Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
J Cell Sci ; 136(20)2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-37870164

RESUMEN

Tumor initiation at either primary or metastatic sites is an inefficient process in which tumor cells must fulfill a series of conditions. One critical condition involves the ability of individual tumor-initiating cells to overcome 'isolation stress', enabling them to survive within harsh isolating microenvironments that can feature nutrient stress, hypoxia, oxidative stress and the absence of a proper extracellular matrix (ECM). In response to isolation stress, tumor cells can exploit various adaptive strategies to develop stress tolerance and gain stemness features. In this Opinion, we discuss how strategies such as the induction of certain cell surface receptors and deposition of ECM proteins enable tumor cells to endure isolation stress, thereby gaining tumor-initiating potential. As examples, we highlight recent findings from our group demonstrating how exposure of tumor cells to isolation stress upregulates the G-protein-coupled receptor lysophosphatidic acid receptor 4 (LPAR4), its downstream target fibronectin and two fibronectin-binding integrins, α5ß1 and αvß3. These responses create a fibronectin-rich niche for tumor cells, ultimately driving stress tolerance, cancer stemness and tumor initiation. We suggest that approaches to prevent cancer cells from adapting to stress by suppressing LPAR4 induction, blocking its downstream signaling or disrupting fibronectin-integrin interactions hold promise as potential strategies for cancer treatment.


Asunto(s)
Fibronectinas , Integrinas , Fibronectinas/metabolismo , Adhesión Celular/fisiología , Regulación hacia Arriba , Integrinas/metabolismo , Integrina alfa5beta1/metabolismo , Matriz Extracelular/metabolismo , Integrina alfaVbeta3/metabolismo
3.
Blood ; 119(9): 2149-58, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22134168

RESUMEN

Vascular development and angiogenesis initially depend on endothelial tip cell invasion, which is followed by a series of maturation steps, including lumen formation and recruitment of perivascular cells. Notch ligands expressed on the endothelium and their cognate receptors expressed on perivascular cells are involved in blood vessel maturation, though little is known regarding the Notch-dependent effectors that facilitate perivascular coverage of nascent vessels. Here, we report that vascular smooth muscle cell (VSMC) recognition of the Notch ligand Jagged1 on endothelial cells leads to expression of integrin αvß3 on VSMCs. Once expressed, integrin αvß3 facilitates VSMC adhesion to VWF in the endothelial basement membrane of developing retinal arteries, leading to vessel maturation. Genetic or pharmacologic disruption of Jagged1, Notch, αvß3, or VWF suppresses VSMC coverage of nascent vessels and arterial maturation during vascular development. Therefore, we define a Notch-mediated interaction between the developing endothelium and VSMCs leading to adhesion of VSMCs to the endothelial basement membrane and arterial maturation.


Asunto(s)
Membrana Basal/metabolismo , Adhesión Celular/fisiología , Endotelio Vascular/metabolismo , Integrinas/metabolismo , Músculo Liso Vascular/metabolismo , Receptores Notch/metabolismo , Animales , Arterias/metabolismo , Proteínas de Unión al Calcio/metabolismo , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Humanos , Integrinas/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/metabolismo , Neovascularización Fisiológica/genética , Unión Proteica , ARN Mensajero/metabolismo , Receptores Notch/genética , Proteínas Serrate-Jagged , Transducción de Señal/fisiología , Factor de von Willebrand/metabolismo
4.
Cancer Res ; 84(10): 1630-1642, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38588407

RESUMEN

Cancer stem/tumor-initiating cells display stress tolerance and metabolic flexibility to survive in a harsh environment with limited nutrient and oxygen availability. The molecular mechanisms underlying this phenomenon could provide targets to prevent metabolic adaptation and halt cancer progression. Here, we showed in cultured cells and live human surgical biopsies of non-small cell lung cancer that nutrient stress drives the expression of the epithelial cancer stem cell marker integrin αvß3 via upregulation of the ß3 subunit, resulting in a metabolic reprogramming cascade that allows tumor cells to thrive despite a nutrient-limiting environment. Although nutrient deprivation is known to promote acute, yet transient, activation of the stress sensor AMP-activated protein kinase (AMPK), stress-induced αvß3 expression via Src activation unexpectedly led to secondary and sustained AMPK activation. This resulted in the nuclear localization of peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC1α) and upregulation of glutamine metabolism, the tricarboxylic acid cycle, and oxidative phosphorylation. Pharmacological or genetic targeting of this axis prevented lung cancer cells from evading the effects of nutrient stress, thereby blocking tumor initiation in mice following orthotopic implantation of lung cancer cells. These findings reveal a molecular pathway driven by nutrient stress that results in cancer stem cell reprogramming to promote metabolic flexibility and tumor initiation. SIGNIFICANCE: Upregulation of integrin αvß3, a cancer stem cell marker, in response to nutrient stress activates sustained AMPK/PGC1α signaling that induces metabolic reprogramming in lung cancer cells to support their survival. See related commentary by Rainero, p. 1543.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Integrina alfaVbeta3 , Neoplasias Pulmonares , Regulación hacia Arriba , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/genética , Animales , Integrina alfaVbeta3/metabolismo , Ratones , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Estrés Fisiológico , Nutrientes/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica
5.
Trends Cell Biol ; 2023 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-37640611

RESUMEN

While the tumor microenvironment is a critical contributor to cancer progression, early steps of tumor initiation and metastasis also rely on the ability of individual tumor cells to survive and thrive at locations where tumor stroma or immune infiltration has yet to be established. In this opinion article, we use the term 'isolation stress' to broadly describe the challenges that individual tumor cells must overcome during the initiation and expansion of the primary tumor beyond permissive boundaries and metastatic spread into distant sites, including a lack of cell-cell contact, adhesion to protumor extracellular matrix proteins, and access to nutrients, oxygen, and soluble factors that support growth. In particular, we highlight the ability of solitary tumor cells to autonomously generate a specialized fibronectin-enriched extracellular matrix to create their own pericellular niche that supports tumor initiation. Cancer cells that can creatively evade the effects of isolation stress not only become more broadly stress tolerant, they also tend to show enhanced stemness, drug resistance, tumor initiation, and metastasis.

6.
Nat Cell Biol ; 25(2): 309-322, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36646789

RESUMEN

Defining drivers of tumour initiation can provide opportunities to control cancer progression. Here we report that lysophosphatidic acid receptor 4 (LPAR4) becomes transiently upregulated on pancreatic cancer cells exposed to environmental stress or chemotherapy where it promotes stress tolerance, drug resistance, self-renewal and tumour initiation. Pancreatic cancer cells gain LPAR4 expression in response to stress by downregulating a tumour suppressor, miR-139-5p. Even in the absence of exogenous lysophosphatidic acid, LPAR4-expressing tumour cells display an enrichment of extracellular matrix genes that are established drivers of cancer stemness. Mechanistically, upregulation of fibronectin via an LPAR4/AKT/CREB axis is indispensable for LPAR4-induced tumour initiation and stress tolerance. Moreover, ligation of this fibronectin-containing matrix via integrins α5ß1 or αVß3 can transfer stress tolerance to LPAR4-negative cells. Therefore, stress- or drug-induced LPAR4 enhances cell-autonomous production of a fibronectin-rich extracellular matrix, allowing cells to survive 'isolation stress' and compensate for the absence of stromal-derived factors by creating their own tumour-initiating niche.


Asunto(s)
MicroARNs , Neoplasias Pancreáticas , Receptores Purinérgicos P2 , Humanos , Fibronectinas/genética , Fibronectinas/metabolismo , Neoplasias Pancreáticas/patología , Matriz Extracelular/metabolismo , Transformación Celular Neoplásica/metabolismo , Receptores Purinérgicos P2/metabolismo , MicroARNs/genética , Neoplasias Pancreáticas
7.
J Clin Invest ; 118(6): 2337-46, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18483622

RESUMEN

Retinal and choroidal vascular diseases, with their associated abnormalities in vascular permeability, account for the majority of patients with vision loss in industrialized nations. VEGF is upregulated in ischemic retinopathies such as diabetes and is known to dramatically alter vascular permeability in a number of nonocular tissues via Src kinase-regulated signaling pathways. VEGF antagonists are currently in clinical use for treating the new blood vessels and retinal edema associated with neovascular eye diseases, but such therapies require repeated intraocular injections. We have found that vascular leakage following intravitreal administration of VEGF in mice was abolished by systemic or topical delivery of what we believe is a novel VEGFR2/Src kinase inhibitor; this was confirmed in rabbits. The relevance of Src inhibition to VEGF-associated alterations in vascular permeability was further substantiated by genetic studies in which VEGF injection or laser-induced vascular permeability failed to augment retinal vascular permeability in Src-/- and Yes-/- mice (Src and Yes are ubiquitously expressed Src kinase family members; Src-/- and Yes-/- mice lacking expression of these kinases show no vascular leak in response to VEGF). These findings establish a role for Src kinase in VEGF-mediated retinal vascular permeability and establish a potentially safe and painless topically applied therapeutic option for treating vision loss due to neovascular-associated retinal edema.


Asunto(s)
Permeabilidad Capilar , Inhibidores Enzimáticos/farmacología , Retina/patología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Modelos Biológicos , Permeabilidad , Conejos , Transducción de Señal , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Familia-src Quinasas/metabolismo
8.
Liver Int ; 31(8): 1222-30, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21745297

RESUMEN

BACKGROUND AND AIMS: Acute liver failure (ALF) is frequently complicated by cerebral oedema, systemic inflammation and multiorgan dysfunction. Vascular endothelial growth factor (VEGF) may stimulate liver regeneration but it can also be pro-inflammatory, activating endothelial cells and increasing permeability, actions mediated through Src kinase signalling. We therefore examined whether a Src inhibitor could have therapeutic potential in ALF. METHODS: Murine ALF was induced with azoxymethane. Liver pathology was graded by a blinded examiner and apoptosis quantified by immunohistochemistry. Cerebral VEGF expression was imaged using VEGF-green fluorescent protein transgenic mice. Circulating and macrophage-secreted VEGF levels were measured. Experimental animals received a Src inhibitor or vehicle controls. RESULTS: VEGF was undetectable in normal plasma but reached a mean of 835 pg/ml at grade III encephalopathy (P<0.001). Ammonia, lipopolysaccharide and interferon-gamma acted synergistically to enhance VEGF secretion by macrophages. Production of VEGF by cerebral cortical astrocytes increased with disease progression. Late treatment with inhibitors of Src or VEGF did not improve liver histology, encephalopathy or survival. However, early use of a Src kinase inhibitor significantly reduced hepatic injury, delayed encephalopathy and allowed 25% of mice to survive an otherwise lethal insult. CONCLUSION: Systemic and cerebral VEGF levels are significantly elevated during experimental ALF and may be exacerbated by hyperammonemia and macrophage activation. Early use of a Src inhibitor reduced hepatocellular injury and enabled survival, indicating such agents may have some promise in the treatment of ALF.


Asunto(s)
Compuestos de Anilina/farmacología , Edema Encefálico/prevención & control , Encéfalo/efectos de los fármacos , Fallo Hepático Agudo/tratamiento farmacológico , Hígado/efectos de los fármacos , Nitrilos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Quinolinas/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Cloruro de Amonio/farmacología , Animales , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Azoximetano , Encéfalo/enzimología , Encéfalo/patología , Edema Encefálico/inducido químicamente , Edema Encefálico/enzimología , Edema Encefálico/patología , Células Cultivadas , Modelos Animales de Enfermedad , Factores de Crecimiento Endotelial/farmacología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Encefalopatía Hepática/enzimología , Encefalopatía Hepática/prevención & control , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Interferón gamma/metabolismo , Lipopolisacáridos/farmacología , Hígado/enzimología , Hígado/patología , Fallo Hepático Agudo/inducido químicamente , Fallo Hepático Agudo/enzimología , Fallo Hepático Agudo/patología , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Péptidos Cíclicos/farmacología , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/sangre , Factor A de Crecimiento Endotelial Vascular/genética , Familia-src Quinasas/metabolismo
9.
Nature ; 437(7058): 497-504, 2005 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-16177780

RESUMEN

Although vascular endothelial growth factor (VEGF) induces angiogenesis, it also disrupts vascular barrier function in diseased tissues. Accordingly, VEGF expression in cancer and ischaemic disease has unexpected pathophysiological consequences. By uncoupling endothelial cell-cell junctions VEGF causes vascular permeability and oedema, resulting in extensive injury to ischaemic tissues after stroke or myocardial infarction. In cancer, VEGF-mediated disruption of the vascular barrier may potentiate tumour cell extravasation, leading to widespread metastatic disease. Therefore, by blocking the vascular permeability promoting effects of VEGF it may be feasible to reduce tissue injury after ischaemic disease and minimize the invasive properties of circulating tumour cells.


Asunto(s)
Permeabilidad Capilar , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Permeabilidad Capilar/efectos de los fármacos , Humanos , Isquemia/metabolismo , Isquemia/patología , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neoplasias/patología , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología
10.
Proc Natl Acad Sci U S A ; 105(27): 9343-8, 2008 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-18607000

RESUMEN

Integrin alphanubeta3 is found on a subset of tumor blood vessels where it is associated with angiogenesis and malignant tumor growth. We designed an alphanubeta3-targeted nanoparticle (NP) encapsulating the cytotoxic drug doxorubicin (Dox) for targeted drug delivery to the alphanubeta3-expressing tumor vasculature. We observed real-time targeting of this NP to tumor vessels and noted selective apoptosis in regions of the alphanubeta3-expressing tumor vasculature. In clinically relevant pancreatic and renal cell orthotopic models of spontaneous metastasis, targeted delivery of Dox produced an antimetastatic effect. In fact, alphanubeta3-mediated delivery of this drug to the tumor vasculature resulted in a 15-fold increase in antimetastatic activity without producing drug-associated weight loss as observed with systemic administration of the free drug. These findings reveal that NP-based delivery of cytotoxic drugs to the alphanubeta3-positive tumor vasculature represents an approach for treating metastatic disease.


Asunto(s)
Doxorrubicina/administración & dosificación , Doxorrubicina/uso terapéutico , Sistemas de Liberación de Medicamentos , Nanopartículas , Metástasis de la Neoplasia/tratamiento farmacológico , Neoplasias/irrigación sanguínea , Neoplasias/patología , Inhibidores de la Angiogénesis/farmacología , Animales , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Doxorrubicina/farmacología , Endotelio/efectos de los fármacos , Endotelio/patología , Integrina alfaVbeta3/metabolismo , Neoplasias Renales/patología , Ratones , Neovascularización Patológica/patología , Oligopéptidos/farmacología , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cell Stem Cell ; 26(2): 187-204.e10, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-31956038

RESUMEN

Zika virus (ZIKV) causes microcephaly by killing neural precursor cells (NPCs) and other brain cells. ZIKV also displays therapeutic oncolytic activity against glioblastoma (GBM) stem cells (GSCs). Here we demonstrate that ZIKV preferentially infected and killed GSCs and stem-like cells in medulloblastoma and ependymoma in a SOX2-dependent manner. Targeting SOX2 severely attenuated ZIKV infection, in contrast to AXL. As mechanisms of SOX2-mediated ZIKV infection, we identified inverse expression of antiviral interferon response genes (ISGs) and positive correlation with integrin αv (ITGAV). ZIKV infection was disrupted by genetic targeting of ITGAV or its binding partner ITGB5 and by an antibody specific for integrin αvß5. ZIKV selectively eliminated GSCs from species-matched human mature cerebral organoids and GBM surgical specimens, which was reversed by integrin αvß5 inhibition. Collectively, our studies identify integrin αvß5 as a functional cancer stem cell marker essential for GBM maintenance and ZIKV infection, providing potential brain tumor therapy.


Asunto(s)
Glioblastoma , Células-Madre Neurales , Infección por el Virus Zika , Virus Zika , Humanos , Receptores de Vitronectina , Factores de Transcripción SOXB1/genética
12.
J Cell Biol ; 167(2): 223-9, 2004 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-15504909

RESUMEN

VEGF is unique among angiogenic growth factors because it disrupts endothelial barrier function. Therefore, we considered whether this property of VEGF might contribute to tumor cell extravasation and metastasis. To test this, mice lacking the Src family kinases Src or Yes, which maintain endothelial barrier function in the presence of VEGF, were injected intravenously with VEGF-expressing tumor cells. We found a dramatic reduction in tumor cell extravasation in lungs or livers of mice lacking Src or Yes. At the molecular level, VEGF compromises the endothelial barrier by disrupting a VE-cadherin-beta-catenin complex in lung endothelium from wild-type, but not Yes-deficient, mice. Disrupting the endothelial barrier directly with anti-VE-cadherin both amplifies metastasis in normal mice and overcomes the genetic resistance in Yes-deficient mice. Pharmacological blockade of VEGF, VEGFR-2, or Src stabilizes endothelial barrier function and suppresses tumor cell extravasation in vivo. Therefore, disrupting Src signaling preserves host endothelial barrier function providing a novel host-targeted approach to control metastatic disease.


Asunto(s)
Endotelio Vascular/metabolismo , Neoplasias/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Familia-src Quinasas/metabolismo , Animales , Antígenos CD , Cadherinas/metabolismo , Adhesión Celular , Línea Celular Tumoral , Proteínas del Citoesqueleto/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Eliminación de Gen , Proteínas Fluorescentes Verdes/metabolismo , Hígado/patología , Pulmón/patología , Ratones , Microscopía Electrónica , Metástasis de la Neoplasia , Neoplasias Ováricas/patología , Proteínas Proto-Oncogénicas c-yes , Retroviridae/genética , Transducción de Señal , Factores de Tiempo , Transactivadores/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , beta Catenina
13.
Cancer Res ; 79(19): 5048-5059, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31416839

RESUMEN

Tumor-associated macrophages (TAM) are highly expressed within the tumor microenvironment of a wide range of cancers, where they exert a protumor phenotype by promoting tumor cell growth and suppressing antitumor immune function. Here, we show that TAM accumulation in human and mouse tumors correlates with tumor cell expression of integrin αvß3, a known driver of epithelial cancer progression and drug resistance. A monoclonal antibody targeting αvß3 (LM609) exploited the coenrichment of αvß3 and TAMs to not only eradicate highly aggressive drug-resistant human lung and pancreas cancers in mice, but also to prevent the emergence of circulating tumor cells. Importantly, this antitumor activity in mice was eliminated following macrophage depletion. Although LM609 had no direct effect on tumor cell viability, it engaged macrophages but not natural killer (NK) cells to induce antibody-dependent cellular cytotoxicity (ADCC) of αvß3-expressing tumor cells despite their expression of the CD47 "don't eat me" signal. In contrast to strategies designed to eliminate TAMs, these findings suggest that anti-αvß3 represents a promising immunotherapeutic approach to redirect TAMs to serve as tumor killers for late-stage or drug-resistant cancers. SIGNIFICANCE: Therapeutic antibodies are commonly engineered to optimize engagement of NK cells as effectors. In contrast, LM609 targets αvß3 to suppress tumor progression and enhance drug sensitivity by exploiting TAMs to trigger ADCC.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Integrina alfaVbeta3/inmunología , Macrófagos/inmunología , Neoplasias Glandulares y Epiteliales/inmunología , Animales , Antineoplásicos/farmacología , Progresión de la Enfermedad , Humanos , Ratones , Neoplasias Glandulares y Epiteliales/patología , Fagocitosis/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
14.
J Clin Invest ; 113(6): 885-94, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15067321

RESUMEN

Ischemia resulting from myocardial infarction (MI) promotes VEGF expression, leading to vascular permeability (VP) and edema, a process that we show here contributes to tissue injury throughout the ventricle. This permeability/edema can be assessed noninvasively by MRI and can be observed at the ultrastructural level as gaps between adjacent endothelial cells. Many of these gaps contain activated platelets adhering to exposed basement membrane, reducing vessel patency. Following MI, genetic or pharmacological blockade of Src preserves endothelial cell barrier function, suppressing VP and infarct volume, providing long-term improvement in cardiac function, fibrosis, and survival. To our surprise, an intravascular injection of VEGF into healthy animals, but not those deficient in Src, induced similar endothelial gaps, VP, platelet plugs, and some myocyte damage. Mechanistically, we show that quiescent blood vessels contain a complex involving Flk, VE-cadherin, and beta-catenin that is transiently disrupted by VEGF injection. Blockade of Src prevents disassociation of this complex with the same kinetics with which it prevents VEGF-mediated VP/edema. These findings define a molecular mechanism to account for the Src requirement in VEGF-mediated permeability and provide a basis for Src inhibition as a therapeutic option for patients with acute MI.


Asunto(s)
Cadherinas/metabolismo , Edema/metabolismo , Infarto del Miocardio/metabolismo , Familia-src Quinasas/metabolismo , Animales , Cadherinas/efectos de los fármacos , Ratas , Factor A de Crecimiento Endotelial Vascular/efectos adversos , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/efectos de los fármacos
15.
Methods Enzymol ; 426: 505-28, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17697897

RESUMEN

All vascular biological processes are influenced to some degree by integrins expressed on endothelial cells, vascular smooth muscle cells, fibroblasts, platelets, or other circulating cells. In particular, angiogenesis requires cells to process signals from their microenvironment and respond by altering their cell-cell and cell-matrix adhesion, events which allow migration and vascular remodeling over the period of days to weeks. On the other hand, endothelial cells can respond to a permeability stimulus and alter their junctional adhesion molecules or vesicular transport machinery within seconds or minutes. This chapter will discuss the current understanding of how integrins participate in these processes, and explore the in vitro and in vivo models available to study the role of integrin function during angiogenesis and vascular leak.


Asunto(s)
Vasos Sanguíneos/fisiología , Permeabilidad Capilar/fisiología , Integrinas/metabolismo , Modelos Biológicos , Neovascularización Fisiológica/fisiología , Animales , Vasos Sanguíneos/citología , Vasos Sanguíneos/crecimiento & desarrollo , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Humanos
16.
Cancer Discov ; 7(12): 1464-1479, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28893801

RESUMEN

Identifying the molecular basis for cancer cell dependence on oncogenes such as KRAS can provide new opportunities to target these addictions. Here, we identify a novel role for the carbohydrate-binding protein galectin-3 as a lynchpin for KRAS dependence. By directly binding to the cell surface receptor integrin αvß3, galectin-3 gives rise to KRAS addiction by enabling multiple functions of KRAS in anchorage-independent cells, including formation of macropinosomes that facilitate nutrient uptake and ability to maintain redox balance. Disrupting αvß3/galectin-3 binding with a clinically active drug prevents their association with mutant KRAS, thereby suppressing macropinocytosis while increasing reactive oxygen species to eradicate αvß3-expressing KRAS-mutant lung and pancreatic cancer patient-derived xenografts and spontaneous tumors in mice. Our work reveals galectin-3 as a druggable target for KRAS-addicted lung and pancreas cancers, and indicates integrin αvß3 as a biomarker to identify susceptible tumors.Significance: There is a significant unmet need for therapies targeting KRAS-mutant cancers. Here, we identify integrin αvß3 as a biomarker to identify mutant KRAS-addicted tumors that are highly sensitive to inhibition of galectin-3, a glycoprotein that binds to integrin αvß3 to promote KRAS-mediated activation of AKT. Cancer Discov; 7(12); 1464-79. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 1355.


Asunto(s)
Galectina 3/genética , Neoplasias Pulmonares/genética , Proteínas ras/genética , Animales , Galectina 3/metabolismo , Humanos , Neoplasias Pulmonares/patología , Ratones , Transducción de Señal
17.
Cancer Cell ; 32(6): 856-868.e5, 2017 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-29198914

RESUMEN

While molecular subtypes of glioblastoma (GBM) are defined using gene expression and mutation profiles, we identify a unique subpopulation based on addiction to the high-affinity glucose transporter, Glut3. Although Glut3 is a known driver of a cancer stem cell phenotype, direct targeting is complicated by its expression in neurons. Using established GBM lines and patient-derived stem cells, we identify a subset of tumors within the "proneural" and "classical" subtypes that are addicted to aberrant signaling from integrin αvß3, which activates a PAK4-YAP/TAZ signaling axis to enhance Glut3 expression. This defined subpopulation of GBM is highly sensitive to agents that disrupt this pathway, including the integrin antagonist cilengitide, providing a targeted therapeutic strategy for this unique subset of GBM tumors.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Transportador de Glucosa de Tipo 3/metabolismo , Integrina alfaVbeta3/metabolismo , Transcriptoma , Animales , Antineoplásicos/farmacología , Neoplasias Encefálicas/mortalidad , Línea Celular Tumoral , Perfilación de la Expresión Génica , Glioblastoma/mortalidad , Humanos , Estimación de Kaplan-Meier , Ratones , Ratones Desnudos , Transducción de Señal , Venenos de Serpiente/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Nat Commun ; 7: 13597, 2016 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-27886180

RESUMEN

Rather than targeting tumour cells directly, elements of the tumour microenvironment can be modulated to sensitize tumours to the effects of therapy. Here we report a unique mechanism by which ectopic microRNA-103 can manipulate tumour-associated endothelial cells to enhance tumour cell death. Using gain-and-loss of function approaches, we show that miR-103 exacerbates DNA damage and inhibits angiogenesis in vitro and in vivo. Local, systemic or vascular-targeted delivery of miR-103 in tumour-bearing mice decreased angiogenesis and tumour growth. Mechanistically, miR-103 regulation of its target gene TREX1 in endothelial cells governs the secretion of pro-inflammatory cytokines into the tumour microenvironment. Our data suggest that this inflammatory milieu may potentiate tumour cell death by supporting immune activation and inducing tumour expression of Fas and TRAIL receptors. Our findings reveal miR-mediated crosstalk between vasculature and tumour cells that can be exploited to improve the efficacy of chemotherapy and radiation.


Asunto(s)
Exodesoxirribonucleasas/genética , MicroARNs/metabolismo , Neoplasias/genética , Neovascularización Patológica/genética , Fosfoproteínas/genética , Microambiente Tumoral/genética , Animales , Línea Celular Tumoral , Regulación hacia Abajo , Exodesoxirribonucleasas/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/administración & dosificación , MicroARNs/genética , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Neovascularización Patológica/patología , Neovascularización Patológica/radioterapia , Fosfoproteínas/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Microambiente Tumoral/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto , Receptor fas/metabolismo
19.
Matrix Biol ; 24(4): 313-24, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15949932

RESUMEN

Because the small leucine-rich proteoglycan decorin has been implicated in regulation of collagen fibrillogenesis leading to proper extracellular matrix assembly, we hypothesized it could play a key role in cardiac fibrosis following myocardial infarction. In this study we ligated the left anterior descending coronary artery in wildtype and decorin-null mice to produce large infarcts in the anterior wall of the left ventricle. At early stages post-coronary occlusion the myocardial infarction size did not appreciably differ between the two genotypes. However, we found a wider distribution of collagen fibril sizes with less organization and loose packing in mature scar from decorin-null mice. Thus, we tested the hypothesis that these abnormal collagen fibrils would adversely affect post-infarction mechanics and ventricular remodeling. Indeed, scar size, right ventricular remote hypertrophy, and left ventricular dilatation were greater in decorin-null animals compared with wildtype littermates 14 days after acute myocardial infarction. Echocardiography revealed depressed left ventricular systolic function between 4 and 8 weeks post-ischemia in the decorin-null animals. These changes indicate that decorin is required for the proper fibrotic evolution of myocardial infarctions, and that its absence leads to abnormal scar tissue formation. This might contribute to aneurysmal ventricular dilatation, remote hypertrophy, and depressed ventricular function.


Asunto(s)
Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Proteoglicanos/metabolismo , Animales , Peso Corporal/genética , Cicatriz/metabolismo , Cicatriz/patología , Colágeno/química , Colágeno/metabolismo , Decorina , Proteínas de la Matriz Extracelular , Fibrosis/metabolismo , Fibrosis/patología , Regulación de la Expresión Génica , Genotipo , Ratones , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/cirugía , Tamaño de los Órganos/genética , Proteoglicanos/deficiencia , Proteoglicanos/genética , Factor de Crecimiento Transformador beta/metabolismo
20.
Trends Cell Biol ; 25(4): 234-40, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25572304

RESUMEN

Interactions between cancer cells and their surroundings can trigger essential signaling cues that determine cell fate and influence the evolution of the malignant phenotype. As the primary receptors involved in cell-matrix adhesion, integrins present on the surface of tumor and stromal cells have a profound impact on the ability to survive in specific locations, but in some cases, these receptors can also function in the absence of ligand binding to promote stemness and survival in the presence of environmental and therapeutic stresses. Understanding how integrin expression and function is regulated in this context will enable the development of new therapeutic approaches to sensitize tumors to therapy and suppress their metastatic phenotype.


Asunto(s)
Resistencia a Medicamentos/efectos de los fármacos , Integrinas/metabolismo , Metástasis de la Neoplasia/terapia , Neoplasias/metabolismo , Células Madre/metabolismo , Adhesión Celular , Humanos , Neoplasias/terapia , Transducción de Señal , Células del Estroma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA