Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 3818, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38740760

RESUMEN

The growing disparity between the demand for transplants and the available donor supply, coupled with an aging donor population and increasing prevalence of chronic diseases, highlights the urgent need for the development of platforms enabling reconditioning, repair, and regeneration of deceased donor organs. This necessitates the ability to preserve metabolically active kidneys ex vivo for days. However, current kidney normothermic machine perfusion (NMP) approaches allow metabolic preservation only for hours. Here we show that human kidneys discarded for transplantation can be preserved in a metabolically active state up to 4 days when perfused with a cell-free perfusate supplemented with TCA cycle intermediates at subnormothermia (25 °C). Using spatially resolved isotope tracing we demonstrate preserved metabolic fluxes in the kidney microenvironment up to Day 4 of perfusion. Beyond Day 4, significant changes were observed in renal cell populations through spatial lipidomics, and increases in injury markers such as LDH, NGAL and oxidized lipids. Finally, we demonstrate that perfused kidneys maintain functional parameters up to Day 4. Collectively, these findings provide evidence that this approach enables metabolic and functional preservation of human kidneys over multiple days, establishing a solid foundation for future clinical investigations.


Asunto(s)
Riñón , Preservación de Órganos , Perfusión , Humanos , Riñón/metabolismo , Preservación de Órganos/métodos , Perfusión/métodos , Trasplante de Riñón , Masculino , Soluciones Preservantes de Órganos , Femenino , Persona de Mediana Edad , Sistema Libre de Células , Ciclo del Ácido Cítrico , Adulto , Nutrientes/metabolismo , Lipidómica/métodos , Anciano
2.
J Immunother Cancer ; 8(1)2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32461350

RESUMEN

BACKGROUND: Tumor ablation techniques, like cryoablation, are successfully used in the clinic to treat tumors. The tumor debris remaining in situ after ablation is a major antigen depot, including neoantigens, which are presented by dendritic cells (DCs) in the draining lymph nodes to induce tumor-specific CD8+ T cells. We have previously shown that co-administration of adjuvants is essential to evoke strong in vivo antitumor immunity and the induction of long-term memory. However, which adjuvants most effectively combine with in situ tumor ablation remains unclear. METHODS AND RESULTS: Here, we show that simultaneous administration of cytidyl guanosyl (CpG) with saponin-based adjuvants following cryoablation affects multifunctional T-cell numbers and interleukin (IL)-1 induced polymorphonuclear neutrophil recruitment in the tumor draining lymph nodes, relative to either adjuvant alone. The combination of CpG and saponin-based adjuvants induces potent DC maturation (mainly CpG-mediated), antigen cross-presentation (mainly saponin-based adjuvant mediated), while excretion of IL-1ß by DCs in vitro depends on the presence of both adjuvants. Most strikingly, CpG/saponin-based adjuvant exposed DCs potentiate antigen-specific T-cell proliferation resulting in multipotent T cells with increased capacity to produce interferon (IFN)γ, IL-2 and tumor necrosis factor-α in vitro. Also in vivo the CpG/saponin-based adjuvant combination plus cryoablation increased the numbers of tumor-specific CD8+ T cells showing enhanced IFNγ production as compared with single adjuvant treatments. CONCLUSIONS: Collectively, these data indicate that co-injection of CpG with saponin-based adjuvants after cryoablation induces an increased amount of tumor-specific multifunctional T cells. The combination of saponin-based adjuvants with toll-like receptor 9 adjuvant CpG in a cryoablative setting therefore represents a promising in situ vaccination strategy.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Interleucina-1/fisiología , Ganglios Linfáticos/inmunología , Melanoma Experimental/terapia , Oligodesoxirribonucleótidos/administración & dosificación , Saponinas/administración & dosificación , Linfocitos T/inmunología , Animales , Ablación por Catéter/métodos , Terapia Combinada , Células Dendríticas/inmunología , Femenino , Ganglios Linfáticos/patología , Activación de Linfocitos/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T/patología
3.
Dev Biol ; 455(1): 42-50, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31265831

RESUMEN

Mammary glands are unique organs in which major adaptive changes occur in morphogenesis and development after birth. Breast cancer is the most common cancer and a major cause of mortality in females worldwide. We have previously identified the loss of expression of the transcription regulator DC-SCRIPT (Zfp366) as a prominent prognostic event in estrogen receptor positive breast cancer patients. DC-SCRIPT affects multiple transcriptional events in breast cancer cells, including estrogen and progesterone receptor-mediated transcription, and promotes CDKN2B-related cell cycle arrest. As loss of DC-SCRIPT expression appears an early event in breast cancer development, we here investigated the role of DC-SCRIPT in mammary gland development using wild-type and DC-SCRIPT knockout mice. Mice lacking DC-SCRIPT exhibited severe breeding problems and showed significant growth delay relative to littermate wild-type mice. Subsequent analysis revealed that DC-SCRIPT was expressed in mouse mammary epithelium and that DC-SCRIPT deficiency delayed mammary gland morphogenesis in vivo. Finally, analysis of 3D mammary gland organoid cultures confirmed that loss of DC-SCRIPT dramatically delayed mammary organoid branching in vitro. The study shows for the first time that DC-SCRIPT deficiency delays mammary gland morphogenesis in vivo and in vitro. These data define DC-SCRIPT as a novel modulator of mammary gland development.


Asunto(s)
Proteínas de Unión al ADN/genética , Glándulas Mamarias Animales/metabolismo , Morfogénesis/genética , Proteínas Nucleares/genética , Organoides/metabolismo , Factores de Transcripción/genética , Animales , Técnicas de Cultivo de Célula/métodos , Puntos de Control del Ciclo Celular/genética , Proteínas de Unión al ADN/deficiencia , Células Epiteliales/metabolismo , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Homeostasis/genética , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/deficiencia , Organoides/citología , Organoides/crecimiento & desarrollo , Factores de Transcripción/deficiencia
4.
Am J Transplant ; 19(5): 1328-1343, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30506641

RESUMEN

The bioengineering of a replacement kidney has been proposed as an approach to address the growing shortage of donor kidneys for the treatment of chronic kidney disease. One approach being investigated is the recellularization of kidney scaffolds. In this study, we present several key advances toward successful re-endothelialization of whole kidney matrix scaffolds from both rodents and humans. Based on the presence of preserved glycosoaminoglycans within the decelullarized kidney scaffold, we show improved localization of delivered endothelial cells after preloading of the vascular matrix with vascular endothelial growth factor and angiopoietin 1. Using a novel simultaneous arteriovenous delivery system, we report the complete re-endothelialization of the kidney vasculature, including the glomerular and peritubular capillaries, using human inducible pluripotent stem cell -derived endothelial cells. Using this source of endothelial cells, it was possible to generate sufficient endothelial cells to recellularize an entire human kidney scaffold, achieving efficient cell delivery, adherence, and endothelial cell proliferation and survival. Moreover, human re-endothelialized scaffold could, in contrast to the non-re-endothelialized human scaffold, be fully perfused with whole blood. These major advances move the field closer to a human bioengineered kidney.


Asunto(s)
Bioingeniería , Endotelio Vascular/citología , Matriz Extracelular/fisiología , Células Madre Pluripotentes Inducidas/citología , Trasplante de Riñón/métodos , Riñón/citología , Andamios del Tejido/química , Animales , Células Cultivadas , Endotelio Vascular/metabolismo , Glicosaminoglicanos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Riñón/metabolismo , Ratas , Ratas Endogámicas Lew
5.
Nat Commun ; 7: 13324, 2016 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-27819292

RESUMEN

Saponin-based adjuvants (SBAs) are being used in animal and human (cancer) vaccines, as they induce protective cellular immunity. Their adjuvant potency is a factor of inflammasome activation and enhanced antigen cross-presentation by dendritic cells (DCs), but how antigen cross-presentation is induced is not clear. Here we show that SBAs uniquely induce intracellular lipid bodies (LBs) in the CD11b+ DC subset in vitro and in vivo. Using genetic and pharmacological interference in models for vaccination and in situ tumour ablation, we demonstrate that LB induction is causally related to the saponin-dependent increase in cross-presentation and T-cell activation. These findings link adjuvant activity to LB formation, aid the application of SBAs as a cancer vaccine component, and will stimulate development of new adjuvants enhancing T-cell-mediated immunity.


Asunto(s)
Vacunas contra el Cáncer/farmacología , Reactividad Cruzada/efectos de los fármacos , Células Dendríticas/inmunología , Melanoma Experimental/terapia , Saponinas/farmacología , Neoplasias Cutáneas/terapia , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/farmacología , Animales , Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/inmunología , Antígeno CD11b/metabolismo , Vacunas contra el Cáncer/química , Línea Celular Tumoral , Reactividad Cruzada/inmunología , Células Dendríticas/metabolismo , Femenino , Humanos , Inmunidad Celular/efectos de los fármacos , Inflamasomas/inmunología , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/inmunología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cultivo Primario de Células , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/inmunología , Saponinas/inmunología , Neoplasias Cutáneas/inmunología
6.
Breast Cancer Res Treat ; 149(3): 693-703, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25663546

RESUMEN

Breast cancer is one of the most common causes of cancer-related deaths in women. The estrogen receptor (ERα) is well known for having growth promoting effects in breast cancer. Recently, we have identified DC-SCRIPT (ZNF366) as a co-suppressor of ERα and as a strong and independent prognostic marker in ESR1 (ERα gene)-positive breast cancer patients. In this study, we further investigated the molecular mechanism on how DC-SCRIPT inhibits breast cancer cell growth. DC-SCRIPT mRNA levels from 190 primary ESR1-positive breast tumors were related to global gene expression, followed by gene ontology and pathway analysis. The effect of DC-SCRIPT on breast cancer cell growth and cell cycle arrest was investigated using novel DC-SCRIPT-inducible MCF7 breast cancer cell lines. Genome-wide expression profiling of DC-SCRIPT-expressing MCF7 cells was performed to investigate the effect of DC-SCRIPT on cell cycle-related gene expression. Findings were validated by real-time PCR in a cohort of 1,132 ESR1-positive breast cancer patients. In the primary ESR1-positive breast tumors, DC-SCRIPT expression negatively correlated with several cell cycle gene ontologies and pathways. DC-SCRIPT expression strongly reduced breast cancer cell growth in vitro, breast tumor growth in vivo, and induced cell cycle arrest. In addition, in the presence of DC-SCRIPT, multiple cell cycles related genes were differentially expressed including the tumor suppressor gene CDKN2B. Moreover, in 1,132 primary ESR1-positive breast tumors, DC-SCRIPT expression also correlated with CDKN2B expression. Collectively, these data show that DC-SCRIPT acts as a novel regulator of CDKN2B and induces cell cycle arrest in ESR1-positive breast cancer cells.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas Portadoras/genética , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Receptor alfa de Estrógeno/genética , Biomarcadores de Tumor/genética , Neoplasias de la Mama/patología , Proteínas Portadoras/metabolismo , Puntos de Control del Ciclo Celular/genética , Proliferación Celular/genética , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Humanos , Células MCF-7 , Proteínas de Neoplasias/biosíntesis , ARN Mensajero/biosíntesis
7.
ACS Nano ; 9(1): 733-45, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25575241

RESUMEN

Sialic acid sugars are overexpressed by cancer cells and contribute to the metastatic cascade at multiple levels. Therapeutic interference of sialic acids, however, has been difficult to pursue because of the absence of dedicated tools. Here we show that a rationally designed sialic acid-blocking glycomimetic (P-3F(ax)-Neu5Ac) successfully prevents cancer metastasis. Formulation of P-3F(ax)--Neu5Ac into poly(lactic-co-glycolic acid nanoparticles coated with antityrosinase-related protein-1 antibodies allowed targeted delivery of P-3F(ax)--Neu5Ac into melanoma cells, slow release, and long-term sialic acid blockade. Most importantly, intravenous injections of melanoma-targeting P-3F(ax)--Neu5Ac nanoparticles prevented metastasis formation in a murine lung metastasis model. These findings stress the importance of sialoglycans in cancer metastasis and advocate that sialic acid blockade using rationally designed glycomimetics targeted to cancer cells can effectively prevent cancer metastases. This targeting strategy to interfere with sialic acid-dependent processes is broadly applicable not only for different types of cancer but also in infection and inflammation.


Asunto(s)
Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Portadores de Fármacos/química , Melanoma Experimental/patología , Ácido N-Acetilneuramínico/antagonistas & inhibidores , Ácidos Siálicos/química , Ácidos Siálicos/farmacología , Animales , Anticuerpos/química , Anticuerpos/inmunología , Femenino , Ácido Láctico/química , Neoplasias Pulmonares/secundario , Glicoproteínas de Membrana/inmunología , Ratones , Nanopartículas/química , Metástasis de la Neoplasia , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico
8.
Mol Cancer Ther ; 12(10): 1935-46, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23974695

RESUMEN

Cancer cells decorate their surface with a dense layer of sialylated glycans by upregulating the expression of sialyltransferases and other glycogenes. Although sialic acids play a vital role in many biologic processes, hypersialylation in particular has been shown to contribute to cancer cell progression and metastasis. Accordingly, selective strategies to interfere with sialic acid synthesis might offer a powerful approach in cancer therapy. In the present study, we assessed the potential of a recently developed fluorinated sialic acid analogue (P-3F(ax)-Neu5Ac) to block the synthesis of sialoglycans in murine melanoma cells and the consequences on cell adhesion, migration, and in vivo growth. The results showed that P-3F(ax)-Neu5Ac readily caused depletion of α2,3-/α2,6-linked sialic acids in B16F10 cells for several days. Long-term inhibition of sialylation for 28 days was feasible without affecting cell viability or proliferation. Moreover, P-3F(ax)-Neu5Ac proved to be a highly potent inhibitor of sialylation even at high concentrations of competing sialyltransferase substrates. P-3F(ax)-Neu5Ac-treated cancer cells exhibited impaired binding to poly-l-lysine, type I collagen, and fibronectin and diminished migratory capacity. Finally, blocking sialylation of B16F10 tumor cells with this novel sialic acid analogue reduced their growth in vivo. These results indicate that P-3F(ax)-Neu5Ac is a powerful glycomimetic capable of inhibiting aberrant sialylation that can potentially be used for anticancer therapy.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Melanoma Experimental/tratamiento farmacológico , Ácido N-Acetilneuramínico/farmacología , Ácidos Siálicos/farmacología , Animales , Adhesión Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Flúor/química , Flúor/farmacología , Humanos , Melanoma Experimental/patología , Ratones , Ácido N-Acetilneuramínico/análogos & derivados , Polisacáridos/biosíntesis
9.
Clin Cancer Res ; 18(14): 3961-3971, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22573349

RESUMEN

PURPOSE: To investigate the angiogenic changes in primary tumor tissue of renal cell carcinoma (RCC) patients treated with VEGF-targeted therapy. EXPERIMENTAL DESIGN: Phase II trials of VEGF pathway-targeted therapy given before cytoreductive surgery were carried out with metastatic RCC patients with the primary tumor in situ to investigate the necessity of nephrectomy. Primary tumor tissues were obtained and assessed for angiogenesis parameters. Results were compared with similar analyses on untreated tumors. RESULTS: Sunitinib or bevacizumab pretreatment resulted in a significant reduction of microvessel density in the primary tumor. Also, an increase in vascular pericyte coverage was found in sunitinib-pretreated tumors, consistent with efficient angiogenesis inhibition. Expression of several key regulators of angiogenesis was found to be suppressed in pretreated tissues, among which VEGFR-1 and VEGFR-2, angiopoietin-1 and angiopoietin-2 and platelet-derived growth factor-B. In addition, apoptosis in tumor and endothelial cells was induced. Interestingly, in sunitinib-pretreated tissues a dramatic increase of the number of proliferating endothelial cells was observed, which was not the case in bevacizumab-pretreated tumors. A positive correlation with the interval between halting the therapy and surgery was found, suggesting a compensatory angiogenic response caused by the discontinuation of sunitinib treatment. CONCLUSION: This study describes, for the first time, the angiostatic response in human primary renal cancers at the tissue level upon treatment with VEGF-targeted therapy. Discontinuation of treatment with tyrosine kinase inhibitors leads to accelerated endothelial cell proliferation. The results of this study contribute important data to the ongoing discussion on the discontinuation of treatment with kinase inhibitors.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Carcinoma de Células Renales/tratamiento farmacológico , Indoles/administración & dosificación , Neovascularización Patológica/tratamiento farmacológico , Pirroles/administración & dosificación , Adulto , Anciano , Apoptosis/efectos de los fármacos , Bevacizumab , Carcinoma de Células Renales/complicaciones , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/secundario , Ensayos Clínicos Fase II como Asunto , Femenino , Humanos , Masculino , Microvasos/efectos de los fármacos , Persona de Mediana Edad , Neovascularización Patológica/complicaciones , Estudios Retrospectivos , Sunitinib
10.
Clin Cancer Res ; 16(14): 3618-27, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20501615

RESUMEN

PURPOSE: In brain tumors, cerebral edema is a significant source of morbidity and mortality. Recent studies have shown that inhibition of vascular endothelial growth factor (VEGF) signaling induces transient vascular normalization and reduces cerebral edema, resulting in a modest survival benefit in glioblastoma patients. During anti-VEGF treatment, circulating levels of angiopoietin (Ang)-2 remained high after an initial minor reduction. It is not known, however, whether Ang-2 can modulate anti-VEGF treatment of glioblastoma. Here, we used an orthotopic glioma model to test the hypothesis that Ang-2 is an additional target for improving the efficacy of current anti-VEGF therapies in glioma patients. EXPERIMENTAL DESIGN: To recapitulate high levels of Ang-2 in glioblastoma patients during anti-VEGF treatment, Ang-2 was ectopically expressed in U87 glioma cells. Animal survival and tumor growth were assessed to determine the effects of Ang-2 and anti-VEGF receptor 2 (VEGFR2) treatment. We also monitored morphologic and functional vascular changes using multiphoton laser scanning microscopy and immunohistochemistry. RESULTS: Ectopic expression of Ang-2 had no effect on vascular permeability, tumor growth, or survival, although it resulted in higher vascular density, with dilated vessels and reduced mural cell coverage. On the other hand, when combined with anti-VEGFR2 treatment, Ang-2 destabilized vessels without affecting vessel regression and compromised the survival benefit of VEGFR2 inhibition by increasing vascular permeability. VEGFR2 inhibition normalized tumor vasculature whereas ectopic expression of Ang-2 diminished the beneficial effects of VEGFR2 blockade by inhibiting vessel normalization. CONCLUSION: Cancer treatment regimens combining anti-VEGF and anti-Ang-2 agents may be an effective strategy to improve the efficacy of current anti-VEGF therapies.


Asunto(s)
Angiopoyetina 2/metabolismo , Glioma/terapia , Neovascularización Patológica/terapia , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Angiopoyetina 2/genética , Animales , Supervivencia Celular , Modelos Animales de Enfermedad , Glioma/irrigación sanguínea , Humanos , Inmunohistoquímica , Ratones , Trasplante de Neoplasias , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...