Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Physiol Plant ; 176(5): e14511, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39279509

RESUMEN

Aspen (Populus tremula L.) is a keystone species and a model system for forest tree genomics. We present an updated resource comprising a chromosome-scale assembly, population genetics and genomics data. Using the resource, we explore the genetic basis of natural variation in leaf size and shape, traits with complex genetic architecture. We generated the genome assembly using long-read sequencing, optical and high-density genetic maps. We conducted whole-genome resequencing of the Umeå Aspen (UmAsp) collection. Using the assembly and re-sequencing data from the UmAsp, Swedish Aspen (SwAsp) and Scottish Aspen (ScotAsp) collections we performed genome-wide association analyses (GWAS) using Single Nucleotide Polymorphisms (SNPs) for 26 leaf physiognomy phenotypes. We conducted Assay of Transposase Accessible Chromatin sequencing (ATAC-Seq), identified genomic regions of accessible chromatin, and subset SNPs to these regions, improving the GWAS detection rate. We identified candidate long non-coding RNAs in leaf samples, quantified their expression in an updated co-expression network, and used this to explore the functions of candidate genes identified from the GWAS. A GWAS found SNP associations for seven traits. The associated SNPs were in or near genes annotated with developmental functions, which represent candidates for further study. Of particular interest was a ~177-kbp region harbouring associations with several leaf phenotypes in ScotAsp. We have incorporated the assembly, population genetics, genomics, and GWAS data into the PlantGenIE.org web resource, including updating existing genomics data to the new genome version, to enable easy exploration and visualisation. We provide all raw and processed data to facilitate reuse in future studies.


Asunto(s)
Genética de Población , Genoma de Planta , Estudio de Asociación del Genoma Completo , Polimorfismo de Nucleótido Simple , Populus , Populus/genética , Genoma de Planta/genética , Polimorfismo de Nucleótido Simple/genética , Cromosomas de las Plantas/genética , Fenotipo , Hojas de la Planta/genética , Genómica/métodos , Mapeo Cromosómico
2.
Mol Cancer ; 22(1): 191, 2023 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-38031106

RESUMEN

Despite major improvements in immunotherapeutic strategies, the immunosuppressive tumor microenvironment remains a major obstacle for the induction of efficient antitumor responses. In this study, we show that local delivery of a bispecific Clec9A-PD-L1 targeted type I interferon (AcTaferon, AFN) overcomes this hurdle by reshaping the tumor immune landscape.Treatment with the bispecific AFN resulted in the presence of pro-immunogenic tumor-associated macrophages and neutrophils, increased motility and maturation profile of cDC1 and presence of inflammatory cDC2. Moreover, we report empowered diversity in the CD8+ T cell repertoire and induction of a shift from naive, dysfunctional CD8+ T cells towards effector, plastic cytotoxic T lymphocytes together with increased presence of NK and NKT cells as well as decreased regulatory T cell levels. These dynamic changes were associated with potent antitumor activity. Tumor clearance and immunological memory, therapeutic immunity on large established tumors and blunted tumor growth at distant sites were obtained upon co-administration of a non-curative dose of chemotherapy.Overall, this study illuminates further application of type I interferon as a safe and efficient way to reshape the suppressive tumor microenvironment and induce potent antitumor immunity; features which are of major importance in overcoming the development of metastases and tumor cell resistance to immune attack. The strategy described here has potential for application across to a broad range of cancer types.


Asunto(s)
Interferón Tipo I , Neoplasias , Humanos , Linfocitos T CD8-positivos , Interferón Tipo I/metabolismo , Microambiente Tumoral , Antígeno B7-H1/metabolismo , Neoplasias/metabolismo , Inmunoterapia , Línea Celular Tumoral
3.
EMBO J ; 38(6)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30745319

