Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell ; 154(3): 651-63, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23911327

RESUMEN

Vessel sprouting by migrating tip and proliferating stalk endothelial cells (ECs) is controlled by genetic signals (such as Notch), but it is unknown whether metabolism also regulates this process. Here, we show that ECs relied on glycolysis rather than on oxidative phosphorylation for ATP production and that loss of the glycolytic activator PFKFB3 in ECs impaired vessel formation. Mechanistically, PFKFB3 not only regulated EC proliferation but also controlled the formation of filopodia/lamellipodia and directional migration, in part by compartmentalizing with F-actin in motile protrusions. Mosaic in vitro and in vivo sprouting assays further revealed that PFKFB3 overexpression overruled the pro-stalk activity of Notch, whereas PFKFB3 deficiency impaired tip cell formation upon Notch blockade, implying that glycolysis regulates vessel branching.


Asunto(s)
Células Endoteliales/metabolismo , Glucólisis , Neovascularización Fisiológica , Fosfofructoquinasa-2/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Células Endoteliales/citología , Femenino , Eliminación de Gen , Silenciador del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfofructoquinasa-2/genética , Seudópodos/metabolismo , Pez Cebra
2.
Blood ; 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38905635

RESUMEN

The interaction between menin and histone-lysine N-methyltransferase 2A (KMT2A) is a critical dependency for KMT2A- or nucleophosmin 1 (NPM1)-altered leukemias and an emerging opportunity for therapeutic development. JNJ-75276617 is a novel, orally bioavailable, potent, and selective protein-protein interaction inhibitor of the binding between menin and KMT2A. In KMT2A-rearranged (KMT2A-r) and NPM1-mutant (NPM1c) AML cells, JNJ-75276617 inhibited the association of the menin-KMT2A complex with chromatin at target gene promoters, resulting in reduced expression of several menin-KMT2A target genes, including MEIS1 and FLT3. JNJ-75276617 displayed potent anti-proliferative activity across several AML and ALL cell lines and patient samples harboring KMT2A- or NPM1-alterations in vitro. In xenograft models of AML and ALL, JNJ-75276617 reduced leukemic burden and provided a significant dose-dependent survival benefit accompanied by expression changes of menin-KMT2A target genes. JNJ-75276617 demonstrated synergistic effects with gilteritinib in vitro in AML cells harboring KMT2A-r. JNJ-75276617 further exhibited synergistic effects with venetoclax and azacitidine in AML cells bearing KMT2A-r in vitro, and significantly increased survival in mice. Interestingly, JNJ-75276617 showed potent anti-proliferative activity in cell lines engineered with recently discovered mutations (MEN1M327I or MEN1T349M) that developed in patients refractory to the menin-KMT2A inhibitor revumenib. A co-crystal structure of menin in complex with JNJ-75276617 indicates a unique binding mode distinct from other menin-KMT2A inhibitors, including revumenib. JNJ-75276617 is being clinically investigated for acute leukemias harboring KMT2A or NPM1 alterations, as a monotherapy for relapsed/refractory (R/R) acute leukemia (NCT04811560), or in combination with AML-directed therapies (NCT05453903).

3.
Nature ; 537(7618): 63-68, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27533040

RESUMEN

Hypermethylation of the promoters of tumour suppressor genes represses transcription of these genes, conferring growth advantages to cancer cells. How these changes arise is poorly understood. Here we show that the activity of oxygen-dependent ten-eleven translocation (TET) enzymes is reduced by tumour hypoxia in human and mouse cells. TET enzymes catalyse DNA demethylation through 5-methylcytosine oxidation. This reduction in activity occurs independently of hypoxia-associated alterations in TET expression, proliferation, metabolism, hypoxia-inducible factor activity or reactive oxygen species, and depends directly on oxygen shortage. Hypoxia-induced loss of TET activity increases hypermethylation at gene promoters in vitro. In patients, tumour suppressor gene promoters are markedly more methylated in hypoxic tumour tissue, independent of proliferation, stromal cell infiltration and tumour characteristics. Our data suggest that up to half of hypermethylation events are due to hypoxia, with these events conferring a selective advantage. Accordingly, increased hypoxia in mouse breast tumours increases hypermethylation, while restoration of tumour oxygenation abrogates this effect. Tumour hypoxia therefore acts as a novel regulator of DNA methylation.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/deficiencia , Oxigenasas de Función Mixta/deficiencia , Oxígeno/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Hipoxia Tumoral/fisiología , 5-Metilcitosina/metabolismo , Animales , Proliferación Celular , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Femenino , Silenciador del Gen/efectos de los fármacos , Genes Supresores de Tumor , Humanos , Masculino , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Oxidación-Reducción/efectos de los fármacos , Oxígeno/farmacología , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Células del Estroma/patología , Hipoxia Tumoral/efectos de los fármacos , Hipoxia Tumoral/genética
4.
Nature ; 511(7508): 167-76, 2014 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-25008522

RESUMEN

Cancer cells have been at the centre of cell metabolism research, but the metabolism of stromal and immune cells has received less attention. Nonetheless, these cells influence the progression of malignant, inflammatory and metabolic disorders. Here we discuss the metabolic adaptations of stromal and immune cells in health and disease, and highlight how metabolism determines their differentiation and function.


Asunto(s)
Macrófagos/metabolismo , Células del Estroma/metabolismo , Linfocitos T/metabolismo , Animales , Diferenciación Celular , Células Endoteliales/citología , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Glucólisis , Humanos , Macrófagos/citología , Neoplasias/metabolismo , Neoplasias/patología , Células del Estroma/citología , Células del Estroma/enzimología , Linfocitos T/citología
5.
Trends Biochem Sci ; 38(1): 3-11, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23200187

RESUMEN

Oxygen-sensing prolyl hydroxylase domain enzymes (PHDs) target hypoxia-inducible factor (HIF)-α subunits for proteasomal degradation in normoxia through hydroxylation. Recently, novel mechanisms of PHD activation and function have been unveiled. Interestingly, PHD3 can unexpectedly amplify HIF signaling through hydroxylation of the glycolytic enzyme pyruvate kinase (PK) muscle isoform 2 (PKM2). Recent studies have also yielded insight into HIF-independent PHD functions, including the control of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor trafficking in synaptic transmission and the activation of transient receptor potential cation channel member A1 (TRPA1) ion channels by oxygen levels in sensory nerves. Finally, PHD activation has been shown to involve the iron chaperoning function of poly(rC) binding protein (PCBP)1 and the (R)-enantiomer of 2-hydroxyglutarate (2-HG). The intersection of these regulatory pathways and interactions highlight the complexity of PHD regulation and function.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Oxígeno/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Transducción de Señal , Animales , Humanos
6.
Nature ; 478(7369): 399-403, 2011 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-22012397

RESUMEN

Angiogenesis is critical during tumour initiation and malignant progression. Different strategies aimed at blocking vascular endothelial growth factor (VEGF) and its receptors have been developed to inhibit angiogenesis in cancer patients. It has become increasingly clear that in addition to its effect on angiogenesis, other mechanisms including a direct effect of VEGF on tumour cells may account for the efficiency of VEGF-blockade therapies. Cancer stem cells (CSCs) have been described in various cancers including squamous tumours of the skin. Here we use a mouse model of skin tumours to investigate the impact of the vascular niche and VEGF signalling on controlling the stemness (the ability to self renew and differentiate) of squamous skin tumours during the early stages of tumour progression. We show that CSCs of skin papillomas are localized in a perivascular niche, in the immediate vicinity of endothelial cells. Furthermore, blocking VEGFR2 caused tumour regression not only by decreasing the microvascular density, but also by reducing CSC pool size and impairing CSC renewal properties. Conditional deletion of Vegfa in tumour epithelial cells caused tumours to regress, whereas VEGF overexpression by tumour epithelial cells accelerated tumour growth. In addition to its well-known effect on angiogenesis, VEGF affected skin tumour growth by promoting cancer stemness and symmetric CSC division, leading to CSC expansion. Moreover, deletion of neuropilin-1 (Nrp1), a VEGF co-receptor expressed in cutaneous CSCs, blocked VEGF's ability to promote cancer stemness and renewal. Our results identify a dual role for tumour-cell-derived VEGF in promoting cancer stemness: by stimulating angiogenesis in a paracrine manner, VEGF creates a perivascular niche for CSCs, and by directly affecting CSCs through Nrp1 in an autocrine loop, VEGF stimulates cancer stemness and renewal. Finally, deletion of Nrp1 in normal epidermis prevents skin tumour initiation. These results may have important implications for the prevention and treatment of skin cancers.


Asunto(s)
Carcinoma de Células Escamosas/irrigación sanguínea , Carcinoma de Células Escamosas/patología , Neuropilina-1/metabolismo , Transducción de Señal , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/citología , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Ratones , Células Madre Neoplásicas , Neuropilina-1/genética , Factor A de Crecimiento Endotelial Vascular/genética
7.
J Hepatol ; 57(1): 61-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22420978

RESUMEN

BACKGROUND & AIMS: The two major primary liver cancers in adults are hepatocellular carcinoma and cholangiocarcinoma. These tumors rapidly outgrow their vascular supply and become hypoxic, resulting in the production of hypoxia inducible factors. Recently, interest has grown in the regulators of these factors. Several reports have been published describing the role of prolyl hydroxylase domains--the key oxygen sensor responsible for the degradation of hypoxia inducible factors--tumor progression and vascularisation. The effect of prolyl hydroxylase domain 2 on the pathogenesis of liver cancer has never been studied. METHODS: A diethylnitrosamine-induced mouse model was used in this study, allowing primary hepatic tumors to occur as a result of chronic liver damage. Several parameters of prolyl hydroxylase domain 2-haplodeficient mice were compared to those of wild type mice, thereby focussing on the expression of angiogenic factors and on the hepatic progenitor cell activation and differentiation. RESULTS: This study shows that inhibiting prolyl hydroxylase domain 2 increases the hepatocarcinogenesis and stimulates the development of cholangiocarcinoma. Furthermore, PHD2 deficiency and the accompanying continuous HIF activation, selected for a more metastatic tumor phenotype. CONCLUSIONS: The effect of prolyl hydroxylase domain 2 deficiency on hepatocarcinogenesis hold a great potential for therapeutic intervention, since hypoxia and the selection for a more aggressive cholangiocarcinoma phenotype might also have a repercussion on patients receiving long-term treatment with anti-angiogenic compounds.


Asunto(s)
Neoplasias de los Conductos Biliares/metabolismo , Conductos Biliares Intrahepáticos/metabolismo , Carcinoma Hepatocelular/metabolismo , Colangiocarcinoma/metabolismo , Neoplasias Hepáticas Experimentales/metabolismo , Procolágeno-Prolina Dioxigenasa/genética , Alquilantes/toxicidad , Animales , Neoplasias de los Conductos Biliares/inducido químicamente , Neoplasias de los Conductos Biliares/genética , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/genética , Colangiocarcinoma/inducido químicamente , Colangiocarcinoma/genética , Dietilnitrosamina/toxicidad , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Genotipo , Hepatitis/genética , Hepatitis/metabolismo , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/genética , Masculino , Ratones , Neovascularización Fisiológica/fisiología , Fenotipo , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Procolágeno-Prolina Dioxigenasa/deficiencia , Procolágeno-Prolina Dioxigenasa/metabolismo , Transducción de Señal/fisiología , Células Madre/fisiología
8.
Arch Pharm (Weinheim) ; 344(7): 431-41, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21618270

RESUMEN

A new series of 16 6-chloro-1,1-dioxo-7-{4-[(4-R(1)-phenyl)imino]-4H-3,1-benzoxazin-2-yl}-3-(substituted amino)-1,4,2-benzodithiazines 7-22 was prepared in order to evaluate the cytotoxic activity against six human cancer cell lines. The structures of the new compounds were confirmed by IR, (1)H-, and (13)C-NMR, elemental analysis and in the cases of 11 and 31 by X-ray crystal structure analysis. This analysis showed that contrary to our earlier report the structures contain a benzoxazine ring instead of the proposed quinazolinone ring. The bioassay indicated that the benzodithiazine derivatives 7-22 possess cancer cell growth-inhibitory properties. Some compounds showed a high level of selectivity for certain cell lines. The most active compounds 11, 12, 16, 19, 21, and 22 exhibited potency higher or comparable to cisplatin. The compounds were particularly effective in LCLC-103H and MCF-7 cell lines with IC(50) values of 0.49-1.60 µM. Quantitative structure activity relationships (QSAR) revealed that a chloro substituent R(1) in the phenyl ring as well as the shape of the substituted amino group at R(2) (e.g., unsaturation is beneficial) are important for potency.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Tiazinas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Cristalografía por Rayos X , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Concentración 50 Inhibidora , Espectroscopía de Resonancia Magnética/métodos , Neoplasias/patología , Relación Estructura-Actividad Cuantitativa , Tiazinas/síntesis química , Tiazinas/química
9.
Molecules ; 15(3): 1113-26, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20335967

RESUMEN

The course of reaction of aryl and heteroaryl sulfonamides with diphenylcarbonate (DPC) and 4-dimethylaminopyridine (DMAP) was found to depend on the pKa of the sulfonamide used. Aryl sulfonamides with pKa approximately 10 gave 4-dimethylamino-pyridinium arylsulfonyl-carbamoylides, while the more acidic heteroaryl sulfonamides (pKa approximately 8) furnished 4-dimethylaminopyridinium heteroarylsulfonyl carbamates. Both the carbamoylides and carbamate salts reacted with aliphatic and aromatic amines with the formation of appropriate aryl(heteroaryl)sulfonyl ureas, and therefore, can be regarded as safe and stable substitutes of the hazardous and difficult to handle aryl(heteroaryl)sulfonyl isocyanates.


Asunto(s)
Compuestos de Sulfonilurea/síntesis química , Línea Celular Tumoral , Cristalografía por Rayos X , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Espectrofotometría Infrarroja , Compuestos de Sulfonilurea/química , Compuestos de Sulfonilurea/farmacología
10.
Nat Commun ; 8: 14206, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28198360

RESUMEN

The secretome of cancer and stromal cells generates a microenvironment that contributes to tumour cell invasion and angiogenesis. Here we compare the secretome of human mammary normal and cancer-associated fibroblasts (CAFs). We discover that the chloride intracellular channel protein 3 (CLIC3) is an abundant component of the CAF secretome. Secreted CLIC3 promotes invasive behaviour of endothelial cells to drive angiogenesis and increases invasiveness of cancer cells both in vivo and in 3D cell culture models, and this requires active transglutaminase-2 (TGM2). CLIC3 acts as a glutathione-dependent oxidoreductase that reduces TGM2 and regulates TGM2 binding to its cofactors. Finally, CLIC3 is also secreted by cancer cells, is abundant in the stromal and tumour compartments of aggressive ovarian cancers and its levels correlate with poor clinical outcome. This work reveals a previously undescribed invasive mechanism whereby the secretion of a glutathione-dependent oxidoreductase drives angiogenesis and cancer progression by promoting TGM2-dependent invasion.


Asunto(s)
Canales de Cloruro/metabolismo , Progresión de la Enfermedad , Glutatión/metabolismo , Animales , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Línea Celular Tumoral , Matriz Extracelular/metabolismo , Femenino , Proteínas de Unión al GTP/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones Endogámicos C57BL , Ratones Desnudos , Modelos Biológicos , Invasividad Neoplásica , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Oxidorreductasas/metabolismo , Unión Proteica , Proteína Glutamina Gamma Glutamiltransferasa 2 , Proteoma/metabolismo , Proteómica , Análisis de Supervivencia , Transglutaminasas/metabolismo , Resultado del Tratamiento
11.
Mol Cell Oncol ; 3(1): e970097, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27308577

RESUMEN

Chloroquine is used clinically as an autophagy blocker to potentiate anticancer treatments. However, whether chloroquine acts solely through autophagy-dependent and cancer cell autonomous mechanisms has remained elusive. In a recent study we found that chloroquine reduced intratumoral hypoxia and metastasis, while improving chemotherapy response, largely through an autophagy-independent, NOTCH1-reliant mechanism of tumor vessel normalization.

12.
Nat Commun ; 7: 12240, 2016 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-27436424

RESUMEN

During vessel sprouting, endothelial cells (ECs) dynamically rearrange positions in the sprout to compete for the tip position. We recently identified a key role for the glycolytic activator PFKFB3 in vessel sprouting by regulating cytoskeleton remodelling, migration and tip cell competitiveness. It is, however, unknown how glycolysis regulates EC rearrangement during vessel sprouting. Here we report that computational simulations, validated by experimentation, predict that glycolytic production of ATP drives EC rearrangement by promoting filopodia formation and reducing intercellular adhesion. Notably, the simulations correctly predicted that blocking PFKFB3 normalizes the disturbed EC rearrangement in high VEGF conditions, as occurs during pathological angiogenesis. This interdisciplinary study integrates EC metabolism in vessel sprouting, yielding mechanistic insight in the control of vessel sprouting by glycolysis, and suggesting anti-glycolytic therapy for vessel normalization in cancer and non-malignant diseases.


Asunto(s)
Glucólisis , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica , Adenosina Trifosfato/metabolismo , Antígenos CD/metabolismo , Cadherinas/antagonistas & inhibidores , Cadherinas/metabolismo , Simulación por Computador , Técnicas de Silenciamiento del Gen , Glucólisis/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Indoles/farmacología , Modelos Biológicos , Neovascularización Fisiológica/efectos de los fármacos , Fosfofructoquinasa-2/antagonistas & inhibidores , Fosfofructoquinasa-2/metabolismo , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Piridinas/farmacología , Pirroles/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
Cancer Cell ; 30(6): 968-985, 2016 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-27866851

RESUMEN

Abnormal tumor vessels promote metastasis and impair chemotherapy. Hence, tumor vessel normalization (TVN) is emerging as an anti-cancer treatment. Here, we show that tumor endothelial cells (ECs) have a hyper-glycolytic metabolism, shunting intermediates to nucleotide synthesis. EC haplo-deficiency or blockade of the glycolytic activator PFKFB3 did not affect tumor growth, but reduced cancer cell invasion, intravasation, and metastasis by normalizing tumor vessels, which improved vessel maturation and perfusion. Mechanistically, PFKFB3 inhibition tightened the vascular barrier by reducing VE-cadherin endocytosis in ECs, and rendering pericytes more quiescent and adhesive (via upregulation of N-cadherin) through glycolysis reduction; it also lowered the expression of cancer cell adhesion molecules in ECs by decreasing NF-κB signaling. PFKFB3-blockade treatment also improved chemotherapy of primary and metastatic tumors.


Asunto(s)
Cisplatino/administración & dosificación , Células Epiteliales/metabolismo , Neoplasias/metabolismo , Fosfofructoquinasa-2/antagonistas & inhibidores , Tamoxifeno/administración & dosificación , Animales , Cadherinas/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Cisplatino/farmacología , Sinergismo Farmacológico , Quimioterapia , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glucólisis/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Tamoxifeno/farmacología
14.
Cell Rep ; 12(6): 992-1005, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26235614

RESUMEN

Several questions about the role of the oxygen sensor prolyl-hydroxylase 2 (PHD2) in cancer have not been addressed. First, the role of PHD2 in metastasis has not been studied in a spontaneous tumor model. Here, we show that global PHD2 haplodeficiency reduced metastasis without affecting tumor growth. Second, it is unknown whether PHD2 regulates cancer by affecting cancer-associated fibroblasts (CAFs). We show that PHD2 haplodeficiency reduced metastasis via two mechanisms: (1) by decreasing CAF activation, matrix production, and contraction by CAFs, an effect that surprisingly relied on PHD2 deletion in cancer cells, but not in CAFs; and (2) by improving tumor vessel normalization. Third, the effect of concomitant PHD2 inhibition in malignant and stromal cells (mimicking PHD2 inhibitor treatment) is unknown. We show that global PHD2 haplodeficiency, induced not only before but also after tumor onset, impaired metastasis. These findings warrant investigation of PHD2's therapeutic potential.


Asunto(s)
Fibroblastos/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Neoplasias/metabolismo , Animales , Línea Celular Tumoral , Femenino , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Immunoblotting , Inmunohistoquímica , Masculino , Ratones , Modelos Biológicos , Metástasis de la Neoplasia , Neoplasias/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Autophagy ; 10(11): 2082-4, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25484095

RESUMEN

Chloroquine (CQ) is exploited in clinical trials as an autophagy blocker to potentiate anticancer therapy, but it is unknown if it solely acts by inhibiting cancer cell-autonomous autophagy. Our recent study shows that besides blocking cancer cell growth, CQ also affects endothelial cells (ECs) and promotes tumor vessel normalization. This vessel normalizing effect of CQ reduces tumor hypoxia, cancer cell intravasation, and metastasis, while improving the delivery and response to chemotherapy. By compromising autophagy in melanoma cells or using mice with a conditional knockout of ATG5 in ECs, we found that the favorable effects of CQ on the tumor vasculature do not rely on autophagy. CQ-induced vessel normalization relies mainly on altered endolysosomal trafficking and sustained NOTCH1 signaling in ECs. Remarkably these CQ-mediated effects are abrogated when tumors are grown in mice harboring EC-specific deletion of NOTCH1. The autophagy-independent vessel normalization by CQ leading to improved delivery and tumor response to chemotherapy further advocates its clinical use in combination with anticancer treatments.


Asunto(s)
Antimaláricos/química , Autofagia , Cloroquina/química , Neoplasias/tratamiento farmacológico , Animales , Endosomas/metabolismo , Eliminación de Gen , Humanos , Hipoxia , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias/patología , Receptor Notch1/metabolismo
16.
Cell Rep ; 6(1): 155-67, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24388748

RESUMEN

Various tumors metastasize via lymph vessels and lymph nodes to distant organs. Even though tumors are hypoxic, the mechanisms of how hypoxia regulates lymphangiogenesis remain poorly characterized. Here, we show that hypoxia reduced vascular endothelial growth factor C (VEGF-C) transcription and cap-dependent translation via the upregulation of hypophosphorylated 4E-binding protein 1 (4E-BP1). However, initiation of VEGF-C translation was induced by hypoxia through an internal ribosome entry site (IRES)-dependent mechanism. IRES-dependent VEGF-C translation was independent of hypoxia-inducible factor 1α (HIF-1α) signaling. Notably, the VEGF-C IRES activity was higher in metastasizing tumor cells in lymph nodes than in primary tumors, most likely because lymph vessels in these lymph nodes were severely hypoxic. Overall, this transcription-independent but translation-dependent upregulation of VEGF-C in hypoxia stimulates lymphangiogenesis in tumors and lymph nodes and may contribute to lymphatic metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinoma/metabolismo , Hipoxia de la Célula , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Biosíntesis de Proteínas , Factor C de Crecimiento Endotelial Vascular/metabolismo , Regiones no Traducidas 3' , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Neoplasias de la Mama/diagnóstico , Carcinoma/diagnóstico , Proteínas de Ciclo Celular , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis Linfática , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Fosfoproteínas/metabolismo , Transcripción Genética , Factor C de Crecimiento Endotelial Vascular/genética
17.
Cell Metab ; 19(1): 37-48, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24332967

RESUMEN

Strategies targeting pathological angiogenesis have focused primarily on blocking vascular endothelial growth factor (VEGF), but resistance and insufficient efficacy limit their success, mandating alternative antiangiogenic strategies. We recently provided genetic evidence that the glycolytic activator phosphofructokinase-2/fructose-2,6-bisphosphatase 3 (PFKFB3) promotes vessel formation but did not explore the antiangiogenic therapeutic potential of PFKFB3 blockade. Here, we show that blockade of PFKFB3 by the small molecule 3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO) reduced vessel sprouting in endothelial cell (EC) spheroids, zebrafish embryos, and the postnatal mouse retina by inhibiting EC proliferation and migration. 3PO also suppressed vascular hyperbranching induced by inhibition of Notch or VEGF receptor 1 (VEGFR1) and amplified the antiangiogenic effect of VEGF blockade. Although 3PO reduced glycolysis only partially and transiently in vivo, this sufficed to decrease pathological neovascularization in ocular and inflammatory models. These insights may offer therapeutic antiangiogenic opportunities.


Asunto(s)
Glucólisis , Neovascularización Patológica/enzimología , Fosfofructoquinasa-2/antagonistas & inhibidores , Inhibidores de la Angiogénesis/farmacología , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Glucólisis/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/genética , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Fosfofructoquinasa-2/metabolismo , Piridinas/farmacología , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/crecimiento & desarrollo , Vasos Retinianos/patología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra
18.
Cancer Cell ; 26(2): 190-206, 2014 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-25117709

RESUMEN

Chloroquine (CQ) has been evaluated as an autophagy blocker for cancer treatment, but it is unknown if it acts solely by inhibiting cancer cell autophagy. We report that CQ reduced tumor growth but improved the tumor milieu. By normalizing tumor vessel structure and function and increasing perfusion, CQ reduced hypoxia, cancer cell invasion, and metastasis, while improving chemotherapy delivery and response. Inhibiting autophagy in cancer cells or endothelial cells (ECs) failed to induce such effects. CQ's vessel normalization activity relied mainly on alterations of endosomal Notch1 trafficking and signaling in ECs and was abrogated by Notch1 deletion in ECs in vivo. Thus, autophagy-independent vessel normalization by CQ restrains tumor invasion and metastasis while improving chemotherapy, supporting the use of CQ for anticancer treatment.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Autofagia , Cloroquina/farmacología , Melanoma Experimental/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Neoplasias Cutáneas/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Proteína 5 Relacionada con la Autofagia , Camptotecina/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cloroquina/uso terapéutico , Sinergismo Farmacológico , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Humanos , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Proteínas Asociadas a Microtúbulos/metabolismo , Invasividad Neoplásica , Neovascularización Patológica/metabolismo , Receptor Notch1/metabolismo , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/patología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
19.
EMBO Mol Med ; 5(10): 1523-36, 2013 10.
Artículo en Inglés | MEDLINE | ID: mdl-24092663

RESUMEN

The role of the fragile X mental retardation protein (FMRP) is well established in brain, where its absence leads to the fragile X syndrome (FXS). FMRP is almost ubiquitously expressed, suggesting that, in addition to its effects in brain, it may have fundamental roles in other organs. There is evidence that FMRP expression can be linked to cancer. FMR1 mRNA, encoding FMRP, is overexpressed in hepatocellular carcinoma cells. A decreased risk of cancer has been reported in patients with FXS while a patient-case with FXS showed an unusual decrease of tumour brain invasiveness. However, a role for FMRP in regulating cancer biology, if any, remains unknown. We show here that FMRP and FMR1 mRNA levels correlate with prognostic indicators of aggressive breast cancer, lung metastases probability and triple negative breast cancer (TNBC). We establish that FMRP overexpression in murine breast primary tumours enhances lung metastasis while its reduction has the opposite effect regulating cell spreading and invasion. FMRP binds mRNAs involved in epithelial mesenchymal transition (EMT) and invasion including E-cadherin and Vimentin mRNAs, hallmarks of EMT and cancer progression.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , ARN Mensajero/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Cadherinas/metabolismo , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Forma de la Célula , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/antagonistas & inhibidores , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Vimentina/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda