Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Biol Cell ; 35(1): br1, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37910204

RESUMO

Fibroblasts migrate discontinuously by generating transient leading-edge protrusions and irregular, abrupt retractions of a narrow trailing edge. In contrast, keratinocytes migrate persistently and directionally via a single, stable, broad protrusion paired with a stable trailing-edge. The Rho GTPases Rac1, Cdc42 and RhoA are key regulators of cell protrusions and retractions. However, how these molecules mediate cell-type specific migration modes is still poorly understood. In fibroblasts, all three Rho proteins are active at the leading edge, suggesting short-range coordination of protrusive Rac1 and Cdc42 signals with RhoA retraction signals. Here, we show that Cdc42 was surprisingly active in the trailing-edge of migrating keratinocytes. Elevated Cdc42 activity colocalized with the effectors MRCK and N-WASP suggesting that Cdc42 controls both myosin activation and actin polymerization in the back. Indeed, Cdc42 was required to maintain the highly dynamic contractile acto-myosin retrograde flow at the trailing edge of keratinocytes, and its depletion induced ectopic protrusions in the back, leading to decreased migration directionality. These findings suggest that Cdc42 is required to stabilize the dynamic cytoskeletal polarization in keratinocytes, to enable persistent, directional migration.


Assuntos
Movimento Celular , Queratinócitos , Proteína cdc42 de Ligação ao GTP , Proteínas rho de Ligação ao GTP , Proteína cdc42 de Ligação ao GTP/metabolismo , Fibroblastos/metabolismo , Queratinócitos/fisiologia , Miosinas/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Humanos
2.
Nat Commun ; 14(1): 8356, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38102112

RESUMO

Rho GTPases play a key role in the spatio-temporal coordination of cytoskeletal dynamics during cell migration. Here, we directly investigate crosstalk between the major Rho GTPases Rho, Rac and Cdc42 by combining rapid activity perturbation with activity measurements in mammalian cells. These studies reveal that Rac stimulates Rho activity. Direct measurement of spatio-temporal activity patterns show that Rac activity is tightly and precisely coupled to local cell protrusions, followed by Rho activation during retraction. Furthermore, we find that the Rho-activating Lbc-type GEFs Arhgef11 and Arhgef12 are enriched at transient cell protrusions and retractions and recruited to the plasma membrane by active Rac. In addition, their depletion reduces activity crosstalk, cell protrusion-retraction dynamics and migration distance and increases migration directionality. Thus, our study shows that Arhgef11 and Arhgef12 facilitate exploratory cell migration by coordinating cell protrusion and retraction by coupling the activity of the associated regulators Rac and Rho.


Assuntos
Tamanho Celular , Proteínas rho de Ligação ao GTP , Animais , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Membrana Celular/metabolismo , Movimento Celular , Citoesqueleto/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Mamíferos/metabolismo , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo
3.
Pflugers Arch ; 475(12): 1439-1452, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37851146

RESUMO

Cell contraction plays an important role in many physiological and pathophysiological processes. This includes functions in skeletal, heart, and smooth muscle cells, which lead to highly coordinated contractions of multicellular assemblies, and functions in non-muscle cells, which are often highly localized in subcellular regions and transient in time. While the regulatory processes that control cell contraction in muscle cells are well understood, much less is known about cell contraction in non-muscle cells. In this review, we focus on the mechanisms that control cell contraction in space and time in non-muscle cells, and how they can be investigated by light-based methods. The review particularly focusses on signal networks and cytoskeletal components that together control subcellular contraction patterns to perform functions on the level of cells and tissues, such as directional migration and multicellular rearrangements during development. Key features of light-based methods that enable highly local and fast perturbations are highlighted, and how experimental strategies can capitalize on these features to uncover causal relationships in the complex signal networks that control cell contraction.


Assuntos
Contração Muscular , Músculo Liso , Músculo Liso/metabolismo , Contração Muscular/fisiologia , Miócitos de Músculo Liso , Fosforilação
4.
iScience ; 25(6): 104355, 2022 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-35601920

RESUMO

The unique threonine protease Tasp1 impacts not only ordered development and cell proliferation but also pathologies. However, its substrates and the underlying molecular mechanisms remain poorly understood. We demonstrate that the unconventional Myo1f is a Tasp1 substrate and unravel the physiological relevance of this proteolysis. We classify Myo1f as a nucleo-cytoplasmic shuttle protein, allowing its unhindered processing by nuclear Tasp1 and an association with chromatin. Moreover, we show that Myo1f induces filopodia resulting in increased cellular adhesion and migration. Importantly, filopodia formation was antagonized by Tasp1-mediated proteolysis, supported by an inverse correlation between Myo1f concentration and Tasp1 expression level. The Tasp1/Myo1f-axis might be relevant in human hematopoiesis as reduced Tasp1 expression coincided with increased Myo1f concentrations and filopodia in macrophages compared to monocytes and vice versa. In sum, we discovered Tasp1-mediated proteolysis of Myo1f as a mechanism to fine-tune filopodia formation, inter alia relevant for cells of the immune system.

5.
Chembiochem ; 23(4): e202100582, 2022 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-34897929

RESUMO

Cells process information via signal networks that typically involve multiple components which are interconnected by feedback loops. The combination of acute optogenetic perturbations and microscopy-based fluorescent response readouts enables the direct investigation of causal links in such networks. However, due to overlaps in spectra of photosensitive and fluorescent proteins, current approaches that combine these methods are limited. Here, we present an improved chemo-optogenetic approach that is based on switch-like perturbations induced by a single, local pulse of UV light. We show that this approach can be combined with parallel monitoring of multiple fluorescent readouts to directly uncover relations between signal network components. We present the application of this technique to directly investigate feedback-controlled regulation in the cell contraction signal network that includes GEF-H1, Rho and Myosin, and functional interactions of this network with tumor relevant RhoA G17 mutants.


Assuntos
Miosinas/genética , Optogenética , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Proteína rhoA de Ligação ao GTP/genética , Linhagem Celular Tumoral , Humanos , Mutação , Raios Ultravioleta
6.
Cell Rep ; 37(8): 110056, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34818551

RESUMO

Statins are among the most commonly prescribed drugs, and around every fourth person above the age of 40 is on statin medication. Therefore, it is of utmost clinical importance to understand the effect of statins on cancer cell plasticity and its consequences to not only patients with cancer but also patients who are on statins. Here, we find that statins induce a partial epithelial-to-mesenchymal transition (EMT) phenotype in cancer cells of solid tumors. Using a comprehensive STRING network analysis of transcriptome, proteome, and phosphoproteome data combined with multiple mechanistic in vitro and functional in vivo analyses, we demonstrate that statins reduce cellular plasticity by enforcing a mesenchymal-like cell state that increases metastatic seeding ability on one side but reduces the formation of (secondary) tumors on the other due to heterogeneous treatment responses. Taken together, we provide a thorough mechanistic overview of the consequences of statin use for each step of cancer development, progression, and metastasis.


Assuntos
Plasticidade Celular/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Neoplasias/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Transição Epitelial-Mesenquimal/genética , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Metástase Neoplásica , Células-Tronco Neoplásicas/patologia
7.
Cell Rep ; 33(9): 108467, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33264629

RESUMO

Local cell contraction pulses play important roles in tissue and cell morphogenesis. Here, we improve a chemo-optogenetic approach and apply it to investigate the signal network that generates these pulses. We use these measurements to derive and parameterize a system of ordinary differential equations describing temporal signal network dynamics. Bifurcation analysis and numerical simulations predict a strong dependence of oscillatory system dynamics on the concentration of GEF-H1, an Lbc-type RhoGEF, which mediates the positive feedback amplification of Rho activity. This prediction is confirmed experimentally via optogenetic tuning of the effective GEF-H1 concentration in individual living cells. Numerical simulations show that pulse amplitude is most sensitive to external inputs into the myosin component at low GEF-H1 concentrations and that the spatial pulse width is dependent on GEF-H1 diffusion. Our study offers a theoretical framework to explain the emergence of local cell contraction pulses and their modulation by biochemical and mechanical signals.


Assuntos
Optogenética/métodos , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Humanos , Transdução de Sinais
8.
Biol Chem ; 399(8): 809-819, 2018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-29664730

RESUMO

Cells need to process multifaceted external cues to steer their dynamic behavior. To efficiently perform this task, cells implement several exploratory mechanisms to actively sample their environment. In particular, cells can use exploratory actin-based cell protrusions and contractions to engage and squeeze the environment and to actively probe its chemical and mechanical properties. Multiple excitable signal networks were identified that can generate local activity pulses to control these exploratory processes. Such excitable signal networks offer particularly efficient mechanisms to process chemical or mechanical signals to steer dynamic cell behavior, such as directional migration, tissue morphogenesis and cell fate decisions.


Assuntos
Movimento Celular , Transdução de Sinais , Animais , Humanos
9.
J Cell Biol ; 216(12): 4271-4285, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29055010

RESUMO

Rho GTPase-based signaling networks control cellular dynamics by coordinating protrusions and retractions in space and time. Here, we reveal a signaling network that generates pulses and propagating waves of cell contractions. These dynamic patterns emerge via self-organization from an activator-inhibitor network, in which the small GTPase Rho amplifies its activity by recruiting its activator, the guanine nucleotide exchange factor GEF-H1. Rho also inhibits itself by local recruitment of actomyosin and the associated RhoGAP Myo9b. This network structure enables spontaneous, self-limiting patterns of subcellular contractility that can explore mechanical cues in the extracellular environment. Indeed, actomyosin pulse frequency in cells is altered by matrix elasticity, showing that coupling of contractility pulses to environmental deformations modulates network dynamics. Thus, our study reveals a mechanism that integrates intracellular biochemical and extracellular mechanical signals into subcellular activity patterns to control cellular contractility dynamics.


Assuntos
Citoesqueleto de Actina/metabolismo , Mecanotransdução Celular , Microtúbulos/metabolismo , Miosinas/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Citoesqueleto de Actina/ultraestrutura , Actomiosina/genética , Actomiosina/metabolismo , Fenômenos Biomecânicos , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Células HeLa , Humanos , Microtúbulos/ultraestrutura , Miosinas/genética , Osteoblastos , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Proteínas rho de Ligação ao GTP/genética
10.
PLoS One ; 10(9): e0137043, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26331477

RESUMO

BACKGROUND: Glomerular podocytes are highly differentiated cells that are key components of the kidney filtration units. The podocyte cytoskeleton builds the basis for the dynamic podocyte cytoarchitecture and plays a central role for proper podocyte function. Recent studies implicate that immunosuppressive agents including the mTOR-inhibitor everolimus have a protective role directly on the stability of the podocyte actin cytoskeleton. In contrast, a potential stabilization of microtubules by everolimus has not been studied so far. METHODS: To elucidate mechanisms underlying mTOR-inhibitor mediated cytoskeletal rearrangements, we carried out microarray gene expression studies to identify target genes and corresponding pathways in response to everolimus. We analyzed the effect of everolimus in a puromycin aminonucleoside experimental in vitro model of podocyte injury. RESULTS: Upon treatment with puromycin aminonucleoside, microarray analysis revealed gene clusters involved in cytoskeletal reorganization, cell adhesion, migration and extracellular matrix composition to be affected. Everolimus was capable of protecting podocytes from injury, both on transcriptional and protein level. Rescued genes included tubulin beta 2B class IIb (TUBB2B) and doublecortin domain containing 2 (DCDC2), both involved in microtubule structure formation in neuronal cells but not identified in podocytes so far. Validating gene expression data, Western-blot analysis in cultured podocytes demonstrated an increase of TUBB2B and DCDC2 protein after everolimus treatment, and immunohistochemistry in healthy control kidneys confirmed a podocyte-specific expression. Interestingly, Tubb2bbrdp/brdp mice revealed a delay in glomerular podocyte development as showed by podocyte-specific markers Wilm's tumour 1, Podocin, Nephrin and Synaptopodin. CONCLUSIONS: Taken together, our study suggests that off-target, non-immune mediated effects of the mTOR-inhibitor everolimus on the podocyte cytoskeleton might involve regulation of microtubules, revealing a potential novel role of TUBB2B and DCDC2 in glomerular podocyte development.


Assuntos
Everolimo/farmacologia , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/efeitos dos fármacos , Podócitos/efeitos dos fármacos , Tubulina (Proteína)/genética , Animais , Adesão Celular , Linhagem Celular Transformada , Humanos , Rim/metabolismo , Camundongos , Camundongos Mutantes , Microtúbulos/metabolismo , Podócitos/metabolismo , Transcriptoma
11.
J Cell Sci ; 127(Pt 7): 1379-93, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24481812

RESUMO

The formin FHOD1 (formin homology 2 domain containing protein 1) can act as a capping and bundling protein in vitro. In cells, active FHOD1 stimulates the formation of ventral stress fibers. However, the cellular mechanisms by which this phenotype is produced and the physiological relevance of FHOD1 function are not currently understood. Here, we first show that FHOD1 controls the formation of two distinct stress fiber precursors differentially. On the one hand, it inhibits dorsal fiber growth, which requires the polymerization of parallel bundles of long actin filaments. On the other hand, it stimulates transverse arcs that are formed by the fusion of short antiparallel actin filaments. This combined action is crucial for the maturation of stress fibers and their spatio-temporal organization, and a lack of FHOD1 function perturbs dynamic cell behavior during cell migration. Furthermore, we show that the GTPase-binding and formin homology 3 domains (GBD and FH3) are responsible for stress fiber association and colocalization with myosin. Surprisingly, a version of FHOD1 that lacks these domains nevertheless retains its full capacity to stimulate arc and ventral stress fiber formation. Based on our findings, we propose a mechanism in which FHOD1 promotes the formation of short actin filaments and transiently associates with transverse arcs, thus providing tight temporal and spatial control of the formation and turnover of transverse arcs into mature ventral stress fibers during dynamic cell behavior.


Assuntos
Citoesqueleto de Actina/metabolismo , Proteínas Fetais/metabolismo , Miosinas/metabolismo , Proteínas Nucleares/metabolismo , Fibras de Estresse/metabolismo , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Forminas , Humanos , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Estrutura Terciária de Proteína
12.
PLoS One ; 8(2): e55980, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23418489

RESUMO

Podocytes are highly differentiated kidney cells playing an important role in maintaining the glomerular filtration barrier. Particularly, the integrity of the actin cytoskeleton is crucial as cytoskeletal damage associated with foot process effacement and loss of slit diaphragms constitutes a major aspect of proteinuria. Previously, the mammalian target of rapamycin (mTOR) was linked to actin regulation and aberrant activity of the kinase was associated with renal disease. In this study, actin-related effects of mTOR inhibition by the immunosuppressant everolimus (EV) were investigated in human podocytes using an in vitro model of puromycin aminonucleoside (PAN) induced proteinuria. EV substantially recovered aberrant podocyte behavior by re-establishing a stationary phenotype with decreased migration efficiency, enhanced cell adhesion and recovery of actin stress fibers. Biochemical studies revealed substantial increase in the activity of RhoA and the effector pathway Rho-associated protein kinase (ROCK) and myosin light chain (MLC) by EV, all known regulators of stress fiber generation. Taken together, we show for the first time cytoskeleton stabilizing effects of the mTOR inhibitor EV and establish RhoA signaling as a key mediator in this process.


Assuntos
Citoesqueleto/efeitos dos fármacos , Imunossupressores/farmacologia , Podócitos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sirolimo/análogos & derivados , Proteína rhoA de Ligação ao GTP/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Células Cultivadas , Citoesqueleto/metabolismo , Everolimo , Humanos , Podócitos/metabolismo , Sirolimo/farmacologia , Fibras de Estresse/efeitos dos fármacos , Fibras de Estresse/metabolismo
13.
Nature ; 461(7260): 99-103, 2009 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-19693013

RESUMO

The GTPases Rac1, RhoA and Cdc42 act together to control cytoskeleton dynamics. Recent biosensor studies have shown that all three GTPases are activated at the front of migrating cells, and biochemical evidence suggests that they may regulate one another: Cdc42 can activate Rac1 (ref. 8), and Rac1 and RhoA are mutually inhibitory. However, their spatiotemporal coordination, at the seconds and single-micrometre dimensions typical of individual protrusion events, remains unknown. Here we examine GTPase coordination in mouse embryonic fibroblasts both through simultaneous visualization of two GTPase biosensors and using a 'computational multiplexing' approach capable of defining the relationships between multiple protein activities visualized in separate experiments. We found that RhoA is activated at the cell edge synchronous with edge advancement, whereas Cdc42 and Rac1 are activated 2 micro-m behind the edge with a delay of 40 s. This indicates that Rac1 and RhoA operate antagonistically through spatial separation and precise timing, and that RhoA has a role in the initial events of protrusion, whereas Rac1 and Cdc42 activate pathways implicated in reinforcement and stabilization of newly expanded protrusions.


Assuntos
Extensões da Superfície Celular/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Técnicas Biossensoriais , Movimento Celular , Forma Celular , Embrião de Mamíferos/citologia , Ativação Enzimática , Fibroblastos/citologia , Fibroblastos/enzimologia , Camundongos , Neuropeptídeos/metabolismo , Transporte Proteico , Fatores de Tempo , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP , Proteína rhoA de Ligação ao GTP
14.
Mol Biol Cell ; 20(18): 4070-82, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19625450

RESUMO

Cell migration involves the cooperative reorganization of the actin and microtubule cytoskeletons, as well as the turnover of cell-substrate adhesions, under the control of Rho family GTPases. RhoA is activated at the leading edge of motile cells by unknown mechanisms to control actin stress fiber assembly, contractility, and focal adhesion dynamics. The microtubule-associated guanine nucleotide exchange factor (GEF)-H1 activates RhoA when released from microtubules to initiate a RhoA/Rho kinase/myosin light chain signaling pathway that regulates cellular contractility. However, the contributions of activated GEF-H1 to coordination of cytoskeletal dynamics during cell migration are unknown. We show that small interfering RNA-induced GEF-H1 depletion leads to decreased HeLa cell directional migration due to the loss of the Rho exchange activity of GEF-H1. Analysis of RhoA activity by using a live cell biosensor revealed that GEF-H1 controls localized activation of RhoA at the leading edge. The loss of GEF-H1 is associated with altered leading edge actin dynamics, as well as increased focal adhesion lifetimes. Tyrosine phosphorylation of focal adhesion kinase and paxillin at residues critical for the regulation of focal adhesion dynamics was diminished in the absence of GEF-H1/RhoA signaling. This study establishes GEF-H1 as a critical organizer of key structural and signaling components of cell migration through the localized regulation of RhoA activity at the cell leading edge.


Assuntos
Movimento Celular , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Pseudópodes/enzimologia , Proteína rhoA de Ligação ao GTP/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Ativação Enzimática , Adesões Focais/enzimologia , Forminas , Fatores de Troca do Nucleotídeo Guanina/deficiência , Células HeLa , Humanos , Microtúbulos/metabolismo , Fosfotirosina/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho , Transdução de Sinais
15.
J Immunol ; 182(6): 3522-9, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19265130

RESUMO

Rho GTPases are essential regulators of signaling networks emanating from many receptors involved in innate or adaptive immunity. The Rho family member RhoA controls cytoskeletal processes as well as the activity of transcription factors such as NF-kappaB, C/EBP, and serum response factor. The multifaceted host cell activation triggered by TLRs in response to soluble and particulate microbial structures includes rapid stimulation of RhoA activity. RhoA acts downstream of TLR2 in HEK-TLR2 and monocytic THP-1 cells, but the signaling pathway connecting TLR2 and RhoA is still unknown. It is also not clear if RhoA activation is dependent on a certain TLR adapter. Using lung epithelial cells, we demonstrate TLR2- and TLR3-triggered recruitment and activation of RhoA at receptor-proximal cellular compartments. RhoA activity was dependent on TLR-mediated stimulation of Src family kinases. Both Src family kinases and RhoA were required for NF-kappaB activation, whereas RhoA was dispensable for type I IFN generation. These results suggest that RhoA plays a role downstream of MyD88-dependent and -independent TLR signaling and acts as a molecular switch downstream of TLR-Src-initiated pathways.


Assuntos
NF-kappa B/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 3 Toll-Like/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Quinases da Família src/metabolismo , Animais , Linhagem Celular , Linhagem Celular Transformada , Linhagem Celular Tumoral , Ativação Enzimática/imunologia , Humanos , Ligantes , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , Fator 88 de Diferenciação Mieloide/fisiologia , NF-kappa B/fisiologia , Transdução de Sinais/imunologia , Receptor 2 Toll-Like/fisiologia , Receptor 3 Toll-Like/fisiologia , Quinases da Família src/fisiologia
16.
Trends Cell Biol ; 18(5): 210-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18394899

RESUMO

The Rho guanine nucleotide exchange factor GEF-H1 is uniquely regulated by microtubule binding and is crucial in coupling microtubule dynamics to Rho-GTPase activation in a variety of normal biological situations. Here, we review the roles of GEF-H1 in epithelial barrier permeability, cell motility and polarization, dendritic spine morphology, antigen presentation, leukemic cell differentiation, cell cycle regulation, and cancer. GEF-H1 might also contribute to pathophysiological signaling involved in leukemias, and in cancers associated with mutated p53 tumor suppressor gene, epithelial and endothelial cell dysfunction, infectious disease, and cardiac hypertrophy. We suggest that GEF-H1 could be a novel therapeutic target in multiple human diseases.


Assuntos
Regulação da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Apresentação de Antígeno , Diferenciação Celular , Movimento Celular , Transformação Celular Neoplásica , Epitélio/patologia , Genes Supressores de Tumor , Humanos , Microtúbulos/metabolismo , Modelos Biológicos , Mutação , Neoplasias/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho
17.
Physiol Genomics ; 34(1): 95-100, 2008 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-18413783

RESUMO

Overlapping sense/antisense RNAs transcribed in opposite directions from the same genomic locus are common in vertebrates. The impact of antisense transcription on gene regulation and cell biology is largely unknown. We show that sense/antisense RNAs of an evolutionarily conserved phosphate transporter gene (Slc34a2a) are coexpressed in a short time window during embryonic development of zebrafish at 48 hours postfertilization (hpf). To address the mechanism by which coexpressed sense/antisense transcripts are processed, we injected sense/antisense RNAs in various combinations into Xenopus oocytes. In the cytoplasm RNAs were stable in whatever combination expressed. In the nucleus coinjected sense/antisense transcripts were degraded into short RNAs of approximately 23 bases within 4 h. A homologous transcript from toad or another isoform (Slc34a2b) from zebrafish failed to trigger processing. In oocytes that were primed with nuclear sense/antisense RNA coinjections, a reporter RNA was rapidly degraded. We produced evidence that the observed processing of complementary transcripts was not restricted to Xenopus oocytes, because Slc34a-related short RNAs were detected in zebrafish embryos by Northern blotting. Signals were observed at stages that showed coexpression of sense/antisense transcripts. Remarkably, strand-specific probes revealed that the orientation of short RNAs was developmentally regulated. In addition, RNA from zebrafish embryos 48 hpf was able to induce degradation of reporter constructs in Xenopus oocytes. Our findings may give important clues to understanding the physiological role of the widespread antisense transcription.


Assuntos
Processamento Pós-Transcricional do RNA , RNA Antissenso/metabolismo , RNA Nuclear Pequeno/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/genética , Transcrição Gênica , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genoma/genética , Camundongos , Oócitos , RNA Antissenso/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Nuclear Pequeno/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/metabolismo , Xenopus , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/metabolismo
18.
Mol Biol Cell ; 19(5): 2147-53, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18287519

RESUMO

The RhoA GTPase plays a vital role in assembly of contractile actin-myosin filaments (stress fibers) and of associated focal adhesion complexes of adherent monolayer cells in culture. GEF-H1 is a microtubule-associated guanine nucleotide exchange factor that activates RhoA upon release from microtubules. The overexpression of GEF-H1 deficient in microtubule binding or treatment of HeLa cells with nocodazole to induce microtubule depolymerization results in Rho-dependent actin stress fiber formation and contractile cell morphology. However, whether GEF-H1 is required and sufficient to mediate nocodazole-induced contractility remains unclear. We establish here that siRNA-mediated depletion of GEF-H1 in HeLa cells prevents nocodazole-induced cell contraction. Furthermore, the nocodazole-induced activation of RhoA and Rho-associated kinase (ROCK) that mediates phosphorylation of myosin regulatory light chain (MLC) is impaired in GEF-H1-depleted cells. Conversely, RhoA activation and contractility are rescued by reintroduction of siRNA-resistant GEF-H1. Our studies reveal a critical role for a GEF-H1/RhoA/ROCK/MLC signaling pathway in mediating nocodazole-induced cell contractility.


Assuntos
Fenômenos Fisiológicos Celulares/efeitos dos fármacos , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Microtúbulos/efeitos dos fármacos , Microtúbulos/enzimologia , Nocodazol/farmacologia , Proteína rhoA de Ligação ao GTP/metabolismo , Diferenciação Celular/efeitos dos fármacos , Fatores de Troca do Nucleotídeo Guanina/deficiência , Células HeLa , Humanos , Cadeias Leves de Miosina/metabolismo , RNA Interferente Pequeno/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho , Transdução de Sinais/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia
19.
J Cell Biol ; 180(1): 187-203, 2008 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-18195107

RESUMO

Integrin binding to matrix proteins such as fibronectin (FN) leads to formation of focal adhesion (FA) cellular contact sites that regulate migration. RhoA GTPases facilitate FA formation, yet FA-associated RhoA-specific guanine nucleotide exchange factors (GEFs) remain unknown. Here, we show that proline-rich kinase-2 (Pyk2) levels increase upon loss of focal adhesion kinase (FAK) in mouse embryonic fibroblasts (MEFs). Additionally, we demonstrate that Pyk2 facilitates deregulated RhoA activation, elevated FA formation, and enhanced cell proliferation by promoting p190RhoGEF expression. In normal MEFs, p190RhoGEF knockdown inhibits FN-associated RhoA activation, FA formation, and cell migration. Knockdown of p190RhoGEF-related GEFH1 does not affect FA formation in FAK(-/-) or normal MEFs. p190RhoGEF overexpression enhances RhoA activation and FA formation in MEFs dependent on FAK binding and associated with p190RhoGEF FA recruitment and tyrosine phosphorylation. These studies elucidate a compensatory function for Pyk2 upon FAK loss and identify the FAK-p190RhoGEF complex as an important integrin-proximal regulator of FA formation during FN-stimulated cell motility.


Assuntos
Movimento Celular/fisiologia , Quinase 1 de Adesão Focal/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Adesões Focais/fisiologia , ras-GRF1/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Proliferação de Células , Quinase 1 de Adesão Focal/genética , Quinase 2 de Adesão Focal/genética , Regulação da Expressão Gênica , Camundongos , Paxilina/metabolismo , Fosforilação , Tirosina/metabolismo , ras-GRF1/genética
20.
J Cell Biol ; 179(3): 403-10, 2007 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-17984318

RESUMO

A fundamental feature of cell polarity in response to spatial cues is asymmetric amplification of molecules generated by positive feedback signaling. We report a positive feedback loop between the guanosine triphosphatase Cdc42, a central determinant in eukaryotic cell polarity, and H(+) efflux by Na-H(+) exchanger 1 (NHE1), which is necessary at the front of migrating cells for polarity and directional motility. In response to migratory cues, Cdc42 is not activated in fibroblasts expressing a mutant NHE1 that lacks H(+) efflux, and wild-type NHE1 is not activated in fibroblasts expressing mutationally inactive Cdc42-N17. H(+) efflux by NHE1 is not necessary for release of Cdc42-guanosine diphosphate (GDP) from Rho GDP dissociation inhibitor or for the membrane recruitment of Cdc42 but is required for GTP binding by Cdc42 catalyzed by a guanine nucleotide exchange factor (GEF). Data indicate that GEF binding to phosphotidylinositol 4,5-bisphosphate is pH dependent, suggesting a mechanism for how H(+) efflux by NHE1 promotes Cdc42 activity to generate a positive feedback signal necessary for polarity in migrating cells.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Animais , Movimento Celular , Cricetinae , Fibroblastos/metabolismo , Nucleotídeos de Guanina/química , Guanosina Trifosfato/química , Concentração de Íons de Hidrogênio , Modelos Biológicos , Mutação , Estrutura Terciária de Proteína , Prótons , Trocador 1 de Sódio-Hidrogênio , Frações Subcelulares
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA