Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
Más filtros

Intervalo de año de publicación
1.
Nat Rev Mol Cell Biol ; 12(8): 493-504, 2011 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-21779026

RESUMEN

The 'invisible hand' is a term originally coined by Adam Smith in The Theory of Moral Sentiments to describe the forces of self-interest, competition and supply and demand that regulate the resources in society. This metaphor continues to be used by economists to describe the self-regulating nature of a market economy. The same metaphor can be used to describe the RHO-specific guanine nucleotide dissociation inhibitor (RHOGDI) family, which operates in the background, as an invisible hand, using similar forces to regulate the RHO GTPase cycle.


Asunto(s)
Inhibidores de Disociación de Guanina Nucleótido/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Evolución Molecular , Inhibidores de Disociación de Guanina Nucleótido/química , Inhibidores de Disociación de Guanina Nucleótido/genética , Humanos , Microdominios de Membrana/metabolismo , Redes y Vías Metabólicas , Modelos Biológicos , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Complejos Multiproteicos , Neoplasias/metabolismo , Fosforilación , Filogenia , Dominios y Motivos de Interacción de Proteínas , Proteínas de Unión al GTP rho/química , Inhibidor alfa de Disociación del Nucleótido Guanina rho , Inhibidores de la Disociación del Nucleótido Guanina rho-Específico
2.
Bioessays ; 41(1): e1800165, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30485463

RESUMEN

Focal adhesions disassemble during mitosis, but surprisingly little is known about how these structures respond to other phases of the cell cycle. Three recent papers reveal unexpected results as they examine adhesions through the cell cycle. A biphasic response is detected where focal adhesions grow during S phase before disassembly begins early in G2. In M phase, activated integrins at the tips of retraction fibers anchor mitotic cells, but these adhesions lack the defining components of focal adhesions, such as talin, paxillin, and zyxin. Re-examining cell-matrix adhesion reveals reticular adhesions, a new class of adhesion. These αVß5 integrin-mediated adhesions also lack conventional focal adhesion components and anchor mitotic cells to the extracellular matrix. As reviewed here, these studies present insight into how adhesion complexes vary through the cell cycle, and how unconventional adhesions maintain attachment during mitosis while providing spatial memory to guide daughter cell re-spreading after cell division.


Asunto(s)
Puntos de Control del Ciclo Celular , Adhesiones Focales/fisiología , Animales , Adhesión Celular , Adhesiones Focales/metabolismo , Humanos
3.
Cell Commun Signal ; 18(1): 129, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32811537

RESUMEN

BACKGROUND: Syndecans regulate cell migration thus having key roles in scarring and wound healing processes. Our previous results have shown that Thy-1/CD90 can engage both αvß3 integrin and Syndecan-4 expressed on the surface of astrocytes to induce cell migration. Despite a well-described role of Syndecan-4 during cell movement, information is scarce regarding specific Syndecan-4 partners involved in Thy-1/CD90-stimulated cell migration. METHODS: Mass spectrometry (MS) analysis of complexes precipitated with the Syndecan-4 cytoplasmic tail peptide was used to identify potential Syndecan-4-binding partners. The interactions found by MS were validated by immunoprecipitation and proximity ligation assays. The conducted research employed an array of genetic, biochemical and pharmacological approaches, including: PAR-3, Syndecan-4 and Tiam1 silencing, active Rac1 GEFs affinity precipitation, and video microscopy. RESULTS: We identified PAR-3 as a Syndecan-4-binding protein. Its interaction depended on the carboxy-terminal EFYA sequence present on Syndecan-4. In astrocytes where PAR-3 expression was reduced, Thy-1-induced cell migration and focal adhesion disassembly was impaired. This effect was associated with a sustained Focal Adhesion Kinase activation in the siRNA-PAR-3 treated cells. Our data also show that Thy-1/CD90 activates Tiam1, a PAR-3 effector. Additionally, we found that after Syndecan-4 silencing, Tiam1 activation was decreased and it was no longer recruited to the membrane. Syndecan-4/PAR-3 interaction and the alteration in focal adhesion dynamics were validated in mouse embryonic fibroblast (MEF) cells, thereby identifying this novel Syndecan-4/PAR-3 signaling complex as a general mechanism for mesenchymal cell migration involved in Thy-1/CD90 stimulation. CONCLUSIONS: The newly identified Syndecan-4/PAR-3 signaling complex participates in Thy-1/CD90-induced focal adhesion disassembly in mesenchymal cells. The mechanism involves focal adhesion kinase dephosphorylation and Tiam1 activation downstream of Syndecan-4/PAR-3 signaling complex formation. Additionally, PAR-3 is defined here as a novel adhesome-associated component with an essential role in focal adhesion disassembly during polarized cell migration. These novel findings uncover signaling mechanisms regulating cell migration, thereby opening up new avenues for future research on Syndecan-4/PAR-3 signaling in processes such as wound healing and scarring.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Adhesiones Focales/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal , Sindecano-4/metabolismo , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Adhesión Celular , Línea Celular , Movimiento Celular , Polaridad Celular , Fibroblastos/metabolismo , Silenciador del Gen , Ratones , Microtúbulos/metabolismo , Unión Proteica , Ratas , Antígenos Thy-1/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 38(10): 2410-2422, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30354217

RESUMEN

Objective- Maintenance of lymphatic permeability is essential for normal lymphatic function during adulthood, but the precise signaling pathways that control lymphatic junctions during development are not fully elucidated. The Gs-coupled AM (adrenomedullin) signaling pathway is required for embryonic lymphangiogenesis and the maintenance of lymphatic junctions during adulthood. Thus, we sought to elucidate the downstream effectors mediating junctional stabilization in lymphatic endothelial cells. Approach and Results- We knocked-down both Rap1A and Rap1B isoforms in human neonatal dermal lymphatic cells (human lymphatic endothelial cells) and genetically deleted the mRap1 gene in lymphatic endothelial cells by producing 2 independent, conditional Rap1a/b knockout mouse lines. Rap1A/B knockdown caused disrupted junctional formation with hyperpermeability and impaired AM-induced lymphatic junctional tightening, as well as rescue of histamine-induced junctional disruption. Less than 60% of lymphatic- Rap1a/b knockout embryos survived to E13.5 exhibiting interstitial edema, blood-filled lymphatics, disrupted lymphovenous valves, and defective lymphangiogenesis. Consistently, inducible lymphatic- Rap1a/b deletion in adult animals prevented AM-rescue of histamine-induced lymphatic leakage and dilation. Conclusions- Rap1 (Ras-related protein) serves as the dominant effector downstream of AM to stabilize lymphatic junctions. Rap1 is required for maintaining lymphatic permeability and driving normal lymphatic development.


Asunto(s)
Adrenomedulina/farmacología , Células Endoteliales/efectos de los fármacos , Endotelio Linfático/efectos de los fármacos , Uniones Intercelulares/efectos de los fármacos , Linfangiogénesis/efectos de los fármacos , Proteínas de Unión al GTP rap/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Animales , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Endoteliales/enzimología , Células Endoteliales/patología , Endotelio Linfático/enzimología , Endotelio Linfático/patología , Histamina/farmacología , Humanos , Uniones Intercelulares/enzimología , Uniones Intercelulares/patología , Ratones , Ratones Noqueados , Permeabilidad , Transducción de Señal , Proteínas de Unión al GTP rap/genética , Proteínas de Unión al GTP rap1/genética
5.
J Immunol ; 198(12): 4823-4836, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28484055

RESUMEN

Inflammation is driven by excessive transmigration (diapedesis) of leukocytes from the blood to the tissue across the endothelial cell monolayer that lines blood vessels. Leukocyte adhesion, crawling, and transmigration are regulated by clustering of the endothelial mechanosensitive receptor intercellular adhesion molecule-1 (ICAM-1). Whereas several proteins are known to promote ICAM-1 function, the molecular mechanisms that limit ICAM-1-mediated adhesion to prevent excessive leukocyte transmigration remain unknown. We identify the endothelial actin-binding protein CD2-associated protein (CD2AP) as a novel interaction partner of ICAM-1. Loss of CD2AP stimulates the dynamics of ICAM-1 clustering, which facilitates the formation of ICAM-1 complexes on the endothelial cell surface. Consequently, neutrophil adhesion is increased, but crawling is decreased. In turn, this promotes the neutrophil preference for the transcellular over the paracellular transmigration route. Mechanistically, CD2AP is required for mechanosensitive ICAM-1 downstream signaling toward activation of the PI3K, and recruitment of F-actin and of the actin-branching protein cortactin. Moreover, CD2AP is necessary for ICAM-1-induced Rac1 recruitment and activation. Mechanical force applied on ICAM-1 impairs CD2AP binding to ICAM-1, suggesting that a tension-induced negative feedback loop promotes ICAM-1-mediated neutrophil crawling and paracellular transmigration. To our knowledge, these data show for the first time that the mechanoreceptor ICAM-1 is negatively regulated by an actin-binding adaptor protein, i.e., CD2AP, to allow a balanced and spatiotemporal control of its adhesive function. CD2AP is important in kidney dysfunction that is accompanied by inflammation. Our findings provide a mechanistic basis for the role of CD2AP in inflamed vessels, identifying this adaptor protein as a potential therapeutic target.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Adhesión Celular , Proteínas del Citoesqueleto/fisiología , Molécula 1 de Adhesión Intercelular/metabolismo , Leucocitos/fisiología , Neutrófilos/fisiología , Transducción de Señal , Migración Transendotelial y Transepitelial , Citoesqueleto de Actina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/inmunología , Movimiento Celular , Proteínas del Citoesqueleto/inmunología , Endotelio Vascular/química , Endotelio Vascular/metabolismo , Humanos , Técnicas In Vitro , Molécula 1 de Adhesión Intercelular/inmunología , Leucocitos/inmunología , Leucocitos/metabolismo , Neutrófilos/inmunología
6.
J Immunol ; 197(9): 3504-3511, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27694496

RESUMEN

The use of nondepleting Abs specific for CD4 and CD8 is an effective strategy to tolerize CD4+ and CD8+ T cells in a tissue-specific manner. We reported that coreceptor therapy reverses diabetes in new onset NOD mice. A striking feature of coreceptor-induced remission is the purging of T cells from the pancreatic lymph nodes (PLN) and islets of NOD mice. Evidence indicates that Abs binding to the coreceptors promotes T cell egress from these tissues. The present study examined how coreceptor therapy affects the migration of CD4+ T cells residing in the PLN of NOD mice. Anti-CD4 Ab treatment resulted in an increased frequency of PLN but not splenic CD4+ T cells that exhibited a polarized morphology consistent with a migratory phenotype. Furthermore, PLN CD4+ T cells isolated from anti-CD4 versus control Ab-treated animals displayed increased in vitro chemotaxis to chemoattractants such as sphingosine-1-phosphate and CXCL12. Notably, the latter was dependent on activation of the small Rho GTPases Rac1 and Rac2. Rac1 and Rac2 activation was increased in Ab-bound CD4+ T cells from the PLN but not the spleen, and knockdown of Rac expression blocked the heightened reactivity of Ab-bound PLN CD4+ T cells to CXCL12. Interestingly, Rac1 and Rac2 activation was independent of Rac guanine nucleotide exchange factors known to regulate T cell activity. Therefore, Ab binding to CD4 initiates a novel pathway that involves inflammation-dependent activation of Rac and establishment of altered T cell migratory properties.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Diabetes Mellitus/terapia , Neuropéptidos/metabolismo , Linfocitos T/efectos de los fármacos , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Antígenos CD4/inmunología , Antígenos CD8/inmunología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL12/metabolismo , Diabetes Mellitus/inmunología , Femenino , Humanos , Tolerancia Inmunológica , Activación de Linfocitos , Lisofosfolípidos/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Neuropéptidos/genética , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Linfocitos T/inmunología , Proteínas de Unión al GTP rac/genética , Proteína de Unión al GTP rac1/genética , Proteína RCA2 de Unión a GTP
7.
Exp Cell Res ; 343(1): 14-20, 2016 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-26519907

RESUMEN

Stress fibers and focal adhesions are complex protein arrays that produce, transmit and sense mechanical tension. Evidence accumulated over many years led to the conclusion that mechanical tension generated within stress fibers contributes to the assembly of both stress fibers themselves and their associated focal adhesions. However, several lines of evidence have recently been presented against this model. Here we discuss the evidence for and against the role of mechanical tension in driving the assembly of these structures. We also consider how their assembly is influenced by the rigidity of the substratum to which cells are adhering. Finally, we discuss the recently identified connections between stress fibers and the nucleus, and the roles that these may play, both in cell migration and regulating nuclear function.


Asunto(s)
Adhesiones Focales/fisiología , Fibras de Estrés/fisiología , Estrés Mecánico , Animales , Humanos , Modelos Biológicos
8.
Stem Cells ; 33(6): 2063-76, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25787126

RESUMEN

A cell's ability to recognize and adapt to the physical environment is central to its survival and function, but how mechanical cues are perceived and transduced into intracellular signals remains unclear. In mesenchymal stem cells (MSCs), high-magnitude substrate strain (HMS, ≥2%) effectively suppresses adipogenesis via induction of focal adhesion (FA) kinase (FAK)/mTORC2/Akt signaling generated at FAs. Physiologic systems also rely on a persistent barrage of low-level signals to regulate behavior. Exposing MSC to extremely low-magnitude mechanical signals (LMS) suppresses adipocyte formation despite the virtual absence of substrate strain (<0.001%), suggesting that LMS-induced dynamic accelerations can generate force within the cell. Here, we show that MSC response to LMS is enabled through mechanical coupling between the cytoskeleton and the nucleus, in turn activating FAK and Akt signaling followed by FAK-dependent induction of RhoA. While LMS and HMS synergistically regulated FAK activity at the FAs, LMS-induced actin remodeling was concentrated at the perinuclear domain. Preventing nuclear-actin cytoskeleton mechanocoupling by disrupting linker of nucleoskeleton and cytoskeleton (LINC) complexes inhibited these LMS-induced signals as well as prevented LMS repression of adipogenic differentiation, highlighting that LINC connections are critical for sensing LMS. In contrast, FAK activation by HMS was unaffected by LINC decoupling, consistent with signal initiation at the FA mechanosome. These results indicate that the MSC responds to its dynamic physical environment not only with "outside-in" signaling initiated by substrate strain, but vibratory signals enacted through the LINC complex enable matrix independent "inside-inside" signaling.


Asunto(s)
Núcleo Celular/metabolismo , Citoesqueleto/metabolismo , Células Madre Mesenquimatosas/citología , Adipocitos/metabolismo , Adipogénesis/fisiología , Animales , Células Cultivadas , Humanos , Ratones Endogámicos C57BL
9.
J Immunol ; 192(7): 3390-8, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24585879

RESUMEN

RhoA-mediated cytoskeletal rearrangements in endothelial cells (ECs) play an active role in leukocyte transendothelial cell migration (TEM), a normal physiological process in which leukocytes cross the endothelium to enter the underlying tissue. Although much has been learned about RhoA signaling pathways downstream from ICAM-1 in ECs, little is known about the consequences of the tractional forces that leukocytes generate on ECs as they migrate over the surface before TEM. We have found that after applying mechanical forces to ICAM-1 clusters, there is an increase in cellular stiffening and enhanced RhoA signaling compared with ICAM-1 clustering alone. We have identified that leukemia-associated Rho guanine nucleotide exchange factor (LARG), also known as Rho GEF 12 (ARHGEF12) acts downstream of clustered ICAM-1 to increase RhoA activity, and that this pathway is further enhanced by mechanical force on ICAM-1. Depletion of LARG decreases leukocyte crawling and inhibits TEM. To our knowledge, this is the first report of endothelial LARG regulating leukocyte behavior and EC stiffening in response to tractional forces generated by leukocytes.


Asunto(s)
Células Endoteliales/inmunología , Molécula 1 de Adhesión Intercelular/inmunología , Mecanotransducción Celular/inmunología , Factores de Intercambio de Guanina Nucleótido Rho/inmunología , Migración Transendotelial y Transepitelial/inmunología , Western Blotting , Células Cultivadas , Citocalasina D/farmacología , Citoesqueleto/efectos de los fármacos , Citoesqueleto/inmunología , Citoesqueleto/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Recién Nacido , Molécula 1 de Adhesión Intercelular/metabolismo , Microscopía Fluorescente , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Interferencia de ARN , Factores de Intercambio de Guanina Nucleótido Rho/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Transducción de Señal/inmunología , Estrés Mecánico
10.
Mol Cell ; 32(1): 43-56, 2008 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-18851832

RESUMEN

Mixed lineage kinase 3 (MLK3) is a MAP3K that activates the JNK-dependent MAPK pathways. Here, we show that MLK3 is required for cell migration in a manner independent of its role as a MAP3K or MLK3 kinase activity. Rather, MLK3 functions in a regulated way to limit levels of the activated GTPase Rho by binding to the Rho activator, p63RhoGEF/GEFT, which, in turn, prevents its activation by Galphaq. These findings demonstrate a scaffolding role for MLK3 in controlling the extent of Rho activation that modulates cell migration. Moreover, they suggest that MLK3 functions as a network hub that links a number of signaling pathways.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Línea Celular , Movimiento Celular/fisiología , Citoesqueleto/ultraestructura , Adhesiones Focales/ultraestructura , Humanos , Técnicas In Vitro , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/genética , Sistema de Señalización de MAP Quinasas , Modelos Biológicos , Unión Proteica , Seudópodos/ultraestructura , ARN Interferente Pequeño/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho , Factor de Respuesta Sérica/metabolismo , Transducción de Señal , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno
11.
Biochemistry ; 53(34): 5526-36, 2014 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-25115937

RESUMEN

Vinculin is an essential structural adaptor protein that localizes to sites of adhesion and is involved in a number of cell processes including adhesion, spreading, motility, force transduction, and cell survival. The C-terminal vinculin tail domain (Vt) contains the necessary structural components to bind and cross-link actin filaments. Actin binding to Vt induces a conformational change that promotes dimerization through the C-terminal hairpin of Vt and enables actin filament cross-linking. Here we show that Src phosphorylation of Y1065 within the C-terminal hairpin regulates Vt-mediated actin bundling and provide a detailed characterization of Y1065 mutations. Furthermore, we show that phosphorylation at Y1065 plays a role in cell spreading and the response to the application of mechanical force.


Asunto(s)
Actinas/química , Vinculina/fisiología , Animales , Células Cultivadas , Dicroismo Circular , Espectroscopía de Resonancia Magnética , Ratones , Microscopía Fluorescente , Mutación , Fosforilación , Unión Proteica , Vinculina/química , Vinculina/genética
12.
Biochim Biophys Acta ; 1833(6): 1409-20, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23481656

RESUMEN

Cell adhesion to the extracellular matrix proteins occurs through interactions with integrins that bind to Arg-Gly-Asp (RGD) tripeptides, and syndecan-4, which recognizes the heparin-binding domain of other proteins. Both receptors trigger signaling pathways, including those that activate RhoGTPases such as RhoA and Rac1. This sequence of events modulates cell adhesion to the ECM and cell migration. Using a neuron-astrocyte model, we have reported that the neuronal protein Thy-1 engages αVß3 integrin and syndecan-4 to induce RhoA activation and strong astrocyte adhesion to their underlying substrate. Thus, because cell-cell interactions and strong cell attachment to the matrix are considered antagonistic to cell migration, we hypothesized that Thy-1 stimulation of astrocytes should preclude cell migration. Here, we studied the effect of Thy-1 expressing neurons on astrocyte polarization and migration using a wound-healing assay and immunofluorescence analysis. Signaling molecules involved were studied by affinity precipitation, western blotting and the usage of specific antibodies. Intriguingly, Thy-1 interaction with its two receptors was found to increase astrocyte polarization and migration. The latter events required interactions of these receptors with both the RGD-like sequence and the heparin-binding domain of Thy-1. Additionally, prolonged Thy-1-receptor interactions inhibited RhoA activation while activating FAK, PI3K and Rac1. Therefore, sustained engagement of integrin and syndecan-4 with the neuronal surface protein Thy-1 induces astrocyte migration. Interestingly we identify here, a cell-cell interaction that despite initially inducing strong cell attachment, favors cell migration upon persistent stimulation by engaging the same signaling receptors and molecules as those utilized by the extracellular matrix proteins to stimulate cell movement.


Asunto(s)
Astrocitos/citología , Comunicación Celular , Movimiento Celular/fisiología , Integrina alfaVbeta3/metabolismo , Oligopéptidos/metabolismo , Sindecano-4/metabolismo , Antígenos Thy-1/metabolismo , Animales , Astrocitos/metabolismo , Western Blotting , Adhesión Celular , Polaridad Celular , Proliferación Celular , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Ratas , Transducción de Señal , Cicatrización de Heridas , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
13.
Stem Cells ; 31(11): 2528-37, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23836527

RESUMEN

Mechanical strain provides an anti-adipogenic, pro-osteogenic stimulus to mesenchymal stem cells (MSC) through generating intracellular signals and via cytoskeletal restructuring. Recently, mTORC2 has been shown to be a novel mechanical target critical for the anti-adipogenic signal leading to preservation of ß-catenin. As mechanical activation of mTORC2 requires focal adhesions (FAs), we asked whether proximal signaling involved Src and FAK, which are early responders to integrin-FA engagement. Application of mechanical strain to marrow-derived MSCs was unable to activate mTORC2 when Src family kinases were inhibited. Fyn, but not Src, was specifically required for mechanical activation of mTORC2 and was recruited to FAs after strain. Activation of mTORC2 was further diminished following FAK inhibition, and as FAK phosphorylation (Tyr-397) required Fyn activity, provided evidence of Fyn/FAK cooperativity. Inhibition of Fyn also prevented mechanical activation of RhoA as well as mechanically induced actin stress fiber formation. We thus asked whether RhoA activation by strain was dependent on mTORC2 downstream of Fyn. Inhibition of mTORC2 or its downstream substrate, Akt, both prevented mechanical RhoA activation, indicating that Fyn/FAK affects cytoskeletal structure via mTORC2. We then sought to ascertain whether this Fyn-initiated signal pathway modulated MSC lineage decisions. siRNA knockdown of Fyn, but not Src, led to rapid attainment of adipogenic phenotype with significant increases in adipocyte protein 2, peroxisome proliferator-activated receptor gamma, adiponectin, and perilipin. As such, Fyn expression in mdMSCs contributes to basal cytoskeletal architecture and, when associated with FAs, functions as a proximal mechanical effector for environmental signals that influence MSC lineage allocation.


Asunto(s)
Adipogénesis/fisiología , Células Madre Mesenquimatosas/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Técnicas de Cultivo de Célula , Humanos , Células MCF-7 , Diana Mecanicista del Complejo 2 de la Rapamicina , Células Madre Mesenquimatosas/citología , Complejos Multiproteicos/genética , Fosforilación , Proteínas Proto-Oncogénicas c-fyn/genética , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Transfección
14.
Blood ; 118(19): 5344-54, 2011 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-21881052

RESUMEN

Kaposi sarcoma-associated herpesvirus (KSHV) is associated with 3 different human malignancies: Kaposi sarcoma (KS), primary effusion lymphoma, and multicentric Castleman disease. The KS lesion is driven by KSHV-infected endothelial cells and is highly dependent on autocrine and paracrine factors for survival and growth. We report that latent KSHV infection increases the vascular permeability of endothelial cells. Endothelial cells with latent KSHV infection display increased Rac1 activation and activation of its downstream modulator, p21-activated kinase 1 (PAK1). The KSHV-infected cells also exhibit increases in tyrosine phosphorylation of vascular endothelial (VE)-cadherin and ß-catenin, whereas total levels of these proteins remained unchanged, suggesting that latent infection disrupted endothelial cell junctions. Consistent with these findings, we found that KSHV-infected endothelial cells displayed increased permeability compared with uninfected endothelial cells. Knockdown of Rac1 and inhibition of reactive oxygen species (ROS) resulted in decreased permeability in the KSHV-infected endothelial cells. We further demonstrate that the KSHV K1 protein can activate Rac1. Rac1 was also highly activated in KSHV-infected endothelial cells and KS tumors. In conclusion, KSHV latent infection increases Rac1 and PAK1 activity in endothelial cells, resulting in the phosphorylation of VE-cadherin and ß-catenin and leading to the disassembly of cell junctions and to increased vascular permeability of the infected endothelial cells.


Asunto(s)
Permeabilidad Capilar , Infecciones por Herpesviridae/fisiopatología , Herpesvirus Humano 8/patogenicidad , Antígenos CD/metabolismo , Secuencia de Bases , Cadherinas/metabolismo , Células Endoteliales/fisiología , Activación Enzimática , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Fosforilación , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , beta Catenina/metabolismo , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/fisiología
15.
Biochemistry ; 51(38): 7420-32, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-22931484

RESUMEN

Throughout their lives, all cells constantly experience and respond to various mechanical forces. These frequently originate externally but can also arise internally as a result of the contractile actin cytoskeleton. Mechanical forces trigger multiple signaling pathways. Several converge and result in the activation of the GTPase RhoA. In this review, we focus on the pathways by which mechanical force leads to RhoA regulation, especially when force is transmitted via cell adhesion molecules that mediate either cell-matrix or cell-cell interactions. We discuss both the upstream signaling events that lead to activation of RhoA and the downstream consequences of this pathway. These include not only cytoskeletal reorganization and, in a positive feedback loop, increased myosin-generated contraction but also profound effects on gene expression and differentiation.


Asunto(s)
Proteína de Unión al GTP rhoA/química , Moléculas de Adhesión Celular/química , Matriz Extracelular/química , Adhesiones Focales , Expresión Génica , Humanos , Neoplasias/patología
16.
J Biol Chem ; 286(45): 39236-46, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-21930699

RESUMEN

The single-celled human parasite Entamoeba histolytica possesses a dynamic actin cytoskeleton vital for its intestinal and systemic pathogenicity. The E. histolytica genome encodes several Rho family GTPases known to regulate cytoskeletal dynamics. EhRho1, the first family member identified, was reported to be insensitive to the Rho GTPase-specific Clostridium botulinum C3 exoenzyme, raising the possibility that it may be a misclassified Ras family member. Here, we report the crystal structures of EhRho1 in both active and inactive states. EhRho1 is activated by a conserved switch mechanism, but diverges from mammalian Rho GTPases in lacking a signature Rho insert helix. EhRho1 engages a homolog of mDia, EhFormin1, suggesting a role in mediating serum-stimulated actin reorganization and microtubule formation during mitosis. EhRho1, but not a constitutively active mutant, interacts with a newly identified EhRhoGDI in a prenylation-dependent manner. Furthermore, constitutively active EhRho1 induces actin stress fiber formation in mammalian fibroblasts, thereby identifying it as a functional Rho family GTPase. EhRho1 exhibits a fast rate of nucleotide exchange relative to mammalian Rho GTPases due to a distinctive switch one isoleucine residue reminiscent of the constitutively active F28L mutation in human Cdc42, which for the latter protein, is sufficient for cellular transformation. Nonconserved, nucleotide-interacting residues within EhRho1, revealed by the crystal structure models, were observed to contribute a moderating influence on fast spontaneous nucleotide exchange. Collectively, these observations indicate that EhRho1 is a bona fide member of the Rho GTPase family, albeit with unique structural and functional aspects compared with mammalian Rho GTPases.


Asunto(s)
Entamoeba histolytica/enzimología , Proteínas Protozoarias/química , Proteínas de Unión al GTP rho/química , ADP Ribosa Transferasas/química , Toxinas Botulínicas/química , Cristalografía por Rayos X , Entamoeba histolytica/genética , Entamoeba histolytica/patogenicidad , Genoma de Protozoos/fisiología , Humanos , Mutación , Estructura Secundaria de Proteína , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Fibras de Estrés/química , Fibras de Estrés/genética , Fibras de Estrés/metabolismo , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
17.
J Biol Chem ; 286(52): 45103-15, 2011 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-22052910

RESUMEN

Vinculin is an essential and highly conserved cell adhesion protein, found at both focal adhesions and adherens junctions, where it couples integrins or cadherins to the actin cytoskeleton. Vinculin is involved in controlling cell shape, motility, and cell survival, and has more recently been shown to play a role in force transduction. The tail domain of vinculin (Vt) contains determinants necessary for binding and bundling of actin filaments. Actin binding to Vt has been proposed to induce formation of a Vt dimer that is necessary for cross-linking actin filaments. Results from this study provide additional support for actin-induced Vt self-association. Moreover, the actin-induced Vt dimer appears distinct from the dimer formed in the absence of actin. To better characterize the role of the Vt strap and carboxyl terminus (CT) in actin binding, Vt self-association, and actin bundling, we employed smaller amino-terminal (NT) and CT deletions that do not perturb the structural integrity of Vt. Although both NT and CT deletions retain actin binding, removal of the CT hairpin (1061-1066) selectively impairs actin bundling in vitro. Moreover, expression of vinculin lacking the CT hairpin in vinculin knock-out murine embryonic fibroblasts affects the number of focal adhesions formed, cell spreading as well as cellular stiffening in response to mechanical force.


Asunto(s)
Actinas/metabolismo , Proteínas Aviares/metabolismo , Adhesiones Focales/metabolismo , Multimerización de Proteína/fisiología , Vinculina/metabolismo , Actinas/genética , Animales , Proteínas Aviares/genética , Células Cultivadas , Pollos , Fibroblastos/citología , Fibroblastos/metabolismo , Adhesiones Focales/genética , Ratones , Ratones Noqueados , Unión Proteica , Estructura Terciaria de Proteína , Vinculina/genética
18.
J Biol Chem ; 286(14): 12141-8, 2011 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-21242305

RESUMEN

SmgGDS is an atypical guanine nucleotide exchange factor (GEF) that promotes both cell proliferation and migration and is up-regulated in several types of cancer. SmgGDS has been previously shown to activate a wide variety of small GTPases, including the Ras family members Rap1a, Rap1b, and K-Ras, as well as the Rho family members Cdc42, Rac1, Rac2, RhoA, and RhoB. In contrast, here we show that SmgGDS exclusively activates RhoA and RhoC among a large panel of purified GTPases. Consistent with the well known properties of GEFs, this activation is catalytic, and SmgGDS preferentially binds to nucleotide-depleted RhoA relative to either GDP- or GTPγS-bound forms. However, mutational analyses indicate that SmgGDS utilizes a distinct exchange mechanism compared with canonical GEFs and in contrast to known GEFs requires RhoA to retain a polybasic region for activation. A homology model of SmgGDS highlights an electronegative surface patch and a highly conserved binding groove. Mutation of either area ablates the ability of SmgGDS to activate RhoA. Finally, the in vitro specificity of SmgGDS for RhoA and RhoC is retained in cells. Together, these results indicate that SmgGDS is a bona fide GEF that specifically activates RhoA and RhoC through a unique mechanism not used by other Rho family exchange factors.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Western Blotting , Línea Celular , Cromatografía en Gel , Dicroismo Circular , Factores de Intercambio de Guanina Nucleótido/química , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas de Unión al GTP rho/química , Proteínas de Unión al GTP rho/genética , Proteína de Unión al GTP rhoA/química , Proteína de Unión al GTP rhoA/genética
19.
Stem Cells ; 29(11): 1829-36, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21898699

RESUMEN

The fate of pluripotent mesenchymal stem cells (MSC) is determined through integration of chemical, spatial, and physical signals. The suppression of MSC adipogenesis by mechanical stimuli, which requires Akt-induced inhibition of glycogen synthase kinase 3ß (GSK3ß) with ß-catenin activation, can be enhanced by repetitive dosing within a single day. Here, we demonstrate that reapplication of cyclic strain within a 24-hour period leads to amplification of both Akt activation and its subsequent inhibition of GSK3ß, such that total cycle number can be reduced while still inhibiting adipogenesis. Amplification of Akt signaling is facilitated by a dynamic restructuring of the cell in response to mechanical signals, as evidenced by a transient increase in focal adhesion (FA) number and increased RhoA activity. Preventing FA assembly or development of tension blocks activation of Akt by mechanical signals, but not by insulin. This indicates that the FA infrastructure is essential to the physical, but not necessarily the chemical, sensitivity, and responsiveness of the cell. Exploiting the transient nature of cytoskeletal remodeling may represent a process to enhance cell responsiveness to mechanical input and ultimately define the fate of MSCs with a minimal input.


Asunto(s)
Adipocitos/citología , Adhesiones Focales/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Pluripotentes/citología , Estrés Mecánico , Adipocitos/metabolismo , Animales , Células Cultivadas , Células Madre Mesenquimatosas/metabolismo , Ratones , Células Madre Pluripotentes/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
20.
J Cell Biol ; 178(7): 1279-93, 2007 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-17875742

RESUMEN

During trans-endothelial migration (TEM), leukocytes use adhesion receptors such as intercellular adhesion molecule-1 (ICAM1) to adhere to the endothelium. In response to this interaction, the endothelium throws up dynamic membrane protrusions, forming a cup that partially surrounds the adherent leukocyte. Little is known about the signaling pathways that regulate cup formation. In this study, we show that RhoG is activated downstream from ICAM1 engagement. This activation requires the intracellular domain of ICAM1. ICAM1 colocalizes with RhoG and binds to the RhoG-specific SH3-containing guanine-nucleotide exchange factor (SGEF). The SH3 domain of SGEF mediates this interaction. Depletion of endothelial RhoG by small interfering RNA does not affect leukocyte adhesion but decreases cup formation and inhibits leukocyte TEM. Silencing SGEF also results in a substantial reduction in RhoG activity, cup formation, and TEM. Together, these results identify a new signaling pathway involving RhoG and its exchange factor SGEF downstream from ICAM1 that is critical for leukocyte TEM.


Asunto(s)
Movimiento Celular , Polaridad Celular , Células Endoteliales/citología , Células Endoteliales/enzimología , Molécula 1 de Adhesión Intercelular/metabolismo , Leucocitos/citología , Proteínas de Unión al GTP rho/metabolismo , Animales , Células COS , Adhesión Celular , Extensiones de la Superficie Celular/metabolismo , Chlorocebus aethiops , Activación Enzimática , Proteínas Fluorescentes Verdes/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HL-60 , Humanos , Molécula 1 de Adhesión Intercelular/química , Leucocitos/enzimología , Leucocitos/ultraestructura , Microesferas , Unión Proteica , Transporte de Proteínas , Proteínas Recombinantes de Fusión/metabolismo , Dominios Homologos src
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA