Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Immunother Cancer ; 12(4)2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38604809

RESUMEN

BACKGROUND: Combining cytotoxic chemotherapy or novel anticancer drugs with T-cell modulators holds great promise in treating advanced cancers. However, the response varies depending on the tumor immune microenvironment (TIME). Therefore, there is a clear need for pharmacologically tractable models of the TIME to dissect its influence on mono- and combination treatment response at the individual level. METHODS: Here we establish a patient-derived explant culture (PDEC) model of breast cancer, which retains the immune contexture of the primary tumor, recapitulating cytokine profiles and CD8+T cell cytotoxic activity. RESULTS: We explored the immunomodulatory action of a synthetic lethal BCL2 inhibitor venetoclax+metformin drug combination ex vivo, discovering metformin cannot overcome the lymphocyte-depleting action of venetoclax. Instead, metformin promotes dendritic cell maturation through inhibition of mitochondrial complex I, increasing their capacity to co-stimulate CD4+T cells and thus facilitating antitumor immunity. CONCLUSIONS: Our results establish PDECs as a feasible model to identify immunomodulatory functions of anticancer drugs in the context of patient-specific TIME.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Compuestos Bicíclicos Heterocíclicos con Puentes , Metformina , Sulfonamidas , Humanos , Femenino , Complejo I de Transporte de Electrón/farmacología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Células Dendríticas , Metformina/farmacología , Metformina/uso terapéutico , Microambiente Tumoral
2.
Mol Oncol ; 18(3): 547-561, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37872868

RESUMEN

Hepsin, a type II transmembrane serine protease, is commonly overexpressed in prostate and breast cancer. The hepsin protein is stabilized by the Ras-MAPK pathway, and, downstream, this protease regulates the degradation of extracellular matrix components and activates growth factor pathways, such as the hepatocyte growth factor (HGF) and transforming growth factor beta (TGFß) pathway. However, how exactly active hepsin promotes cell proliferation machinery to sustain tumor growth is not fully understood. Here, we show that genetic deletion of the gene encoding hepsin (Hpn) in a WAP-Myc model of aggressive MYC-driven breast cancer inhibits tumor growth in the primary syngrafted sites and the growth of disseminated tumors in the lungs. The suppression of tumor growth upon loss of hepsin was accompanied by downregulation of TGFß and EGFR signaling together with a reduction in epidermal growth factor receptor (EGFR) protein levels. We further demonstrate in 3D cultures of patient-derived breast cancer explants that both basal TGFß signaling and EGFR protein expression are inhibited by neutralizing antibodies or small-molecule inhibitors of hepsin. The study demonstrates a role for hepsin as a regulator of cell proliferation and tumor growth through TGFß and EGFR pathways, warranting consideration of hepsin as a potential indirect upstream target for therapeutic inhibition of TGFß and EGFR pathways in cancer.


Asunto(s)
Neoplasias de la Mama , Factor de Crecimiento Epidérmico , Serina Endopeptidasas , Humanos , Masculino , Neoplasias de la Mama/genética , Línea Celular Tumoral , Receptores ErbB/genética , Receptores de Factores de Crecimiento Transformadores beta , Factor de Crecimiento Transformador beta
4.
J Cell Sci ; 135(20)2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36148554

RESUMEN

SHARPIN is involved in several cellular processes and promotes cancer progression. However, how the choice between different functions of SHARPIN is post-translationally regulated is unclear. Here, we characterized SHARPIN phosphorylation by mass spectrometry and in vitro kinase assay. Focusing on S131 and S146, we demonstrate that they have a role in SHARPIN-ARP2/3 complex interaction, but play no role in integrin inhibition or LUBAC activation. Consistent with its novel role in ARP2/3 regulation, S146 phosphorylation of SHARPIN promoted lamellipodia formation. We also demonstrate that SHARPIN S146 phosphorylation-mediated ARP2/3 interaction is sensitive to inhibition of ERK1/2 or reactivation of protein phosphatase 2A (PP2A). Notably, CRISPR/Cas9-mediated knockout of SHARPIN abrogated three-dimensional (3D) invasion of several cancer cell lines. The 3D invasion of cancer cells was rescued by overexpression of the wild-type SHARPIN, but not by SHARPIN S146A mutant. Finally, we demonstrate that inhibition of phosphorylation at S146 significantly reduces in vivo metastasis in a zebrafish model. Collectively, these results map SHARPIN phosphorylation sites and identify S146 as a novel phosphorylation switch defining ARP2/3 interaction and cancer cell invasion. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Proteína Fosfatasa 2 , Pez Cebra , Animales , Integrinas , Invasividad Neoplásica , Proteínas del Tejido Nervioso , Fosforilación
5.
Mol Cancer Ther ; 21(7): 1236-1245, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35364610

RESUMEN

Identification of ovarian cancer patient subpopulations with increased sensitivity to targeted therapies could offer significant clinical benefit. We report that 22% of the high-grade ovarian cancer tumors at diagnosis express CIP2A oncoprotein at low levels. Furthermore, regardless of their significantly lower likelihood of disease relapse after standard chemotherapy, a portion of relapsed tumors retain their CIP2A-deficient phenotype. Through a screen for therapeutics that would preferentially kill CIP2A-deficient ovarian cancer cells, we identified reactive oxygen species inducer APR-246, tested previously in ovarian cancer clinical trials. Consistent with CIP2A-deficient ovarian cancer subtype in humans, CIP2A is dispensable for development of MISIIR-Tag-driven mouse ovarian cancer tumors. Nevertheless, CIP2A-null ovarian cancer tumor cells from MISIIR-Tag mice displayed APR-246 hypersensitivity both in vitro and in vivo. Mechanistically, the lack of CIP2A expression hypersensitizes the ovarian cancer cells to APR-246 by inhibition of NF-κB activity. Accordingly, combination of APR-246 and NF-κB inhibitor compounds strongly synergized in killing of CIP2A-positive ovarian cancer cells. Collectively, the results warrant consideration of clinical testing of APR-246 for CIP2A-deficient ovarian cancer tumor subtype patients. Results also reveal CIP2A as a candidate APR-246 combination therapy target for ovarian cancer.


Asunto(s)
FN-kappa B , Neoplasias Ováricas , Animales , Autoantígenos/genética , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Femenino , Humanos , Ratones , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Quinuclidinas
6.
Nat Commun ; 12(1): 6967, 2021 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-34845227

RESUMEN

Breast cancer is now globally the most frequent cancer and leading cause of women's death. Two thirds of breast cancers express the luminal estrogen receptor-positive (ERα + ) phenotype that is initially responsive to antihormonal therapies, but drug resistance emerges. A major barrier to the understanding of the ERα-pathway biology and therapeutic discoveries is the restricted repertoire of luminal ERα + breast cancer models. The ERα + phenotype is not stable in cultured cells for reasons not fully understood. We examine 400 patient-derived breast epithelial and breast cancer explant cultures (PDECs) grown in various three-dimensional matrix scaffolds, finding that ERα is primarily regulated by the matrix stiffness. Matrix stiffness upregulates the ERα signaling via stress-mediated p38 activation and H3K27me3-mediated epigenetic regulation. The finding that the matrix stiffness is a central cue to the ERα phenotype reveals a mechanobiological component in breast tissue hormonal signaling and enables the development of novel therapeutic interventions. Subject terms: ER-positive (ER + ), breast cancer, ex vivo model, preclinical model, PDEC, stiffness, p38 SAPK.


Asunto(s)
Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/genética , Mecanotransducción Celular/genética , Transcriptoma , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Estudios de Casos y Controles , Línea Celular Tumoral , Cinamatos/farmacología , Colágeno/química , Colágeno/farmacología , Combinación de Medicamentos , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Femenino , Fulvestrant/farmacología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Histonas/genética , Histonas/metabolismo , Humanos , Indazoles/farmacología , Laminina/química , Laminina/farmacología , Glándulas Mamarias Humanas/efectos de los fármacos , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Fenotipo , Proteoglicanos/química , Proteoglicanos/farmacología , Tamoxifeno/farmacología , Técnicas de Cultivo de Tejidos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
EMBO Rep ; 22(11): e52532, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34515392

RESUMEN

Transforming growth factor-beta (TGFß) is a multifunctional cytokine with a well-established role in mammary gland development and both oncogenic and tumor-suppressive functions. The extracellular matrix (ECM) indirectly regulates TGFß activity by acting as a storage compartment of latent-TGFß, but how TGFß is released from the ECM via proteolytic mechanisms remains largely unknown. In this study, we demonstrate that hepsin, a type II transmembrane protease overexpressed in 70% of breast tumors, promotes canonical TGFß signaling through the release of latent-TGFß from the ECM storage compartment. Mammary glands in hepsin CRISPR knockout mice showed reduced TGFß signaling and increased epithelial branching, accompanied by increased levels of fibronectin and latent-TGFß1, while overexpression of hepsin in mammary tumors increased TGFß signaling. Cell-free and cell-based experiments showed that hepsin is capable of direct proteolytic cleavage of fibronectin but not latent-TGFß and, importantly, that the ability of hepsin to activate TGFß signaling is dependent on fibronectin. Altogether, this study demonstrates a role for hepsin as a regulator of the TGFß pathway in the mammary gland via a novel mechanism involving proteolytic downmodulation of fibronectin.


Asunto(s)
Fibronectinas , Factor de Crecimiento Transformador beta , Animales , Fibronectinas/metabolismo , Ratones , Proteolisis , Serina Endopeptidasas/genética , Factor de Crecimiento Transformador beta/metabolismo
8.
Sci Rep ; 10(1): 18733, 2020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-33127915

RESUMEN

Breast cancer is the most common form of cancer in women. Despite significant therapeutic advances in recent years, breast cancer also still causes the greatest number of cancer-related deaths in women, the vast majority of which (> 90%) are caused by metastases. However, very few mouse mammary cancer models exist that faithfully recapitulate the multistep metastatic process in human patients. Here we assessed the suitability of a syngrafting protocol for a Myc-driven mammary tumor model (WAP-Myc) to study autochthonous metastasis. A moderate but robust spontaneous lung metastasis rate of around 25% was attained. In addition, increased T cell infiltration was observed in metastatic tumors compared to donor and syngrafted primary tumors. Thus, the WAP-Myc syngrafting protocol is a suitable tool to study the mechanisms of metastasis in MYC-driven breast cancer.


Asunto(s)
Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Proteínas de la Leche/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Ratones , Proteínas de la Leche/genética , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas c-myc/genética
9.
PLoS One ; 12(10): e0186628, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29040328

RESUMEN

SHARPIN (Shank-Associated RH Domain-Interacting Protein) is a component of the linear ubiquitin chain assembly complex (LUBAC), which enhances TNF-induced NF-κB activity. SHARPIN-deficient (Sharpincpdm/cpdm) mice display multi-organ inflammation and chronic proliferative dermatitis (cpdm) due to TNF-induced keratinocyte apoptosis. In cells, SHARPIN also inhibits integrins independently of LUBAC, but it has remained enigmatic whether elevated integrin activity levels in the dermis of Sharpincpdm/cpdm mice is due to increased integrin activity or is secondary to inflammation. In addition, the functional contribution of increased integrin activation to the Sharpincpdm/cpdm phenotype has not been investigated. Here, we find increased integrin activity in keratinocytes from Tnfr1-/- Sharpincpdm/cpdm double knockout mice, which do not display chronic inflammation or proliferative dermatitis, thus suggesting that SHARPIN indeed acts as an integrin inhibitor in vivo. In addition, we present evidence for a functional contribution of integrin activity to the Sharpincpdm/cpdm skin phenotype. Treatment with an integrin beta 1 function blocking antibody reduced epidermal hyperproliferation and epidermal thickness in Sharpincpdm/cpdm mice. Our data indicate that, while TNF-induced cell death triggers the chronic inflammation and proliferative dermatitis, absence of SHARPIN-dependent integrin inhibition exacerbates the epidermal hyperproliferation in Sharpincpdm/cpdm mice.


Asunto(s)
Proteínas Portadoras/genética , Dermatitis/tratamiento farmacológico , Epidermis/efectos de los fármacos , Integrina beta1/genética , Queratinocitos/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Animales , Anticuerpos Neutralizantes/farmacología , Apoptosis , Proteínas Portadoras/inmunología , Proliferación Celular , Enfermedad Crónica , Dermatitis/genética , Dermatitis/inmunología , Dermatitis/patología , Epidermis/inmunología , Epidermis/patología , Femenino , Eliminación de Gen , Regulación de la Expresión Génica , Inflamación , Integrina beta1/inmunología , Péptidos y Proteínas de Señalización Intracelular , Queratinocitos/inmunología , Queratinocitos/patología , Masculino , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/inmunología , Fenotipo , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Transducción de Señal , Ubiquitina/genética , Ubiquitina/inmunología
10.
J Cell Sci ; 130(18): 3094-3107, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28775156

RESUMEN

Sharpin, a multifunctional adaptor protein, regulates several signalling pathways. For example, Sharpin enhances signal-induced NF-κB signalling as part of the linear ubiquitin assembly complex (LUBAC) and inhibits integrins, the T cell receptor, caspase 1 and PTEN. However, despite recent insights into Sharpin and LUBAC function, a systematic approach to identify the signalling pathways regulated by Sharpin has not been reported. Here, we present the first 'Sharpin interactome', which identifies a large number of novel potential Sharpin interactors in addition to several known ones. These data suggest that Sharpin and LUBAC might regulate a larger number of biological processes than previously identified, such as endosomal trafficking, RNA processing, metabolism and cytoskeleton regulation. Importantly, using the Sharpin interactome, we have identified a novel role for Sharpin in lamellipodium formation. We demonstrate that Sharpin interacts with Arp2/3, a protein complex that catalyses actin filament branching. We have identified the Arp2/3-binding site in Sharpin and demonstrate using a specific Arp2/3-binding deficient mutant that the Sharpin-Arp2/3 interaction promotes lamellipodium formation in a LUBAC-independent fashion.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Mapas de Interacción de Proteínas , Seudópodos/metabolismo , Movimiento Celular , Citoesqueleto/metabolismo , Ontología de Genes , Células HeLa , Humanos , Espectrometría de Masas , Unión Proteica , Imagen de Lapso de Tiempo
11.
Nat Cell Biol ; 19(4): 292-305, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28263956

RESUMEN

SHANK3, a synaptic scaffold protein and actin regulator, is widely expressed outside of the central nervous system with predominantly unknown function. Solving the structure of the SHANK3 N-terminal region revealed that the SPN domain is an unexpected Ras-association domain with high affinity for GTP-bound Ras and Rap G-proteins. The role of Rap1 in integrin activation is well established but the mechanisms to antagonize it remain largely unknown. Here, we show that SHANK1 and SHANK3 act as integrin activation inhibitors by sequestering active Rap1 and R-Ras via the SPN domain and thus limiting their bioavailability at the plasma membrane. Consistently, SHANK3 silencing triggers increased plasma membrane Rap1 activity, cell spreading, migration and invasion. Autism-related mutations within the SHANK3 SPN domain (R12C and L68P) disrupt G-protein interaction and fail to counteract integrin activation along the Rap1-RIAM-talin axis in cancer cells and neurons. Altogether, we establish SHANKs as critical regulators of G-protein signalling and integrin-dependent processes.


Asunto(s)
Integrina beta1/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Proteínas ras/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Celular , Línea Celular , Movimiento Celular , Extensiones de la Superficie Celular/metabolismo , Femenino , Citometría de Flujo , Ratones Endogámicos C57BL , Modelos Biológicos , Mutación/genética , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Reacción en Cadena de la Polimerasa , Unión Proteica , Dominios Proteicos , Ratas Wistar , Alineación de Secuencia , Talina/metabolismo , Ubiquitinas/genética
13.
Nat Struct Mol Biol ; 23(2): 172-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26779610

RESUMEN

Integrins are heterodimeric cell-surface adhesion molecules comprising one of 18 possible α-chains and one of eight possible ß-chains. They control a range of cell functions in a matrix- and ligand-specific manner. Integrins can be internalized by clathrin-mediated endocytosis (CME) through ß subunit-based motifs found in all integrin heterodimers. However, whether specific integrin heterodimers can be selectively endocytosed was unknown. Here, we found that a subset of α subunits contain an evolutionarily conserved and functional YxxΦ motif directing integrins to selective internalization by the most abundant endocytic clathrin adaptor, AP2. We determined the structure of the human integrin α4-tail motif in complex with the AP2 C-µ2 subunit and confirmed the interaction by isothermal titration calorimetry. Mutagenesis of the motif impaired selective heterodimer endocytosis and attenuated integrin-mediated cell migration. We propose that integrins evolved to enable selective integrin-receptor turnover in response to changing matrix conditions.


Asunto(s)
Complejo 2 de Proteína Adaptadora/metabolismo , Endocitosis , Integrina alfa2/metabolismo , Integrina alfa4/metabolismo , Complejo 2 de Proteína Adaptadora/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Adhesión Celular , Movimiento Celular , Humanos , Integrina alfa2/química , Integrina alfa4/química , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína , Alineación de Secuencia
14.
PLoS One ; 10(11): e0143423, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26600301

RESUMEN

SHANK-associated RH domain interactor (SHARPIN) inhibits integrins through interaction with the integrin α-subunit. In addition, SHARPIN enhances nuclear factor-kappaB (NF-κB) activity as a component of the linear ubiquitin chain assembly complex (LUBAC). However, it is currently unclear how regulation of these seemingly different roles is coordinated. Here, we show that SHARPIN binds integrin and LUBAC in a mutually exclusive manner. We map the integrin binding site on SHARPIN to the ubiquitin-like (UBL) domain, the same domain implicated in SHARPIN interaction with LUBAC component RNF31 (ring finger protein 31), and identify two SHARPIN residues (V267, L276) required for both integrin and RNF31 regulation. Accordingly, the integrin α-tail is capable of competing with RNF31 for SHARPIN binding in vitro. Importantly, the full SHARPIN RNF31-binding site contains residues (F263A/I272A) that are dispensable for SHARPIN-integrin interaction. Importantly, disrupting SHARPIN interaction with integrin or RNF31 abolishes SHARPIN-mediated regulation of integrin or NF-κB activity, respectively. Altogether these data suggest that the roles of SHARPIN in inhibiting integrin activity and supporting linear ubiquitination are (molecularly) distinct.


Asunto(s)
Integrinas/metabolismo , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Movimiento Celular , Humanos , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas del Tejido Nervioso/química , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Ubiquitina-Proteína Ligasas
15.
Dev Cell ; 34(4): 475-83, 2015 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-26256210

RESUMEN

Regulated turnover of integrin receptors is essential for cell adhesion and migration. Pathways selectively regulating ß1-integrin recycling are implicated in cancer invasion and metastasis, yet proteins required for the internalization of this pro-invasive integrin remain to be identified. Here, we uncover formin-like 2 (FMNL2) as a critical regulator of ß1-integrin internalization downstream of protein kinase C (PKC). PKCα associates with and phosphorylates FMNL2 at S1072 within its Diaphanous autoregulatory region, leading to the release of formin autoinhibition. Phosphorylation of FMNL2 triggers its rapid relocation and promotes its interaction with the cytoplasmic tails of the α-integrin subunits for ß1-integrin endocytosis. FMNL2 drives ß1-integrin internalization and invasive motility in a phosphorylation-dependent manner, while a FMNL2 mutant defective in actin assembly interferes with ß1-integrin endocytosis and cancer cell invasion. Our data establish a role for FMNL2 in the regulation of ß1-integrin and provide a mechanistic understanding of the function of FMNL2 in cancer invasiveness.


Asunto(s)
Movimiento Celular , Integrina beta1/metabolismo , Proteína Quinasa C-alfa/metabolismo , Proteínas/metabolismo , Secuencia de Aminoácidos , Citoplasma/metabolismo , Endocitosis , Endosomas/metabolismo , Activación Enzimática , Forminas , Células HEK293 , Células HeLa , Humanos , Integrina beta1/química , Membranas Intracelulares/metabolismo , Datos de Secuencia Molecular , Invasividad Neoplásica , Fosforilación , Fosfoserina/metabolismo , Unión Proteica , Proteínas/química
16.
Cell Rep ; 5(3): 619-28, 2013 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-24210817

RESUMEN

SHARPIN-deficient mice display a multiorgan chronic inflammatory phenotype suggestive of altered leukocyte migration. We therefore studied the role of SHARPIN in lymphocyte adhesion, polarization, and migration. We found that SHARPIN localizes to the trailing edges (uropods) of both mouse and human chemokine-activated lymphocytes migrating on intercellular adhesion molecule-1 (ICAM-1), which is one of the major endothelial ligands for migrating leukocytes. SHARPIN-deficient cells adhere better to ICAM-1 and show highly elongated tails when migrating. The increased tail lifetime in SHARPIN-deficient lymphocytes decreases the migration velocity. The adhesion, migration, and uropod defects in SHARPIN-deficient lymphocytes were rescued by reintroducing SHARPIN into the cells. Mechanistically, we show that SHARPIN interacts directly with lymphocyte-function-associated antigen-1 (LFA-1), a leukocyte counterreceptor for ICAM-1, and inhibits the expression of intermediate and high-affinity forms of LFA-1. Thus, SHARPIN controls lymphocyte migration by endogenously maintaining LFA-1 inactive to allow adjustable detachment of the uropods in polarized cells.


Asunto(s)
Proteínas Portadoras/metabolismo , Movimiento Celular/fisiología , Extensiones de la Superficie Celular/metabolismo , Linfocitos/citología , Linfocitos/metabolismo , Ubiquitinas/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Celular/fisiología , Polaridad Celular/fisiología , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular
17.
PLoS One ; 8(8): e70696, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23967087

RESUMEN

Recently, we demonstrated that integrin adhesion to the extracellular matrix at the cleavage furrow is essential for cytokinesis of adherent cells. Here, we report that tight junction protein ZO-1 (Zonula Occludens-1) is required for successful cytokinesis in NCI-H460 cells plated on fibronectin. This function of ZO-1 involves interaction with the cytoplasmic domain of α5-integrin to facilitate recruitment of active fibronectin-binding integrins to the base of the cleavage furrow. In the absence of ZO-1, or a functional ZO-1/α5ß1-integrin complex, proper actin-dependent constriction between daughter cells is impaired and cells fail cytokinesis. Super-resolution microscopy reveals that in ZO-1 depleted cells the furrow becomes delocalized from the matrix. We also show that PKCε-dependent phosphorylation at Serine168 is required for ZO-1 localization to the furrow and successful cell division. Altogether, our results identify a novel regulatory pathway involving the interplay between ZO-1, α5-integrin and PKCε in the late stages of mammalian cell division.


Asunto(s)
Citocinesis , Integrina alfa5beta1/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Proteína de la Zonula Occludens-1/metabolismo , Adhesión Celular , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Células Cultivadas , Fibronectinas/metabolismo , Humanos , Modelos Biológicos , Fosfatidilinositol 4,5-Difosfato/metabolismo , Unión Proteica , Transducción de Señal
18.
Nat Rev Mol Cell Biol ; 14(7): 430-42, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23719537

RESUMEN

Integrins mediate cell-matrix and cell-cell interactions and integrate extracellular cues to the cytoskeleton and cellular signalling pathways. Integrin function on the cell surface is regulated by their activity switching such that intracellular proteins interacting with the integrin cytoplasmic domains increase or decrease integrin-ligand binding affinity. It is widely accepted that integrin activation by specific proteins is essential for cell adhesion and integrin linkage to the actin cytoskeleton. However, there is also increasing evidence that integrin-inactivating proteins are crucial for appropriate integrin function in vitro and in vivo and that the regulation of integrin-ligand interactions is a fine-tuned balancing act between inactivation and activation.


Asunto(s)
Integrinas/fisiología , Transducción de Señal , Secuencia de Aminoácidos , Animales , Humanos , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Datos de Secuencia Molecular , Neoplasias/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional
19.
Carcinogenesis ; 34(2): 436-45, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23104179

RESUMEN

Mitosis represents a clinically important determination point in the life cycle of proliferating cells. One potential drug target within the mitotic machinery is the spindle assembly checkpoint (SAC), an evolutionarily conserved signaling pathway that monitors the connections between microtubules (MTs) and chromosomes. Mistakes in SAC signaling may lead to cell division errors that can trigger elimination of cancer cells at M phase or soon after exit from mitosis. In this study, we describe the cellular effects of a novel pyrimidine-2,4-diamine derivative that we discovered to inhibit the activity of SAC. The compound caused rapid escape from the mitotic arrest induced by lack of interkinetochore tension but not by lack of MT-kinetochore attachments. In cycling cells, the compound disrupted the architecture of mitotic spindle that triggered a transient M-phase arrest that was rapidly followed by a forced mitotic exit. The premature termination of M phase was found to be a consequence of precocious inactivation of SAC caused by a direct inhibitory effect of the compound on Aurora B kinase in vitro and in cells. The compound also targets Aurora A kinase and tubulin in vitro and in cells, which can explain the observed spindle anomalies. The reduced activity of Aurora B kinase resulted in polyploidy and suppression of cancer cell viability. Our data suggest that this new pharmacophore possesses interesting anticancer properties that could be exploited in development of mitosis-targeting therapies.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Neoplasias/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Huso Acromático/efectos de los fármacos , Aurora Quinasa B , Aurora Quinasas , Western Blotting , Técnica del Anticuerpo Fluorescente , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
20.
J Cell Sci ; 125(Pt 14): 3271-80, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22822081

RESUMEN

Integrins are heterodimeric transmembrane adhesion receptors composed of α- and ß-subunits. They are ubiquitously expressed and have key roles in a number of important biological processes, such as development, maintenance of tissue homeostasis and immunological responses. The activity of integrins, which indicates their affinity towards their ligands, is tightly regulated such that signals inside the cell cruicially regulate the switching between active and inactive states. An impaired ability to activate integrins is associated with many human diseases, including bleeding disorders and immune deficiencies, whereas inappropriate integrin activation has been linked to inflammatory disorders and cancer. In recent years, the molecular details of integrin 'inside-out' activation have been actively investigated. Binding of cytoplasmic proteins, such as talins and kindlins, to the cytoplasmic tail of ß-integrins is widely accepted as being the crucial step in integrin activation. By contrast, much less is known with regard to the counteracting mechanism involved in switching integrins into an inactive conformation. In this Commentary, we aim to discuss the known mechanisms of integrin inactivation and the molecules involved.


Asunto(s)
Integrinas/antagonistas & inhibidores , Integrinas/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Celular/fisiología , Humanos , Cadenas alfa de Integrinas/metabolismo , Cadenas beta de Integrinas/metabolismo , Ratones , Datos de Secuencia Molecular , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...