RESUMEN

DSCAM and DSCAML1 are immunoglobulin and cell adhesion-type receptors serving important neurodevelopmental functions including control of axon growth, branching, neurite self-avoidance, and neuronal cell death. The signal transduction mechanisms or effectors of DSCAM receptors, however, remain poorly characterized. We used a human ORFeome library to perform a high-throughput screen in mammalian cells and identified novel cytoplasmic signaling effector candidates including the Down syndrome kinase Dyrk1a, STAT3, USP21, and SH2D2A. Unexpectedly, we also found that the intracellular domains (ICDs) of DSCAM and DSCAML1 specifically and directly interact with IPO5, a nuclear import protein of the importin beta family, via a conserved nuclear localization signal. The DSCAM ICD is released by γ-secretase-dependent cleavage, and both the DSCAM and DSCAML1 ICDs efficiently translocate to the nucleus. Furthermore, RNA sequencing confirms that expression of the DSCAM as well as the DSCAML1 ICDs alone can profoundly alter the expression of genes associated with neuronal differentiation and apoptosis, as well as synapse formation and function. Gain-of-function experiments using primary cortical neurons show that increasing the levels of either the DSCAM or the DSCAML1 ICD leads to an impairment of neurite growth. Strikingly, increased expression of either full-length DSCAM or the DSCAM ICD, but not the DSCAML1 ICD, significantly decreases synapse numbers in primary hippocampal neurons. Taken together, we identified a novel membrane-to-nucleus signaling mechanism by which DSCAM receptors can alter the expression of regulators of neuronal differentiation and synapse formation and function. Considering that chromosomal duplications lead to increased DSCAM expression in trisomy 21, our findings may help uncover novel mechanisms contributing to intellectual disability in Down syndrome.


Asunto(s)
Transporte Activo de Núcleo Celular , Moléculas de Adhesión Celular/metabolismo , Núcleo Celular/metabolismo , Neuritas/fisiología , Sinapsis/fisiología , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Adhesión Celular , Moléculas de Adhesión Celular/genética , Núcleo Celular/genética , Células HEK293 , Hipocampo/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Neurogénesis , Neuronas/metabolismo , Dominios Proteicos , Dominios y Motivos de Interacción de Proteínas , beta Carioferinas/genética , beta Carioferinas/metabolismo
4.
Bioinformatics ; 33(17): 2740-2742, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28472230

RESUMEN

MOTIVATION: The Bionano Genomics platform allows for the optical detection of short sequence patterns in very long DNA molecules (up to 2.5 Mbp). Molecules with overlapping patterns can be assembled to generate a consensus optical map of the entire genome. In turn, these optical maps can be used to validate or improve de novo genome assembly projects or to detect large-scale structural variation in genomes. Simulated optical map data can assist in the development and benchmarking of tools that operate on those data, such as alignment and assembly software. Additionally, it can help to optimize the experimental setup for a genome of interest. Such a simulator is currently not available. RESULTS: We have developed a simulator, OMSim, that produces synthetic optical map data that mimics real Bionano Genomics data. These simulated data have been tested for compatibility with the Bionano Genomics Irys software system and the Irys-scaffolding scripts. OMSim is capable of handling very large genomes (over 30 Gbp) with high throughput and low memory requirements. AVAILABILITY AND IMPLEMENTATION: The Python simulation tool and a cross-platform graphical user interface are available as open source software under the GNU GPL v2 license ( http://www.bioinformatics.intec.ugent.be/omsim ). CONTACT: jan.fostier@ugent.be. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Genoma Humano , Análisis de Secuencia de ADN/métodos , Programas Informáticos , Genómica/métodos , Humanos
5.
J Pathol ; 242(1): 24-38, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28035683

RESUMEN

Although p53 protein aggregates have been observed in cancer cell lines and tumour tissue, their impact in cancer remains largely unknown. Here, we extensively screened for p53 aggregation phenotypes in tumour biopsies, and identified nuclear inclusion bodies (nIBs) of transcriptionally inactive mutant or wild-type p53 as the most frequent aggregation-like phenotype across six different cancer types. p53-positive nIBs co-stained with nuclear aggregation markers, and shared molecular hallmarks of nIBs commonly found in neurodegenerative disorders. In cell culture, tumour-associated stress was a strong inducer of p53 aggregation and nIB formation. This was most prominent for mutant p53, but could also be observed in wild-type p53 cell lines, for which nIB formation correlated with the loss of p53's transcriptional activity. Importantly, protein aggregation also fuelled the dysregulation of the proteostasis network in the tumour cell by inducing a hyperactivated, oncogenic heat-shock response, to which tumours are commonly addicted, and by overloading the proteasomal degradation system, an observation that was most pronounced for structurally destabilized mutant p53. Patients showing tumours with p53-positive nIBs suffered from a poor clinical outcome, similar to those with loss of p53 expression, and tumour biopsies showed a differential proteostatic expression profile associated with p53-positive nIBs. p53-positive nIBs therefore highlight a malignant state of the tumour that results from the interplay between (1) the functional inactivation of p53 through mutation and/or aggregation, and (2) microenvironmental stress, a combination that catalyses proteostatic dysregulation. This study highlights several unexpected clinical, biological and therapeutically unexplored parallels between cancer and neurodegeneration. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Neoplasias del Colon/genética , Glioblastoma/genética , Cuerpos de Inclusión Intranucleares/metabolismo , Agregación Patológica de Proteínas/genética , Deficiencias en la Proteostasis/genética , Proteína p53 Supresora de Tumor/genética , Biopsia , Línea Celular Tumoral , Neoplasias del Colon/complicaciones , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Citoplasma/metabolismo , Glioblastoma/complicaciones , Glioblastoma/metabolismo , Glioblastoma/patología , Respuesta al Choque Térmico/genética , Respuesta al Choque Térmico/fisiología , Humanos , Estimación de Kaplan-Meier , Mutación , Agregación Patológica de Proteínas/etiología , Agregación Patológica de Proteínas/metabolismo , Deficiencias en la Proteostasis/etiología , Deficiencias en la Proteostasis/metabolismo , Receptores sigma/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
6.
Nucleic Acids Res ; 40(10): 4320-33, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22275523

RESUMEN

Most bacteria control oxidative stress through the H(2)O(2)-responsive transactivator OxyR, a member of the LTTR family (LysR Type Transcriptional Regulators), which activates the expression of defensive genes such as those encoding catalases, alkyl hydroperoxide reductases and superoxide dismutases. In the human opportunistic pathogen Pseudomonas aeruginosa, OxyR positively regulates expression of the oxidative stress response genes katA, katB, ahpB and ahpCF. To identify additional targets of OxyR in P. aeruginosa PAO1, we performed chromatin immunoprecipitation in combination with whole genome tiling array analyses (ChIP-chip). We detected 56 genes including all the previously identified defensive genes and a battery of novel direct targets of OxyR. Electrophoretic mobility shift assays (EMSAs) for selected newly identified targets indicated that ∼70% of those were bound by purified oxidized OxyR and their regulation was confirmed by quantitative real-time polymerase chain reaction. Furthermore, a thioredoxin system was identified to enzymatically reduce OxyR under oxidative stress. Functional classification analysis showed that OxyR controls a core regulon of oxidative stress defensive genes, and other genes involved in regulation of iron homeostasis (pvdS), quorum-sensing (rsaL), protein synthesis (rpsL) and oxidative phosphorylation (cyoA and snr1). Collectively, our results indicate that OxyR is involved in oxidative stress defense and regulates other aspects of cellular metabolism as well.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Estrés Oxidativo/genética , Pseudomonas aeruginosa/genética , Transactivadores/metabolismo , Sitios de Unión , Inmunoprecipitación de Cromatina , Genoma Bacteriano , Análisis de Secuencia por Matrices de Oligonucleótidos , Pseudomonas aeruginosa/enzimología , Pseudomonas aeruginosa/metabolismo , Regulón , Tiorredoxinas/metabolismo
7.
N Engl J Med ; 361(4): 345-57, 2009 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-19625716

RESUMEN

BACKGROUND: The hemolytic-uremic syndrome consists of the triad of microangiopathic hemolytic anemia, thrombocytopenia, and renal failure. The common form of the syndrome is triggered by infection with Shiga toxin-producing bacteria and has a favorable outcome. The less common form of the syndrome, called atypical hemolytic-uremic syndrome, accounts for about 10% of cases, and patients with this form of the syndrome have a poor prognosis. Approximately half of the patients with atypical hemolytic-uremic syndrome have mutations in genes that regulate the complement system. Genetic factors in the remaining cases are unknown. We studied the role of thrombomodulin, an endothelial glycoprotein with anticoagulant, antiinflammatory, and cytoprotective properties, in atypical hemolytic-uremic syndrome. METHODS: We sequenced the entire thrombomodulin gene (THBD) in 152 patients with atypical hemolytic-uremic syndrome and in 380 controls. Using purified proteins and cell-expression systems, we investigated whether thrombomodulin regulates the complement system, and we characterized the mechanisms. We evaluated the effects of thrombomodulin missense mutations associated with atypical hemolytic-uremic syndrome on complement activation by expressing thrombomodulin variants in cultured cells. RESULTS: Of 152 patients with atypical hemolytic-uremic syndrome, 7 unrelated patients had six different heterozygous missense THBD mutations. In vitro, thrombomodulin binds to C3b and factor H (CFH) and negatively regulates complement by accelerating factor I-mediated inactivation of C3b in the presence of cofactors, CFH or C4b binding protein. By promoting activation of the plasma procarboxypeptidase B, thrombomodulin also accelerates the inactivation of anaphylatoxins C3a and C5a. Cultured cells expressing thrombomodulin variants associated with atypical hemolytic-uremic syndrome had diminished capacity to inactivate C3b and to activate procarboxypeptidase B and were thus less protected from activated complement. CONCLUSIONS: Mutations that impair the function of thrombomodulin occur in about 5% of patients with atypical hemolytic-uremic syndrome.


Asunto(s)
Activación de Complemento/genética , Síndrome Hemolítico-Urémico/genética , Mutación Missense , Trombomodulina/genética , Adolescente , Adulto , Niño , Complemento C3b , Factor I de Complemento , Vía Alternativa del Complemento/fisiología , Análisis Mutacional de ADN , Síndrome Hemolítico-Urémico/inmunología , Heterocigoto , Humanos , Persona de Mediana Edad , Linaje , Polimorfismo de Nucleótido Simple , Trombomodulina/metabolismo , Adulto Joven
8.
Blood ; 115(11): 2264-73, 2010 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-19965679

RESUMEN

The transforming and tumor growth-promoting properties of Axl, a member of the Tyro3, Axl, and Mer (TAM) family of receptor tyrosine kinases (TAMRs), are well recognized. In contrast, little is known about the role of the TAMR ligand growth arrest-specific gene 6 (Gas6) in tumor biology. By using Gas6-deficient (Gas6(-/-)) mice, we show that bone marrow-derived Gas6 promotes growth and metastasis in different experimental cancer models, including one resistant to vascular endothelial growth factor inhibitors. Mechanistic studies reveal that circulating leukocytes produce minimal Gas6. However, once infiltrated in the tumor, leukocytes up-regulate Gas6, which is mitogenic for tumor cells. Consistent herewith, impaired tumor growth in Gas6(-/-) mice is rescued by transplantation of wild-type bone marrow and, conversely, mimicked by transplantation of Gas6(-/-) bone marrow into wild-type hosts. These findings highlight a novel role for Gas6 in a positive amplification loop, whereby tumors promote their growth by educating infiltrating leukocytes to up-regulate the production of the mitogen Gas6. Hence, inhibition of Gas6 might offer novel opportunities for the treatment of cancer.


Asunto(s)
Movimiento Celular , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Leucocitos/metabolismo , Leucocitos/patología , Mitógenos/biosíntesis , Neoplasias/patología , Animales , Trasplante de Médula Ósea , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Inflamación/complicaciones , Inflamación/patología , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Interleucina-10/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Metástasis de la Neoplasia , Neoplasias/irrigación sanguínea , Neoplasias/enzimología , Neoplasias/genética , Neovascularización Patológica/complicaciones , Neovascularización Patológica/genética , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Células del Estroma/metabolismo , Células del Estroma/patología , Regulación hacia Arriba/genética
9.
Nat Med ; 11(9): 998-1004, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16116431

RESUMEN

Lymph vessels control fluid homeostasis, immunity and metastasis. Unraveling the molecular basis of lymphangiogenesis has been hampered by the lack of a small animal model that can be genetically manipulated. Here, we show that Xenopus tadpoles develop lymph vessels from lymphangioblasts or, through transdifferentiation, from venous endothelial cells. Lymphangiography showed that these lymph vessels drain lymph, through the lymph heart, to the venous circulation. Morpholino-mediated knockdown of the lymphangiogenic factor Prox1 caused lymph vessel defects and lymphedema by impairing lymphatic commitment. Knockdown of vascular endothelial growth factor C (VEGF-C) also induced lymph vessel defects and lymphedema, but primarily by affecting migration of lymphatic endothelial cells. Knockdown of VEGF-C also resulted in aberrant blood vessel formation in tadpoles. This tadpole model offers opportunities for the discovery of new regulators of lymphangiogenesis.


Asunto(s)
Linfangiogénesis/fisiología , Xenopus laevis/crecimiento & desarrollo , Xenopus laevis/genética , Animales , Proteínas de Homeodominio/fisiología , Larva/genética , Larva/crecimiento & desarrollo , Linfangiogénesis/genética , Sistema Linfático/anatomía & histología , Sistema Linfático/crecimiento & desarrollo , Proteínas Supresoras de Tumor
10.
Cells ; 11(11)2022 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-35681479

RESUMEN

One important facet of glaucoma pathophysiology is axonal damage, which ultimately disrupts the connection between the retina and its postsynaptic brain targets. The concurrent loss of retrograde support interferes with the functionality and survival of the retinal ganglion cells (RGCs). Previous research has shown that stimulation of neuronal activity in a primary retinal target area-i.e., the superior colliculus-promotes RGC survival in an acute mouse model of glaucoma. To build further on this observation, we applied repeated chemogenetics in the superior colliculus of a more chronic murine glaucoma model-i.e., the microbead occlusion model-and performed bulk RNA sequencing on collicular lysates and isolated RGCs. Our study revealed that chronic target stimulation upon glaucomatous injury phenocopies the a priori expected molecular response: growth factors were pinpointed as essential transcriptional regulators both in the locally stimulated tissue and in distant, unstimulated RGCs. Strikingly, and although the RGC transcriptome revealed a partial reversal of the glaucomatous signature and an enrichment of pro-survival signaling pathways, functional rescue of injured RGCs was not achieved. By postulating various explanations for the lack of RGC neuroprotection, we aim to warrant researchers and drug developers for the complexity of chronic neuromodulation and growth factor signaling.


Asunto(s)
Glaucoma , Colículos Superiores , Animales , Modelos Animales de Enfermedad , Glaucoma/metabolismo , Ratones , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo
11.
J Clin Invest ; 118(2): 583-96, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18188450

RESUMEN

Many patients with anemia fail to respond to treatment with erythropoietin (Epo), a commonly used hormone that stimulates erythroid progenitor production and maturation by human BM or by murine spleen. The protein product of growth arrest-specific gene 6 (Gas6) is important for cell survival across several cell types, but its precise physiological role remains largely enigmatic. Here, we report that murine erythroblasts released Gas6 in response to Epo and that Gas6 enhanced Epo receptor signaling by activating the serine-threonine kinase Akt in these cells. In the absence of Gas6, erythroid progenitors and erythroblasts were hyporesponsive to the survival activity of Epo and failed to restore hematocrit levels in response to anemia. In addition, Gas6 may influence erythropoiesis via paracrine erythroblast-independent mechanisms involving macrophages. When mice with acute anemia were treated with Gas6, the protein normalized hematocrit levels without causing undesired erythrocytosis. In a transgenic mouse model of chronic anemia caused by insufficient Epo production, Gas6 synergized with Epo in restoring hematocrit levels. These findings may have implications for the treatment of patients with anemia who fail to adequately respond to Epo.


Asunto(s)
Anemia/tratamiento farmacológico , Anemia/genética , Eritropoyesis/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Animales , Adhesión Celular/genética , Supervivencia Celular , Modelos Animales de Enfermedad , Resistencia a Medicamentos , Eritroblastos/efectos de los fármacos , Eritroblastos/metabolismo , Eritropoyetina/genética , Eritropoyetina/farmacología , Eritropoyetina/uso terapéutico , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Mutantes , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Eritropoyetina/agonistas , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Tirosina Quinasa c-Mer , Tirosina Quinasa del Receptor Axl
12.
Nat Med ; 8(7): 702-10, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12053176

RESUMEN

Respiratory distress syndrome (RDS) due to insufficient production of surfactant is a common and severe complication of preterm delivery. Here, we report that loss of the hypoxia-inducible transcription factor-2alpha (HIF-2alpha) caused fatal RDS in neonatal mice due to insufficient surfactant production by alveolar type 2 cells. VEGF, a target of HIF-2alpha, regulates fetal lung maturation: because VEGF levels in alveolar cells were reduced in HIF-2alpha-deficient fetuses; mice with a deficiency of the VEGF(164) and VEGF(188) isoforms or of the HIF-binding site in the VEGF promotor died of RDS; intrauterine delivery of anti-VEGF-receptor-2 antibodies caused RDS and VEGF stimulated production of surfactant proteins by cultured type 2 pneumocytes. Intrauterine delivery or postnatal intratracheal instillation of VEGF stimulated conversion of glycogen to surfactant and protected preterm mice against RDS. The pneumotrophic effect of VEGF may have therapeutic potential for lung maturation in preterm infants.


Asunto(s)
Factores de Crecimiento Endotelial/fisiología , Enfermedades Pulmonares/prevención & control , Linfocinas/fisiología , Transactivadores/fisiología , Envejecimiento , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Modelos Animales de Enfermedad , Factores de Crecimiento Endotelial/farmacología , Pulmón/efectos de los fármacos , Pulmón/crecimiento & desarrollo , Pulmón/patología , Pulmón/fisiopatología , Enfermedades Pulmonares/patología , Enfermedades Pulmonares/fisiopatología , Linfocinas/farmacología , Ratones , Ratones Noqueados , Ratones Transgénicos , Alveolos Pulmonares/crecimiento & desarrollo , Alveolos Pulmonares/patología , Síndrome de Dificultad Respiratoria/patología , Mucosa Respiratoria/patología , Mucosa Respiratoria/fisiopatología , Transactivadores/deficiencia , Transactivadores/genética , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
13.
Nat Med ; 9(2): 173-82, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12539040

RESUMEN

Hemizygous deletion of chromosome 22q11 (del22q11) causes thymic, parathyroid, craniofacial and life-threatening cardiovascular birth defects in 1 in 4,000 infants. The del22q11 syndrome is likely caused by haploinsufficiency of TBX1, but its variable expressivity indicates the involvement of additional modifiers. Here, we report that absence of the Vegf164 isoform caused birth defects in mice, reminiscent of those found in del22q11 patients. The close correlation of birth and vascular defects indicated that vascular dysgenesis may pathogenetically contribute to the birth defects. Vegf interacted with Tbx1, as Tbx1 expression was reduced in Vegf164-deficient embryos and knocked-down vegf levels enhanced the pharyngeal arch artery defects induced by tbx1 knockdown in zebrafish. Moreover, initial evidence suggested that a VEGF promoter haplotype was associated with an increased risk for cardiovascular birth defects in del22q11 individuals. These genetic data in mouse, fish and human indicate that VEGF is a modifier of cardiovascular birth defects in the del22q11 syndrome.


Asunto(s)
Deleción Cromosómica , Síndrome de DiGeorge/genética , Factores de Crecimiento Endotelial/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Linfocinas/genética , Animales , Vasos Sanguíneos/anomalías , Anomalías Congénitas/genética , Cara/anomalías , Ratones , Ratones Noqueados , Neuropilina-1/genética , Isoformas de Proteínas/genética , Cráneo/anomalías , Proteínas de Dominio T Box/genética , Timo/anomalías , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Pez Cebra
14.
Nat Med ; 9(7): 936-43, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12796773

RESUMEN

Therapeutic angiogenesis is likely to require the administration of factors that complement each other. Activation of the receptor tyrosine kinase (RTK) Flk1 by vascular endothelial growth factor (VEGF) is crucial, but molecular interactions of other factors with VEGF and Flk1 have been studied to a limited extent. Here we report that placental growth factor (PGF, also known as PlGF) regulates inter- and intramolecular cross talk between the VEGF RTKs Flt1 and Flk1. Activation of Flt1 by PGF resulted in intermolecular transphosphorylation of Flk1, thereby amplifying VEGF-driven angiogenesis through Flk1. Even though VEGF and PGF both bind Flt1, PGF uniquely stimulated the phosphorylation of specific Flt1 tyrosine residues and the expression of distinct downstream target genes. Furthermore, the VEGF/PGF heterodimer activated intramolecular VEGF receptor cross talk through formation of Flk1/Flt1 heterodimers. The inter- and intramolecular VEGF receptor cross talk is likely to have therapeutic implications, as treatment with VEGF/PGF heterodimer or a combination of VEGF plus PGF increased ischemic myocardial angiogenesis in a mouse model that was refractory to VEGF alone.


Asunto(s)
Proteínas Gestacionales/metabolismo , Receptor Cross-Talk/fisiología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Cultivadas , Dimerización , Factores de Crecimiento Endotelial/metabolismo , Factores de Crecimiento Endotelial/farmacología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Activación Enzimática , Perfilación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Linfocinas/metabolismo , Linfocinas/farmacología , Ratones , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Neovascularización Patológica/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Fosforilación , Factor de Crecimiento Placentario , Proteínas Gestacionales/farmacología , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Proteínas Virales/metabolismo , Proteínas Virales/farmacología
15.
J Immunother Cancer ; 9(11)2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34772757

RESUMEN

BACKGROUND: Clinical success of therapeutic cancer vaccines depends on the ability to mount strong and durable antitumor T cell responses. To achieve this, potent cellular adjuvants are highly needed. Interleukin-1ß (IL-1ß) acts on CD8+ T cells and promotes their expansion and effector differentiation, but toxicity and undesired tumor-promoting side effects hamper efficient clinical application of this cytokine. METHODS: This 'cytokine problem' can be solved by use of AcTakines (Activity-on-Target cytokines), which represent fusions between low-activity cytokine mutants and cell type-specific single-domain antibodies. AcTakines deliver cytokine activity to a priori selected cell types and as such evade toxicity and unwanted off-target side effects. Here, we employ subcutaneous melanoma and lung carcinoma models to evaluate the antitumor effects of AcTakines. RESULTS: In this work, we use an IL-1ß-based AcTakine to drive proliferation and effector functionality of antitumor CD8+ T cells without inducing measurable toxicity. AcTakine treatment enhances diversity of the T cell receptor repertoire and empowers adoptive T cell transfer. Combination treatment with a neovasculature-targeted tumor necrosis factor (TNF) AcTakine mediates full tumor eradication and establishes immunological memory that protects against secondary tumor challenge. Interferon-γ was found to empower this AcTakine synergy by sensitizing the tumor microenvironment to TNF. CONCLUSIONS: Our data illustrate that anticancer cellular immunity can be safely promoted with an IL-1ß-based AcTakine, which synergizes with other immunotherapies for efficient tumor destruction.


Asunto(s)
Inmunoterapia/métodos , Interleucina-1/metabolismo , Neoplasias/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Linfocitos T CD8-positivos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Ratones
16.
Blood ; 112(5): 1740-9, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18474726

RESUMEN

The importance of the lymphangiogenic factor VEGF-D and its receptor VEGFR-3 in early lymphatic development remains largely unresolved. We therefore investigated their role in Xenopus laevis tadpoles, a small animal model allowing chemicogenetic dissection of developmental lymphangiogenesis. Single morpholino antisense oligo knockdown of xVEGF-D did not affect lymphatic commitment, but transiently impaired lymphatic endothelial cell (LEC) migration. Notably, combined knockdown of xVEGF-D with xVEGF-C at suboptimal morpholino concentrations resulted in more severe migration defects and lymphedema formation than the corresponding single knockdowns. Knockdown of VEGFR-3 or treatment with the VEGFR-3 inhibitor MAZ51 similarly impaired lymph vessel formation and function and caused pronounced edema. VEGFR-3 silencing by morpholino knockdown, MAZ51 treatment, or xVEGF-C/D double knockdown also resulted in dilation and dysfunction of the lymph heart. These findings document a critical role of VEGFR-3 in embryonic lymphatic development and function, and reveal a previously unrecognized modifier role of VEGF-D in the regulation of embryonic lymphangiogenesis in frog embryos.


Asunto(s)
Linfangiogénesis/fisiología , Factor D de Crecimiento Endotelial Vascular/fisiología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/fisiología , Proteínas de Xenopus/fisiología , Xenopus laevis/crecimiento & desarrollo , Xenopus laevis/fisiología , Animales , Silenciador del Gen , Larva/crecimiento & desarrollo , Linfangiogénesis/genética , Vasos Linfáticos/anomalías , Vasos Linfáticos/embriología , Oligodesoxirribonucleótidos Antisentido/genética , Factor D de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor D de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Proteínas de Xenopus/genética , Xenopus laevis/embriología , Xenopus laevis/genética
17.
Transl Oncol ; 13(3): 100743, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32145636

RESUMEN

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is a very lethal disease that can develop therapy resistance over time. The dense stroma in PDAC plays a critical role in tumor progression and resistance. How this stroma interacts with the tumor cells and how this is influenced by chemotherapy remain poorly understood. METHODS: The backbone of this study is the parallel transcriptome analysis of human tumor and mouse stroma in two molecular and clinical representative patient-derived tumor xenografts models. Mice (8 animals per group) were treated for 4 weeks with gemcitabine or control. We studied tumor growth, RNA expression in the stroma, tumor-associated macrophages (TAMs) with immunofluorescence, and cytokines in the serum. RESULTS: A method for parallel transcriptome analysis was optimized. We found that the tumor (differentiation, gene expression) determines the infiltration of macrophages into the stroma. In aggressive PDAC (epithelial-to-mesenchymal transition high), we find more M2 polarized TAMs and the activation of cytokines and growth factors (TNFα, TGFß1, and IL6). There are increased stromal glycolysis, reduced fatty acid oxidation, and reduced mitochondrial oxidation (tricarboxylic acid cycle and oxidative phosphorylation). Treatment with gemcitabine results in a shift of innate immune cells, especially additional infiltration of protumoral M2 TAMs (P < .001) and metabolic reprogramming. CONCLUSIONS: Gemcitabine treatment of PDAC xenografts stimulates a protumoral macrophage phenotype, and this, in combination with a shift of the tumor cells to a mesenchymal phenotype that we reported previously, contributes to tumor progression and therapeutic resistance. Targeting M2-polarized TAMs may benefit PDAC patients at risk to become refractory to current anticancer regimens.

18.
J Cell Mol Med ; 13(11-12): 4587-95, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19210287

RESUMEN

Proteinases have been implicated in the mobilization of haematopoietic progenitor cells (HPCs) from the bone marrow (BM). Here, we report the involvement of the plasminogen (Plg) system in the haematopoietic recovery following chemotherapy. By using gene-deficient mice, we found that plasmin and its activators tPA and uPA play a role in the haematopoietic recovery upon delivery of the cytotoxic agent 5-fluoro-uracil (5-FU). The impaired haematopoietic recovery of Plg-deficient (Plg(-/-)) mice after 5-FU was not rescued by depletion of fibrinogen, indicating that it was not due to defective fibrinolysis. Instead, loss of Plg impaired breakdown of fibronectin, VCAM-1 and laminin-BM matrix proteins involved in adhesion of HPCs to their BM microenvironment and in transendothelial migration of HPCs. These findings provide novel insights in how plasmin regulates haematopoietic recovery upon cytotoxic myeloablation.


Asunto(s)
Purgación de la Médula Ósea/métodos , Fibrinolisina/metabolismo , Fibrinólisis , Hematopoyesis , Activador de Tejido Plasminógeno/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibrinógeno/metabolismo , Fibrinólisis/efectos de los fármacos , Fluorouracilo/farmacología , Hematopoyesis/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Plasminógeno/deficiencia , Plasminógeno/metabolismo
19.
J Clin Invest ; 115(2): 237-46, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15650770

RESUMEN

Mechanisms regulating thrombus stabilization remain largely unknown. Here, we report that loss of any 1 of the Gas6 receptors (Gas6-Rs), i.e., Tyro3, Axl, or Mer, or delivery of a soluble extracellular domain of Axl that traps Gas6 protects mice against life-threatening thrombosis. Loss of a Gas6-R does not prevent initial platelet aggregation but impairs subsequent stabilization of platelet aggregates, at least in part by reducing "outside-in" signaling and platelet granule secretion. Gas6, through its receptors, activates PI3K and Akt and stimulates tyrosine phosphorylation of the beta3 integrin, thereby amplifying outside-in signaling via alphaIIbbeta3. Blocking the Gas6-R-alphaIIbbeta3 integrin cross-talk might be a novel approach to the reduction of thrombosis.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Agregación Plaquetaria/fisiología , Transducción de Señal/fisiología , Trombosis/metabolismo , Animales , Integrina beta3/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/metabolismo , Agregación Plaquetaria/genética , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/genética , Trombosis/tratamiento farmacológico , Trombosis/genética , Trombosis/patología
20.
Thromb Haemost ; 87(4): 666-73, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12008950

RESUMEN

Staphylokinase is a potent highly fibrin-selective thrombolytic agent, but it induces a humoral immune response in most treated patients. Staphylokinase-specific T-lymphocytes can be found in normal healthy individuals, from whom a large panel of staphylokinase-specific T-cells were cloned. The staphylokinase amino acid sequence 71-87 was widely recognized, as it induced proliferation of T-cell clones isolated from 90% of the donors. Computer modeling of this area, threaded as 11-mer peptides within the peptide-binding groove of the major HLA-DR alleles, indicated two putative partially overlapping binding sequences. The region-(71-87)-specific T-cell clones recognized either one or the other minimal peptide, confirming that both sequences could be functional T-cell epitopes. Furthermore, to guide the mutagenesis to eliminate T-cell reactivity, the contribution of each residue to the HLA-DR-anchoring and T-cell receptor exposure was evaluated for both binding motifs. Computer calculations combined with functional assays resulted in the design of staphylokinase-variants, including 2 to 4 amino acid substitutions in the region 71-87. These variants were no longer recognized by the region-(71-87)-specific T-cell clones, and importantly no new staphylokinase-variant-specific cellular immune response could be measured.


Asunto(s)
Antígenos Bacterianos/inmunología , Simulación por Computador , Fibrinolíticos/inmunología , Metaloendopeptidasas/inmunología , Modelos Moleculares , Subgrupos de Linfocitos T/inmunología , Secuencia de Aminoácidos , Antígenos Bacterianos/química , Antígenos Bacterianos/genética , Sitios de Unión , Diseño de Fármacos , Epítopos/química , Epítopos/inmunología , Fibrinolíticos/química , Antígenos HLA-DR/inmunología , Humanos , Activación de Linfocitos , Metaloendopeptidasas/química , Metaloendopeptidasas/genética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Oligopéptidos/química , Oligopéptidos/inmunología , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA