Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Carcinogenesis ; 44(1): 54-64, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-36548952

RESUMEN

Loss of heterozygosity and promoter hypermethylation of APC is frequently observed in human endometrial cancer, which is the most common gynecological cancer in the USA, but its carcinogenic driver status in the endometrial epithelium has not been confirmed. We have identified a novel population of progenitor endometrial epithelial cells (EECs) in mice that express lysozyme M (LysM) and give rise to approximately 15% of all EECs in adult mice. LysM is a glycoside hydrolase that is encoded by Lyz2 and functions to protect cells from bacteria as part of the innate immune system. Its expression has been shown in a subset of hematopoietic stem cells and in specialized lung and small intestinal epithelial cells. Conditional deletion of Apc in LysM + EECs results in significantly more epithelial cells compared to wild-type mice. At 5 months of age, the ApccKO mice have enlarged uterine horns with pathology that is consistent with endometrial hyperplasia with cystic endometrial glands, non-villous luminal papillae and nuclear atypia. Nuclear accumulation of ß-catenin and ERα, both of which are known to induce endometrial hyperplasia, was observed in the EECs of the ApccKO mice. These results confirm that loss of APC in EECs can result in a phenotype similar to endometrial hyperplasia.


Asunto(s)
Hiperplasia Endometrial , Neoplasias Endometriales , Adulto , Femenino , Humanos , Ratones , Animales , Hiperplasia Endometrial/genética , Hiperplasia Endometrial/patología , Células Epiteliales/patología , Endometrio/patología , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Células Madre/metabolismo
2.
Blood ; 124(26): 3967-77, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25298036

RESUMEN

Megakaryocytes are highly specialized precursor cells that produce platelets via cytoplasmic extensions called proplatelets. Proplatelet formation (PPF) requires profound changes in microtubule and actin organization. In this work, we demonstrated that DIAPH1 (mDia1), a mammalian homolog of Drosophila diaphanous that works as an effector of the small GTPase Rho, negatively regulates PPF by controlling the dynamics of the actin and microtubule cytoskeletons. Moreover, we showed that inhibition of both DIAPH1 and the Rho-associated protein kinase (Rock)/myosin pathway increased PPF via coordination of both cytoskeletons. We provide evidence that 2 major effectors of the Rho GTPase pathway (DIAPH1 and Rock/myosin II) are involved not only in Rho-mediated stress fibers assembly, but also in the regulation of microtubule stability and dynamics during PPF.


Asunto(s)
Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Citoesqueleto/metabolismo , Megacariocitos/citología , Microtúbulos/metabolismo , Antígenos CD34/metabolismo , Plaquetas/citología , Plaquetas/metabolismo , Diferenciación Celular , Clonación Molecular , Forminas , GTP Fosfohidrolasas/metabolismo , Humanos , Lentivirus/genética , Miosina Tipo II/metabolismo , ARN Interferente Pequeño/metabolismo , Trombopoyetina/química , Tubulina (Proteína)/química
3.
Nat Cell Biol ; 9(12): 1347-59, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18026093

RESUMEN

Extension of neurites from a cell body is essential to form a functional nervous system; however, the mechanisms underlying neuritogenesis are poorly understood. Ena/VASP proteins regulate actin dynamics and modulate elaboration of cellular protrusions. We recently reported that cortical axon-tract formation is lost in Ena/VASP-null mice and Ena/VASP-null cortical neurons lack filopodia and fail to elaborate neurites. Here, we report that neuritogenesis in Ena/VASP-null neurons can be rescued by restoring filopodia formation through ectopic expression of the actin nucleating protein mDia2. Conversely, wild-type neurons in which filopodia formation is blocked fail to elaborate neurites. We also report that laminin, which promotes the formation of filopodia-like actin-rich protrusions, rescues neuritogenesis in Ena/VASP-deficient neurons. Therefore, filopodia formation is a key prerequisite for neuritogenesis in cortical neurons. Neurite initiation also requires microtubule extension into filopodia, suggesting that interactions between actin-filament bundles and dynamic microtubules within filopodia are crucial for neuritogenesis.


Asunto(s)
Corteza Cerebral/citología , Neuritas/fisiología , Neuronas/fisiología , Seudópodos/fisiología , Actinas/metabolismo , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/fisiología , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Laminina/fisiología , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/fisiología , Proteínas Asociadas a Microtúbulos , Microtúbulos/fisiología , Mutación , Miosina Tipo II/antagonistas & inhibidores , Miosinas/biosíntesis , NADPH Deshidrogenasa/biosíntesis , Neuronas/ultraestructura , Fosfoproteínas/genética , Fosfoproteínas/fisiología
4.
Circ Res ; 110(10): 1279-93, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22511750

RESUMEN

RATIONALE: The mammalian diaphanous-related formin (mDia1), governs microtubule and microfilament dynamics while functioning as an effector for Rho small GTP-binding proteins during key cellular processes such as adhesion, cytokinesis, cell polarity, and morphogenesis. The cytoplasmic domain of the receptor for advanced glycation endproducts binds to the formin homology 1 domain of mDia1; mDia1 is required for receptor for advanced glycation endproducts ligand-induced cellular migration in transformed cells. OBJECTIVE: Because a key mechanism in vascular remodeling is the induction of smooth muscle cell migration, we tested the role of mDia1 in this process. METHODS AND RESULTS: We report that endothelial denudation injury to the murine femoral artery significantly upregulates mDia1 mRNA transcripts and protein in the injured vessel, particularly in vascular smooth muscle cells within the expanding neointima. Loss of mDia1 expression significantly reduces pathological neointimal expansion consequent to injury. In primary murine aortic smooth muscle cells, mDia1 is required for receptor for advanced glycation endproducts ligand-induced membrane translocation of c-Src, which leads to Rac1 activation, redox phosphorylation of AKT/glycogen synthase kinase 3ß, and consequent smooth muscle cell migration. CONCLUSIONS: We conclude that mDia1 integrates oxidative and signal transduction pathways triggered, at least in part, by receptor for advanced glycation endproducts ligands, thereby regulating pathological neointimal expansion.


Asunto(s)
Proteínas Portadoras/metabolismo , Músculo Liso Vascular/metabolismo , Neointima/patología , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología , Citoesqueleto de Actina/fisiología , Animales , Proteínas Portadoras/genética , Movimiento Celular/fisiología , Células Cultivadas , Arteria Femoral/lesiones , Arteria Femoral/metabolismo , Arteria Femoral/patología , Forminas , Productos Finales de Glicación Avanzada/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microtúbulos/fisiología , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/patología , NADH NADPH Oxidorreductasas/genética , NADH NADPH Oxidorreductasas/metabolismo , NADPH Oxidasa 1 , Neointima/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/metabolismo
5.
Cancer Cell ; 4(5): 405-13, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14667507

RESUMEN

By positional cloning, we identified two breakpoint-spanning genes in a familial clear cell renal cell carcinoma (CCRCC)-associated t(1;3)(q32.1;q13.3): LSAMP and NORE1 (RASSF1 homolog). Both genes are downregulated in 9 of 9 RCC cell lines. While the NORE1A promoter predominantly presents partial methylation in 6 of the cell lines and 17/53 (32%) primary tumors, the LSAMP promoter is completely methylated in 5 of 9 cell lines and in 14/53 (26%) sporadic and 4 familial CCRCCs. Expression of LSAMP and NORE1A proteins in CCRCC cell lines inhibited cell proliferation. These characteristics indicate that LSAMP and NORE1A may represent new candidate tumor suppressors for CCRCC.


Asunto(s)
Adenocarcinoma de Células Claras/genética , Carcinoma de Células Renales/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Adenocarcinoma de Células Claras/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Secuencia de Bases , Carcinoma de Células Renales/metabolismo , Moléculas de Adhesión Celular Neuronal/genética , División Celular/fisiología , Células Cultivadas , Clonación Molecular , Metilación de ADN , Proteínas Ligadas a GPI , Humanos , Datos de Secuencia Molecular , Proteínas de Unión al GTP Monoméricas/genética
6.
Biochim Biophys Acta ; 1803(2): 226-33, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19941910

RESUMEN

Formins are a conserved family of proteins that play key roles in cytoskeletal remodeling. They nucleate and processively elongate non-branched actin filaments and also modulate microtubule dynamics. Despite their significant contributions to cell biology and development, few studies have directly implicated formins in disease pathogenesis. This review highlights the roles of formins in cell division, migration, immunity, and microvesicle formation in the context of human disease. In addition, we discuss the importance of controlling formin activity and protein expression to maintain cell homeostasis.


Asunto(s)
Enfermedad , Proteínas Fetales , Proteínas de Microfilamentos , Proteínas Nucleares , Secuencia de Aminoácidos , Animales , Movimiento Celular/fisiología , Citocinesis/fisiología , Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Proteínas Fetales/genética , Proteínas Fetales/metabolismo , Forminas , Humanos , Sistema Inmunológico/fisiología , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Datos de Secuencia Molecular , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Alineación de Secuencia
7.
Curr Biol ; 18(14): R605-8, 2008 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-18644336

RESUMEN

Mammalian Diaphanous-related (mDia) formins are well known for their actin nucleation and filament elongation activities. They have since emerged as microtubule-binding proteins, and a recent study shows that mDia2 stabilizes microtubules independently of its actin nucleation activity.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Actinas/química , Actinas/metabolismo , Animales , Proteínas Portadoras/química , Humanos , Proteínas Asociadas a Microtúbulos/química , Modelos Biológicos , Unión Proteica
8.
Nat Cell Biol ; 6(9): 820-30, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15311282

RESUMEN

Lysophosphatidic acid (LPA) stimulates Rho GTPase and its effector, the formin mDia, to capture and stabilize microtubules in fibroblasts. We investigated whether mammalian EB1 and adenomatous polyposis coli (APC) function downstream of Rho-mDia in microtubule stabilization. A carboxy-terminal APC-binding fragment of EB1 (EB1-C) functioned as a dominant-negative inhibitor of microtubule stabilization induced by LPA or active mDia. Knockdown of EB1 with small interfering RNAs also prevented microtubule stabilization. Expression of either full-length EB1 or APC, but not an APC-binding mutant of EB1, was sufficient to stabilize microtubules. Binding and localization studies showed that EB1, APC and mDia may form a complex at stable microtubule ends. Furthermore, EB1-C, but not an APC-binding mutant, inhibited fibroblast migration in an in vitro wounding assay. These results show an evolutionarily conserved pathway for microtubule capture, and suggest that mDia functions as a scaffold protein for EB1 and APC to stabilize microtubules and promote cell migration.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Proteínas Portadoras/metabolismo , Movimiento Celular , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/fisiología , Animales , Proteínas Portadoras/fisiología , Fibroblastos/fisiología , Forminas , Lisofosfolípidos/farmacología , Ratones , Proteínas Asociadas a Microtúbulos/fisiología , Células 3T3 NIH , Unión Proteica , Transfección , Proteínas de Unión al GTP rho
9.
J Immunol ; 182(6): 3837-45, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19265163

RESUMEN

Neutrophil chemotaxis depends on actin dynamics, but the roles for specific cytoskeleton regulators in this response remain unclear. By analysis of mammalian diaphanous-related formin 1 (mDia1)-deficient mice, we have identified an essential role for this actin nucleator in neutrophil chemotaxis. Lack of mDia1 was associated with defects in chemoattractant-induced neutrophil actin polymerization, polarization, and directional migration, and also with impaired activation of RhoA, its downstream target p160-Rho-associated coil-containing protein kinase (ROCK), and the leukemia-associated RhoA guanine nucleotide exchange factor (LARG). Our data also revealed mDia1 to be associated with another cytoskeletal regulator, Wiskott-Aldrich syndrome protein (WASp), at the leading edge of chemotaxing neutrophils and revealed polarized morphology and chemotaxis to be more mildly impaired in WAS(-/-) than in mDia1(-/-) neutrophils, but essentially abrogated by combined mDia1/WASp deficiency. Thus, mDia1 roles in neutrophil chemotaxis appear to be subserved in concert with WASp and are realized at least in part by activation of the LARG/RhoA/ROCK signaling pathway.


Asunto(s)
Proteínas Portadoras/fisiología , Polaridad Celular/inmunología , Quimiotaxis de Leucocito/inmunología , Factores de Intercambio de Guanina Nucleótido/fisiología , Neutrófilos/inmunología , Transducción de Señal/inmunología , Proteínas de Unión al GTP rho/fisiología , Quinasas Asociadas a rho/fisiología , Animales , Proteínas Portadoras/genética , Movimiento Celular/inmunología , Retroalimentación Fisiológica/inmunología , Proteínas Fetales/deficiencia , Proteínas Fetales/genética , Proteínas Fetales/fisiología , Forminas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/fisiología , Neutrófilos/citología , Neutrófilos/metabolismo , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Factores de Intercambio de Guanina Nucleótido Rho , Proteína del Síndrome de Wiskott-Aldrich/fisiología , Proteína de Unión al GTP rhoA
10.
Curr Biol ; 17(7): 579-91, 2007 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-17398099

RESUMEN

BACKGROUND: Mammalian Diaphanous (mDia)-related formins and the N-WASP-activated Arp2/3 complex initiate the assembly of filamentous actin. Dia-interacting protein (DIP) binds via its amino-terminal SH3 domain to the proline-rich formin homology 1 (FH1) domain of mDia1 and mDia2 and to the N-WASp proline-rich region. RESULTS: Here, we investigated an interaction between a conserved leucine-rich region (LRR) in DIP and the mDia FH2 domain that nucleates, processively elongates, and bundles actin filaments. DIP binding to mDia2 was regulated by the same Rho-GTPase-controlled autoinhibitory mechanism modulating formin-mediated actin assembly. DIP was previously shown to interact with and stimulate N-WASp-dependent branched filament assembly via Arp2/3. Despite direct binding to both mDia1 and mDia2 FH2 domains, DIP LRR inhibited only mDia2-dependent filament assembly and bundling in vitro. DIP expression interfered with filopodia formation, consistent with a role for mDia2 in assembly of these structures. After filopodia retraction into the cell body, DIP expression induced excessive nonapoptotic membrane blebbing, a physiological process involved in both cytokinesis and amoeboid cell movement. DIP-induced blebbing was dependent on mDia2 but did not require the activities of either mDia1 or Arp2/3. CONCLUSIONS: These observations point to a pivotal role for DIP in the control of nonbranched and branched actin-filament assembly that is mediated by Diaphanous-related formins and activators of Arp2/3, respectively. The ability of DIP to trigger blebbing also suggests a role for mDia2 in the assembly of cortical actin necessary for maintaining plasma-membrane integrity.


Asunto(s)
Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Musculares/metabolismo , Citoesqueleto de Actina/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Línea Celular , Membrana Celular/metabolismo , Forminas , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Datos de Secuencia Molecular , Proteínas Musculares/química , Proteínas Musculares/genética , Mutación , Estructura Terciaria de Proteína , Seudópodos/metabolismo , Seudópodos/ultraestructura , ARN Interferente Pequeño/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Proteína de Unión al GTP cdc42/metabolismo
11.
Trends Cell Biol ; 13(8): 435-46, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12888296

RESUMEN

Evolutionarily conserved in eukaryotes, formin homology (FH) proteins, or formins, exert their effects on the actin and microtubule (MT) networks during meiosis, mitosis, the maintenance of cell polarity, vesicular trafficking, signaling to the nucleus and embryonic development. Once thought to be only molecular scaffolds that indirectly affected cellular functions through the binding of other proteins, recent in vitro studies have illustrated that they can function as actin nucleators in the formation of new filaments. The connection between formins and MTs is less well understood. In yeast, the MT effects appear to be dependent on the ability of formins to generate polarized actin cables whereas, in mammalian cells, formin signals that cause MT stabilization and polarization might be more direct. A subclass of formins, the Diaphanous-related formins (Drfs), can act as effectors for Rho small GTPases, yet it is not clear what GTPase binding contributes to formin function.


Asunto(s)
Proteínas del Citoesqueleto/fisiología , Citoesqueleto/fisiología , Actinas/fisiología , Animales , Proteínas Portadoras/fisiología , Polaridad Celular/fisiología , Proteínas Contráctiles/fisiología , Proteínas de Unión al GTP/fisiología , Humanos , Ratones , Proteínas de Microfilamentos/fisiología , Microtúbulos/fisiología , Modelos Biológicos , Profilinas , Familia-src Quinasas/fisiología
12.
Cancer Res ; 67(16): 7565-71, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17699759

RESUMEN

Rho GTPase-effector mammalian diaphanous (mDia)-related formins assemble nonbranched actin filaments as part of cellular processes, including cell division, filopodia assembly, and intracellular trafficking. Whereas recent efforts have led to thorough characterization of formins in cytoskeletal remodeling and actin assembly in vitro, little is known about the role of mDia proteins in vivo. To fill this knowledge gap, the Drf1 gene, which encodes the canonical formin mDia1, was targeted by homologous recombination. Upon birth, Drf1+/- and Drf1-/- mice were developmentally and morphologically indistinguishable from their wild-type littermates. However, both Drf1+/- and Drf1-/- developed age-dependent myeloproliferative defects. The phenotype included splenomegaly, fibrotic and hypercellular bone marrow, extramedullary hematopoiesis in both spleen and liver, and the presence of immature myeloid progenitor cells with high nucleus-to-cytoplasm ratios. Analysis of cell surface markers showed an age-dependent increase in the percentage of CD11b+-activated and CD14+-activated monocytes/macrophages in both spleen and bone marrow in Drf1+/- and Drf1-/- animals. Analysis of the erythroid compartment showed a significant increase in the proportion of splenic cells in S phase and an expansion of erythroid precursors (TER-119+ and CD71+) in Drf1-targeted mice. Overall, knocking out mDia1 expression in mice leads to a phenotype similar to human myeloproliferative syndrome (MPS) and myelodysplastic syndromes (MDS). These observations suggest that defective DRF1 expression or mDia1 function may contribute to myeloid malignancies and point to mDia1 as an attractive therapeutic target in MDS and MPS.


Asunto(s)
Proteínas Portadoras/fisiología , Síndromes Mielodisplásicos/genética , Trastornos Mieloproliferativos/genética , Animales , Células de la Médula Ósea/fisiología , Antígeno CD11b/biosíntesis , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Forminas , Expresión Génica , Marcación de Gen , Genes Supresores de Tumor , Homeostasis/genética , Receptores de Lipopolisacáridos/biosíntesis , Ratones , Ratones Noqueados , Síndromes Mielodisplásicos/metabolismo , Mielopoyesis/genética , Trastornos Mieloproliferativos/metabolismo
13.
Cancers (Basel) ; 11(3)2019 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-30897774

RESUMEN

High-grade glioma (HGG, WHO Grade III⁻IV) accounts for the majority of adult primary malignant brain tumors. Failure of current therapies to target invasive glioma cells partly explains the minimal survival advantages: invasive tumors lack easily-defined surgical margins, and are inherently more chemo- and radioresistant. Much work centers upon Rho GTPase-mediated glioma invasion, yet downstream Rho effector roles are poorly understood and represent potential therapeutic targets. The roles for the mammalian Diaphanous (mDia)-related formin family of Rho effectors have emerged in invasive/metastatic disease. mDias assemble linear F-actin to promote protrusive cytoskeletal structures underlying tumor cell invasion. Small molecule mDia intramimic (IMM) agonists induced mDia functional activities including F-actin polymerization. mDia agonism inhibited polarized migration in Glioblastoma (WHO Grade IV) cells in three-dimensional (3D) in vitro and rat brain slice models. Here, we evaluate whether clinically-relevant high-grade glioma patient-derived neuro-sphere invasion is sensitive to formin agonism. Surgical HGG samples were dissociated, briefly grown as monolayers, and spontaneously formed non-adherent neuro-spheres. IMM treatment dramatically inhibited HGG patient neuro-sphere invasion, both at neuro-sphere embedding and mid-invasion assay, inducing an amoeboid morphology in neuro-sphere edge cells, while inhibiting actin- and tubulin-enriched tumor microtube formation. Thus, mDia agonism effectively disrupts multiple aspects of patient-derived HGG neuro-sphere invasion.

14.
Curr Biol ; 15(22): 2007-12, 2005 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-16303559

RESUMEN

Macrophages, dendritic cells, and neutrophils use phagocytosis to capture and clear off invading pathogens. The process is triggered by the interaction of ligands on the pathogens' surface with specific phagocytic receptors, including immunoglobulin (FcR) and complement C3bi (CR3) receptors (integrin alpha(M)beta2, Mac1) . Localized actin-filament assembly that acts as the driving force for particle engulfment is controlled by Rho-family small GTPases . RhoA regulates CR3-mediated phagocytosis through a mechanism that is still unclear . Mammalian Diaphanous-related (mDia) formins participate in the generation of a diverse set of actin-remodeling events downstream of RhoA , and mDia1 is recruited around fibronectin-coated beads in a RhoA-dependent manner in fibroblasts . Here, we set out to examine whether mDia proteins are involved in CR3-mediated phagocytosis in macrophages. We show that the RhoA effector mDia1 is recruited early during CR3-mediated phagocytosis and colocalizes with polymerized actin in the phagocytic cup. Interfering with mDia activity inhibits CR3-mediated phagocytosis while having no effect on FcR-mediated phagocytosis. These results indicate a new function for mDia proteins in the regulation of actin polymerization during CR3-mediated phagocytosis.


Asunto(s)
Actinas/metabolismo , Proteínas Portadoras/metabolismo , Antígeno de Macrófago-1/metabolismo , Macrófagos/fisiología , Fagocitosis/fisiología , Animales , Western Blotting , Línea Celular , Técnica del Anticuerpo Fluorescente , Forminas , Macrófagos/ultraestructura , Ratones , Microscopía Electrónica de Rastreo , ARN Interferente Pequeño/genética
15.
Front Pharmacol ; 9: 340, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29692731

RESUMEN

The mammalian Diaphanous-related (mDia) formins are cytoskeletal regulators that assemble and, in some cases, bundle filamentous actin (F-actin), as well as stabilize microtubules. The development of small molecule antagonists and agonists that interrogate mDia formin function has allowed us to investigate the roles of formins in disease states. A small molecule inhibitor of FH2 domain (SMIFH2) inhibits mDia-dependent actin dynamics and abrogates tumor cell migration and cell division in vitro and ex vivo tissue explants. mDia formin activation with small molecule intramimics IMM01/02 and mDia2-DAD peptides inhibited glioblastoma motility and invasion in vitro and ex vivo rat brain slices. However, SMIFH2, IMMs, and mDia2 DAD efficacy in vivo remains largely unexplored and potential toxicity across a range of developmental phenotypes has not been thoroughly characterized. In this study, we performed an in vivo screen of early life-stage toxicity in Danio rerio zebrafish embryos 2 days post-fertilization (dpf) in response to SMIFH2, IMM01/02, and mDia2 DAD. SMIFH2 at concentrations ≥5-10 µM induced significant defects in developing zebrafish, including shorter body lengths, tail curvature and defective tail cellularity, craniofacial malformations, pericardial edema, absent and/or compromised vasculature function and flow, depressed heart rates and increased mortality. Conversely, IMM and mDia2 DAD peptides were minimally toxic at concentrations up to 10-20 and 50 µM, respectively. SMIFH2's therapeutic potential may therefore be limited by its substantial in vivo toxicity at functional concentrations. mDia formin agonism with IMMs and mDia2 DADs may therefore be a more effective and less toxic anti-invasive therapeutic approach.

16.
Curr Biol ; 13(7): 534-45, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12676083

RESUMEN

BACKGROUND: Mammalian Diaphanous-related formins (Drfs) act as Rho small GTPase effectors during growth factor-induced cytoskeletal remodeling and cell division. While both p140 mDia1 (herein called Drf1) and p134 mDia2 (Drf3) have been shown to bind in vitro to activated RhoA-C, and Drf3 has also been shown to bind to Cdc42, little is known about the cellular function of these GTPase effector pairs. Thus, we have begun targeting the murine Drf genes to address their various contributions to small GTPase signaling in cytoskeletal remodeling and development. RESULTS: Drf1 +/+, +/-, and -/- cell lines were derived from embryonic stem cells. While some Drf1 +/- lines had fewer actin stress fibers, several Drf1 +/- and -/- cells were more motile and had more abundant lamella and filopodia. Because the apparent "gain-of-function" corresponded with elevated levels of Drf3 protein expression, we hypothesized that the effects on the actin cytoskeleton were due to Cdc42 utilization of Drf3 as an effector. In this study, we found that inactive Drf3 variants and microinjected Drf3 antibodies interfered with Cdc42-induced filopodia. In addition, we observed that Drf3 contains a previously unidentified CRIB-like motif within its GTPase binding domain (GBD). By fluorescent resonance energy transfer (FRET) analysis, we demonstrate that this motif is required for Cdc42 binding and Drf3 recruitment to the leading edge and, surprisingly, to the microtubule organizing center (MTOC) of migrating fibroblasts. CONCLUSIONS: Our observations extend the role of the mammalian Drfs in cell signaling and demonstrate that Cdc42 not only activates Drf3, but guides the effector to sites at the cell cortex where it remodels the actin cytoskeleton.


Asunto(s)
Actinas/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Citoesqueleto/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Actinas/genética , Actinas/ultraestructura , Animales , Southern Blotting , Técnicas de Cultivo de Célula , Mapeo Cromosómico , Citoesqueleto/genética , Transferencia Resonante de Energía de Fluorescencia , Forminas , Regulación de la Expresión Génica , Ratones , Microinyecciones , Seudópodos/metabolismo , Transducción de Señal , Células Madre , Proteína de Unión al GTP cdc42/genética
17.
EBioMedicine ; 26: 165-174, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29239839

RESUMEN

The biochemical, ionic, and signaling changes that occur within cardiomyocytes subjected to ischemia are exacerbated by reperfusion; however, the precise mechanisms mediating myocardial ischemia/reperfusion (I/R) injury have not been fully elucidated. The receptor for advanced glycation end-products (RAGE) regulates the cellular response to cardiac tissue damage in I/R, an effect potentially mediated by the binding of the RAGE cytoplasmic domain to the diaphanous-related formin, DIAPH1. The aim of this study was to investigate the role of DIAPH1 in the physiological response to experimental myocardial I/R in mice. After subjecting wild-type mice to experimental I/R, myocardial DIAPH1 expression was increased, an effect that was echoed following hypoxia/reoxygenation (H/R) in H9C2 and AC16 cells. Further, compared to wild-type mice, genetic deletion of Diaph1 reduced infarct size and improved contractile function after I/R. Silencing Diaph1 in H9C2 cells subjected to H/R downregulated actin polymerization and serum response factor-regulated gene expression. Importantly, these changes led to increased expression of sarcoplasmic reticulum Ca2+ ATPase and reduced expression of the sodium calcium exchanger. This work demonstrates that DIAPH1 is required for the myocardial response to I/R, and that targeting DIAPH1 may represent an adjunctive approach for myocardial salvage after acute infarction.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Daño por Reperfusión Miocárdica/genética , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Forminas , Regulación de la Expresión Génica , Humanos , Ratones , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/patología , Miocitos Cardíacos/patología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Transducción de Señal/genética , Intercambiador de Sodio-Calcio/genética
19.
Nat Commun ; 7: 10997, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26975831

RESUMEN

Cell migration has two opposite faces: although necessary for physiological processes such as immune responses, it can also have detrimental effects by enabling metastatic cells to invade new organs. In vivo, migration occurs in complex environments and often requires a high cellular deformability, a property limited by the cell nucleus. Here we show that dendritic cells, the sentinels of the immune system, possess a mechanism to pass through micrometric constrictions. This mechanism is based on a rapid Arp2/3-dependent actin nucleation around the nucleus that disrupts the nuclear lamina, the main structure limiting nuclear deformability. The cells' requirement for Arp2/3 to pass through constrictions can be relieved when nuclear stiffness is decreased by suppressing lamin A/C expression. We propose a new role for Arp2/3 in three-dimensional cell migration, allowing fast-moving cells such as leukocytes to rapidly and efficiently migrate through narrow gaps, a process probably important for their function.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Actinas/metabolismo , Movimiento Celular , Núcleo Celular/metabolismo , Células Dendríticas , Neutrófilos , Lámina Nuclear/metabolismo , Animales , Immunoblotting , Lamina Tipo A/metabolismo , Ratones , Polimerizacion
20.
Nat Cell Biol ; 18(1): 43-53, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26641718

RESUMEN

Dendritic cell (DC) migration in peripheral tissues serves two main functions: antigen sampling by immature DCs, and chemokine-guided migration towards lymphatic vessels (LVs) on maturation. These migratory events determine the efficiency of the adaptive immune response. Their regulation by the core cell locomotion machinery has not been determined. Here, we show that the migration of immature DCs depends on two main actin pools: a RhoA-mDia1-dependent actin pool located at their rear, which facilitates forward locomotion; and a Cdc42-Arp2/3-dependent actin pool present at their front, which limits migration but promotes antigen capture. Following TLR4-MyD88-induced maturation, Arp2/3-dependent actin enrichment at the cell front is markedly reduced. Consequently, mature DCs switch to a faster and more persistent mDia1-dependent locomotion mode that facilitates chemotactic migration to LVs and lymph nodes. Thus, the differential use of actin-nucleating machineries optimizes the migration of immature and mature DCs according to their specific function.


Asunto(s)
Actinas/metabolismo , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Quimiotaxis/fisiología , Células Dendríticas/metabolismo , Animales , Células Cultivadas , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA