Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 7(3)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-34941574

RESUMEN

Discoidin domain receptor 1 (DDR1), a receptor tyrosine kinase activated by collagen, contributes to chronic kidney disease. However, its role in acute kidney injury and subsequent development of kidney fibrosis is not clear. Thus, we performed a model of severe ischemia/reperfusion-induced acute kidney injury that progressed to kidney fibrosis in WT and Ddr1-null mice. We showed that Ddr1-null mice had reduced acute tubular injury, inflammation, and tubulointerstitial fibrosis with overall decreased renal monocyte chemoattractant protein (MCP-1) levels and STAT3 activation. We identified breakpoint cluster region (BCR) protein as a phosphorylated target of DDR1 that controls MCP-1 production in renal proximal tubule epithelial cells. DDR1-induced BCR phosphorylation or BCR downregulation increased MCP-1 secretion, suggesting that BCR negatively regulates the levels of MCP-1. Mechanistically, phosphorylation or downregulation of BCR increased ß-catenin activity and in turn MCP-1 production. Finally, we showed that DDR1-mediated STAT3 activation was required to stimulate the secretion of TGF-ß. Thus, DDR1 contributes to acute and chronic kidney injury by regulating BCR and STAT3 phosphorylation and in turn the production of MCP-1 and TGF-ß. These findings identify DDR1 an attractive therapeutic target for ameliorating both proinflammatory and profibrotic signaling in kidney disease.


Asunto(s)
Receptor con Dominio Discoidina 1/genética , Regulación de la Expresión Génica , Inflamación/complicaciones , Túbulos Renales Proximales/metabolismo , Proteínas Proto-Oncogénicas c-bcr/genética , ARN/genética , Factor de Transcripción STAT3/genética , Lesión Renal Aguda , Animales , Línea Celular , Células Cultivadas , Receptor con Dominio Discoidina 1/biosíntesis , Femenino , Fibrosis/complicaciones , Fibrosis/genética , Fibrosis/patología , Inflamación/genética , Inflamación/patología , Túbulos Renales Proximales/patología , Masculino , Ratones , Ratones Noqueados , Fosforilación , Proteínas Proto-Oncogénicas c-bcr/biosíntesis , Factor de Transcripción STAT3/biosíntesis , Transducción de Señal
2.
JCI Insight ; 6(17)2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34237033

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) tumors are characterized by a desmoplastic reaction resulting in dense deposition of collagen that is known to promote cancer progression. A central mediator of protumorigenic collagen signaling is the receptor tyrosine kinase discoid domain receptor 1 (DDR1). DDR1 is a critical driver of a mesenchymal and invasive cancer cell PDAC phenotype. Previous studies have demonstrated that genetic or pharmacologic inhibition of DDR1 reduces PDAC tumorigenesis and metastasis. Here, we investigated whether DDR1 signaling has cancer cell nonautonomous effects that promote PDAC progression and metastasis. We demonstrate that collagen-induced DDR1 activation in cancer cells is a major stimulus for CXCL5 production, resulting in the recruitment of tumor-associated neutrophils (TANs), the formation of neutrophil extracellular traps (NETs), and subsequent cancer cell invasion and metastasis. Moreover, we have identified that collagen-induced CXCL5 production was mediated by a DDR1/PKCθ/SYK/NF-κB signaling cascade. Together, these results highlight the critical contribution of the collagen I-DDR1 interaction in the formation of an immune microenvironment that promotes PDAC metastasis.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Receptor con Dominio Discoidina 1/genética , Trampas Extracelulares/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Experimentales , Neutrófilos/patología , Neoplasias Pancreáticas/genética , Animales , Carcinogénesis , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/secundario , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , ADN de Neoplasias/genética , Receptor con Dominio Discoidina 1/biosíntesis , Trampas Extracelulares/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Metástasis de la Neoplasia , Neutrófilos/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Transducción de Señal , Microambiente Tumoral
3.
PLoS One ; 14(7): e0211670, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31260458

RESUMEN

Tolvaptan is the only drug approved to slow cyst growth and preserve kidney function in patients with autosomal dominant polycystic kidney disease (ADPKD). However, its limited efficacy combined with significant side effects underscores the need to identify new and safe therapeutic drug targets to slow progression to end stage kidney disease. We identified Discoidin Domain Receptor 1 (DDR1) as receptor tyrosine kinase upregulated in vivo in 3 mouse models of ADPKD using a novel mass spectrometry approach to identify kinases upregulated in ADPKD. Previous studies demonstrating critical roles for DDR1 to cancer progression, its potential role in the pathogenesis of a variety of other kidney disease, along with the possibility that DDR1 could provide new insight into how extracellular matrix impacts cyst growth led us to study the role of DDR1 in ADPKD pathogenesis. However, genetic deletion of DDR1 using CRISPR/Cas9 failed to slow cyst growth or preserve kidney function in both a rapid and slow mouse model of ADPKD demonstrating that DDR1 does not play a role in PKD pathogenesis and is thus a not viable drug target. In spite of the negative results, our studies will be of interest to the nephrology community as it will prevent others from potentially conducting similar experiments on DDR1 and reinforces the potential of performing unbiased screens coupled with in vivo gene editing using CRISPR/Cas9 to rapidly identify and confirm new potential drug targets for ADPKD.


Asunto(s)
Receptor con Dominio Discoidina 1/biosíntesis , Regulación Enzimológica de la Expresión Génica , Riñón/enzimología , Enfermedades Renales Poliquísticas/enzimología , Regulación hacia Arriba , Animales , Receptor con Dominio Discoidina 1/genética , Modelos Animales de Enfermedad , Riñón/patología , Ratones , Ratones Transgénicos , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/patología
4.
Int J Oncol ; 51(6): 1639-1650, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29039472

RESUMEN

In the present study, knockdown of E2F1 impaired the migration and invasion of osteosarcoma cells. Further analysis showed that E2F1 knockdown decreased the expression of discoidin domain receptor 1 (DDR1) which plays a crucial role in many fundamental processes such as cell differentiation, adhesion, migration and invasion. Luciferase and ChIP assays confirmed that E2F1 silencing attenuated the expression of DDR1 through disrupting E2F1-mediated transcription of DDR1 in osteosarcoma cells. Similarly with the effect of E2F1 silencing, DDR1 knockdown weakened the migratory and invasive capabilities of osteosarcoma cells; while overexpression of DDR1 resulted in a significant increase of cell motility and invasiveness, even after knocking down E2F1. Interestingly, inactivation of E2F1/DDR1 pathway by shRNA weakened STAT3 signaling and subsequently suppressed the epithelial-mesenchymal transition (EMT) of osteosarcoma cells, as shown with decreased vimentin, MMP2, MMP9, and increased E­cadherin. Consistently, high expressions of E2F1 and DDR1 observed in osteosarcoma tissues were related to TNM stage and metastasis. In addition, high level of E2F1 or DDR1 was associated with poor prognosis in osteosarcoma patients. These results suggest that E2F1/DDR1/STAT3 pathway is critical for malignancy of osteosarcoma, which may provide a novel prognostic indicator or approach for osteosarcoma therapy.


Asunto(s)
Neoplasias Óseas/metabolismo , Receptor con Dominio Discoidina 1/biosíntesis , Factor de Transcripción E2F1/metabolismo , Osteosarcoma/metabolismo , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Línea Celular Tumoral , Movimiento Celular , Receptor con Dominio Discoidina 1/genética , Factor de Transcripción E2F1/biosíntesis , Factor de Transcripción E2F1/genética , Transición Epitelial-Mesenquimal , Femenino , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Osteosarcoma/genética , Osteosarcoma/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Transcripción STAT3/metabolismo , Transcripción Genética
5.
Oncotarget ; 8(26): 43248-43270, 2017 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-28591735

RESUMEN

The fetal isoform A of the insulin receptor (IR-A) is frequently overexpressed in a variety of malignancies including breast cancer. IR overexpression has a recognized role in cancer progression and resistance to anticancer therapies. In particular, IR-A has a peculiar mitogenic potential and is activated not only by insulin but also by IGF-2. Previously, we identified discoidin domain receptor 1 (DDR1) as a new IR-A interacting protein. DDR1, a non-integrin collagen tyrosine kinase receptor, is overexpressed in several malignancies and plays a role in cancer progression and metastasis.We now evaluated whether DDR1 is able to exert a role in breast cancer biology by functionally cross-talking with IR. In MCF-7 human breast cancer cells, IR and DDR1 co-immunoprecipitated and co-localized after insulin or IGF-2 stimulation. In a panel of breast cancer cells, DDR1 knockdown by specific siRNAs markedly inhibited IR downstream signaling as well as proliferation, migration and colony formation in response to insulin and IGF-2. These effects were accompanied by reduction of IR protein and mRNA expression, which involved both transcriptional and post-transcriptional effects. DDR1 overexpression elicited opposite effects. Bioinformatics analysis of public domain databases showed that IR and DDR1 co-expression significantly correlates with several clinically relevant histopathological and molecular features of human breast carcinomas.These findings demonstrate that, in human breast cancer cells, DDR1 regulates IR expression and ligand dependent biological actions. This novel functional crosstalk is likely clinically relevant and may become a new molecular target in breast cancer.


Asunto(s)
Antígenos CD/metabolismo , Neoplasias de la Mama/metabolismo , Receptor con Dominio Discoidina 1/metabolismo , Receptor de Insulina/metabolismo , Antígenos CD/biosíntesis , Antígenos CD/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Receptor con Dominio Discoidina 1/biosíntesis , Receptor con Dominio Discoidina 1/genética , Femenino , Humanos , Insulina/farmacología , Factor II del Crecimiento Similar a la Insulina/farmacología , Células MCF-7 , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Cross-Talk , Receptor de Insulina/biosíntesis , Receptor de Insulina/genética , Transducción de Señal , Transfección
6.
Eur J Cell Biol ; 95(11): 503-512, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27720259

RESUMEN

Transforming growth factor-ß1 (TGF-ß1) is an important player in chronic liver diseases inducing fibrogenesis and hepatocellular carcinoma (HCC) development. TGF-ß1 promotes pleiotropic modifications at the cellular and matrix microenvironment levels. TGF-ß1 was described to enhance production of type I collagen and its associated cross-linking enzyme, the lysyl oxidase-like2 (LOXL2). In addition, TGF-ß1 and type I collagen are potent inducers of invadosomes. Indeed, type I collagen fibers induce the formation of active linear invadosomes through the discoidin domain receptor 1 (DDR1). The goal of our study was to address the role of TGF-ß1 in collagen cross-linking and its impact on the formation of linear invadosomes in liver cancer cells. We first report a significant correlation between expressions of TGF-ß1, and type I collagen, LOXL2, DDR1 and MT1-MMP in human HCCs. We demonstrate that TGF-ß1 promotes a Smad4-dependent up-regulation of DDR1, together with LOXL2, in cultured HCC cells. Moreover, we show that LOXL2-induced collagen cross-linking enhances linear invadosome formation. Altogether, our data demonstrate that TGF-ß1 favors linear invadosome formation through the expressions of both the inducers, such as collagen and LOXL2, and the components such as DDR1 and MT1-MMP of linear invadosomes in cancer cells. Meanwhile, our data uncover a new TGF-ß1-dependent regulation of DDR1 expression.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Colágeno Tipo I/metabolismo , Receptor con Dominio Discoidina 1/biosíntesis , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Colágeno Tipo I/genética , Receptor con Dominio Discoidina 1/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Factor de Crecimiento Transformador beta1/genética
7.
Adv Healthc Mater ; 5(12): 1481-92, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27125602

RESUMEN

This study reports how extracellular matrix (ECM) ligand self-assembly on biomaterial surfaces and the resulting nanoscale architecture can drive stem cell behavior. To isolate the biological effects of surface wettability on protein deposition, folding, and ligand activity, a polydimethylsiloxane (PDMS)-based platform was developed and characterized with the ability to tune wettability of elastomeric substrates with otherwise equivalent topology, ligand loading, and mechanical properties. Using this platform, markedly different assembly of covalently bound type I collagen monomers was observed depending on wettability, with hydrophobic substrates yielding a relatively rough layer of collagen aggregates compared to a smooth collagen layer on more hydrophilic substrates. Cellular and molecular investigations with human bone marrow stromal cells revealed higher osteogenic differentiation and upregulation of focal adhesion-related components on the resulting smooth collagen layer coated substrates. The initial collagen assembly driven by the PDMS surface directly affected α1ß1 integrin/discoidin domain receptor 1 signaling, activation of the extracellular signal-regulated kinase/mitogen activated protein kinase pathway, and ultimately markers of osteogenic stem cell differentiation. We demonstrate for the first time that surface-driven ligand assembly on material surfaces, even on materials with otherwise identical starting topographies and mechanical properties, can dominate the biomaterial surface-driven cell response.


Asunto(s)
Matriz Extracelular/química , Osteogénesis , Transducción de Señal , Células Madre/metabolismo , Diferenciación Celular , Colágeno/química , Receptor con Dominio Discoidina 1/biosíntesis , Humanos , Integrina alfa1beta1/biosíntesis , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...