Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
EJHaem ; 4(3): 631-638, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37601846

RESUMEN

The α4ß1 integrin regulates the trafficking of multiple myeloma (MM) cells and contributes to MM disease progression. MicroRNAs (miRNAs) can have both tumor suppressor and oncogenic roles and thus are key controllers of tumor evolution, and have been associated with different phases of MM pathogenesis. Using small RNAseq analysis, we show here that α4ß1-dependent MM cell adhesion regulates the expression of forty different miRNAs, therefore expanding our current view of the α4ß1 involvement in MM cell biology. Specific upregulation of miR-324-5p and miR-331-3p in cells attached to α4ß1 ligands was confirmed upon silencing the α4 integrin subunit, and their increased levels found to be dependent on Erk1/2- and PI3K-Akt-, but not Src-dependent signaling. Enhanced miR-324-5p expression upon α4ß1-mediated MM cell adhesion aimed the hedgehog (Hh) component SMO, revealing that the miR-324-5p-SMO module represents a α4ß1-regulated pathway that could control Hh-dependent cellular responses in myeloma. Our results open new therapy research avenues around the α4ß1 contribution to MM progression that deserve to be investigated.

2.
Eur J Immunol ; 52(8): 1228-1242, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35491946

RESUMEN

ICAP-1 regulates ß1-integrin activation and cell adhesion. Here, we used ICAP-1-null mice to study ICAP-1 potential involvement during immune cell development and function. Integrin α4ß1-dependent adhesion was comparable between ICAP-1-null and control thymocytes, but lack of ICAP-1 caused a defective single-positive (SP) CD8+ cell generation, thus, unveiling an ICAP-1 involvement in SP thymocyte development. ICAP-1 bears a nuclear localization signal and we found it displayed a strong nuclear distribution in thymocytes. Interestingly, there was a direct correlation between the lack of ICAP-1 and reduced levels in SP CD8+ thymocytes of Runx3, a transcription factor required for CD8+ thymocyte generation. In the spleen, ICAP-1 was found evenly distributed between cytoplasm and nuclear fractions, and ICAP-1-/- spleen T and B cells displayed upregulation of α4ß1-mediated adhesion, indicating that ICAP-1 negatively controls their attachment. Furthermore, CD3+ - and CD19+ -selected spleen cells from ICAP-1-null mice showed reduced proliferation in response to T- and B-cell stimuli, respectively. Finally, loss of ICAP-1 caused a remarkable decrease in marginal zone B- cell frequencies and a moderate increase in follicular B cells. Together, these data unravel an ICAP-1 involvement in the generation of SP CD8+ thymocytes and in the control of marginal zone B-cell numbers.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Linfocitos B , Linfocitos T CD8-positivos , Activación de Linfocitos , Timocitos , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Linfocitos B/citología , Linfocitos T CD8-positivos/citología , Diferenciación Celular , Integrina beta1/metabolismo , Ratones , Ratones Noqueados , Bazo/citología , Timocitos/citología , Timo/citología
3.
Cancers (Basel) ; 13(2)2021 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-33435306

RESUMEN

Multiple myeloma (MM) is a hematologic cancer characterized by clonal proliferation of plasma cells in the bone marrow (BM). The progression, from the early stages of the disease as monoclonal gammopathy of undetermined significance (MGUS) and smoldering multiple myeloma (SMM) to MM and occasionally extramedullary disease, is drastically affected by the tumor microenvironment (TME). Soluble factors and direct cell-cell interactions regulate MM plasma cell trafficking and homing to the BM niche. Mesenchymal stromal cells, osteoclasts, osteoblasts, myeloid and lymphoid cells present in the BM create a unique milieu that favors MM plasma cell immune evasion and promotes disease progression. Moreover, TME is implicated in malignant cell protection against anti-tumor therapy. This review describes the main cellular and non-cellular components located in the BM, which condition the immunosuppressive environment and lead the MM establishment and progression.

4.
J Pathol ; 252(1): 29-40, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32501543

RESUMEN

The interaction of multiple myeloma (MM) cells with the bone marrow (BM) microenvironment promotes MM cell retention, survival, and resistance to different anti-MM agents, including proteasome inhibitors (PIs) such as bortezomib (BTZ). The α4ß1 integrin is a main adhesion receptor mediating MM cell-stroma interactions and MM cell survival, and its expression and function are downregulated by BTZ, leading to inhibition of cell adhesion-mediated drug resistance (CAM-DR) and MM cell apoptosis. Whether decreased α4ß1 expression and activity are maintained or recovered upon development of resistance to BTZ represents an important question, as a potential rescue of α4ß1 function could boost MM cell survival and disease progression. Using BTZ-resistant MM cells, we found that they not only rescue their α4ß1 expression, but its levels were higher than in parental cells. Increased α4ß1 expression in resistant cells correlated with enhanced α4ß1-mediated cell lodging in the BM, and with disease progression. BTZ-resistant MM cells displayed enhanced NF-κB pathway activation relative to parental counterparts, which contributed to upregulated α4 expression and to α4ß1-dependent MM cell adhesion. These data emphasize the upregulation of α4ß1 expression and function as a key event during resistance to BTZ in MM, which might indirectly contribute to stabilize this resistance, as stronger MM cell attachment to BM stroma will regain CAM-DR and MM cell growth and survival. Finally, we found a strong correlation between high ITGB1 (integrin ß1) expression in MM and poor progression-free survival (PFS) and overall survival (OS) during treatment of MM patients with BTZ and IMIDs, and combination of high ITGB1 levels and presence of the high-risk genetic factor amp1q causes low PFS and OS. These results unravel a novel prognostic value for ITGB1 in myeloma. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Antineoplásicos/administración & dosificación , Bortezomib/administración & dosificación , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Integrina alfa4beta1/metabolismo , Mieloma Múltiple/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Integrina alfa4beta1/genética , Ratones , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Microambiente Tumoral
6.
Cancer Res ; 79(9): 2244-2256, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30833419

RESUMEN

Combined treatment of metastatic melanoma with BRAF and MEK inhibitors has improved survival, but the emergence of resistance represents an important clinical challenge. Targeting ERK is a suitable strategy currently being investigated in melanoma and other cancers. To anticipate possible resistance to ERK inhibitors (ERKi), we used SCH772984 (SCH) as a model ERKi to characterize resistance mechanisms in two BRAF V600E melanoma cell lines. The ERKi-resistant cells were also resistant to vemurafenib (VMF), trametinib (TMT), and combined treatment with either VMF and SCH or TMT and SCH. Resistance to SCH involved stimulation of the IGF1R-MEK5-Erk5 signaling pathway, which counteracted inhibition of Erk1/2 activation and cell growth. Inhibition of IGF1R with linsitinib blocked Erk5 activation in SCH-resistant cells and decreased their growth in 3D spheroid growth assays as well as in NOD scid gamma (NSG) mice. Cells doubly resistant to VMF and TMT or to VMF and SCH also exhibited downregulated Erk1/2 activation linked to stimulation of the IGF1R-MEK5-Erk5 pathway, which accounted for resistance. In addition, we found that the decreased Erk1/2 activation in SCH-resistant cells involved reduced expression and function of TGFα. These data reveal an escape signaling route that melanoma cells use to bypass Erk1/2 blockade during targeted melanoma treatment and offer several possible targets whose disruption may circumvent resistance. SIGNIFICANCE: Activation of the IGF1R-MEK5-Erk5 signaling pathway opposes pharmacologic inhibition of Erk1/2 in melanoma, leading to the reactivation of cell proliferation and acquired resistance.


Asunto(s)
Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Indazoles/farmacología , MAP Quinasa Quinasa 5/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Melanoma/patología , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Piperazinas/farmacología , Receptor IGF Tipo 1/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor , Proliferación Celular , Femenino , Humanos , MAP Quinasa Quinasa 5/genética , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteína Quinasa 7 Activada por Mitógenos/genética , Receptor IGF Tipo 1/genética , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Front Immunol ; 10: 156, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30787933

RESUMEN

The trafficking of neoplastic cells represents a key process that contributes to progression of hematologic malignancies. Diapedesis of neoplastic cells across endothelium and perivascular cells is facilitated by adhesion molecules and chemokines, which act in concert to tightly regulate directional motility. Intravital microscopy provides spatio-temporal views of neoplastic cell trafficking, and is crucial for testing and developing therapies against hematologic cancers. Multiple myeloma (MM), chronic lymphocytic leukemia (CLL), and acute lymphoblastic leukemia (ALL) are hematologic malignancies characterized by continuous neoplastic cell trafficking during disease progression. A common feature of these neoplasias is the homing and infiltration of blood cancer cells into the bone marrow (BM), which favors growth and survival of the malignant cells. MM cells traffic between different BM niches and egress from BM at late disease stages. Besides the BM, CLL cells commonly home to lymph nodes (LNs) and spleen. Likewise, ALL cells also infiltrate extramedullary organs, such as the central nervous system, spleen, liver, and testicles. The α4ß1 integrin and the chemokine receptor CXCR4 are key molecules for MM, ALL, and CLL cell trafficking into and out of the BM. In addition, the chemokine receptor CCR7 controls CLL cell homing to LNs, and CXCR4, CCR7, and CXCR3 contribute to ALL cell migration across endothelia and the blood brain barrier. Some of these receptors are used as diagnostic markers for relapse and survival in ALL patients, and their level of expression allows clinicians to choose the appropriate treatments. In CLL, elevated α4ß1 expression is an established adverse prognostic marker, reinforcing its role in the disease expansion. Combining current chemotherapies with inhibitors of malignant cell trafficking could represent a useful therapy against these neoplasias. Moreover, immunotherapy using humanized antibodies, CAR-T cells, or immune check-point inhibitors together with agents targeting the migration of tumor cells could also restrict their survival. In this review, we provide a view of the molecular players that regulate the trafficking of neoplastic cells during development and progression of MM, CLL, and ALL, together with current therapies that target the malignant cells.


Asunto(s)
Movimiento Celular , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patología , ADP-Ribosil Ciclasa 1/metabolismo , Animales , Quimiocinas/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Integrinas/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Selectinas/metabolismo
10.
Cancer Res ; 78(4): 1017-1030, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29229605

RESUMEN

Melanoma treatment with the BRAF V600E inhibitor vemurafenib provides therapeutic benefits but the common emergence of drug resistance remains a challenge. We generated A375 melanoma cells resistant to vemurafenib with the goal of investigating changes in miRNA expression patterns that might contribute to resistance. Increased expression of miR-204-5p and miR-211-5p occurring in vemurafenib-resistant cells was determined to impact vemurafenib response. Their expression was rapidly affected by vemurafenib treatment through RNA stabilization. Similar effects were elicited by MEK and ERK inhibitors but not AKT or Rac inhibitors. Ectopic expression of both miRNA in drug-naïve human melanoma cells was sufficient to confer vemurafenib resistance and more robust tumor growth in vivo Conversely, silencing their expression in resistant cells inhibited cell growth. Joint overexpression of miR-204-5p and miR-211-5p durably stimulated Ras and MAPK upregulation after vemurafenib exposure. Overall, our findings show how upregulation of miR-204-5p and miR-211-5p following vemurafenib treatment enables the emergence of resistance, with potential implications for mechanism-based strategies to improve vemurafenib responses.Significance: Identification of miRNAs that enable resistance to BRAF inhibitors in melanoma suggests a mechanism-based strategy to limit resistance and improve clinical outcomes. Cancer Res; 78(4); 1017-30. ©2017 AACR.


Asunto(s)
Melanoma/tratamiento farmacológico , Melanoma/genética , MicroARNs/biosíntesis , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Vemurafenib/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Resistencia a Antineoplásicos , Células HEK293 , Humanos , Melanoma/enzimología , Melanoma/patología , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Distribución Aleatoria , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Int J Biochem Cell Biol ; 95: 121-131, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29288743

RESUMEN

Chemokines are chemotactic cytokines that promote cell migration and activation under homeostatic and inflammatory conditions. Chemokines bind to seven transmembrane-spanning receptors that are coupled to heterotrimeric guanine nucleotide-binding (G) proteins, which are the responsible for intracellularly transmitting the activating signals for cell migration. Hematopoiesis, vascular development, lymphoid organ morphogenesis, cardiogenesis and neural differentiation are amongst the processes involving chemokine function. In addition, immune cell trafficking from bone marrow to blood circulation, and from blood and lymph to lymphoid and inflamed tissues, is tightly regulated by chemokines both under physiological conditions and also in autoimmune diseases. Furthermore, chemokine binding to their receptors stimulate trafficking to and positioning of cancer cells into target tissues and organs during tumour dissemination. The CXCL12 chemokine (also known as stromal-cell derived factor-1α, SDF-1α) plays key roles in hematopoiesis and lymphoid tissue architecture, in cardiogenesis, vascular formation and neurogenesis, as well as in the trafficking of solid and hematological cancer cell types. CXCL12 binds to the CXCR4 receptor, a multi-facetted molecule which tightly mirrors CXCL12 functions in homeostasis and disease. This review addresses the important roles of the CXCR4-CXCL12 axis in homeostasis, specially focusing in hematopoiesis, as well as it provides a picture of CXCR4 as mediator of cancer cell spreading, and a view of the available CXCR4 antagonists in different cancer types.


Asunto(s)
Quimiocina CXCL12/metabolismo , Hematopoyesis , Modelos Biológicos , Modelos Moleculares , Receptores CXCR4/metabolismo , Transducción de Señal , Animales , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Quimiocina CXCL12/química , Drogas en Investigación/uso terapéutico , Hematopoyesis/efectos de los fármacos , Humanos , Ligandos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Conformación Proteica , Multimerización de Proteína , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/química , Transducción de Señal/efectos de los fármacos
12.
Oncotarget ; 8(1): 215-227, 2017 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-27966446

RESUMEN

We have investigated the role of vascular-endothelial (VE)-cadherin in melanoma and breast cancer metastasis. We found that VE-cadherin is expressed in highly aggressive melanoma and breast cancer cell lines. Remarkably, inactivation of VE-cadherin triggered a significant loss of malignant traits (proliferation, adhesion, invasion and transendothelial migration) in melanoma and breast cancer cells. These effects, except transendothelial migration, were induced by the VE-cadherin RGD motifs. Co-immunoprecipitation experiments demonstrated an interaction between VE-cadherin and α2ß1 integrin, with the RGD motifs found to directly affect ß1 integrin activation. VE-cadherin-mediated integrin signaling occurred through specific activation of SRC, ERK and JNK, including AKT in melanoma. Knocking down VE-cadherin suppressed lung colonization capacity of melanoma or breast cancer cells inoculated in mice, while pre-incubation with VE-cadherin RGD peptides promoted lung metastasis for both cancer types. Finally, an in silico study revealed the association of high VE-cadherin expression with poor survival in a subset of melanoma patients and breast cancer patients showing low CD34 expression. These findings support a general role for VE-cadherin and other RGD cadherins as critical regulators of lung and liver metastasis in multiple solid tumours. These results pave the way for cadherin-specific RGD targeted therapies to control disseminated metastasis in multiple cancers.


Asunto(s)
Antígenos CD/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Cadherinas/metabolismo , Melanoma/metabolismo , Melanoma/patología , Oligopéptidos , Dominios y Motivos de Interacción de Proteínas , Animales , Antígenos CD/química , Antígenos CD/genética , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Cadherinas/antagonistas & inhibidores , Cadherinas/química , Cadherinas/genética , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Xenoinjertos , Humanos , Integrinas/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/mortalidad , Ratones , Modelos Biológicos , Terapia Molecular Dirigida , Metástasis de la Neoplasia , Pronóstico , Unión Proteica , Mapas de Interacción de Proteínas , Transducción de Señal
13.
Blood ; 128(18): 2241-2252, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27625360

RESUMEN

Tumor-associated macrophages (TAM) are important components of the multiple myeloma (MM) microenvironment that support malignant plasma cell survival and resistance to therapy. It has been proposed that macrophages (MØ) retain the capacity to change in response to stimuli that can restore their antitumor functions. Here, we investigated several approaches to reprogram MØ as a novel therapeutic strategy in MM. First, we found tumor-limiting and tumor-supporting capabilities for monocyte-derived M1-like MØ and M2-like MØ, respectively, when mixed with MM cells, both in vitro and in vivo. Multicolor confocal microscopy revealed that MM-associated MØ displayed a predominant M2-like phenotype in the bone marrow of MM patient samples, and a high expression of the pro-M2 cytokine macrophage migration inhibitory factor (MIF). To reprogram the protumoral M2-like MØ present in MM toward antitumoral M1-like MØ, we tested the pro-M1 cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) plus blockade of the M2 cytokines macrophage colony-stimulating factor or MIF. The combination of GM-CSF plus the MIF inhibitor 4-iodo-6-phenyl-pyrimidine achieved the best reprogramming responses toward an M1 profile, at both gene and protein expression levels, as well as remarkable tumoricidal effects. Furthermore, this combined treatment elicited MØ-dependent therapeutic responses in MM xenograft mouse models, which were linked to upregulation of M1 and reciprocal downregulation of M2 MØ markers. Our results reveal the therapeutic potential of reprogramming MØ in the context of MM.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Técnicas de Reprogramación Celular/métodos , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Macrófagos/patología , Mieloma Múltiple/inmunología , Animales , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Microscopía Confocal , Pirimidinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Biol Chem ; 291(40): 21053-21062, 2016 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-27481944

RESUMEN

Chemokine stimulation of integrin α4ß1-dependent T lymphocyte adhesion is a key step during lymphocyte trafficking. A central question regarding α4ß1 function is how its lateral mobility and organization influence its affinity and avidity following cell stimulation with chemokines and/or ligands. Using single particle tracking and superresolution imaging approaches, we explored the lateral mobility and spatial arrangement of individual α4ß1integrins on T cells exposed to different activating stimuli. We show that CXCL12 stimulation leads to rapid and transient α4ß1activation, measured by induction of the activation epitope recognized by the HUTS-21 anti-ß1antibody and by increased talin-ß1 association. CXCL12-dependent α4ß1 activation directly correlated with restricted lateral diffusion and integrin immobilization. Moreover, co-stimulation by CXCL12 together with soluble VCAM-1 potentiated integrin immobilization with a 5-fold increase in immobile integrins compared with unstimulated conditions. Our data indicate that docking by talin of the chemokine-activated α4ß1 to the actin cytoskeleton favors integrin immobilization, which likely facilitates ligand interaction and increased adhesiveness. Superresolution imaging showed that the nanoscale organization of high-affinity α4ß1 remains unaffected following chemokine and/or ligand addition. Instead, newly activated α4ß1 integrins organize on the cell membrane as independent units without joining pre-established integrin sites to contribute to cluster formation. Altogether, our results provide a rationale to understand how the spatiotemporal organization of activated α4ß1 integrins regulates T lymphocyte adhesion.


Asunto(s)
Quimiocina CXCL12/metabolismo , Integrina alfa4beta1/metabolismo , Linfocitos T/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Adhesión Celular/fisiología , Línea Celular , Integrina alfa4beta1/genética , Transporte de Proteínas/fisiología , Talina/genética , Talina/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética
15.
Mol Biol Cell ; 26(18): 3215-28, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26202465

RESUMEN

Stimulation by chemokines of integrin α4ß1-dependent T-lymphocyte adhesion is a crucial step for lymphocyte trafficking. The adaptor Vav1 is required for chemokine-activated T-cell adhesion mediated by α4ß1. Conceivably, proteins associating with Vav1 could potentially modulate this adhesion. Correlating with activation by the chemokine CXCL12 of T-lymphocyte attachment to α4ß1 ligands, a transient stimulation in the association of Vav1 with SLP-76, Pyk2, and ADAP was observed. Using T-cells depleted for SLP-76, ADAP, or Pyk2, or expressing Pyk2 kinase-inactive forms, we show that SLP-76 and ADAP stimulate chemokine-activated, α4ß1-mediated adhesion, whereas Pyk2 opposes T-cell attachment. While CXCL12-promoted generation of high-affinity α4ß1 is independent of SLP-76, ADAP, and Pyk2, the strength of α4ß1-VCAM-1 interaction and cell spreading on VCAM-1 are targets of regulation by these three proteins. GTPase assays, expression of activated or dominant-negative Rac1, or combined ADAP and Pyk2 silencing indicated that Rac1 activation by CXCL12 is a common mediator response in SLP-76-, ADAP-, and Pyk2-regulated cell adhesion involving α4ß1. Our data strongly suggest that chemokine-stimulated associations between Vav1, SLP-76, and ADAP facilitate Rac1 activation and α4ß1-mediated adhesion, whereas Pyk2 opposes this adhesion by limiting Rac1 activation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Integrina alfa4beta1/metabolismo , Fosfoproteínas/metabolismo , Linfocitos T/metabolismo , Adhesión Celular/fisiología , Línea Celular , Quimiocina CXCL12/metabolismo , Humanos , Células Jurkat , Ligandos , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-vav/metabolismo , Transducción de Señal , Linfocitos T/citología , Molécula 1 de Adhesión Celular Vascular/metabolismo , Proteína de Unión al GTP rac1/metabolismo
16.
Cell Signal ; 26(11): 2551-61, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25025568

RESUMEN

Activation of the GTPase RhoA linked to cell invasion can be tightly regulated following Gα13 stimulation. We have used a cellular model displaying Gα13-dependent inhibition of RhoA activation associated with defective cell invasion to the chemokine CXCL12 to characterize the molecular players regulating these processes. Using both RNAi transfection approaches and protein overexpression experiments here we show that the Src kinase Blk is involved in Gα13-activated tyrosine phosphorylation of p190RhoGAP, which causes RhoA inactivation and ultimately leads to deficient cell invasion. Characterization of molecular interplays between Gα13, Blk and p190RhoGAP revealed that Blk binds Gα13, and that Blk-mediated p190RhoGAP phosphorylation upon Gα13 activation correlates with weakening of Gα13-Blk association connected to increased Blk-p190RhoGAP assembly. These results place Blk upstream of the p190RhoGAP-RhoA pathway in Gα13-activated cells, overall representing an opposing signaling module during CXCL12-triggered invasion. In addition, analyses with Blk- or Gα13-knockdown cells indicated that Blk can also mediate CXCL12-triggered phosphorylation of p190RhoGAP independently of Gα13. However, even if CXCL12 induces the Blk-mediated GAP phosphorylation, the simultaneous stimulation of the guanine-nucleotide exchange factor Vav1 by the chemokine, as earlier reported, leads to a net increase in RhoA activation. Therefore, when Gα13 is concurrently stimulated with CXCL12 there appears to be sufficient Blk activity to promote adequate levels of p190RhoGAP tyrosine phosphorylation to inactivate RhoA and to impair cell invasiveness.


Asunto(s)
Quimiocina CXCL12/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteínas Represoras/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Familia-src Quinasas/metabolismo , Línea Celular Tumoral , Quimiocina CXCL12/genética , Activación Enzimática/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patología , Fosforilación/genética , Proteínas Represoras/genética , Proteína de Unión al GTP rhoA/genética , Familia-src Quinasas/genética
17.
PLoS One ; 8(5): e63695, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23696849

RESUMEN

Fibroblast growth factor receptor 4 (FGFR4) is vital in early development and tissue repair. FGFR4 expression levels are very restricted in adult tissues, except in several solid tumors including colorectal cancer, which showed overexpression of FGFR4. Here, FGFR4 mutation analysis discarded the presence of activating mutations, other than Arg(388), in different colorectal cancer cell lines and tumoral samples. Stable shRNA FGFR4-silencing in SW480 and SW48 cell lines resulted in a significant decrease in cell proliferation, adhesion, cell migration and invasion. This decrease in the tumorigenic and invasive capabilities of colorectal cancer cells was accompanied by a decrease of Snail, Twist and TGFß gene expression levels and an increase of E-cadherin, causing a reversion to a more epithelial phenotype, in three different cell lines. In addition, FGFR4-signaling activated the oncogenic SRC, ERK1/2 and AKT pathways in colon cancer cells and promoted an increase in cell survival. The relevance of FGFR4 in tumor growth was supported by two different strategies. Kinase inhibitors abrogated FGFR4-related cell growth and signaling pathways at the same extent than FGFR4-silenced cells. Specific FGFR4-targeting using antibodies provoked a similar reduction in cell growth. Moreover, FGFR4 knock-down cells displayed a reduced capacity for in vivo tumor formation and angiogenesis in nude mice. Collectively, our data support a crucial role for FGFR4 in tumorigenesis, invasion and survival in colorectal cancer. In addition, FGFR4 targeting demonstrated its applicability for colorectal cancer therapy.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Western Blotting , Adhesión Celular/genética , Adhesión Celular/fisiología , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/genética , Transición Epitelial-Mesenquimal/genética , Humanos , Ratones , Ratones Desnudos , Polimorfismo de Nucleótido Simple/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética
18.
Cell Signal ; 25(4): 848-59, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23333462

RESUMEN

Recent studies have suggested a regulatory role for the dioxin receptor (AhR) in cell adhesion and migration. Following our previous work, we report here that the C-terminal Src kinase-binding protein (Cbp) signaling pathway controls ß1 integrin activation and that this mechanism is AhR dependent. T-FGM AhR-/- fibroblasts displayed higher integrin ß1 activation, revealed by the increased binding of the activation reporter 9EG7 anti-ß1 mAb and of a soluble fibronectin fragment, as well as by enhanced talin-ß1 association. AhR-/- fibroblasts also showed increased fibronectin secretion and impaired directional migration. Notably, interfering Cbp expression in AhR-/- fibroblasts reduced ß1 integrin activation, improved cell migration and rescued wild-type cell morphology. Cbp over-expression in T-FGM AhR-/- cells enhanced the formation of inhibitory Csk-Cbp complexes which in turn reduced c-Src p-Tyr(416) activation and focal adhesion kinase (FAK) phosphorylation at the c-Src-responsive residues p-Tyr(576) and p-Tyr(577). The c-Src target and migration-related protein Cav1 was also hypophosphorylated at p-Tyr(14) in AhR-/- cells, and such effect was rescued by down-modulating Cbp levels. Thus, AhR regulates fibroblast migration by modulating ß1 integrin activation via Cbp-dependent, Src-mediated signaling.


Asunto(s)
Integrina beta1/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Sialoglicoproteínas/metabolismo , Familia-src Quinasas/metabolismo , Actinas/metabolismo , Animales , Proteína Tirosina Quinasa CSK , Caveolina 1/metabolismo , Adhesión Celular , Movimiento Celular , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Ratones , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Receptores de Hidrocarburo de Aril/genética , Transducción de Señal
19.
J Pathol ; 229(1): 36-48, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22711564

RESUMEN

Myeloma cell adhesion dependent on α4ß1 integrin is crucial for the progression of multiple myeloma (MM). The α4ß1-dependent myeloma cell adhesion is up-regulated by the chemokine CXCL12, and pharmacological blockade of the CXCL12 receptor CXCR4 leads to defective myeloma cell homing to bone marrow (BM). Sphingosine-1-phosphate (S1P) regulates immune cell trafficking upon binding to G-protein-coupled receptors. Here we show that myeloma cells express S1P1, a receptor for S1P. We found that S1P up-regulated the α4ß1-mediated myeloma cell adhesion and transendothelial migration stimulated by CXCL12. S1P promoted generation of high-affinity α4ß1 that efficiently bound the α4ß1 ligand VCAM-1, a finding that was associated with S1P-triggered increase in talin-ß1 integrin association. Furthermore, S1P cooperated with CXCL12 for enhancement of α4ß1-dependent adhesion strengthening and spreading. CXCL12 and S1P activated the DOCK2-Rac1 pathway, which was required for stimulation of myeloma cell adhesion involving α4ß1. Moreover, in vivo analyses indicated that S1P contributes to optimizing the interactions of MM cells with the BM microvasculture and for their lodging inside the bone marrow. The regulation of α4ß1-dependent adhesion and migration of myeloma cells by CXCL12-S1P combined activities might have important consequences for myeloma disease progression.


Asunto(s)
Médula Ósea/metabolismo , Adhesión Celular , Quimiocina CXCL12/metabolismo , Integrina alfa4beta1/metabolismo , Lisofosfolípidos/metabolismo , Mieloma Múltiple/metabolismo , Esfingosina/análogos & derivados , Células del Estroma/metabolismo , Migración Transendotelial y Transepitelial , Animales , Médula Ósea/irrigación sanguínea , Médula Ósea/inmunología , Médula Ósea/patología , Forma de la Célula , Técnicas de Cocultivo , Proteínas Activadoras de GTPasa , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Integrina alfa5beta1/metabolismo , Células K562 , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mieloma Múltiple/genética , Mieloma Múltiple/inmunología , Mieloma Múltiple/patología , Interferencia de ARN , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Esfingosina/metabolismo , Células del Estroma/inmunología , Células del Estroma/patología , Talina/metabolismo , Factores de Tiempo , Transfección , Células Tumorales Cultivadas , Molécula 1 de Adhesión Celular Vascular/metabolismo , Proteína de Unión al GTP rac1/metabolismo
20.
J Cell Sci ; 125(Pt 22): 5338-52, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22946047

RESUMEN

Cell migration and invasion require regulated turnover of integrin-dependent adhesion complexes. Rap1-GTP-interacting adaptor molecule (RIAM) is an adaptor protein that mediates talin recruitment to the cell membrane, and whose depletion leads to defective melanoma cell migration and invasion. In this study, we investigated the potential involvement of RIAM in focal adhesion (FA) dynamics. RIAM-depleted melanoma and breast carcinoma cells displayed an increased number, size and stability of FAs, which accumulated centrally at the ventral cell surface, a phenotype caused by defective FA disassembly. Impairment in FA disassembly resulting from RIAM knockdown correlated with deficient integrin-dependent mitogen-activated protein kinase kinase (MEK)-Erk1/2 activation and, importantly, overexpression of constitutively active MEK resulted in rescue of FA disassembly and recovery of cell invasion. Furthermore, RIAM-promoted Ras homologue gene family, member A (RhoA) activation following integrin engagement was needed for subsequent Erk1/2 activation. In addition, RhoA overexpression partially rescued the FA phenotype in RIAM-depleted cells, also suggesting a functional role for RhoA downstream of RIAM, but upstream of Erk1/2. RIAM knockdown also led to enhanced phosphorylation of paxillin Tyr118 and Tyr31. However, expression of phosphomimetic and nonphosphorylatable mutants at these paxillin residues indicated that paxillin hyperphosphorylation is a subsequent consequence of the blockade of FA disassembly, but does not cause the FA phenotype. RIAM depletion also weakened the association between FA proteins, suggesting that it has important adaptor roles in the correct assembly of adhesion complexes. Our data suggest that integrin-triggered, RIAM-dependent MEK activation represents a key feedback event required for efficient FA disassembly, which could help explain the role of RIAM in cell migration and invasion.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adhesiones Focales/metabolismo , MAP Quinasa Quinasa 1/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana/metabolismo , Animales , Línea Celular Tumoral , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Melanoma/enzimología , Melanoma/patología , Ratones , Modelos Biológicos , Paxillin/metabolismo , Fosforilación , Fosfotirosina/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Regulación hacia Arriba , Proteína de Unión al GTP rhoA/metabolismo , Familia-src Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA