein and kinesin-1 requires dynactin, the substitution of full-length p150(glued) with DeltaN-p150(glued) has no effect on the rate, processivity, or step size of transport. However, truncation of the microtubule-binding domain of p150(glued) has a dramatic effect on cell division, resulting in the generation of multipolar spindles and free microtubule-organizing centers. Thus, dynactin binding to microtubules is required for organizing spindle microtubule arrays but not cargo motility in vivo.


Assuntos
Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Sequência de Aminoácidos , Animais , Sítios de Ligação , Transporte Biológico/fisiologia , Linhagem Celular , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Drosophila melanogaster/ultraestrutura , Complexo Dinactina , Dineínas/metabolismo , Dineínas/fisiologia , Cinesinas/fisiologia , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/química , Centro Organizador dos Microtúbulos/metabolismo , Centro Organizador dos Microtúbulos/ultraestrutura , Peroxissomos/fisiologia , Estrutura Terciária de Proteína , Interferência de RNA , Deleção de Sequência , Fuso Acromático/metabolismo , Fuso Acromático/ultraestrutura
11.
Mol Biol Cell ; 33(5): ar19, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35235367

RESUMO

The spectraplakin family of proteins includes ACF7/MACF1 and BPAG1/dystonin in mammals, VAB-10 in Caenorhabditis elegans, Magellan in zebrafish, and Short stop (Shot), the sole Drosophila member. Spectraplakins are giant cytoskeletal proteins that cross-link actin, microtubules, and intermediate filaments, coordinating the activity of the entire cytoskeleton. We examined the role of Shot during cell migration using two systems: the in vitro migration of Drosophila tissue culture cells and in vivo through border cell migration. RNA interference (RNAi) depletion of Shot increases the rate of random cell migration in Drosophila tissue culture cells as well as the rate of wound closure during scratch-wound assays. This increase in cell migration prompted us to analyze focal adhesion dynamics. We found that the rates of focal adhesion assembly and disassembly were faster in Shot-depleted cells, leading to faster adhesion turnover that could underlie the increased migration speeds. This regulation of focal adhesion dynamics may be dependent on Shot being in an open confirmation. Using Drosophila border cells as an in vivo model for cell migration, we found that RNAi depletion led to precocious border cell migration. Collectively, these results suggest that spectraplakins not only function to cross-link the cytoskeleton but may regulate cell-matrix adhesion.


Assuntos
Actinas , Proteínas de Drosophila , Actinas/metabolismo , Animais , Movimento Celular , Proteínas do Citoesqueleto/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Adesões Focais/metabolismo , Mamíferos/metabolismo , Proteínas dos Microfilamentos/metabolismo , Microtúbulos/metabolismo , Peixe-Zebra/metabolismo
12.
J Neurosci ; 30(16): 5653-67, 2010 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-20410118

RESUMO

Slit/Roundabout (Robo) signaling controls midline repulsive axon guidance. However, proteins that interact with Slit/Robo at the cell surface remain largely uncharacterized. Here, we report that the Drosophila transmembrane septate junction-specific protein Neurexin IV (Nrx IV) functions in midline repulsive axon guidance. Nrx IV is expressed in the neurons of the developing ventral nerve cord, and nrx IV mutants show crossing and circling of ipsilateral axons and fused commissures. Interestingly, the axon guidance defects observed in nrx IV mutants seem independent of its other binding partners, such as Contactin and Neuroglian and the midline glia protein Wrapper, which interacts in trans with Nrx IV. nrx IV mutants show diffuse Robo localization, and dose-dependent genetic interactions between nrx IV/robo and nrx IV/slit indicate that they function in a common pathway. In vivo biochemical studies reveal that Nrx IV associates with Robo, Slit, and Syndecan, and interactions between Robo and Slit, or Nrx IV and Slit, are affected in nrx IV and robo mutants, respectively. Coexpression of Nrx IV and Robo in mammalian cells confirms that these proteins retain the ability to interact in a heterologous system. Furthermore, we demonstrate that the extracellular region of Nrx IV is sufficient to rescue Robo localization and axon guidance phenotypes in nrx IV mutants. Together, our studies establish that Nrx IV is essential for proper Robo localization and identify Nrx IV as a novel interacting partner of the Slit/Robo signaling pathway.


Assuntos
Axônios/fisiologia , Moléculas de Adesão Celular Neuronais/fisiologia , Proteínas de Drosophila/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/fisiologia , Receptores Imunológicos/metabolismo , Animais , Células CHO , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Linhagem Celular , Células Cultivadas , Cricetinae , Cricetulus , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Humanos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Receptores Imunológicos/genética , Receptores Imunológicos/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Proteínas Roundabout
13.
MicroPubl Biol ; 20212021 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-33537561

RESUMO

We have previously adapted a select number of Drosophila cell lines to grow in serum-free media supplemented with fly extract. This condition is arguably more representative of a native growth environment. Here, we validated that the fly extract adapted line, S2R+ (FEx 2.5%) is amenable to RNAi. RNAi against Rho1 in both S2R+ and S2R+ (FEx 2.5%) produced phenotypes similar to ones previously described in Drosophila S2 cells.

14.
J Cell Biol ; 168(4): 587-98, 2005 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-15699215

RESUMO

EB1 is a member of a conserved protein family that localizes to growing microtubule plus ends. EB1 proteins also recruit cell polarity and signaling molecules to microtubule tips. However, the mechanism by which EB1 recognizes cargo is unknown. Here, we have defined a repeat sequence in adenomatous polyposis coli (APC) that binds to EB1's COOH-terminal domain and identified a similar sequence in members of the microtubule actin cross-linking factor (MACF) family of spectraplakins. We show that MACFs directly bind EB1 and exhibit EB1-dependent plus end tracking in vivo. To understand how EB1 recognizes APC and MACFs, we solved the crystal structure of the EB1 COOH-terminal domain. The structure reveals a novel homodimeric fold comprised of a coiled coil and four-helix bundle motif. Mutational analysis reveals that the cargo binding site for MACFs maps to a cluster of conserved residues at the junction between the coiled coil and four-helix bundle. These results provide a structural understanding of how EB1 binds two regulators of microtubule-based cell polarity.


Assuntos
Proteína da Polipose Adenomatosa do Colo/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Modelos Moleculares , Proteína da Polipose Adenomatosa do Colo/genética , Motivos de Aminoácidos/fisiologia , Sequência de Aminoácidos , Animais , Sítios de Ligação/fisiologia , Células Cultivadas , Drosophila , Dados de Sequência Molecular , Mutagênese/genética , Ligação Proteica
15.
Nature ; 427(6972): 364-70, 2004 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-14681690

RESUMO

During anaphase identical sister chromatids separate and move towards opposite poles of the mitotic spindle. In the spindle, kinetochore microtubules have their plus ends embedded in the kinetochore and their minus ends at the spindle pole. Two models have been proposed to account for the movement of chromatids during anaphase. In the 'Pac-Man' model, kinetochores induce the depolymerization of kinetochore microtubules at their plus ends, which allows chromatids to move towards the pole by 'chewing up' microtubule tracks. In the 'poleward flux' model, kinetochores anchor kinetochore microtubules and chromatids are pulled towards the poles through the depolymerization of kinetochore microtubules at the minus ends. Here, we show that two functionally distinct microtubule-destabilizing KinI kinesin enzymes (so named because they possess a kinesin-like ATPase domain positioned internally within the polypeptide) are responsible for normal chromatid-to-pole motion in Drosophila. One of them, KLP59C, is required to depolymerize kinetochore microtubules at their kinetochore-associated plus ends, thereby contributing to chromatid motility through a Pac-Man-based mechanism. The other, KLP10A, is required to depolymerize microtubules at their pole-associated minus ends, thereby moving chromatids by means of poleward flux.


Assuntos
Anáfase , Cromátides/fisiologia , Segregação de Cromossomos , Proteínas de Drosophila/metabolismo , Cinesinas/metabolismo , Mitose , Anáfase/efeitos dos fármacos , Animais , Cromátides/efeitos dos fármacos , Pareamento Cromossômico/efeitos dos fármacos , Segregação de Cromossomos/efeitos dos fármacos , Cromossomos/efeitos dos fármacos , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Cinesinas/antagonistas & inibidores , Cinesinas/genética , Mitose/efeitos dos fármacos , Fuso Acromático/efeitos dos fármacos , Fuso Acromático/metabolismo
16.
Mol Biol Cell ; 31(21): 2379-2397, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32816624

RESUMO

To identify novel regulators of nonmuscle myosin II (NMII) we performed an image-based RNA interference screen using stable Drosophila melanogaster S2 cells expressing the enhanced green fluorescent protein (EGFP)-tagged regulatory light chain (RLC) of NMII and mCherry-Actin. We identified the Rab-specific GTPase-activating protein (GAP) RN-tre as necessary for the assembly of NMII RLC into contractile actin networks. Depletion of RN-tre led to a punctate NMII phenotype, similar to what is observed following depletion of proteins in the Rho1 pathway. Depletion of RN-tre also led to a decrease in active Rho1 and a decrease in phosphomyosin-positive cells by immunostaining, while expression of constitutively active Rho or Rho-kinase (Rok) rescues the punctate phenotype. Functionally, RN-tre depletion led to an increase in actin retrograde flow rate and cellular contractility in S2 and S2R+ cells, respectively. Regulation of NMII by RN-tre is only partially dependent on its GAP activity as overexpression of constitutively active Rabs inactivated by RN-tre failed to alter NMII RLC localization, while a GAP-dead version of RN-tre partially restored phosphomyosin staining. Collectively, our results suggest that RN-tre plays an important regulatory role in NMII RLC distribution, phosphorylation, and function, likely through Rho1 signaling and putatively serving as a link between the secretion machinery and actomyosin contractility.


Assuntos
Citoesqueleto de Actina/metabolismo , Drosophila melanogaster/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Miosina Tipo II/metabolismo , Transdução de Sinais , Animais , Proteínas de Drosophila/metabolismo , Miosina Tipo II/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo
17.
Curr Biol ; 30(4): 610-623.e5, 2020 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-31928876

RESUMO

Neuronal axons terminate as synaptic boutons that form stable yet plastic connections with their targets. Synaptic bouton development relies on an underlying network of both long-lived and dynamic microtubules that provide structural stability for the boutons while also allowing for their growth and remodeling. However, a molecular-scale mechanism that explains how neurons appropriately balance these two microtubule populations remains a mystery. We hypothesized that α-tubulin acetyltransferase (αTAT), which both stabilizes long-lived microtubules against mechanical stress via acetylation and has been implicated in promoting microtubule dynamics, could play a role in this process. Using the Drosophila neuromuscular junction as a model, we found that non-enzymatic dαTAT activity limits the growth of synaptic boutons by affecting dynamic, but not stable, microtubules. Loss of dαTAT results in the formation of ectopic boutons. These ectopic boutons can be similarly suppressed by resupplying enzyme-inactive dαTAT or by treatment with a low concentration of the microtubule-targeting agent vinblastine, which acts to suppress microtubule dynamics. Biophysical reconstitution experiments revealed that non-enzymatic αTAT1 activity destabilizes dynamic microtubules but does not substantially impact the stability of long-lived microtubules. Further, during microtubule growth, non-enzymatic αTAT1 activity results in increasingly extended tip structures, consistent with an increased rate of acceleration of catastrophe frequency with microtubule age, perhaps via tip structure remodeling. Through these mechanisms, αTAT enriches for stable microtubules at the expense of dynamic ones. We propose that the specific suppression of dynamic microtubules by non-enzymatic αTAT activity regulates the remodeling of microtubule networks during synaptic bouton development.


Assuntos
Acetiltransferases/metabolismo , Drosophila melanogaster/metabolismo , Junção Neuromuscular/fisiologia , Terminações Pré-Sinápticas/fisiologia , Animais , Drosophila melanogaster/enzimologia , Drosophila melanogaster/crescimento & desenvolvimento , Larva/enzimologia , Larva/crescimento & desenvolvimento , Larva/metabolismo
18.
J Cell Biol ; 162(6): 1079-88, 2003 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12975351

RESUMO

Cell migration occurs through the protrusion of the actin-enriched lamella. Here, we investigated the effects of RNAi depletion of approximately 90 proteins implicated in actin function on lamella formation in Drosophila S2 cells. Similar to in vitro reconstitution studies of actin-based Listeria movement, we find that lamellae formation requires a relatively small set of proteins that participate in actin nucleation (Arp2/3 and SCAR), barbed end capping (capping protein), filament depolymerization (cofilin and Aip1), and actin monomer binding (profilin and cyclase-associated protein). Lamellae are initiated by parallel and partially redundant signaling pathways involving Rac GTPases and the adaptor protein Nck, which stimulate SCAR, an Arp2/3 activator. We also show that RNAi of three proteins (kette, Abi, and Sra-1) known to copurify with and inhibit SCAR in vitro leads to SCAR degradation, revealing a novel function of this protein complex in SCAR stability. Our results have identified an essential set of proteins involved in actin dynamics during lamella formation in Drosophila S2 cells.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Movimento Celular/fisiologia , Proteínas Contráteis , Células Eucarióticas/metabolismo , Pseudópodes/metabolismo , Citoesqueleto de Actina/ultraestrutura , Fatores de Despolimerização de Actina , Proteína 2 Relacionada a Actina , Actinas/antagonistas & inibidores , Actinas/deficiência , Actinas/genética , Animais , Proteínas de Caenorhabditis elegans/antagonistas & inibidores , Proteínas de Caenorhabditis elegans/genética , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Proteínas do Citoesqueleto/antagonistas & inibidores , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Destrina , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Células Eucarióticas/citologia , Proteínas dos Microfilamentos/antagonistas & inibidores , Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Profilinas , Pseudópodes/ultraestrutura , Interferência de RNA , Proteínas rac de Ligação ao GTP/metabolismo
19.
J Cell Biol ; 158(5): 873-84, 2002 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-12213835

RESUMO

EB1 is an evolutionarily conserved protein that localizes to the plus ends of growing microtubules. In yeast, the EB1 homologue (BIM1) has been shown to modulate microtubule dynamics and link microtubules to the cortex, but the functions of metazoan EB1 proteins remain unknown. Using a novel preparation of the Drosophila S2 cell line that promotes cell attachment and spreading, we visualized dynamics of single microtubules in real time and found that depletion of EB1 by RNA-mediated inhibition (RNAi) in interphase cells causes a dramatic increase in nondynamic microtubules (neither growing nor shrinking), but does not alter overall microtubule organization. In contrast, several defects in microtubule organization are observed in RNAi-treated mitotic cells, including a drastic reduction in astral microtubules, malformed mitotic spindles, defocused spindle poles, and mispositioning of spindles away from the cell center. Similar phenotypes were observed in mitotic spindles of Drosophila embryos that were microinjected with anti-EB1 antibodies. In addition, live cell imaging of mitosis in Drosophila embryos reveals defective spindle elongation and chromosomal segregation during anaphase after antibody injection. Our results reveal crucial roles for EB1 in mitosis, which we postulate involves its ability to promote the growth and interactions of microtubules within the central spindle and at the cell cortex.


Assuntos
Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Fuso Acromático/metabolismo , Anáfase , Animais , Linhagem Celular , Segregação de Cromossomos , Drosophila melanogaster/embriologia , Interfase , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/deficiência , Microtúbulos/metabolismo , Mitose , Fatores de Tempo
20.
Neuron ; 39(5): 779-91, 2003 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-12948445

RESUMO

Actin and microtubules (MTs) are tightly coordinated during neuronal growth cone navigation and are dynamically regulated in response to guidance cues; however, little is known about the underlying molecular mechanisms. Here, we characterize Drosophila pod-1 (dpod1) and show that purified Dpod1 can crosslink both actin and MTs. In cultured S2 cells, Dpod1 colocalizes with lamellar actin and MTs, and overexpression remodels the cytoskeleton to promote dynamic neurite-like actin-dependent projections. Consistent with these observations, Dpod1 localizes to the tips of growing axons, regions where actin and MTs interact, and is especially abundant at navigational choice points. In either the absence or overabundance of Dpod1, growth cone targeting but not outgrowth is disrupted. Taken together, these results reveal novel activities for pod-1 and show that proper levels of Dpod1, an actin/MT crosslinker, must be maintained in the growth cone for correct axon guidance.


Assuntos
Actinas/metabolismo , Proteínas de Drosophila , Cones de Crescimento/fisiologia , Microtúbulos/metabolismo , Sistema Nervoso/embriologia , Fatores de Transcrição/metabolismo , Actinas/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Células Cultivadas , Citoesqueleto/efeitos dos fármacos , Drosophila , Embrião de Mamíferos , Embrião não Mamífero , Humanos , Imuno-Histoquímica , Hibridização In Situ , Microtúbulos/efeitos dos fármacos , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/química , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Fatores de Transcrição/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa