Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 339
Filtrar
1.
J Cell Biol ; 221(10)2022 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-35976098

RESUMEN

Modulation of presynaptic actin dynamics is fundamental to synaptic growth and functional plasticity; yet the underlying molecular and cellular mechanisms remain largely unknown. At Drosophila NMJs, the presynaptic Rac1-SCAR pathway mediates BMP-induced receptor macropinocytosis to inhibit BMP growth signaling. Here, we show that the Rho-type GEF Vav acts upstream of Rac1 to inhibit synaptic growth through macropinocytosis. We also present evidence that Vav-Rac1-SCAR signaling has additional roles in tetanus-induced synaptic plasticity. Presynaptic inactivation of Vav signaling pathway components, but not regulators of macropinocytosis, impairs post-tetanic potentiation (PTP) and enhances synaptic depression depending on external Ca2+ concentration. Interfering with the Vav-Rac1-SCAR pathway also impairs mobilization of reserve pool (RP) vesicles required for tetanus-induced synaptic plasticity. Finally, treatment with an F-actin-stabilizing drug completely restores RP mobilization and plasticity defects in Vav mutants. We propose that actin-regulatory Vav-Rac1-SCAR signaling independently regulates structural and functional presynaptic plasticity by driving macropinocytosis and RP mobilization, respectively.


Asunto(s)
Actinas , Proteínas de Drosophila , Factores de Intercambio de Guanina Nucleótido , Plasticidad Neuronal , Sinapsis , Actinas/fisiología , Animales , Receptores de Proteínas Morfogenéticas Óseas/fisiología , Calcio , Drosophila/fisiología , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas de Microfilamentos/fisiología , Unión Neuromuscular/fisiología , Transducción de Señal , Sinapsis/fisiología , Tétanos/metabolismo , Proteínas de Unión al GTP rac/fisiología
2.
Sci Rep ; 11(1): 23285, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34857846

RESUMEN

Machine learning approaches have shown great promise in biology and medicine discovering hidden information to further understand complex biological and pathological processes. In this study, we developed a deep learning-based machine learning algorithm to meaningfully process image data and facilitate studies in vascular biology and pathology. Vascular injury and atherosclerosis are characterized by neointima formation caused by the aberrant accumulation and proliferation of vascular smooth muscle cells (VSMCs) within the vessel wall. Understanding how to control VSMC behaviors would promote the development of therapeutic targets to treat vascular diseases. However, the response to drug treatments among VSMCs with the same diseased vascular condition is often heterogeneous. Here, to identify the heterogeneous responses of drug treatments, we created an in vitro experimental model system using VSMC spheroids and developed a machine learning-based computational method called HETEROID (heterogeneous spheroid). First, we established a VSMC spheroid model that mimics neointima-like formation and the structure of arteries. Then, to identify the morphological subpopulations of drug-treated VSMC spheroids, we used a machine learning framework that combines deep learning-based spheroid segmentation and morphological clustering analysis. Our machine learning approach successfully showed that FAK, Rac, Rho, and Cdc42 inhibitors differentially affect spheroid morphology, suggesting that multiple drug responses of VSMC spheroid formation exist. Overall, our HETEROID pipeline enables detailed quantitative drug characterization of morphological changes in neointima formation, that occurs in vivo, by single-spheroid analysis.


Asunto(s)
Aprendizaje Automático , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología , Aterosclerosis/patología , Células Cultivadas , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/fisiología , Humanos , Neointima/patología , Esferoides Celulares/fisiología , Lesiones del Sistema Vascular/patología , Proteína de Unión al GTP cdc42/antagonistas & inhibidores , Proteína de Unión al GTP cdc42/fisiología , Proteínas de Unión al GTP rac/antagonistas & inhibidores , Proteínas de Unión al GTP rac/fisiología
3.
Mol Cell Endocrinol ; 518: 110993, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32814070

RESUMEN

The type 1 diabetes (T1D) risk locus on chromosome 15q25.1 harbors the candidate gene CTSH (cathepsin H). We previously demonstrated that CTSH regulates ß-cell function in vitro and in vivo. CTSH overexpression protected insulin-secreting INS-1 cells against cytokine-induced apoptosis. The purpose of the present study was to identify the genes through which CTSH mediates its protective effects. Microarray analysis identified 63 annotated genes differentially expressed between CTSH-overexpressing INS-1 cells and control cells treated with interleukin-1ß and interferon-γ for up to 16h. Permutation test identified 10 significant genes across all time-points: Elmod1, Fam49a, Gas7, Gna15, Msrb3, Nox1, Ptgs1, Rac2, Scn7a and Ttn. Pathway analysis identified the "Inflammation mediated by chemokine and cytokine signaling pathway" with Gna15, Ptgs1 and Rac2 as significant. Knockdown of Rac2 abolished the protective effect of CTSH overexpression on cytokine-induced apoptosis, suggesting that the small GTPase and T1D candidate gene Rac2 contributes to the anti-apoptotic effect of CTSH.


Asunto(s)
Apoptosis , Catepsina H/genética , Citocinas/farmacología , Células Secretoras de Insulina/fisiología , Proteínas de Unión al GTP rac/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Catepsina H/fisiología , Células Cultivadas , Citoprotección/efectos de los fármacos , Citoprotección/genética , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Ratas , Proteína RCA2 de Unión a GTP
4.
Mol Med Rep ; 22(2): 1187-1194, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32626999

RESUMEN

The treatment of renal cell carcinoma (RCC) with chemotherapy remains a challenge; therefore, improving the knowledge of the molecular mechanisms underlying RCC chemoresistance and developing novel therapeutic strategies is important. Dedicator of cytokinesis 1 (DOCK1), the first member of the DOCK family to be discovered, displays various roles during tumorigenesis; however, its role during RCC progression is not completely understood. Therefore, the present study aimed to clarify the function of DOCK1 and 1­[2­(3'­(trifluoromethyl)­(1,1'­biphenyl)­4­yl)­2­oxoethyl]­5­pyrrolidinylsulfonyl­2 (1H)­pyridone (TBOPP), a DOCK1­sensitive inhibitor, during RCC development and chemoresistance. The results of CCK­8 and EdU assay indicated that TBOPP decreased RCC cell viability and proliferation compared with the control group, and sensitized RCC cells to cisplatin. Moreover, RCC cells with high DOCK1 expression levels displayed increased resistance to cisplatin, whereas DOCK1 knockdown enhanced the lethal effects of cisplatin on RCC cells. Furthermore, the results determined by western blotting, CCK­8 and cell apoptosis assay indicated that TBOPP effectively reduced DOCK1 expression levels compared with the control group, and the TBOPP­mediated cisplatin sensitizing effect was mediated by DOCK1 inhibition. The present study suggests that DOCK1 plays a vital role in RCC cell chemoresistance to cisplatin; therefore, TBOPP may serve as a novel therapeutic agent for RCC chemoresistance.


Asunto(s)
Carcinogénesis , Carcinoma de Células Renales , Cisplatino , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Renales , Piridonas/farmacología , Proteínas de Unión al GTP rac , Apoptosis/efectos de los fármacos , Carcinogénesis/efectos de los fármacos , Carcinogénesis/metabolismo , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/metabolismo , Proteínas de Unión al GTP rac/antagonistas & inhibidores , Proteínas de Unión al GTP rac/fisiología
5.
Sci Rep ; 10(1): 7024, 2020 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-32341385

RESUMEN

Vav1 regulates Rac activation as a hematopoietic-specific Rho/Rac-family guanine nucleotide exchange factor. Rac is a subfamily of Rho GTPases that regulates the bone-resorbing capacity of osteoclasts (OCs). In this study, we show that hematopoietic-specific Rac2 and Vav1 play opposing roles by enhancing or attenuating OC differentiation, respectively. This was demonstrated by higher and lower bone density in the femurs from Rac2-deficient (Rac2-/-) and Vav1-deficient (Vav1-/-) mice, respectively, compared to the wild-type (WT) mice. Accordingly, Rac2-/- cells displayed low numbers of tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cells (41%) compared to WT cells, whereas, Vav1-/- cells showed high TRAP-positive cell numbers (150%), and the double-knockout Rac2-/-Vav1-/- mice nullified the effects on OC numbers achieved by the individual knockouts. These reciprocal roles of Rac2 and Vav1 in OC differentiation were confirmed by reduced and increased levels of OC-specific markers, such as TRAP, calcitonin receptor, cathepsin K, and DC-STAMP in the Rac2-/- and Vav1-/- OCs, respectively. Our findings of decrease and increase in actin ring formation and αvß3 integrin-mediated adhesion in Rac2-/- and Vav1-/- mice, respectively, suggest that Vav1 and its downstream GTPase, Rac2, may counteract to fine-tune OC differentiation and bone resorption.


Asunto(s)
Diferenciación Celular/fisiología , Hematopoyesis , Osteoclastos/citología , Proteínas Proto-Oncogénicas c-vav/fisiología , Proteínas de Unión al GTP rac/fisiología , Animales , Ratones Noqueados , Osteoclastos/metabolismo , Proteína RCA2 de Unión a GTP
6.
PLoS One ; 14(8): e0220496, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31369617

RESUMEN

The Rac1 and Rac3 GTPases are co-expressed in the developing nervous system, where they are involved in different aspects of neuronal development, including the formation of synapses. The deletion of both Rac genes determines a stronger reduction of dendritic spines in vitro compared to the knockout of either gene, indicating that Rac1 and Rac3 play a synergistic role in the formation of these structures. Here, we have addressed the role of each GTPase in the formation of dendritic spines by overexpressing either Rac1 or Rac3 in wildtype neurons, or by re-expressing either GTPase in double knockout hippocampal cultures. We show that the Rac3 protein is expressed with Rac1 in developing hippocampal neurons. Overexpression of either GTPase in WT neurons increases the density of dendritic spines, suggesting the involvement of both GTPases in their formation. We also found that the re-expression of either Rac1 or Rac3 in double knockout neurons is sufficient to restore spinogenesis. Rac1 is significantly more efficient than Rac3 in restoring the formation of spines. On the other hand the quantitative analysis in neurons overexpressing or re-expressing either GTPase shows that Rac3 induces a more pronounced increase in the size of the spines compared to Rac1. These enlarged spines form morphological synapses identified by the juxtaposition of postsynaptic and presynaptic markers. Thus, while Rac1 appears more efficient in inducing the formation of mature spines, Rac3 is more efficient in promoting their enlargement. Our study highlights specific roles of Rac1 and Rac3, which may be functionally relevant also to synaptic plasticity.


Asunto(s)
Espinas Dendríticas/enzimología , Hipocampo/citología , Neuronas/enzimología , Neuropéptidos/fisiología , Proteínas de Unión al GTP rac/fisiología , Proteína de Unión al GTP rac1/fisiología , Animales , Espinas Dendríticas/fisiología , Técnica del Anticuerpo Fluorescente , Hipocampo/anatomía & histología , Hipocampo/enzimología , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/fisiología , Imagen de Lapso de Tiempo
7.
Eur J Clin Invest ; 48 Suppl 2: e12939, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29682742

RESUMEN

Rac-GTPases and their Rac-GEF activators play important roles in the recruitment and host defence functions of neutrophils. These proteins control the activation of adhesion molecules and the cytoskeletal dynamics that enable the adhesion, migration and tissue recruitment of neutrophils. They also regulate the effector functions that allow neutrophils to kill bacterial and fungal pathogens, and to clear debris. This review focuses on the roles of Rac-GTPases and Rac-GEFs in neutrophil adhesion, migration and recruitment.


Asunto(s)
Neutrófilos/fisiología , Factores de Intercambio de Guanina Nucleótido Rho/fisiología , Proteína de Unión al GTP rac1/fisiología , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Factores de Intercambio de Guanina Nucleótido/fisiología , Humanos , Infiltración Neutrófila/fisiología , Neutrófilos/enzimología , Proteínas Proto-Oncogénicas c-vav/fisiología , Transducción de Señal/fisiología , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/fisiología , Proteínas de Unión al GTP rac/fisiología
8.
Proc Natl Acad Sci U S A ; 115(16): E3731-E3740, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29610331

RESUMEN

Developmental programs often rely on parallel morphogenetic mechanisms that guarantee precise tissue architecture. While redundancy constitutes an obvious selective advantage, little is known on how novel morphogenetic mechanisms emerge during evolution. In zebrafish, rhombomeric boundaries behave as an elastic barrier, preventing cell intermingling between adjacent compartments. Here, we identify the fundamental role of the small-GTPase Rac3b in actomyosin cable assembly at hindbrain boundaries. We show that the novel rac3b/rfng/sgca regulatory cluster, which is specifically expressed at the boundaries, emerged in the Ostariophysi superorder by chromosomal rearrangement that generated new cis-regulatory interactions. By combining 4C-seq, ATAC-seq, transgenesis, and CRISPR-induced deletions, we characterized this regulatory domain, identifying hindbrain boundary-specific cis-regulatory elements. Our results suggest that the capacity of boundaries to act as an elastic mesh for segregating rhombomeric cells evolved by cooption of critical genes to a novel regulatory block, refining the mechanisms for hindbrain segmentation.


Asunto(s)
Actomiosina/fisiología , Regulación del Desarrollo de la Expresión Génica , Rombencéfalo/embriología , Sarcoglicanos/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra/embriología , Proteínas de Unión al GTP rac/fisiología , Animales , Tipificación del Cuerpo/genética , Sistemas CRISPR-Cas , Movimiento Celular , Characidae/genética , Characidae/fisiología , Cromatina/genética , Cromatina/ultraestructura , Evolución Molecular , Peces/clasificación , Peces/genética , Morfogénesis , Mutagénesis Sitio-Dirigida , Neurogénesis , Filogenia , Sarcoglicanos/genética , Especificidad de la Especie , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Unión al GTP rac/genética
9.
PLoS One ; 13(3): e0194003, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29518139

RESUMEN

Netrin receptors of the DCC/NEO/UNC-40/Frazzled family have well established roles in cell migration and axon guidance but can also regulate epithelial features such as adhesion, polarity and adherens junction (AJ) stability. Previously, we have shown that overexpression of Drosophila Frazzled (Fra) in the peripodial epithelium (PE) inhibits wing disc eversion and also generates cellular protrusions typical of motile cells. Here, we tested whether the molecular pathways by which Fra inhibits eversion are distinct from those driving motility. We show that in disc proper (DP) epithelial cells Fra, in addition to inducing F-Actin rich protrusions, can affect localization of AJ components and columnar cell shape. We then show that these phenotypes have different requirements for the three conserved Fra cytoplasmic P-motifs and for downstream genes. The formation of protrusions required the P3 motif of Fra, as well as integrins (mys and mew), the Rac pathway (Rac1, wave and, arpc3) and myosin regulatory light chain (Sqh). In contrast, apico-basal cell shape change, which was accompanied by increased myosin phosphorylation, was critically dependent upon the P1 motif and was promoted by RhoGef2 but inhibited by Rac1. Fra also caused a loss of AJ proteins (DE-Cad and Arm) from basolateral regions of epithelial cells. This phenotype required all 3 P-motifs, and was dependent upon the polarity factor par6. par6 was not required for protrusions or cell shape change, but was required to block eversion suggesting that control of AJ components may underlie the ability of Fra to promote epithelial stability. The results imply that multiple molecular pathways act downstream of Fra in epithelial cells.


Asunto(s)
Cadherinas/metabolismo , Proteínas de Drosophila/fisiología , Drosophila melanogaster/metabolismo , Células Epiteliales/citología , Receptores de Netrina/fisiología , Uniones Adherentes/metabolismo , Secuencias de Aminoácidos , Animales , Animales Modificados Genéticamente , Proteínas del Dominio Armadillo/metabolismo , Proteínas de Ciclo Celular , Movimiento Celular , Polaridad Celular , Forma de la Célula , Extensiones de la Superficie Celular/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Discos Imaginales/citología , Integrinas/fisiología , Larva , Miosinas/metabolismo , Receptores de Netrina/química , Receptores de Netrina/genética , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Transgenes , Proteínas de Unión al GTP rac/fisiología , Proteínas de Unión al GTP rho/fisiología
10.
Am J Hematol ; 93(2): 269-276, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29124783

RESUMEN

NET formation in mice (NETosis) is supported by reactive oxygen species (ROS) production by NADPH oxidase and histone hypercitrullination by peptidylarginine deiminase 4 (PAD4). Rac1 and Rac2, expressed in polymorphonuclear neutrophils (PMNs), regulate the cytoskeleton, cell shape, adhesion, and migration and are also essential components of the NADPH oxidase complex. We aimed to explore the role of the Rac signaling pathway including the upstream guanosine exchange factor (GEF) activator, Vav, and a downstream effector, the p21-activated kinase, Pak, on NETosis in PMNs using a previously described flow-cytometry-based assay. Rac2-/- PMNs showed reduced levels of citrullinated histone H3 (H3Cit)-positive cells and defective NETosis. Rac1Δ/Δ ; Rac2-/- PMNs demonstrated a further reduction in PMA-induced H3Cit levels and a more profound impairment of NETosis than deletion of Rac2 alone, suggesting an overlapping role of these two highly related proteins. Genetic knockouts of Vav1, or Vav2, did not impair H3Cit response to phorbol myristate ester (PMA) or NETosis. Combined, Vav1 and Vav3 deletions decreased H3Cit response and caused a modest but significant impairment of NETosis. Pharmacologic inhibition of Pak by two inhibitors with distinct mechanisms of action, led to reduced H3Cit levels after PMA stimulation, as well as significant inhibition of NETosis. We validated the importance of Pak using Pak2Δ/Δ PMNs, which demonstrated significantly impaired histone H3 citrullination and NETosis. These data confirm and more comprehensively define the key role of the Rac signaling pathway in PMN NETosis. The Rac signaling cascade may represent a valuable target for inhibition of NETosis and related pathological processes.


Asunto(s)
Trampas Extracelulares/metabolismo , Transducción de Señal , Quinasas p21 Activadas/fisiología , Proteínas de Unión al GTP rac/fisiología , Animales , Citrulinación , Histonas/metabolismo , Ratones , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Quinasas p21 Activadas/metabolismo , Proteínas de Unión al GTP rac/metabolismo
11.
Development ; 144(10): 1863-1875, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28512198

RESUMEN

Rac signaling impacts a relatively large number of downstream targets; however, few studies have established an association between Rac pathways and pathological conditions. In the present study, we generated mice with double knockout of Rac1 and Rac3 (Atoh1-Cre;Rac1flox/flox;Rac3-/- ) in cerebellar granule neurons (CGNs). We observed impaired tangential migration at E16.5, as well as numerous apoptotic CGNs at the deepest layer of the external granule layer (EGL) in the medial cerebellum of Atoh1-Cre;Rac1flox/flox;Rac3-/- mice at P8. Atoh1-Cre;Rac1flox/flox;Rac3-/- CGNs differentiated normally until expression of p27kip1 and NeuN in the deep EGL at P5. Primary CGNs and cerebellar microexplants from Atoh1-Cre;Rac1flox/flox;Rac3-/- mice exhibited impaired neuritogenesis, which was more apparent in Map2-positive dendrites. Such findings suggest that impaired tangential migration and final differentiation of CGNs have resulted in decreased cerebellum size and agenesis of the medial internal granule layer, respectively. Furthermore, Rac depleted/deleted cells exhibited decreased levels of Mid1 and impaired mTORC1 signaling. Mid1 depletion in CGNs produced mild impairments in neuritogenesis and reductions in mTORC1 signaling. Thus, a novel Rac-signaling pathway (Rac1-Mid1-mTORC1) may be involved in medial cerebellar development.


Asunto(s)
Cerebelo/embriología , Proteínas/fisiología , Proteínas de Unión al GTP rac/fisiología , Animales , Diferenciación Celular/genética , Células Cultivadas , Cerebelo/metabolismo , Células HEK293 , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Complejos Multiproteicos/fisiología , Neurogénesis/genética , Organogénesis/genética , Proteínas/genética , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/fisiología , Ubiquitina-Proteína Ligasas , Proteínas de Unión al GTP rac/genética
12.
Mol Microbiol ; 104(3): 487-498, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28164413

RESUMEN

Rac proteins are involved in a variety of cellular processes. Effector proteins that interact with active Rac convey the GTPase-generated signal to downstream developmental cascades and processes. Here we report on the analysis of the main effector and signal cascade downstream of BcRac, the Rac homolog of the grey mold fungus Botrytis cinerea. Several lines of evidence highlighted the p21-activated kinase Cla4 as an important effector of Rac in fungi. Analysis of Δbccla4 strains revealed that the BcCla4 protein was sufficient to mediate all of the examined BcRac-driven processes, including hyphal growth and morphogenesis, conidia production and pathogenicity. In addition, the Δbccla4 strains had altered nuclei content, a phenomenon that was previously observed in Δbcrac isolates, thus connecting the BcRac/BcCla4 module with cell cycle control. Further analyses revealed that BcRac/BcCla4 control mitotic entry through changes in phosphorylation status of the cyclin dependent kinase BcCdk1. The complete cascade includes the kinase BcWee1, which is downstream of BcCla4 and upstream of BcCdk1. These results provide a mechanistic insight on the connection of cell cycle, morphogenesis and pathogenicity in fungi, and position BcCla4 as the most essential effector and central regulator of all of these processes downstream of BcRac.


Asunto(s)
Botrytis/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas de Unión al GTP rac/fisiología , Botrytis/enzimología , Botrytis/crecimiento & desarrollo , Botrytis/metabolismo , Ciclo Celular/fisiología , División Celular/fisiología , Proteínas Fúngicas/metabolismo , Morfogénesis , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Esporas Fúngicas/metabolismo , Quinasas p21 Activadas/metabolismo , Proteínas de Unión al GTP rac/metabolismo
13.
Bioessays ; 38(12): 1246-1254, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27790724

RESUMEN

Small GTPases in the Rho family act as major nodes with functions beyond cytoskeletal rearrangements shaping the Caenorhabditis elegans embryo during development. These small GTPases are key signal transducers that integrate diverse developmental signals to produce a coordinated response in the cell. In C. elegans, the best studied members of these highly conserved Rho family small GTPases, RHO-1/RhoA, CED-10/Rac, and CDC-42, are crucial in several cellular processes dealing with cytoskeletal reorganization. In this review, we update the functions described for the Rho family small GTPases in spindle orientation and cell division, engulfment, and cellular movements during C. elegans embryogenesis, focusing on the Rho subfamily Rac. Please also see the video abstract here.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Caenorhabditis elegans/embriología , Proteínas de Ciclo Celular/fisiología , Citoesqueleto/metabolismo , Proteínas de Unión al GTP/fisiología , Proteínas de Unión al GTP rac/fisiología , Proteínas de Unión al GTP rho/fisiología , Animales , Caenorhabditis elegans/metabolismo , Transducción de Señal
14.
Tumour Biol ; 37(9): 12597-12607, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27402308

RESUMEN

Lung cancer is still the leading cause of malignant deaths in the world. It is of great importance to find novel functional genes for the tumorigenesis of lung cancer. We demonstrated that Rac3 could promote cell proliferation and inhibit apoptosis in lung adenocarcinoma cell line A549 previously. The aim of this study was to investigate the function and mechanism of Rac3 in lung adenocarcinoma cell lines. Immunohistochemistry staining was performed in 107 lung adenocarcinoma tissues and matched non-tumor tissues. Multivariate analysis and Kaplan-Meier analysis were used to investigate the correlation between Rac3 expression and the clinical outcomes. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, colony formation assay, and flow cytometry analysis were employed to determine the proliferative ability, cell cycle distribution, and apoptosis in H1299 and H1975 cell lines. Gene expression microarray and pathway analysis between the Rac3-siRNA group and the control group in A549 cells were performed to investigate the pathways and mechanism of Rac3 regulation. Rac3 was shown to be positively expressed in lung adenocarcinoma tissues, and the expression of Rac3 associates with longer survival in lung adenocarcinoma patients. Silencing of Rac3 significantly induced cell growth inhibition, colony formation decrease, cell cycle arrest, and apoptosis of lung adenocarcinoma cell lines, which accompanied by obvious downregulation of CCND1, MYC, and TFDP1 of cell cycle pathway involving in the tumorigenesis of lung adenocarcinoma based on the gene expression microarray. In conclusion, these findings suggest that Rac3 has the potential of being a therapeutic target for lung adenocarcinoma.


Asunto(s)
Adenocarcinoma/patología , Neoplasias Pulmonares/patología , Proteínas de Unión al GTP rac/fisiología , Adenocarcinoma/mortalidad , Adenocarcinoma del Pulmón , Adulto , Anciano , Apoptosis , Puntos de Control del Ciclo Celular , Proliferación Celular , Ciclina D1/genética , Femenino , Humanos , Neoplasias Pulmonares/mortalidad , Masculino , Persona de Mediana Edad , Pronóstico , Proteínas Proto-Oncogénicas c-myc/genética , Factor de Transcripción DP1/genética , Proteínas de Unión al GTP rac/análisis
15.
Cell Death Dis ; 6: e1902, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26469953

RESUMEN

Receptor-associated coactivator 3 (RAC3) is a nuclear receptor coactivator usually overexpressed in tumors that exerts oncogenic functions in the cytoplasm and the nucleus. Although as part of its oncogenic actions it was previously identified as an inhibitor of apoptosis and autophagy, its expression is required in order to preserve the pluripotency and embryonic stem cell self-renewal. In this work we investigated its role in cellular senescence. We found that RAC3 overexpression in the nontumoral HEK293 cells inhibits the premature senescence induced by hydrogen peroxide or rapamycin. The mechanism involves not only the inhibition of autophagy early induced by these stimuli in the pathway to senescence, but also the increase in levels and nuclear localization of both the cell cycle suppressors p53/p21 and the longevity promoters FOXO1A, FOXO3A and SIRT1. Furthermore, we found that RAC3 overexpression is required in order to maintain the telomerase activity. In tumoral HeLa cells its activity was inhibited by depletion of RAC3 inducing replicative senescence. Moreover, we demonstrated that in vivo, levels of RAC3 are downregulated in the liver from aged as compared with young rats, whereas the levels of p21 are increased, correlating with the expected senescent cell contents in aged tissues. A similar downregulation of RAC3 was observed in the premature and replicative senescence of human fetal WI-38 cells and premature senescence of hepatocyte HepG2 cell line. Taken together, all these results demonstrate that RAC3 is an inhibitor of senescence whose downregulation in aged individuals could be probably a tumor suppressor mechanism, avoiding the clonal expansion of risky old cells having damaged DNA.


Asunto(s)
Proteínas de Unión al GTP rac/fisiología , Envejecimiento , Animales , Proliferación Celular , Senescencia Celular , Regulación hacia Abajo , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Humanos , Peróxido de Hidrógeno/farmacología , Ratas Wistar , Sirolimus/farmacología
16.
Proc Natl Acad Sci U S A ; 111(34): 12544-9, 2014 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-25099352

RESUMEN

Nervous system injury or disease leads to activation of glia, which govern postinjury responses in the nervous system. Axonal injury in Drosophila results in transcriptional up-regulation of the glial engulfment receptor Draper; there is extension of glial membranes to the injury site (termed activation), and then axonal debris is internalized and degraded. Loss of the small GTPase Rac1 from glia completely suppresses glial responses to injury, but upstream activators remain poorly defined. Loss of the Rac guanine nucleotide exchange factor (GEF) Crk/myoblast city (Mbc)/dCed-12 has no effect on glial activation, but blocks internalization and degradation of debris. Here we show that the signaling molecules downstream of receptor kinase (DRK) and daughter of sevenless (DOS) (mammalian homologs, Grb2 and Gab2, respectively) and the GEF son of sevenless (SOS) (mammalian homolog, mSOS) are required for efficient activation of glia after axotomy and internalization/degradation of axonal debris. At the earliest steps of glial activation, DRK/DOS/SOS function in a partially redundant manner with Crk/Mbc/dCed-12, with blockade of both complexes strongly suppressing all glial responses, similar to loss of Rac1. This work identifies DRK/DOS/SOS as the upstream Rac GEF complex required for glial responses to axonal injury, and demonstrates a critical requirement for multiple GEFs in efficient glial activation after injury and internalization/degradation of axonal debris.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila melanogaster/citología , Drosophila melanogaster/fisiología , Proteínas del Ojo/fisiología , Neuroglía/fisiología , Proteína Son Of Sevenless Drosofila/fisiología , Proteínas de Unión al GTP rac/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Animales Modificados Genéticamente , Axones/fisiología , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/fisiología , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas del Ojo/genética , Genes de Insecto , Mutación , Degeneración Nerviosa , Fagosomas/fisiología , Proteínas Proto-Oncogénicas c-crk/genética , Proteínas Proto-Oncogénicas c-crk/fisiología , Proteína Son Of Sevenless Drosofila/genética , Proteínas de Unión al GTP rac/genética , Proteínas ras/genética , Proteínas ras/fisiología
17.
PLoS One ; 9(4): e95893, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24770346

RESUMEN

Although it is well-established that the macrophage M1 to M2 transition plays a role in tumor progression, the molecular basis for this process remains incompletely understood. Herein, we demonstrate that the small GTPase, Rac2 controls macrophage M1 to M2 differentiation and the metastatic phenotype in vivo. Using a genetic approach, combined with syngeneic and orthotopic tumor models we demonstrate that Rac2-/- mice display a marked defect in tumor growth, angiogenesis and metastasis. Microarray, RT-PCR and metabolomic analysis on bone marrow derived macrophages isolated from the Rac2-/- mice identify an important role for Rac2 in M2 macrophage differentiation. Furthermore, we define a novel molecular mechanism by which signals transmitted from the extracellular matrix via the α4ß1 integrin and MCSF receptor lead to the activation of Rac2 and potentially regulate macrophage M2 differentiation. Collectively, our findings demonstrate a macrophage autonomous process by which the Rac2 GTPase is activated downstream of the α4ß1 integrin and the MCSF receptor to control tumor growth, metastasis and macrophage differentiation into the M2 phenotype. Finally, using gene expression and metabolomic data from our Rac2-/- model, and information related to M1-M2 macrophage differentiation curated from the literature we executed a systems biologic analysis of hierarchical protein-protein interaction networks in an effort to develop an iterative interactome map which will predict additional mechanisms by which Rac2 may coordinately control macrophage M1 to M2 differentiation and metastasis.


Asunto(s)
Diferenciación Celular , Neoplasias Pulmonares/enzimología , Macrófagos/fisiología , Melanoma Experimental/enzimología , Neovascularización Patológica/enzimología , Proteínas de Unión al GTP rac/fisiología , Animales , Línea Celular Tumoral , Activación Enzimática , Integrina alfa4beta1/metabolismo , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/secundario , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Invasividad Neoplásica , Trasplante de Neoplasias , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Transducción de Señal , Carga Tumoral , Proteína RCA2 de Unión a GTP
18.
Gene ; 544(2): 241-7, 2014 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-24769554

RESUMEN

Cell polarity is fundamentally important to growth and development in higher plants, from pollen tubes to root hairs. Basal land plants (mosses and ferns) also have cell polarity, developing protonemal apical cells that show polar tip growth. Flowering plants have a distinct group of Rho GTPases that regulate polarity in polarized cell growth. Rop/RAC signaling module components have been identified in non-flowering plants, but their roles remain unclear. To understand the importance and evolution of Rop/RAC signaling in polarity regulation in land plants, we examined the functions of PpRop and PpRopGEF in protonemal apical cells of the moss Physcomitrella patens. Inducible overexpression of PpRop2 or PpRopGEF3 caused depolarized growth of tip-growing apical cells. PpRop2 overexpression also caused aberrant cross wall formation. Fluorescent protein-tagged PpRop2 localized to the plasma membrane, including the cross wall membrane, and fluorescent-tagged PpRopGEF3 showed polarized localization to the tip region in apical cells. Thus, our results suggest common functions of PpRop and PpRopGEF in the tip-growing apical cells and the importance of a conserved Rop/RAC signaling module in the control of cell polarity in land plants.


Asunto(s)
Bryopsida/citología , Bryopsida/genética , Polaridad Celular/genética , Proteínas de Unión al GTP rac/fisiología , Secuencia de Aminoácidos , Plantas Modificadas Genéticamente , Alineación de Secuencia , Transducción de Señal/genética , Proteínas de Unión al GTP rac/genética
19.
Proc Natl Acad Sci U S A ; 110(49): E4723-32, 2013 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-24248334

RESUMEN

During chemotaxis, cells sense extracellular chemical gradients and position Ras GTPase activation and phosphatidylinositol (3,4,5)-triphosphate (PIP3) production toward chemoattractants. These two major signaling events are visualized by biosensors in a crescent-like zone at the plasma membrane. Here, we show that a Dictyostelium Rho GTPase, RacE, and a guanine nucleotide exchange factor, GxcT, stabilize the orientation of Ras activation and PIP3 production in response to chemoattractant gradients, and this regulation occurred independently of the actin cytoskeleton and cell polarity. Cells lacking RacE or GxcT fail to persistently direct Ras activation and PIP3 production toward chemoattractants, leading to lateral pseudopod extension and impaired chemotaxis. Constitutively active forms of RacE and human RhoA are located on the portion of the plasma membrane that faces lower concentrations of chemoattractants, opposite of PIP3 production. Mechanisms that control the localization of the constitutively active form of RacE require its effector domain, but not PIP3. Our findings reveal a critical role for Rho GTPases in positioning Ras activation and thereby establishing the accuracy of directional sensing.


Asunto(s)
Factores Quimiotácticos/metabolismo , Quimiotaxis/fisiología , Fosfatos de Fosfatidilinositol/metabolismo , Transducción de Señal/fisiología , Proteínas de Unión al GTP rac/metabolismo , Southern Blotting , Técnicas de Inactivación de Genes , Factores de Intercambio de Guanina Nucleótido/metabolismo , Microscopía Confocal , Proteínas de Unión al GTP rac/fisiología
20.
J Clin Invest ; 123(10): 4449-63, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24091327

RESUMEN

An acquired somatic mutation at codon 816 in the KIT receptor tyrosine kinase is associated with poor prognosis in patients with systemic mastocytosis and acute myeloid leukemia (AML). Treatment of leukemic cells bearing this mutation with an allosteric inhibitor of p21-activated kinase (Pak) or its genetic inactivation results in growth repression due to enhanced apoptosis. Inhibition of the upstream effector Rac abrogates the oncogene-induced growth and activity of Pak. Although both Rac1 and Rac2 are constitutively activated via the guanine nucleotide exchange factor (GEF) Vav1, loss of Rac1 or Rac2 alone moderately corrected the growth of KIT-bearing leukemic cells, whereas the combined loss resulted in 75% growth repression. In vivo, the inhibition of Vav or Rac or Pak delayed the onset of myeloproliferative neoplasms (MPNs) and corrected the associated pathology in mice. To assess the role of Rac GEFs in oncogene-induced transformation, we used an inhibitor of Rac, EHop-016, which specifically targets Vav1 and found that EHop-016 was a potent inhibitor of human and murine leukemic cell growth. These studies identify Pak and Rac GTPases, including Vav1, as potential therapeutic targets in MPN and AML involving an oncogenic form of KIT.


Asunto(s)
Aminoquinolinas/farmacología , Antineoplásicos/farmacología , Carbazoles/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-kit/genética , Pirimidinas/farmacología , Quinasas p21 Activadas/fisiología , Proteínas de Unión al GTP rac/fisiología , Animales , Carcinogénesis/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Activación Enzimática , Humanos , Mastocitosis/tratamiento farmacológico , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Mutación Missense , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Proto-Oncogénicas c-vav/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasas p21 Activadas/antagonistas & inhibidores , Proteínas de Unión al GTP rac/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA