Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Nat Commun ; 15(1): 5638, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38965263

RESUMEN

Triple Negative Breast Cancer (TNBC) is the most aggressive breast cancer subtype suffering from limited targeted treatment options. Following recent reports correlating Fibroblast growth factor-inducible 14 (Fn14) receptor overexpression in Estrogen Receptor (ER)-negative breast cancers with metastatic events, we show that Fn14 is specifically overexpressed in TNBC patients and associated with poor survival. We demonstrate that constitutive Fn14 signalling rewires the transcriptomic and epigenomic landscape of TNBC, leading to enhanced tumour growth and metastasis. We further illustrate that such mechanisms activate TNBC-specific super enhancers (SE) to drive the transcriptional activation of cancer dependency genes via chromatin looping. In particular, we uncover the SE-driven upregulation of Nicotinamide phosphoribosyltransferase (NAMPT), which promotes NAD+ and ATP metabolic reprogramming critical for filopodia formation and metastasis. Collectively, our study details the complex mechanistic link between TWEAK/Fn14 signalling and TNBC metastasis, which reveals several vulnerabilities which could be pursued for the targeted treatment of TNBC patients.


Asunto(s)
Citocina TWEAK , Regulación Neoplásica de la Expresión Génica , Nicotinamida Fosforribosiltransferasa , Transducción de Señal , Receptor de TWEAK , Neoplasias de la Mama Triple Negativas , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Humanos , Receptor de TWEAK/metabolismo , Receptor de TWEAK/genética , Femenino , Citocina TWEAK/metabolismo , Citocina TWEAK/genética , Nicotinamida Fosforribosiltransferasa/metabolismo , Nicotinamida Fosforribosiltransferasa/genética , Animales , Línea Celular Tumoral , Ratones , Metástasis de la Neoplasia , Citocinas/metabolismo , Elementos de Facilitación Genéticos/genética
2.
Int J Mol Sci ; 24(1)2023 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-36614196

RESUMEN

Large numbers of neutrophils infiltrate tumors and comprise a notable component of the inflammatory tumor microenvironment. While it is established that tumor cells exhibit the Warburg effect for energy production, the contribution of the neutrophil metabolic state to tumorigenesis is unknown. Here, we investigated whether neutrophil infiltration and metabolic status promotes tumor progression in an orthotopic mouse model of pancreatic ductal adenocarcinoma (PDAC). We observed a large increase in the proportion of neutrophils in the blood and tumor upon orthotopic transplantation. Intriguingly, these tumor-infiltrating neutrophils up-regulated glycolytic factors and hypoxia-inducible factor 1-alpha (HIF-1α) expression compared to neutrophils from the bone marrow and blood of the same mouse. This enhanced glycolytic signature was also observed in human PDAC tissue samples. Strikingly, neutrophil-specific deletion of HIF-1α (HIF-1αΔNφ) significantly reduced tumor burden and improved overall survival in orthotopic transplanted mice, by converting the pro-tumorigenic neutrophil phenotype to an anti-tumorigenic phenotype. This outcome was associated with elevated reactive oxygen species production and activated natural killer cells and CD8+ cytotoxic T cells compared to littermate control mice. These data suggest a role for HIF-1α in neutrophil metabolism, which could be exploited as a target for metabolic modulation in cancer.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Animales , Ratones , Neutrófilos/metabolismo , Línea Celular Tumoral , Ratones Noqueados , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/patología , Carcinogénesis , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Microambiente Tumoral/genética , Neoplasias Pancreáticas
3.
FEBS J ; 290(13): 3336-3354, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-35816016

RESUMEN

The actin cytoskeleton (AC) undergoes rapid remodelling to coordinate cellular processes during signal transduction, including changes in actin nucleation, crosslinking, and depolymerization in a time- and space-dependent manner. Switching the initial actin nucleation often provides timely control of the entire actin network formation. Located at the cell surface, the plant class I formin family is a major class of actin nucleators that rapidly respond to exterior chemical and environmental cues. Plant class I formins are structurally integrated within the plant cell wall-plasma membrane-actin cytoskeleton (CW-PM-AC) continuum, sharing similar biophysical properties to mammalian integrins that are embedded within the extracellular matrix-PM-AC continuum. In plants, perturbation of structural components of the CW-PM-AC continuum changes the biophysical properties of two dimensional-scaffolding structures, which results in uncontrolled molecular diffusion and interactions of class I formins, as well as their clustering and activities in the nucleation of the AC. Emerging studies have shown that the PM-integrated formins are highly responsive to the mechanical perturbation of CW and AC integrity changes that tune the oligomerization and condensation of formin on the cell surface. However, during diverse signalling transductions, the molecular mechanisms that spatiotemporally underlie the mechanosensing and mechanoregulation of formin for remodelling actin remain unclear. Here, the emphasis will be placed on recent developments in understanding how the molecular condensation of class I formin regulates the biochemical activities in tuning actin polymerization during plant immune signalling, as well as how the plant structural components of the CW-PM-AC continuum control formin condensation at a nanometre scale.


Asunto(s)
Actinas , Proteínas de Microfilamentos , Animales , Actinas/metabolismo , Forminas/metabolismo , Proteínas de Microfilamentos/metabolismo , Integrinas/metabolismo , Citoesqueleto de Actina/metabolismo , Plantas/metabolismo , Mamíferos/metabolismo
4.
Leukemia ; 37(2): 441-452, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36446947

RESUMEN

DLBCL is the most common lymphoma with high tumor heterogeneity. Treatment refractoriness and relapse from R-CHOP therapy in patients remain a clinical problem. Activation of the non-canonical NF-κB pathway is associated with R-CHOP resistance. However, downstream targets of non-canonical NF-κB mediating R-CHOP-induced resistance remains uncharacterized. Here, we identify the common mechanisms underlying both intrinsic and acquired resistance that are induced by doxorubicin, the main cytotoxic component of R-CHOP. We performed global transcriptomic analysis of (1) a panel of resistant versus sensitive and (2) isogenic acquired doxorubicin-resistant DLBCL cell lines following short and chronic exposure to doxorubicin respectively. Doxorubicin-induced stress in resistant cells activates a distinct transcriptional signature that is enriched in metabolic reprogramming and oncogenic signalling. Selective and sustained activation of non-canonical NF-κB signalling in these resistant cells exacerbated their survival by augmenting glycolysis. In response to doxorubicin, p52-RelB complexes transcriptionally activated multiple glycolytic regulators with prognostic significance through increased recruitment at their gene promoters. Targeting p52-RelB and their targets in resistant cells increased doxorubicin sensitivity in vitro and in vivo. Collectively, our study uncovered novel molecular drivers of doxorubicin-induced resistance that are regulated by non-canonical NF-κB pathway. We reveal new avenues of therapeutic targeting for R-CHOP-treated refractory/relapsed DLBCL patients.


Asunto(s)
Antineoplásicos , Linfoma de Células B Grandes Difuso , Humanos , FN-kappa B/metabolismo , Recurrencia Local de Neoplasia/tratamiento farmacológico , Transducción de Señal , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Antineoplásicos/uso terapéutico , Rituximab/farmacología , Rituximab/uso terapéutico , Ciclofosfamida/uso terapéutico , Vincristina/farmacología , Vincristina/uso terapéutico , Prednisona/uso terapéutico , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico
5.
Int J Mol Sci ; 24(1)2022 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-36613707

RESUMEN

Many antibiotics are ineffective in killing Gram-negative bacteria due to the permeability barrier of the outer-membrane LPS. Infections caused by multi-drug-resistant Gram-negative pathogens require new antibiotics, which are often difficult to develop. Antibiotic potentiators disrupt outer-membrane LPS and can assist the entry of large-scaffold antibiotics to the bacterial targets. In this work, we designed a backbone-cyclized ultra-short, six-amino-acid-long (WKRKRY) peptide, termed cWY6 from LPS binding motif of ß-boomerang bactericidal peptides. The cWY6 peptide does not exhibit any antimicrobial activity; however, it is able to permeabilize the LPS outer membrane. Our results demonstrate the antibiotic potentiator activity in the designed cWY6 peptide for several conventional antibiotics (vancomycin, rifampicin, erythromycin, novobiocin and azithromycin). Remarkably, the short cWY6 peptide exhibits wound-healing activity in in vitro assays. NMR, computational docking and biophysical studies describe the atomic-resolution structure of the peptide in complex with LPS and mode of action in disrupting the outer membrane. The dual activities of cWY6 peptide hold high promise for further translation to therapeutics.


Asunto(s)
Antibacterianos , Lipopolisacáridos , Lipopolisacáridos/metabolismo , Antibacterianos/farmacología , Antibacterianos/química , Péptidos Catiónicos Antimicrobianos/química , Azitromicina/farmacología , Rifampin/farmacología , Pruebas de Sensibilidad Microbiana , Bacterias Gramnegativas
6.
Biochim Biophys Acta Gen Subj ; 1865(12): 130005, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34509570

RESUMEN

BACKGROUND: Cytoskeletal protein filamin A is critical for the outside-in signaling of integrins. Although molecular mechanisms of filamin-integrin interactions are not fully understood. Mostly, the membrane distal (MD) part of the cytosolic tail (CT) of ß subunit of integrin is known to interact with filamin A domain 21 (FLNa-Ig2). However, binary and ternary complexes of full-length CTs of leucocyte specific ß2 integrins with FLNa-Ig21 are yet to be elucidated. METHODS: Binding interactions of the CTs of integrin αMß2 with FLNa-Ig21 are extensively investigated by NMR, ITC, cell-based functional assays and computational docking. RESULTS: The αM CT demonstrates interactions with FLNa-Ig21 forming a binary complex. Filamin/αM interface is mediated by sidechain-sidechain interactions among non-polar and aromatic residues involving MP helix of αM and the canonical CD face of FLNa-Ig21. Functional assays delineated an interfacial residue Y1137 of αM CT is critical for in-cell binding to FLNa-Ig2. In addition, full-length ß2 CT occupies two distinct binding sites in complex with FLNa-Ig21. A ternary complex of FLNa-Ig21 with CTs has been characterized. In the ternary complex, αM CT moves away to a distal site of FLNa-Ig21 with fewer interactions. CONCLUSION: Our findings demonstrate a plausible dual role of filamin in integrin regulation. The molecular interactions of the ternary complex are critical for the resting state of integrins whereas stable FLNa-Ig21/αM CT binary complex perhaps be required for the activated state. GENERAL SIGNIFICANCE: Filamin binding to both α and ß CTs of other integrins could be essential in regulating bidirectional signaling mechanisms.


Asunto(s)
Citosol , Comunicación Celular , Filaminas
7.
J Cell Sci ; 134(8)2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33912917

RESUMEN

Integrin-mediated cell-extracellular matrix (ECM) interactions play crucial roles in a broad range of physiological and pathological processes. Kindlins are important positive regulators of integrin activation. The FERM-domain-containing kindlin family comprises three members, kindlin-1, kindlin-2 and kindlin-3 (also known as FERMT1, FERMT2 and FERMT3), which share high sequence similarity (identity >50%), as well as domain organization, but exhibit diverse tissue-specific expression patterns and cellular functions. Given the significance of kindlins, analysis of their atomic structures has been an attractive field for decades. Recently, the structures of kindlin and its ß-integrin-bound form have been obtained, which greatly advance our understanding of the molecular functions that involve kindlins. In particular, emerging evidence indicates that oligomerization of kindlins might affect their integrin binding and focal adhesion localization, positively or negatively. In this Review, we presented an update on the recent progress of obtaining kindlin structures, and discuss the implication for integrin activation based on kindlin oligomerization, as well as the possible regulation of this process.


Asunto(s)
Proteínas de la Membrana , Proteínas de Neoplasias , Adhesión Celular , Adhesiones Focales , Integrinas/genética , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética
8.
PLoS Biol ; 18(7): e3000755, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32644996

RESUMEN

Kindlin-1, -2, and -3 directly bind integrin ß cytoplasmic tails to regulate integrin activation and signaling. Despite their functional significance and links to several diseases, structural information on full-length kindlin proteins remains unknown. Here, we report the crystal structure of human full-length kindlin-3, which reveals a novel homotrimer state. Unlike kindlin-3 monomer, which is the major population in insect and mammalian cell expression systems, kindlin-3 trimer does not bind integrin ß cytoplasmic tail as the integrin-binding pocket in the F3 subdomain of 1 protomer is occluded by the pleckstrin homology (PH) domain of another protomer, suggesting that kindlin-3 is auto-inhibited upon trimer formation. This is also supported by functional assays in which kindlin-3 knockout K562 erythroleukemia cells reconstituted with the mutant kindlin-3 containing trimer-disrupting mutations exhibited an increase in integrin-mediated adhesion and spreading on fibronectin compared with those reconstituted with wild-type kindlin-3. Taken together, our findings reveal a novel mechanism of kindlin auto-inhibition that involves its homotrimer formation.


Asunto(s)
Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/química , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Multimerización de Proteína , Movimiento Celular , Humanos , Integrinas/metabolismo , Células K562 , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Proteínas de Neoplasias/metabolismo , Unión Proteica , Dominios Proteicos , Homología Estructural de Proteína , Relación Estructura-Actividad
9.
J Biol Chem ; 295(18): 5928-5943, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32169902

RESUMEN

Kindlins are focal adhesion proteins that regulate integrin activation and outside-in signaling. The kindlin family consists of three members, kindlin-1, -2, and -3. Kindlin-2 is widely expressed in multiple cell types, except those from the hematopoietic lineage. A previous study has reported that the Drosophila Fit1 protein (an ortholog of kindlin-2) prevents abnormal spindle assembly; however, the mechanism remains unknown. Here, we show that kindlin-2 maintains spindle integrity in mitotic human cells. The human neuroblastoma SH-SY5Y cell line expresses only kindlin-2, and we found that when SH-SY5Y cells are depleted of kindlin-2, they exhibit pronounced spindle abnormalities and delayed mitosis. Of note, acetylation of α-tubulin, which maintains microtubule flexibility and stability, was diminished in the kindlin-2-depleted cells. Mechanistically, we found that kindlin-2 maintains α-tubulin acetylation by inhibiting the microtubule-associated deacetylase histone deacetylase 6 (HDAC6) via a signaling pathway involving AKT Ser/Thr kinase (AKT)/glycogen synthase kinase 3ß (GSK3ß) or paxillin. We also provide evidence that prolonged hypoxia down-regulates kindlin-2 expression, leading to spindle abnormalities not only in the SH-SY5Y cell line, but also cell lines derived from colon and breast tissues. The findings of our study highlight that kindlin-2 regulates mitotic spindle assembly and that this process is perturbed in cancer cells in a hypoxic environment.


Asunto(s)
Histona Desacetilasa 6/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Mitosis , Proteínas de Neoplasias/metabolismo , Huso Acromático/metabolismo , Tubulina (Proteína)/metabolismo , Acetilación , Línea Celular Tumoral , Regulación hacia Abajo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Paxillin/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Hipoxia Tumoral
10.
J Mol Biol ; 430(21): 4419-4430, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30243836

RESUMEN

Integrins are hetero-dimeric (α and ß subunits) type I transmembrane proteins that facilitate cell adhesion and migration. The cytoplasmic tails (CTs) of integrins interact with a plethora of intra-cellular proteins that are required for integrin bidirectional signaling. In particular, the ß CTs of integrins are known to recruit a variety of cytosolic proteins that often have overlapping recognition sites. However, the chronological sequence of ß CTs/cytosolic proteins interactions remains to be fully characterized. Previous studies have shown that the scaffold protein 14-3-3ζ binds to phosphorylated ß CTs in activated integrins, whereas interactions of Dok-1 with phosphorylated ß CTs maintained integrins in the resting state. In this study, we examined the binding interactions between 14-3-3ζ, Dok1, and phosphorylated integrin ß2 and ß3 CTs. We show that the scaffold protein 14-3-3ζ interacts with the phosphotyrosine binding (PTB) domain of Dok1 even in the absence of the phosphorylated integrin ß CTs. The interactions were mapped onto the ß-sheet region of the PTB domain of Dok1. Furthermore, we provide evidence that the 14-3-3ζ/Dok1 binary complex is able to bind to their cognate phosphorylated sequence motifs in the integrin ß CTs. We demonstrate that Thr phosphorylated pTTT ß2 CT or pTST ß3 CT can bind to 14-3-3ζ that is in complex with the Dok1 PTB domain, whereas Ser phosphorylated ß2 CT or Tyr phosphorylated ß3 CT interacted with Dok1 in 14-3-3ζ/Dok1 complex. Based on these data, we propose that 14-3-3ζ/Dok1 complex could serve as a molecular switch providing novel molecular insights into the regulating integrin activation.


Asunto(s)
Proteínas 14-3-3/metabolismo , Proteínas de Unión al ADN/metabolismo , Cadenas beta de Integrinas/metabolismo , Integrina beta3/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas 14-3-3/química , Sitios de Unión , Citoplasma/metabolismo , Proteínas de Unión al ADN/química , Humanos , Cadenas beta de Integrinas/química , Integrina beta3/química , Modelos Moleculares , Fosfoproteínas/química , Fosforilación , Unión Proteica , Conformación Proteica en Lámina beta , Dominios Proteicos , Proteínas de Unión al ARN/química , Treonina/metabolismo
11.
Sci Rep ; 8(1): 5490, 2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29615775

RESUMEN

Integrins are transmembrane proteins that mediate cell adhesion and migration. Each integrin is a heterodimer formed by an α and a ß subunit. A large number of cytoplasmic proteins interact with the cytoplasmic tails (CTs) of integrins. The actin-binding cytoskeletal protein filamin A is a negative regulator of integrin activation. The IgFLNa21 domain of filamin A binds to the C-terminus of ß2 CT that contains a TTT-motif. Based on x-ray crystallography, it has been reported that the integrin ß2 CT forms a ß strand that docks into the ß strands C and D of IgFLNa21. In this study, we performed solution NMR analyses of IgFLNa21 in the presence of integrin ß2 CT peptides, and hybrid IgFLNa21, a construct of covalently linked IgFLNa21 and ß2 CT. The atomic resolution structure of the hybrid IgFLNa21 demonstrated conserved binding mode with ß2 CT. Although, 15N relaxation, model free analyses and H-D exchange studies have uncovered important insights into the conformational dynamics and stability of ß2 CT in complex with IgFLNa21. Such dynamical characteristics are likely to be necessary for the TTT-motif to serve as a phosphorylation switch that regulates filamin A binding to integrin ß2 CT.


Asunto(s)
Antígenos CD18/química , Antígenos CD18/metabolismo , Citoplasma/metabolismo , Filaminas/química , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Unión Proteica , Dominios Proteicos
12.
FEBS Lett ; 592(1): 112-121, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29237230

RESUMEN

Focal adhesion (FA) proteins, kindlin-2 and integrin-linked kinase (ILK), regulate cell adhesion and migration. ILK interacts with and promotes kindlin-2 targeting to FAs. Leu353 and Leu357 in kindlin-2 have been reported to be important for the interaction between kindlin-2 and ILK. However, the binding interface between kindlin-2 and ILK remains unclear. Using molecular modeling and molecular dynamics simulations, we show that Asp344, Asp352, and Thr356 in kindlin-2 and Arg243 and Arg334 in ILK kinase domain (KD) are important in kindlin-2/ILK complex formation. Mutations that disrupt these interactions abrogate kindlin-2 and ILK colocalization in HeLa cells. The interactions are direct based on data from pull-down assays using purified recombinant kindlin-2 F2-pleckstrin homology and ILK KDs. These data provide additional insights into the binding interface between kindlin-2 and ILK.


Asunto(s)
Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Adhesión Celular/genética , Adhesión Celular/fisiología , Movimiento Celular/genética , Movimiento Celular/fisiología , Células HeLa , Humanos , Proteínas de la Membrana/genética , Modelos Moleculares , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Proteínas de Neoplasias/genética , Dominios y Motivos de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Electricidad Estática
13.
J Immunol ; 198(2): 883-894, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27974454

RESUMEN

The leukocyte integrin αMß2 (CR3 or Mac-1) has both proinflammatory and immune regulatory functions. Genome-wide association studies have identified several ITGAM (αM subunit) single nucleotide polymorphisms that are associated with systemic lupus erythematosus. The single nucleotide polymorphism rs1143678 substitutes Pro1146 for Ser in the integrin αM cytoplasmic tail. A detailed functional characterization of this substitution is lacking. Using transfected human cell lines, reconstituted mouse bone marrow neutrophils, and bone marrow-derived macrophages (BMDMs), we showed that P1146S (PS) substitution promoted integrin αMß2-mediated adhesion, spreading, and migration of cells on iC3b and fibrinogen. In the presence of LPS together with iC3b or fibrinogen, the expression levels of IL-6 and TNF-α in integrin αM(PS)ß2 BMDMs were significantly higher than those of integrin αM(wild-type)ß2 BMDMs, and they showed faster kinetics of Erk1/2 activation through the src family kinase(s)-Syk signaling pathway. Integrin αM(PS)ß2 BMDMs also exhibited higher levels of active RhoA and phagocytic activity. Mechanistically, P1146S substitution in the αM cytoplasmic tail generates a noncanonical 14-3-3ζ binding site that modulates integrin αM(PS)ß2 outside-in signaling.


Asunto(s)
Proteínas 14-3-3/metabolismo , Lupus Eritematoso Sistémico/genética , Antígeno de Macrófago-1/genética , Macrófagos/metabolismo , Animales , Sitios de Unión , Citometría de Flujo , Transferencia Resonante de Energía de Fluorescencia , Humanos , Immunoblotting , Inmunoprecipitación , Lupus Eritematoso Sistémico/metabolismo , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple
14.
Cell Adh Migr ; 11(5-6): 419-433, 2017 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-27715393

RESUMEN

Kindlins are a small family of 4.1-ezrin-radixin-moesin (FERM)-containing cytoplasmic proteins. Kindlin-3 is expressed in platelets, hematopoietic cells, and endothelial cells. Kindlin-3 promotes integrin activation, clustering and outside-in signaling. Aberrant expression of kindlin-3 was reported in melanoma and breast cancer. Intriguingly, kindlin-3 has been reported to either positively or negatively regulate cancer cell metastasis. In this study, we sought to clarify the expression of kindlin-3 in melanoma cells and its role in melanoma metastasis. Two widely used metastatic mouse and human melanoma cell lines B16-F10 and M10, respectively, were examined and found to lack kindlin-3 mRNA and protein expression. When kindlin-3 was ectopically expressed in these cells, cell migration was markedly reduced. These are attributed to aberrant Rac1 and RhoA activation and overt membrane ruffling. Our data demonstrate for the first time that despite its well established role as a positive regulator of integrin-mediated cell adhesion, aberrant expression of kindlin-3 could lead to imbalanced RhoGTPases signaling that impedes rather than promotes cell migration.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Melanoma/metabolismo , Proteínas de la Membrana/metabolismo , Metástasis de la Neoplasia/genética , Proteínas de Neoplasias/metabolismo , Animales , Adhesión Celular/genética , Adhesión Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/fisiología , Proteínas del Citoesqueleto/genética , Humanos , Melanoma/genética , Proteínas de la Membrana/genética , Ratones , Proteínas de Neoplasias/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
15.
J Exp Med ; 213(11): 2293-2314, 2016 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-27811056

RESUMEN

It is well established that Ly6Chi monocytes develop from common monocyte progenitors (cMoPs) and reside in the bone marrow (BM) until they are mobilized into the circulation. In our study, we found that BM Ly6Chi monocytes are not a homogenous population, as current data would suggest. Using computational analysis approaches to interpret multidimensional datasets, we demonstrate that BM Ly6Chi monocytes consist of two distinct subpopulations (CXCR4hi and CXCR4lo subpopulations) in both mice and humans. Transcriptome studies and in vivo assays revealed functional differences between the two subpopulations. Notably, the CXCR4hi subset proliferates and is immobilized in the BM for the replenishment of functionally mature CXCR4lo monocytes. We propose that the CXCR4hi subset represents a transitional premonocyte population, and that this sequential step of maturation from cMoPs serves to maintain a stable pool of BM monocytes. Additionally, reduced CXCR4 expression on monocytes, upon their exit into the circulation, does not reflect its diminished role in monocyte biology. Specifically, CXCR4 regulates monocyte peripheral cellular activities by governing their circadian oscillations and pulmonary margination, which contributes toward lung injury and sepsis mortality. Together, our study demonstrates the multifaceted role of CXCR4 in defining BM monocyte heterogeneity and in regulating their function in peripheral tissues.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular , Monocitos/citología , Receptores CXCR4/metabolismo , Animales , Antígenos Ly/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Ritmo Circadiano/genética , Endotoxinas/toxicidad , Femenino , Perfilación de la Expresión Génica , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/metabolismo
16.
J Mol Biol ; 428(20): 4129-4142, 2016 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-27545410

RESUMEN

Integrins, which are heterodimeric (α and ß subunits) signal-transducer proteins, are essential for cell adhesion and migration. ß cytosolic tails (ß-CTs) of integrins interact with a number of cytosolic proteins including talin, Dok1, and 14-3-3ζ. The formation of multiprotein complexes with ß-CTs is involved in the activation and regulation of integrins. The leukocyte-specific ß2 integrins are essential for leukocyte trafficking, phagocytosis, antigen presentation, and proliferation. In this study, we examined the binding interactions between integrin ß2-CT and T758-phosphorylated ß2-CT with positive regulators talin and 14-3-3ζ and negative regulator Dok1. Residues of the F3 domain of talin belonging to the C-terminal helix, ß-strand 5, and the adjacent loop were found to be involved in the binding interactions with ß2-CT. The binding affinity between talin F3 and ß2-CT was reduced when ß2 T758 was phosphorylated, but this modification promoted 14-3-3ζ binding. However, we were able to detect stable ternary complex formation of T758-phosphorylated ß2-CT, talin F3, and 14-3-3ζ that involved the repositioning of talin F3 on ß2-CT. We showed that Dok1 binding to ß2-CT was reduced in the presence of 14-3-3ζ and when ß2 T758 was phosphorylated. Based on these data, we propose a sequential model of ß2 integrin activation involving these molecules. Our study provides for the first time insights toward ß2 integrin activation that involves a multiprotein complex.


Asunto(s)
Proteínas 14-3-3/metabolismo , Antígenos CD18/metabolismo , Multimerización de Proteína , Talina/metabolismo , Modelos Biológicos , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional
17.
PLoS One ; 11(4): e0153501, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27101375

RESUMEN

Kindlins-1,2 and 3 are FERM domain-containing cytosolic proteins involved in the activation and regulation of integrin-mediated cell adhesion. Apart from binding to integrin ß cytosolic tails, kindlins and the well characterized integrin-activator talin bind membrane phospholipids. The ubiquitin-like F1 sub-domain of the FERM domain of talin contains a short loop that binds to the lipid membrane. By contrast, the F1 sub-domain of kindlins contains a long loop demonstrated binding to the membrane. Here, we report structural characterization and lipid interactions of the 83-residue F1 loop of kindlin-3 using NMR and optical spectroscopy methods. NMR studies demonstrated that the F1 loop of kindlin-3 is globally unfolded but stretches of residues assuming transient helical conformations could be detected in aqueous solution. We mapped membrane binding interactions of the F1 loop with small unilamellar vesicles (SUVs) containing either zwitterionic lipids or negatively charged lipids using 15N-1H HSQC titrations. These experiments revealed that the F1 loop of kindlin-3 preferentially interacted with the negatively charged SUVs employing almost all of the residues. By contrast, only fewer residues appeared to be interacted with SUVs containing neutral lipids. Further, CD and NMR data suggested stabilization of helical conformations and predominant resonance perturbations of the F1 loop in detergent containing solutions. Conformations of an isolated N-terminal peptide fragment, or EK21, of the F1 loop, containing a poly-Lys sequence motif, important for membrane interactions, were also investigated in detergent solutions. EK21 adopted a rather extended or ß-type conformations in complex with negatively charged SDS micelles. To our knowledge, this is the first report describing the conformations and residue-specific interactions of kindlin F1 loop with lipids. These data therefore provide important insights into the interactions of kindlin FERM domain with membrane lipids that contribute toward the integrin activating property.


Asunto(s)
Lípidos de la Membrana/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Liposomas Unilamelares/metabolismo , Secuencia de Aminoácidos , Humanos , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Desplegamiento Proteico
18.
Data Brief ; 6: 803-10, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26937451

RESUMEN

Fibronectin and collagen type I are abundant extracellular matrix proteins that modulate cell mechanics and they regulate angiogenic sprouting. In this data article, fibronectin- or collagen type I-coated micro-posts were used to examine the traction force, cell spread area and directional contraction of human umbilical vein endothelial cells (HUVECs).

19.
J Invest Dermatol ; 136(2): 416-424, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26802238

RESUMEN

Deposition of immune complexes (ICs) in tissues triggers acute inflammatory pathology characterized by massive neutrophil influx leading to edema and hemorrhage, and is especially associated with vasculitis of the skin, but the mechanisms that regulate this type III hypersensitivity process remain poorly understood. Here, using a combination of multiphoton intravital microscopy and genomic approaches, we re-examined the cutaneous reverse passive Arthus reaction and observed that IC-activated neutrophils underwent transmigration, triggered further IC formation, and transported these ICs into the interstitium, whereas neutrophil depletion drastically reduced IC formation and ameliorated vascular leakage in vivo. Thereafter, we show that these neutrophils expressed high levels of CXCL2, which further amplified neutrophil recruitment and activation in an autocrine and/or paracrine manner. Notably, CXCL1 expression was restricted to tissue-resident cell types, but IC-activated neutrophils may also indirectly, via soluble factors, modulate macrophage CXCL1 expression. Consistent with their distinct cellular origins and localization, only neutralization of CXCL2 but not CXCL1 in the interstitium effectively reduced neutrophil recruitment. In summary, our study establishes that neutrophils are able to self-regulate their own recruitment and responses during IC-mediated inflammation through a CXCL2-driven feed forward loop.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Quimiocina CXCL2/metabolismo , Dermatitis/inmunología , Enfermedades del Complejo Inmune/inmunología , Neutrófilos/inmunología , Animales , Células Cultivadas , Quimiocina CXCL2/inmunología , Dermatitis/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Enfermedades del Complejo Inmune/fisiopatología , Mediadores de Inflamación/metabolismo , Macrófagos/inmunología , Masculino , Mastocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila/inmunología , Neutrófilos/metabolismo , ARN Mensajero/análisis
20.
Biochem Biophys Rep ; 7: 214-217, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28955909

RESUMEN

We showed that the αLß2 integrin with the non-functional mutation G150D cannot be induced with Mg/EGTA to express the mAb KIM127 epitope, which reports the leg-extended conformation. We extended the study to the αIIbß3, an integrin without an αI domain. The equivalent mutation, i.e. G161D, also resulted in an expressible, but non-adhesive αIIbß3 integrin. An NMR study of synthetic peptides spanning the α1-α1' helix of the ß3 I domain shows that both wild-type and mutant peptides are α-helical. However, whereas in the wild-type peptide this helix is continuous, the mutant presents a discontinuity, or kink, precisely at the site of mutation G161D. Our results suggest that the mutation may lock integrin heterodimers in a bent conformation that prevents integrin activation via conformational extension.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA