Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
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.
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.

3.
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.

4.
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
5.
Mol Biol Cell ; 26(21): 3704-18, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26354425

RESUMEN

The extensive invasive capacity of glioblastoma (GBM) makes it resistant to surgery, radiotherapy, and chemotherapy and thus makes it lethal. In vivo, GBM invasion is mediated by Rho GTPases through unidentified downstream effectors. Mammalian Diaphanous (mDia) family formins are Rho-directed effectors that regulate the F-actin cytoskeleton to support tumor cell motility. Historically, anti-invasion strategies focused upon mDia inhibition, whereas activation remained unexplored. The recent development of small molecules directly inhibiting or activating mDia-driven F-actin assembly that supports motility allows for exploration of their role in GBM. We used the formin inhibitor SMIFH2 and mDia agonists IMM-01/-02 and mDia2-DAD peptides, which disrupt autoinhibition, to examine the roles of mDia inactivation versus activation in GBM cell migration and invasion in vitro and in an ex vivo brain slice invasion model. Inhibiting mDia suppressed directional migration and spheroid invasion while preserving intrinsic random migration. mDia agonism abrogated both random intrinsic and directional migration and halted U87 spheroid invasion in ex vivo brain slices. Thus mDia agonism is a superior GBM anti-invasion strategy. We conclude that formin agonism impedes the most dangerous GBM component-tumor spread into surrounding healthy tissue. Formin activation impairs novel aspects of transformed cells and informs the development of anti-GBM invasion strategies.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/agonistas , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Forminas , Glioblastoma/metabolismo , Glioblastoma/patología , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Humanos , Invasividad Neoplásica , Ratas , Esferoides Celulares
6.
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
7.
PLoS One ; 9(1): e85409, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24465552

RESUMEN

Epithelial plasticity plays a critical role during physiological processes, such as wound healing and tissue regeneration, and dysregulation of epithelial plasticity can lead to pathological conditions, such as cancer. Cell-cell junctions are a critical feature of epithelial cells and loss of junctions is associated with acquisition of mesenchymal features, such as enhanced protrusion and migration. Although Rho has been implicated in regulation of junctions in epithelial cells, the role of Rho signaling in the regulation of epithelial plasticity has not been understood. We show that members of the RGS RhoGEFs family play a critical role in regulation of epithelial cell-cell junctions in breast epithelial cells. We identify a novel role for p115RhoGEF in regulation of epithelial plasticity. Loss of p115RhoGEF leads to decreased junctional E-cadherin and enhanced protrusiveness and migration. Conversely, overexpression of p115RhoGEF enhanced junctional E-cadherin and inhibited cell protrusion and migration. siRNA screen of 23 Rho effectors showed that members of the Diaphanous-Related Formin (DRF) family are required for p115RhoGEF-mediated changes in epithelial plasticity. Thus, our data indicates a novel role for p115RhoGEF in regulation of epithelial plasticity, which is dependent on Rho-DRF signaling module.


Asunto(s)
Células Epiteliales/fisiología , Uniones Adherentes/metabolismo , Antígenos CD , Cadherinas/metabolismo , Proteínas Portadoras/metabolismo , Movimiento Celular , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Células MCF-7 , Factores de Intercambio de Guanina Nucleótido Rho/fisiología
8.
Cancer Res ; 73(22): 6793-803, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24242070

RESUMEN

Although the cancer cell cytoskeleton is a clinically validated target, few new strategies have emerged for selectively targeting cell division by modulating the cytoskeletal structure, particularly ways that could avoid the cardiotoxic and neurotoxic effects of current agents such as taxanes. We address this gap by describing a novel class of small-molecule agonists of the mammalian Diaphanous (mDia)-related formins, which act downstream of Rho GTPases to assemble actin filaments, and their organization with microfilaments to establish and maintain cell polarity during migration and asymmetric division. GTP-bound Rho activates mDia family members by disrupting the interaction between the DID and DAD autoregulatory domains, which releases the FH2 domain to modulate actin and microtubule dynamics. In screening for DID-DAD disruptors that activate mDia, we identified two molecules called intramimics (IMM-01 and -02) that were sufficient to trigger actin assembly and microtubule stabilization, serum response factor-mediated gene expression, cell-cycle arrest, and apoptosis. In vivo analysis of IMM-01 and -02 established their ability to slow tumor growth in a mouse xenograft model of colon cancer. Taken together, our work establishes the use of intramimics and mDia-related formins as a new general strategy for therapeutic targeting of the cytoskeletal remodeling machinery of cancer cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Antineoplásicos/uso terapéutico , Citoesqueleto/efectos de los fármacos , Retroalimentación Fisiológica/efectos de los fármacos , Proteínas de Microfilamentos/antagonistas & inhibidores , Imitación Molecular , Neoplasias/tratamiento farmacológico , Proteínas Adaptadoras Transductoras de Señales/química , Animales , Antineoplásicos/farmacología , Citoesqueleto/metabolismo , Femenino , Forminas , Ratones , Ratones Desnudos , Proteínas de Microfilamentos/química , Terapia Molecular Dirigida , Células 3T3 NIH , Neoplasias/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
9.
PLoS One ; 8(11): e80500, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24260404

RESUMEN

The mammalian diaphanous-related formin (mDia1), a Rho-regulated cytoskeletal modulator, has been shown to promote T lymphocyte chemotaxis and interaction with antigen presenting cells, but the mechanisms underpinning mDia1 roles in these processes have not been defined. Here we show that mDia1(-/-) T cells exhibit impaired lymphocyte function-associated antigen 1 (LFA-1)-mediated T cell adhesion, migration and in vivo trafficking. These defects are associated with impaired microtubule (MT) polarization and stabilization, altered MT dynamics and reduced peripheral clustering of the MT plus-end-protein, adenomatous polyposis coli (APC) in migrating T cells following LFA-1-engagement. Loss of mDia1 also leads to impaired inducible inactivation of the glycogen synthase kinase (GSK) 3ß as well as hyperphosphorylation and reduced levels of APC in migrating T cells. These findings identify essential roles for the mDia1 formin in modulating GSK3ß-dependent MT contributions to induction of T-cell polarity, adhesion and motility.


Asunto(s)
Proteínas Portadoras/metabolismo , Movimiento Celular , Glucógeno Sintasa Quinasa 3/metabolismo , Microtúbulos/metabolismo , Linfocitos T/metabolismo , Proteína de la Poliposis Adenomatosa del Colon , Animales , Proteínas Portadoras/genética , Movimiento Celular/genética , Movimiento Celular/inmunología , Polaridad Celular/genética , Polaridad Celular/inmunología , Activación Enzimática , Forminas , Glucógeno Sintasa Quinasa 3 beta , Molécula 1 de Adhesión Intercelular/metabolismo , Activación de Linfocitos/inmunología , Antígeno-1 Asociado a Función de Linfocito/genética , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Ratones , Ratones Noqueados , Fosforilación , Unión Proteica , Linfocitos T/inmunología
10.
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
11.
Sci Signal ; 4(180): ra44, 2011 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-21730326

RESUMEN

The antimalaria drug chloroquine has been used as an anti-inflammatory agent for treating systemic lupus erythematosus and rheumatoid arthritis. We report that chloroquine promoted the transrepression of proinflammatory cytokines by the glucocorticoid receptor (GR). In a mouse collagen-induced arthritis model, chloroquine enhanced the therapeutic effects of glucocorticoid treatment. By inhibiting lysosome function, chloroquine synergistically activated glucocorticoid signaling. Lysosomal inhibition by either bafilomycin A1 (an inhibitor of the vacuolar adenosine triphosphatase) or knockdown of transcription factor EB (TFEB, a master activator of lysosomal biogenesis) mimicked the effects of chloroquine. The abundance of the GR, as well as that of the androgen receptor and estrogen receptor, correlated with changes in lysosomal biogenesis. Thus, we showed that glucocorticoid signaling is regulated by lysosomes, which provides a mechanistic basis for treating inflammation and autoimmune diseases with a combination of glucocorticoids and lysosomal inhibitors.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Cloroquina/uso terapéutico , Glucocorticoides/metabolismo , Lisosomas/efectos de los fármacos , Transducción de Señal , Animales , Antirreumáticos , Artritis Experimental/metabolismo , Artritis Experimental/patología , Cloroquina/farmacología , Citocinas , Glucocorticoides/uso terapéutico , Inflamación , Lisosomas/metabolismo , Lisosomas/fisiología , Ratones , Receptores de Glucocorticoides
12.
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
13.
PLoS One ; 4(9): e7102, 2009 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-19768111

RESUMEN

Myelodysplastic syndrome (MDS) is characterized by ineffective hematopoiesis and hyperplastic bone marrow. Complete loss or interstitial deletions of the long arm of chromosome 5 occur frequently in MDS. One candidate tumor suppressor on 5q is the mammalian Diaphanous (mDia)-related formin mDia1, encoded by DIAPH1 (5q31.3). mDia-family formins act as effectors for Rho-family small GTP-binding proteins including RhoB, which has also been shown to possess tumor suppressor activity. Mice lacking the Drf1 gene that encodes mDia1 develop age-dependent myelodysplastic features. We crossed mDia1 and RhoB knockout mice to test whether the additional loss of RhoB expression would compound the myelodysplastic phenotype. Drf1(-/-)RhoB(-/-) mice are fertile and develop normally. Relative to age-matched Drf1(-/-)RhoB(+/-) mice, the age of myelodysplasia onset was earlier in Drf1(-/-)RhoB(-/-) animals--including abnormally shaped erythrocytes, splenomegaly, and extramedullary hematopoiesis. In addition, we observed a statistically significant increase in the number of activated monocytes/macrophages in both the spleen and bone marrow of Drf1(-/-)RhoB(-/-) mice relative to Drf1(-/-)RhoB(+/-) mice. These data suggest a role for RhoB-regulated mDia1 in the regulation of hematopoietic progenitor cells.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Fetales/metabolismo , Regulación de la Expresión Génica , Proteínas de Microfilamentos/metabolismo , Síndromes Mielodisplásicos/metabolismo , Proteínas Nucleares/metabolismo , Proteína de Unión al GTP rhoB/biosíntesis , Proteína de Unión al GTP rhoB/fisiología , Animales , Células de la Médula Ósea/metabolismo , Forminas , Células Madre Hematopoyéticas/citología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Monocitos/metabolismo , Síndromes Mielodisplásicos/patología , Fenotipo , Bazo/metabolismo
14.
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
15.
Biochem Cell Biol ; 87(1): 207-16, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19234535

RESUMEN

The haemopoietic cell kinase (Hck) plays an important but poorly understood role in coupling chemoattractant stimuli to the actin cytoskeletal rearrangement required for neutrophil polarization and chemotaxis. Here, we show that Hck coimmunoprecipitates with the cytoskeletal regulatory Wiskott-Aldrich syndrome protein (WASp) and mammalian diaphanous-related formin 1 (mDia1) in chemoattractant-stimulated neutrophils, and that the 3 proteins inducibly colocalize with one another at the leading edge of chemotaxing cells. Hck interaction with WASp was found to be mediated by the Hck SH3 domain binding to the WASp proline-rich region, while Hck interaction with mDia1 was indirect but was required for binding to WASp. In contrast to wild-type cells, both WASp- and mDia1-deficient neutrophils showed severe impairment of chemokine-induced Hck membrane translocation and induction of Hck binding to WASp, and Hck activation and WASp tyrosine phosphorylation were impaired in mDia1-/- cells. Thus, chemotactic stimulation appears to induce an mDia1/Hck/WASp complex required for Hck membrane targeting and for induction of the Hck-mediated WASp tyrosine phosphorylation thought to be required for WASp-driven actin polymerization. These findings reveal that Hck functions in neutrophils to be realized, at least in part, via its interaction with mDia1 and WASp, and identifies the mDia1/Hck/WASp axis as a cytoskeletal signaling interface linking tyrosine phosphorylation to chemotactic and, possibly, other actin-based neutrophil responses.


Asunto(s)
Proteínas Portadoras/metabolismo , Membrana Celular/enzimología , Factores Quimiotácticos/farmacología , Citoesqueleto/metabolismo , Neutrófilos/enzimología , Proteínas Proto-Oncogénicas c-hck/metabolismo , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Animales , Células COS , Membrana Celular/efectos de los fármacos , Quimiocinas/farmacología , Quimiotaxis/efectos de los fármacos , Chlorocebus aethiops , Citoesqueleto/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Forminas , Células HL-60 , Humanos , Ratones , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Fosfotirosina/metabolismo , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Seudópodos/efectos de los fármacos , Seudópodos/enzimología
16.
Mol Cell Biol ; 28(9): 2930-40, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18212049

RESUMEN

Parafibromin, a transcription factor associated with the PAF complex, is encoded by the HRPT2 gene, mutations of which cause the hyperparathyroidism-jaw tumor syndrome (OMIM145001). To elucidate the function of parafibromin, we generated conventional and conditional Hrpt2 knockout mice and found that Hrpt2(-/-) mice were embryonic lethal by embryonic day 6.5 (E6.5). Controlled deletion of Hrpt2 after E8.5 resulted in apoptosis and growth retardation. Deletion of Hrpt2 in adult mice led to severe cachexia and death within 20 days. To explore the mechanism underlying the embryonic lethality and death of adult mice, mouse embryonic fibroblasts (MEFs) were cultured and Hrpt2 was deleted in vitro. Hrpt2(-/-) MEFs underwent apoptosis, while Hrpt2(+/+) and Hrpt2(+/-) MEFs grew normally. To study the mechanism of this apoptosis, Hrpt2(+/+) and Hrpt2(-/-) MEFs were used in cDNA microarray, semiquantitative reverse transcription-PCR, and chromatin immunoprecipitation assays to identify genes regulated by parafibromin. These revealed that Hrpt2 expression and the parafibromin/PAF complex directly regulate genes involved in cell growth and survival, including H19, Igf1, Igf2, Igfbp4, Hmga1, Hmga2, and Hmgcs2. Thus, our results show that expression of Hrpt2 and parafibromin is pivotal in mammalian development and survival in adults and that these functions are likely mediated by the transcriptional regulation of growth factors.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Riñón/embriología , Hígado/embriología , Glándulas Salivales/embriología , Proteínas Supresoras de Tumor/fisiología , Animales , Apoptosis/fisiología , Caquexia/metabolismo , Caquexia/patología , Supervivencia Celular/fisiología , Células Cultivadas , Pérdida del Embrión/genética , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Riñón/metabolismo , Riñón/patología , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Glándulas Salivales/metabolismo , Glándulas Salivales/patología , Proteínas Supresoras de Tumor/genética
17.
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
18.
J Cell Sci ; 120(Pt 19): 3475-87, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17855386

RESUMEN

Cell migration requires spatial and temporal regulation of filamentous actin (F-actin) dynamics. This regulation is achieved by distinct actin-associated proteins, which mediate polymerization, depolymerization, severing, contraction, bundling or engagement to the membrane. Mammalian Diaphanous-related (mDia) formins, which nucleate, processively elongate, and in some cases bundle actin filaments, have been extensively studied in vitro, but their function in the cell has been less well characterized. Here we study the role of mDia2 activity in the dynamic organization of F-actin in migrating epithelial cells. We find that mDia2 localizes in the lamella of migrating epithelial cells, where it is involved in the formation of a stable pool of cortical actin and in maintenance of polymerization-competent free filament barbed ends at focal adhesions. Specific inhibition of mDia2 alters focal adhesion turnover and reduces migration velocity. We suggest that the regulation of filament assembly dynamics at focal adhesions may be necessary for the formation of a stable pool of cortical lamella actin and the proper assembly and disassembly dynamics of focal adhesions, making mDia2 an important factor in epithelial cell migration.


Asunto(s)
Actinas/metabolismo , Proteínas Portadoras/metabolismo , Movimiento Celular/fisiología , Extensiones de la Superficie Celular/metabolismo , Células Epiteliales , Adhesiones Focales/metabolismo , Animales , Proteínas Portadoras/genética , Forma de la Célula , Extensiones de la Superficie Celular/ultraestructura , Células Epiteliales/citología , Células Epiteliales/fisiología , Forminas , Humanos
19.
Cancer Biol Ther ; 6(9): 1422-7, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17786040

RESUMEN

Farnesyl transferase inhibitors (FTIs) exhibit limited cytotoxic effects against human cancer cells, perhaps explaining the limited efficacy of FTIs in clinical trials. Learning how these well-tolerated drugs trigger p53-independent apoptosis in mouse models of cancer might therefore benefit efforts to leverage their utility in clinic. Recent clinical findings indicate that the oncogenic Rho guanine nucleotide exchange factor AKAP13/Lbc is associated with clinical responsiveness, in support of an earlier genetic proof in mice that gain of the geranylgeranylated isoform of RhoB which blocks oncogenic Rho signaling is essential for FTI-induced apoptosis. Here we offer evidence that the RhoB effector mDia is a critical downstream player in this death program. Dominant inhibition of mDia ablated FTI-induced apoptosis but not actin reorganization or growth inhibition, the latter of which has been linked previously to interactions with a RhoB effector kinase pathway that downregulates c-Myc. In nude mice, dominant inhibition of mDia promoted tumor formation and ablated FTI antitumor efficacy. Our findings suggest that the RhoB-mDia pathway is critical for the cell death mechanism engaged by FTI. Further, they suggest that mDia may be important for Rho-dependent survival of oncogenically transformed cells, perhaps driven by AKAP13/Lbc.


Asunto(s)
Proteínas Portadoras/metabolismo , Inhibidores Enzimáticos/farmacología , Farnesiltransferasa/antagonistas & inhibidores , Animales , Apoptosis , Muerte Celular , Línea Celular Transformada , Fibroblastos/metabolismo , Forminas , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Desnudos , Microscopía Fluorescente , Modelos Biológicos , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína de Unión al GTP rhoB/metabolismo
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA