Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Sci Rep ; 12(1): 6255, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35428815

RESUMEN

Bone marrow megakaryocytes (MKs) undergo a maturation involving contacts with the microenvironment before extending proplatelets through sinusoids to deliver platelets in the bloodstream. We demonstrated that MKs assemble linear F-actin-enriched podosomes on collagen I fibers. Microscopy analysis evidenced an inverse correlation between the number of dot-like versus linear podosomes over time. Confocal videomicroscopy confirmed that they derived from each-other. This dynamics was dependent on myosin IIA. Importantly, MKs progenitors expressed the Tks4/5 adaptors, displayed a strong gelatinolytic ability and did not form linear podosomes. While maturing, MKs lost Tks expression together with digestive ability. However, those MKs were still able to remodel the matrix by exerting traction on collagen I fibers through a collaboration between GPVI, ß1 integrin and linear podosomes. Our data demonstrated that a change in structure and composition of podosomes accounted for the shift of function during megakaryopoiesis. These data highlight the fact that members of the invadosome family could correspond to different maturation status of the same entity, to adapt to functional responses required by differentiation stages of the cell that bears them.


Asunto(s)
Megacariocitos , Podosomas , Plaquetas/metabolismo , Colágeno Tipo I/metabolismo , Megacariocitos/metabolismo , Trombopoyesis
2.
Cancer Res ; 81(6): 1472-1485, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33414172

RESUMEN

Invadopodia are actin-based proteolytic membrane protrusions required for invasive behavior and tumor growth. In this study, we used our high-content screening assay to identify kinases whose activity affects invadopodia formation. Among the top hits selected for further analysis was TAO3, an STE20-like kinase of the GCK subfamily. TAO3 was overexpressed in many human cancers and regulated invadopodia formation in melanoma, breast, and bladder cancers. Furthermore, TAO3 catalytic activity facilitated melanoma growth in three-dimensional matrices and in vivo. A novel, potent catalytic inhibitor of TAO3 was developed that inhibited invadopodia formation and function as well as tumor cell extravasation and growth. Treatment with this inhibitor demonstrated that TAO3 activity is required for endosomal trafficking of TKS5α, an obligate invadopodia scaffold protein. A phosphoproteomics screen for TAO3 substrates revealed the dynein subunit protein LIC2 as a relevant substrate. Knockdown of LIC2 or expression of a phosphomimetic form promoted invadopodia formation. Thus, TAO3 is a new therapeutic target with a distinct mechanism of action. SIGNIFICANCE: An unbiased screening approach identifies TAO3 as a regulator of invadopodia formation and function, supporting clinical development of this class of target.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Endosomas/metabolismo , Invasividad Neoplásica/patología , Podosomas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Dineínas Citoplasmáticas/genética , Dineínas Citoplasmáticas/metabolismo , Conjuntos de Datos como Asunto , Matriz Extracelular , Femenino , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ensayos Analíticos de Alto Rendimiento , Humanos , Masculino , Melanoma/tratamiento farmacológico , Melanoma/patología , Ratones , Invasividad Neoplásica/prevención & control , Podosomas/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Imagen de Lapso de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Eur J Cell Biol ; 99(7): 151122, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33070041

RESUMEN

The scaffold protein Tks5α is required for invadopodia-mediated cancer invasion both in vitro and in vivo. We have previously also revealed a role for Tks5 in tumor cell growth using three-dimensional (3D) culture model systems and mouse transplantation experiments. Here we use both 3D and high-density fibrillar collagen (HDFC) culture to demonstrate that native collagen-I, but not a form lacking the telopeptides, stimulated Tks5-dependent growth, which was dependent on the DDR collagen receptors. We used microenvironmental microarray (MEMA) technology to determine that laminin, fibronectin and tropoelastin also stimulated invadopodia formation. A Tks5α-specific monoclonal antibody revealed its expression both on microtubules and at invadopodia. High- and super-resolution microscopy of cells in and on collagen was then used to place Tks5α at the base of invadopodia, separated from much of the actin and cortactin, but coincident with both matrix metalloprotease and cathepsin proteolytic activity. Inhibition of the Src family kinases, cathepsins or metalloproteases all reduced invadopodia length but each had distinct effects on Tks5α localization. These studies highlight the crosstalk between invadopodia and extracellular matrix components, and reveal the invadopodium to be a spatially complex structure.


Asunto(s)
Matriz Extracelular/metabolismo , Podosomas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Humanos , Ratones , Isoformas de Proteínas
4.
Sci Adv ; 4(9): eaat7828, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30214939

RESUMEN

High lethality rates associated with metastatic cancer highlight an urgent medical need for improved understanding of biologic mechanisms driving metastatic spread and identification of biomarkers predicting late-stage progression. Numerous neoplastic cell intrinsic and extrinsic mechanisms fuel tumor progression; however, mechanisms driving heterogeneity of neoplastic cells in solid tumors remain obscure. Increased mutational rates of neoplastic cells in stressed environments are implicated but cannot explain all aspects of tumor heterogeneity. We present evidence that fusion of neoplastic cells with leukocytes (for example, macrophages) contributes to tumor heterogeneity, resulting in cells exhibiting increased metastatic behavior. Fusion hybrids (cells harboring hematopoietic and epithelial properties) are readily detectible in cell culture and tumor-bearing mice. Further, hybrids enumerated in peripheral blood of human cancer patients correlate with disease stage and predict overall survival. This unique population of neoplastic cells provides a novel biomarker for tumor staging, as well as a potential therapeutic target for intervention.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Células Neoplásicas Circulantes/patología , Neoplasias Pancreáticas/patología , Animales , Biomarcadores de Tumor/sangre , Carcinoma Ductal Pancreático/mortalidad , Fusión Celular , Línea Celular Tumoral , Supervivencia Celular , Células Epiteliales/patología , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células Híbridas , Cariotipificación , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias Pancreáticas/mortalidad , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Biol Ther ; 19(10): 921-933, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29856687

RESUMEN

Head and neck squamous cell carcinoma (HNSCC) currently only has one FDA-approved cancer intrinsic targeted therapy, the epidermal growth factor receptor (EGFR) inhibitor cetuximab, to which only approximately 10% of tumors are sensitive. In order to extend therapy options, we subjected patient-derived HNSCC cells to small-molecule inhibitor and siRNA screens, first, to find effective combination therapies with an EGFR inhibitor, and second, to determine a potential mechanistic basis for repurposing the FDA approved agents for HNSCC. The combinations of EGFR inhibitor with anaplastic lymphoma kinase (ALK) inhibitors demonstrated synergy at the highest ratio in our cohort, 4/8 HNSCC patients' derived tumor cells, and this corresponded with an effectiveness of siRNA targeting ALK combined with the EGFR inhibitor gefitinib. Co-targeting EGFR and ALK decreased HNSCC cell number and colony formation ability and increased annexin V staining. Because ALK expression is low and ALK fusions are infrequent in HNSCC, we hypothesized that gefitinib treatment could induce ALK expression. We show that ALK expression was induced in HNSCC patient-derived cells both in 2D and 3D patient-derived cell culture models, and in patient-derived xenografts in mice. Four different ALK inhibitors, including two (ceritinib and brigatinib) FDA approved for lung cancer, were effective in combination with gefitinib. Together, we identified induction of ALK by EGFR inhibitor as a novel mechanism potentially relevant to resistance to EGFR inhibitor, a high ratio of response of HNSCC patient-derived tumor cells to a combination of ALK and EGFR inhibitors, and applicability of repurposing ALK inhibitors to HNSCC that lack ALK aberrations.


Asunto(s)
Quinasa de Linfoma Anaplásico/genética , Antineoplásicos/farmacología , Resistencia a Antineoplásicos/genética , Receptores ErbB/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Quinasa de Linfoma Anaplásico/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Cell Sci ; 131(1)2018 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-29311151

RESUMEN

Tyrosine kinase substrate (Tks) adaptor proteins are considered important regulators of various physiological and/or pathological processes, particularly cell migration and invasion, and cancer progression. These proteins contain PX and SH3 domains, and act as scaffolds, bringing membrane and cellular components in close proximity in structures known as invadopodia or podosomes. Tks proteins, analogous to the related proteins p47phox, p40phox and NoxO1, also facilitate local generation of reactive oxygen species (ROS), which aid in signaling at invadopodia and/or podosomes to promote their activity. As their name suggests, Tks adaptor proteins are substrates for tyrosine kinases, especially Src. In this Cell Science at a Glance article and accompanying poster, we discuss the known structural and functional aspects of Tks adaptor proteins. As the science of Tks proteins is evolving, this article will point out where we stand and what still needs to be explored. We also underscore pathological conditions involving these proteins, providing a basis for future research to develop therapies for treatment of these diseases.


Asunto(s)
Movimiento Celular , Neoplasias/metabolismo , Podosomas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Línea Celular Tumoral , Extensiones de la Superficie Celular/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , NADPH Oxidasas/metabolismo , Podosomas/patología , Proteínas Tirosina Quinasas/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
7.
Mol Cell Biol ; 38(6)2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29263157

RESUMEN

The transcription factor gene MYC is important in breast cancer, and its mRNA is maintained at a high level even in the absence of gene amplification. The mechanism(s) underlying increased MYC mRNA expression is unknown. Here, we demonstrate that MYC mRNA was stabilized upon estrogen stimulation of estrogen receptor-positive breast cancer cells via SRC-dependent effects on a recently described RNA-binding protein, IMP1 with an N-terminal deletion (ΔN-IMP1). We also show that loss of the tumor suppressor p53 increased MYC mRNA levels even in the absence of estrogen stimulation. However, in cells with wild-type p53, SRC acted to overcome p53-mediated inhibition of estrogen-stimulated cell cycle entry and progression. SRC thus promotes cell proliferation in two ways: by stabilizing MYC mRNA and by inhibiting p53 function. Since estrogen receptor-positive breast cancers typically express wild-type p53, these studies establish a rationale for p53 status to be predictive for effective SRC inhibitor treatment in this subtype of breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Familia-src Quinasas/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Ciclo Celular/fisiología , Línea Celular Tumoral , Proliferación Celular/fisiología , Estradiol/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Xenoinjertos , Humanos , Células MCF-7 , Ratones Desnudos , Proteínas Proto-Oncogénicas c-myc/biosíntesis , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Receptores de Estrógenos/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Familia-src Quinasas/genética
8.
FEBS J ; 285(1): 8-27, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28548369

RESUMEN

Invadopodia and podosomes are discrete, actin-based molecular protrusions that form in cancer cells and normal cells, respectively, in response to diverse signaling pathways and extracellular matrix cues. Although they participate in a host of different cellular processes, they share a common functional theme of controlling pericellular proteolytic activity, which sets them apart from other structures that function in migration and adhesion, including focal adhesions, lamellipodia, and filopodia. In this review, we highlight research that explores the function of these complex structures, including roles for podosomes in embryonic and postnatal development, in angiogenesis and remodeling of the vasculature, in maturation of the postsynaptic membrane, in antigen sampling and recognition, and in cell-cell fusion mechanisms, as well as the involvement of invadopodia at multiple steps of the metastatic cascade, and how all of this may apply in the treatment of human disease states. Finally, we explore recent research that implicates a novel role for exosomes and microvesicles in invadopodia-dependent and invadopodia-independent mechanisms of invasion, respectively.


Asunto(s)
Extensiones de la Superficie Celular/metabolismo , Neoplasias/metabolismo , Podosomas/metabolismo , Seudópodos/metabolismo , Animales , Exosomas/metabolismo , Adhesiones Focales/metabolismo , Humanos , Invasividad Neoplásica , Neoplasias/patología
9.
J Cell Sci ; 130(12): 2036-2048, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28468988

RESUMEN

The Twist1 transcription factor promotes tumor invasion and metastasis by inducing epithelial-mesenchymal transition (EMT) and invadopodia-mediated extracellular matrix (ECM) degradation. The critical transcription targets of Twist1 for mediating these events remain to be uncovered. Here, we report that Twist1 strongly induces expression of a disintegrin and metalloproteinase 12 (ADAM12). We observed that the expression levels of Twist1 mRNA and ADAM12 mRNA are tightly correlated in human breast tumors. Knocking down ADAM12 blocked cell invasion in a 3D mammary organoid culture. Suppression of ADAM12 also inhibited Twist1-induced tumor invasion and metastasis in human breast tumor xenografts, without affecting primary tumor formation. Mechanistically, knockdown of ADAM12 in breast cancer cells significantly reduced invadopodia formation and matrix degradation, and simultaneously increased overall cell adhesion to the ECM. Live-imaging analysis showed that knockdown of ADAM12 significantly inhibited focal adhesion turnover. Mechanistically, both the disintegrin and metalloproteinase domains of ADAM12 are required for its function at invadopodia, whereas the metalloproteinase domain is dispensable for its function at focal adhesions. Taken together, these data suggest that ADAM12 plays a crucial role in tumor invasion and metastasis by regulating both invadopodia and focal adhesions.


Asunto(s)
Proteína ADAM12/metabolismo , Adhesiones Focales/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas Nucleares/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Adhesión Celular , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Tamaño de la Célula , Células Cultivadas , Transición Epitelial-Mesenquimal , Matriz Extracelular/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Neoplasias Mamarias Animales/metabolismo , Ratones , Mutagénesis Sitio-Dirigida , Trasplante de Neoplasias , Dominios Proteicos , Transducción de Señal
10.
Oncotarget ; 7(48): 78473-78486, 2016 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-27802184

RESUMEN

Metastatic cancer cells are characterized by their ability to degrade and invade through extracellular matrix. We previously showed that the Tks adaptor proteins, Tks4 and Tks5, are required for invadopodia formation and/or function in Src-transformed fibroblasts and a number of human cancer cell types. In this study, we investigated the role of Tks adaptor proteins in melanoma cell invasion and metastasis. Knockdown of either Tks4 or Tks5 in both mouse and human melanoma cell lines resulted in a decreased ability to form invadopodia and degrade extracellular matrix. In addition, Tks-knockdown melanoma cells had decreased proliferation in a 3-dimensional type l collagen matrix, but not in 2-dimensional culture conditions. We also investigated the role of Tks proteins in melanoma progression in vivo using xenografts and experimental metastasis assays. Consistent with our in vitro results, reduction of Tks proteins markedly reduced subcutaneous melanoma growth as well as metastatic growth in the lung. We explored the clinical relevance of Tks protein expression in human melanoma specimens using a tissue microarray. Compared to non-malignant nevi, both Tks proteins were highly expressed in melanoma tissues. Moreover, metastatic melanoma cases showed higher expression of Tks5 than primary melanoma cases. Taken together, these findings suggest the importance of Tks adaptor proteins in melanoma growth and metastasis in vivo, likely via functional invadopodia formation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Movimiento Celular , Proliferación Celular , Neoplasias Pulmonares/metabolismo , Melanoma/metabolismo , Fosfoproteínas/metabolismo , Podosomas/metabolismo , Neoplasias Cutáneas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Melanoma/genética , Melanoma/secundario , Ratones , Ratones Desnudos , Invasividad Neoplásica , Proteínas de Unión a Fosfato , Fosfoproteínas/genética , Podosomas/patología , Interferencia de ARN , Transducción de Señal , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Factores de Tiempo , Transfección , Carga Tumoral
11.
PLoS One ; 10(3): e0121003, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25826475

RESUMEN

The ability of cancer cells to invade underlies metastatic progression. One mechanism by which cancer cells can become invasive is through the formation of structures called invadopodia, which are dynamic, actin-rich membrane protrusions that are sites of focal extracellular matrix degradation. While there is a growing consensus that invadopodia are instrumental in tumor metastasis, less is known about whether they are involved in tumor growth, particularly in vivo. The adaptor protein Tks5 is an obligate component of invadopodia, and is linked molecularly to both actin-remodeling proteins and pericellular proteases. Tks5 appears to localize exclusively to invadopodia in cancer cells, and in vitro studies have demonstrated its critical requirement for the invasive nature of these cells, making it an ideal surrogate to investigate the role of invadopodia in vivo. In this study, we examined how Tks5 contributes to human breast cancer progression. We used immunohistochemistry and RNA sequencing data to evaluate Tks5 expression in clinical samples, and we characterized the role of Tks5 in breast cancer progression using RNA interference and orthotopic implantation in SCID-Beige mice. We found that Tks5 is expressed to high levels in approximately 50% of primary invasive breast cancers. Furthermore, high expression was correlated with poor outcome, particularly in those patients with late relapse of stage I/II disease. Knockdown of Tks5 expression in breast cancer cells resulted in decreased growth, both in 3D in vitro cultures and in vivo. Moreover, our data also suggest that Tks5 is important for the integrity and permeability of the tumor vasculature. Together, this work establishes an important role for Tks5 in tumor growth in vivo, and suggests that invadopodia may play broad roles in tumor progression.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/fisiología , Neoplasias de la Mama/patología , División Celular/fisiología , Animales , Xenoinjertos , Humanos , Técnicas In Vitro , Ratones , Ratones SCID
12.
PLoS One ; 9(9): e107674, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25259869

RESUMEN

Tks5 is a scaffold protein and Src substrate involved in cell migration and matrix degradation through its essential role in invadosome formation and function. We have previously described that Tks5 is fundamental for zebrafish neural crest cell migration in vivo. In the present study, we sought to investigate the function of Tks5 in mammalian development by analyzing mice mutant for sh3pxd2a, the gene encoding Tks5. Homozygous disruption of the sh3pxd2a gene by gene-trapping in mouse resulted in neonatal death and the presence of a complete cleft of the secondary palate. Interestingly, embryonic fibroblasts from homozygous gene-trap sh3pxd2a mice lacked only the highest molecular weight band of the characteristic Tks5 triplet observed in protein extracts, leaving the lower molecular weight bands unaffected. This finding, together with the existence of two human Expressed Sequence Tags lacking the first 5 exons of SH3PXD2A, made us hypothesize about the presence of a second alternative transcription start site located in intron V. We performed 5'RACE on mouse fibroblasts and isolated a new transcript of the sh3pxd2a gene encoding a novel Tks5 isoform, that we named Tks5ß. This novel isoform diverges from the long form of Tks5 in that it lacks the PX-domain, which confers affinity for phosphatidylinositol-3,4-bisphosphate. Instead, Tks5ß has a short unique amino terminal sequence encoded by the newly discovered exon 6ß; this exon includes a start codon located 29 bp from the 5'-end of exon 6. Tks5ß mRNA is expressed in MEFs and all mouse adult tissues analyzed. Tks5ß is a substrate for the Src tyrosine kinase and its expression is regulated through the proteasome degradation pathway. Together, these findings indicate the essentiality of the larger Tks5 isoform for correct mammalian development and the transcriptional complexity of the sh3pxd2a gene.


Asunto(s)
Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Línea Celular , Fisura del Paladar/genética , Fisura del Paladar/patología , Femenino , Orden Génico , Sitios Genéticos , Homocigoto , Masculino , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación , Fenotipo , Proteínas de Unión a Fosfato , Fosfoproteínas/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Isoformas de Proteínas , Proteolisis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Transcripción Genética , Familia-src Quinasas/metabolismo
13.
Cell Rep ; 8(5): 1558-70, 2014 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-25176655

RESUMEN

Tumor cell extravasation is a key step during cancer metastasis, yet the precise mechanisms that regulate this dynamic process are unclear. We utilized a high-resolution time-lapse intravital imaging approach to visualize the dynamics of cancer cell extravasation in vivo. During intravascular migration, cancer cells form protrusive structures identified as invadopodia by their enrichment of MT1-MMP, cortactin, Tks4, and importantly Tks5, which localizes exclusively to invadopodia. Cancer cells extend invadopodia through the endothelium into the extravascular stroma prior to their extravasation at endothelial junctions. Genetic or pharmacological inhibition of invadopodia initiation (cortactin), maturation (Tks5), or function (Tks4) resulted in an abrogation of cancer cell extravasation and metastatic colony formation in an experimental mouse lung metastasis model. This provides direct evidence of a functional role for invadopodia during cancer cell extravasation and distant metastasis and reveals an opportunity for therapeutic intervention in this clinically important process.


Asunto(s)
Extensiones de la Superficie Celular/metabolismo , Neoplasias Pulmonares/metabolismo , Células Madre Neoplásicas/metabolismo , Migración Transcelular de la Célula , Proteínas Adaptadoras Transductoras de Señales , Animales , Antineoplásicos/farmacología , Benzodioxoles/farmacología , Línea Celular Tumoral , Extensiones de la Superficie Celular/efectos de los fármacos , Embrión de Pollo , Cortactina/genética , Cortactina/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/patología , Metaloproteinasa 14 de la Matriz/metabolismo , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Células Madre Neoplásicas/fisiología , Proteínas de Unión a Fosfato , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología
14.
Cell Adh Migr ; 8(3): 226-35, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24714597

RESUMEN

The tumor microenvironment consists of stromal cells, extracellular matrix (ECM), and signaling molecules that communicate with cancer cells. As tumors grow and develop, the tumor microenvironment changes. In addition, the tumor microenvironment is not only influenced by signals from tumor cells, but also stromal components contribute to tumor progression and metastasis by affecting cancer cell function. One of the mechanisms that cancer cells use to invade and metastasize is mediated by actin-rich, proteolytic structures called invadopodia. Here, we discuss how signals from the tumor environment, including growth factors, hypoxia, pH, metabolism, and stromal cell interactions, affect the formation and function of invadopodia to regulate cancer cell invasion and metastasis. Understanding how the tumor microenvironment affects invadopodia biology could aid in the development of effective therapeutics to target cancer cell invasion and metastasis.


Asunto(s)
Invasividad Neoplásica/patología , Animales , Movimiento Celular/fisiología , Matriz Extracelular/metabolismo , Humanos , Microambiente Tumoral/fisiología
15.
Prostate ; 74(2): 134-48, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24174371

RESUMEN

BACKGROUND: The Src tyrosine kinase substrate and adaptor protein Tks5 had previously been implicated in the invasive phenotype of normal and transformed cell types via regulation of cytoskeletal structures called podosomes/invadopodia. The role of Src-Tks5 signaling in invasive prostate cancer, however, had not been previously evaluated. METHODS: We measured the relative expression of Tks5 in normal (n = 20) and cancerous (n = 184, from 92 patients) prostate tissue specimens by immunohistochemistry using a commercially available tumor microarray. We also manipulated the expression and activity of wild-type and mutant Src and Tks5 constructs in the LNCaP and PC-3 prostate cancer cell lines in order to ascertain the role of Src-Tks5 signaling in invadopodia development, matrix-remodeling activity, motility, and invasion. RESULTS: Our studies demonstrated that Src was activated and Tks5 upregulated in high Gleason score prostate tumor specimens and in invasive prostate cancer cell lines. Remarkably, overexpression of Tks5 in LNCaP cells was sufficient to induce invadopodia formation and associated matrix degradation. This Tks5-dependent increase in invasive behavior further depended on Src tyrosine kinase activity and the phosphorylation of Tks5 at tyrosine residues 557 and 619. In PC-3 cells we demonstrated that Tks5 phosphorylation at these sites was necessary and sufficient for invadopodia-associated matrix degradation and invasion. CONCLUSIONS: Our results suggest a general role for Src-Tks5 signaling in prostate tumor progression and the utility of Tks5 as a marker protein for the staging of this disease.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/fisiología , Adenocarcinoma/patología , Movimiento Celular/fisiología , Citoesqueleto/fisiología , Neoplasias de la Próstata/patología , Familia-src Quinasas/fisiología , Adenocarcinoma/fisiopatología , Biomarcadores de Tumor/fisiología , Estudios de Casos y Controles , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Inmunohistoquímica , Masculino , Fosforilación/fisiología , Neoplasias de la Próstata/fisiopatología , Transducción de Señal/fisiología
16.
J Cell Biol ; 201(2): 279-92, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23589494

RESUMEN

Notch regulates cell-cell contact-dependent signaling and is activated by hypoxia, a microenvironmental condition that promotes cellular invasion during both normal physiology and disease. The mechanisms by which hypoxia and Notch regulate cellular invasion are not fully elucidated. In this paper, we show that, in cancer cells, hypoxia increased the levels and activity of the ADAM12 metalloprotease in a Notch signaling-dependent manner, leading to increased ectodomain shedding of the epidermal growth factor (EGF) receptor (EGFR) ligand heparin-binding EGF-like growth factor. Released HB-EGF induced the formation of invadopodia, cellular structures that aid cancer cell invasion. Thus, we describe a signaling pathway that couples cell contact-dependent signaling with the paracrine activation of the EGFR, indicating cross talk between the Notch and EGFR pathways in promoting cancer cell invasion. This signaling pathway might regulate the coordinated acquisition of invasiveness by neighboring cells and mediate the communication between normoxic and hypoxic areas of tumors to facilitate cancer cell invasion.


Asunto(s)
Proteínas ADAM/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Seudópodos/metabolismo , Receptores Notch/metabolismo , Proteínas ADAM/genética , Proteína ADAM12 , Comunicación Celular/efectos de los fármacos , Comunicación Celular/genética , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor de Crecimiento Similar a EGF de Unión a Heparina , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/farmacología , Proteínas de la Membrana/genética , Modelos Biológicos , Estructura Terciaria de Proteína , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
17.
Sci Transl Med ; 4(127): 127ra36, 2012 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-22461640

RESUMEN

The invasive nature of glioblastoma (GBM) represents a major clinical challenge contributing to poor outcomes. Invasion of GBM into healthy tissue restricts chemotherapeutic access and complicates surgical resection. Here, we test the hypothesis that an effective anti-invasive agent can "contain" GBM and increase the efficacy of chemotherapy. We report a new anti-invasive small molecule, Imipramine Blue (IB), which inhibits invasion of glioma in vitro when tested against several models. IB inhibits NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidase-mediated reactive oxygen species generation and alters expression of actin regulatory elements. In vivo, liposomal IB (nano-IB) halts invasion of glioma, leading to a more compact tumor in an aggressively invasive RT2 syngeneic astrocytoma rodent model. When nano-IB therapy was followed by liposomal doxorubicin (nano-DXR) chemotherapy, the combination therapy prolonged survival compared to nano-IB or nano-DXR alone. Our data demonstrate that nano-IB-mediated containment of diffuse glioma enhanced the efficacy of nano-DXR chemotherapy, demonstrating the promise of an anti-invasive compound as an adjuvant treatment for glioma.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Glioma/tratamiento farmacológico , Glioma/patología , Imipramina/uso terapéutico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Proliferación Celular , Quimioterapia Adyuvante , Doxorrubicina/uso terapéutico , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Humanos , Imipramina/síntesis química , Imipramina/química , Imipramina/farmacocinética , Liposomas/química , Nanopartículas/química , Invasividad Neoplásica , Ratas , Resultado del Tratamiento
18.
Biochem Soc Trans ; 40(1): 129-32, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22260678

RESUMEN

Cell invasion plays a central role in a wide variety of biological phenomena and is the cause of tumour growth and metastasis. Understanding the biochemical mechanisms that control cell invasion is one of the major goals of our laboratory. Podosomes and invadopodia are specialized cellular structures present in cells with physiological or pathological invasive behaviours. These transient structures are localized at the ventral cell surface, contain an array of different proteins and facilitate cell-substrate adhesion, as well as the local proteolytic activity necessary for extracellular matrix remodelling and subsequent cellular invasion. We have shown previously that the adaptor proteins and Src substrates Tks4 and Tks5 are required for podosome and invadopodia formation, for cancer cell invasion in vitro, and for tumour growth in vivo. We have also defined a role for the Tks-mediated generation of ROS (reactive oxygen species) in both podosome and invadopodia formation, and invasive behaviour. Tks4 and Tks5 are also required for proper embryonic development, probably because of their roles in cell migration. Finally, we recently implicated podosome formation as part of the synthetic phenotype of vascular smooth muscle cells. Inhibitors of podosome and invadopodia formation might have utility in the treatment of vascular diseases and cancer. We have therefore developed a high-content cell-based high-throughput screening assay that allows us to identify inhibitors and activators of podosome/invadopodia formation. We have used this assay to screen for small-molecule inhibitors and defined novel regulators of invadopodia formation. In the present paper, I review these recent findings.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Movimiento Celular , Neoplasias/metabolismo , Animales , Extensiones de la Superficie Celular/metabolismo , Extensiones de la Superficie Celular/patología , Desarrollo Embrionario , Humanos , Invasividad Neoplásica , Neoplasias/patología , Transducción de Señal
19.
Free Radic Biol Med ; 52(2): 247-56, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22033009

RESUMEN

Redox signaling contributes to the regulation of cancer cell proliferation, survival, and invasion and participates in the adaptation of cancer cells to their microenvironment. NADPH oxidases are important mediators of redox signaling in normal and cancer cells. Redox signal specificity in normal cells is in part achieved by targeting enzymes that generate reactive oxygen species to specific subcellular microdomains such as focal adhesions, dorsal ruffles, lipid rafts, or caveolae. In a similar fashion, redox signal specificity during cancer cell invasion can be regulated by targeting reactive oxygen generation to invasive microdomains such as invadopodia. Here we summarize recent advances in the understanding of the redox signaling processes that control the cancer cell proinvasive program by modulating cell adhesion, migration, and proteolysis as well as the interaction of cancer cells with the tumor microenvironment. We focus on redox signaling events mediated by invadopodia NADPH oxidase complexes and their contribution to cancer cell invasion.


Asunto(s)
Microdominios de Membrana/enzimología , Neoplasias/patología , Transducción de Señal , Animales , Adhesión Celular , Movimiento Celular , Extensiones de la Superficie Celular/enzimología , Extensiones de la Superficie Celular/metabolismo , Humanos , Microdominios de Membrana/metabolismo , NADPH Oxidasas/metabolismo , Invasividad Neoplásica , Neoplasias/enzimología , Neoplasias/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Microambiente Tumoral
20.
PLoS One ; 6(7): e22499, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21799874

RESUMEN

In the adult organism, cell migration is required for physiological processes such as angiogenesis and immune surveillance, as well as pathological events such as tumor metastasis. The adaptor protein and Src substrate Tks5 is necessary for cancer cell migration through extracellular matrix in vitro and tumorigenicity in vivo. However, a role for Tks5 during embryonic development, where cell migration is essential, has not been examined. We used morpholinos to reduce Tks5 expression in zebrafish embryos, and observed developmental defects, most prominently in neural crest-derived tissues such as craniofacial structures and pigmentation. The Tks5 morphant phenotype was rescued by expression of mammalian Tks5, but not by a variant of Tks5 in which the Src phosphorylation sites have been mutated. We further evaluated the role of Tks5 in neural crest cells and neural crest-derived tissues and found that loss of Tks5 impaired their ventral migration. Inhibition of Src family kinases also led to abnormal ventral patterning of neural crest cells and their derivatives. We confirmed that these effects were likely to be cell autonomous by shRNA-mediated knockdown of Tks5 in a murine neural crest stem cell line. Tks5 was required for neural crest cell migration in vitro, and both Src and Tks5 were required for the formation of actin-rich structures with similarity to podosomes. Additionally, we observed that neural crest cells formed Src-Tks5-dependent cell protrusions in 3-D culture conditions and in vivo. These results reveal an important and novel role for the Src-Tks5 pathway in neural crest cell migration during embryonic development. Furthermore, our data suggests that this pathway regulates neural crest cell migration through the generation of actin-rich pro-migratory structures, implying that similar mechanisms are used to control cell migration during embryogenesis and cancer metastasis.


Asunto(s)
Movimiento Celular , Cresta Neural/citología , Cresta Neural/embriología , Fosfoproteínas/metabolismo , Familia-src Quinasas/metabolismo , Actinas/metabolismo , Animales , Matriz Extracelular/metabolismo , Humanos , Ratones , Cresta Neural/metabolismo , Proteínas de Unión a Fosfato , Pez Cebra/embriología , Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA