Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Cell ; 180(4): 764-779.e20, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-32059779

RESUMEN

The heterogeneity of endothelial cells (ECs) across tissues remains incompletely inventoried. We constructed an atlas of >32,000 single-EC transcriptomes from 11 mouse tissues and identified 78 EC subclusters, including Aqp7+ intestinal capillaries and angiogenic ECs in healthy tissues. ECs from brain/testis, liver/spleen, small intestine/colon, and skeletal muscle/heart pairwise expressed partially overlapping marker genes. Arterial, venous, and lymphatic ECs shared more markers in more tissues than did heterogeneous capillary ECs. ECs from different vascular beds (arteries, capillaries, veins, lymphatics) exhibited transcriptome similarity across tissues, but the tissue (rather than the vessel) type contributed to the EC heterogeneity. Metabolic transcriptome analysis revealed a similar tissue-grouping phenomenon of ECs and heterogeneous metabolic gene signatures in ECs between tissues and between vascular beds within a single tissue in a tissue-type-dependent pattern. The EC atlas taxonomy enabled identification of EC subclusters in public scRNA-seq datasets and provides a powerful discovery tool and resource value.


Asunto(s)
Células Endoteliales/metabolismo , Análisis de la Célula Individual , Transcriptoma , Animales , Encéfalo/citología , Sistema Cardiovascular/citología , Células Endoteliales/clasificación , Células Endoteliales/citología , Tracto Gastrointestinal/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Músculos/citología , Especificidad de Órganos , RNA-Seq , Testículo/citología
2.
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
3.
Nature ; 561(7721): 63-69, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30158707

RESUMEN

Glutamine synthetase, encoded by the gene GLUL, is an enzyme that converts glutamate and ammonia to glutamine. It is expressed by endothelial cells, but surprisingly shows negligible glutamine-synthesizing activity in these cells at physiological glutamine levels. Here we show in mice that genetic deletion of Glul in endothelial cells impairs vessel sprouting during vascular development, whereas pharmacological blockade of glutamine synthetase suppresses angiogenesis in ocular and inflammatory skin disease while only minimally affecting healthy adult quiescent endothelial cells. This relies on the inhibition of endothelial cell migration but not proliferation. Mechanistically we show that in human umbilical vein endothelial cells GLUL knockdown reduces membrane localization and activation of the GTPase RHOJ while activating other Rho GTPases and Rho kinase, thereby inducing actin stress fibres and impeding endothelial cell motility. Inhibition of Rho kinase rescues the defect in endothelial cell migration that is induced by GLUL knockdown. Notably, glutamine synthetase palmitoylates itself and interacts with RHOJ to sustain RHOJ palmitoylation, membrane localization and activation. These findings reveal that, in addition to the known formation of glutamine, the enzyme glutamine synthetase shows unknown activity in endothelial cell migration during pathological angiogenesis through RHOJ palmitoylation.


Asunto(s)
Células Endoteliales/enzimología , Células Endoteliales/patología , Glutamato-Amoníaco Ligasa/metabolismo , Glutamina/biosíntesis , Neovascularización Patológica , Actinas/metabolismo , Animales , Movimiento Celular , Células Endoteliales/metabolismo , Femenino , Glutamato-Amoníaco Ligasa/deficiencia , Glutamato-Amoníaco Ligasa/genética , Glutamato-Amoníaco Ligasa/fisiología , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Lipoilación , Ratones , Ácido Palmítico/metabolismo , Procesamiento Proteico-Postraduccional , Fibras de Estrés/metabolismo , Proteínas de Unión al GTP rho/química , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo
4.
Nature ; 542(7639): 49-54, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28024299

RESUMEN

Lymphatic vessels are lined by lymphatic endothelial cells (LECs), and are critical for health. However, the role of metabolism in lymphatic development has not yet been elucidated. Here we report that in transgenic mouse models, LEC-specific loss of CPT1A, a rate-controlling enzyme in fatty acid ß-oxidation, impairs lymphatic development. LECs use fatty acid ß-oxidation to proliferate and for epigenetic regulation of lymphatic marker expression during LEC differentiation. Mechanistically, the transcription factor PROX1 upregulates CPT1A expression, which increases acetyl coenzyme A production dependent on fatty acid ß-oxidation. Acetyl coenzyme A is used by the histone acetyltransferase p300 to acetylate histones at lymphangiogenic genes. PROX1-p300 interaction facilitates preferential histone acetylation at PROX1-target genes. Through this metabolism-dependent mechanism, PROX1 mediates epigenetic changes that promote lymphangiogenesis. Notably, blockade of CPT1 enzymes inhibits injury-induced lymphangiogenesis, and replenishing acetyl coenzyme A by supplementing acetate rescues this process in vivo.


Asunto(s)
Ácidos Grasos/química , Ácidos Grasos/metabolismo , Linfangiogénesis , Vasos Linfáticos/citología , Vasos Linfáticos/metabolismo , Acetatos/farmacología , Acetilcoenzima A/metabolismo , Acetilación/efectos de los fármacos , Animales , Carnitina O-Palmitoiltransferasa/antagonistas & inhibidores , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Epigénesis Genética , Femenino , Histonas/metabolismo , Proteínas de Homeodominio/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Linfangiogénesis/efectos de los fármacos , Linfangiogénesis/genética , Vasos Linfáticos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción/efectos de los fármacos , Biosíntesis de Proteínas , Transcripción Genética , Proteínas Supresoras de Tumor/metabolismo , Arterias Umbilicales/citología , Regulación hacia Arriba
5.
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
6.
Nucleic Acids Res ; 48(W1): W385-W394, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32392297

RESUMEN

The amount of biological data, generated with (single cell) omics technologies, is rapidly increasing, thereby exacerbating bottlenecks in the data analysis and interpretation of omics experiments. Data mining platforms that facilitate non-bioinformatician experimental scientists to analyze a wide range of experimental designs and data types can alleviate such bottlenecks, aiding in the exploration of (newly generated or publicly available) omics datasets. Here, we present BIOMEX, a browser-based software, designed to facilitate the Biological Interpretation Of Multi-omics EXperiments by bench scientists. BIOMEX integrates state-of-the-art statistical tools and field-tested algorithms into a flexible but well-defined workflow that accommodates metabolomics, transcriptomics, proteomics, mass cytometry and single cell data from different platforms and organisms. The BIOMEX workflow is accompanied by a manual and video tutorials that provide the necessary background to navigate the interface and get acquainted with the employed methods. BIOMEX guides the user through omics-tailored analyses, such as data pretreatment and normalization, dimensionality reduction, differential and enrichment analysis, pathway mapping, clustering, marker analysis, trajectory inference, meta-analysis and others. BIOMEX is fully interactive, allowing users to easily change parameters and generate customized plots exportable as high-quality publication-ready figures. BIOMEX is open source and freely available at https://www.vibcancer.be/software-tools/biomex.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Análisis de la Célula Individual/métodos , Programas Informáticos , Algoritmos , Neoplasias de los Conductos Biliares/genética , Colangiocarcinoma/genética , Gráficos por Computador , Células Endoteliales/metabolismo , Humanos , Metabolómica/métodos , Neoplasias/mortalidad , Proteómica/métodos , Análisis de Supervivencia , Flujo de Trabajo
7.
EMBO J ; 36(16): 2334-2352, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28659375

RESUMEN

Endothelial cell (EC) metabolism is emerging as a regulator of angiogenesis, but the precise role of glutamine metabolism in ECs is unknown. Here, we show that depriving ECs of glutamine or inhibiting glutaminase 1 (GLS1) caused vessel sprouting defects due to impaired proliferation and migration, and reduced pathological ocular angiogenesis. Inhibition of glutamine metabolism in ECs did not cause energy distress, but impaired tricarboxylic acid (TCA) cycle anaplerosis, macromolecule production, and redox homeostasis. Only the combination of TCA cycle replenishment plus asparagine supplementation restored the metabolic aberrations and proliferation defect caused by glutamine deprivation. Mechanistically, glutamine provided nitrogen for asparagine synthesis to sustain cellular homeostasis. While ECs can take up asparagine, silencing asparagine synthetase (ASNS, which converts glutamine-derived nitrogen and aspartate to asparagine) impaired EC sprouting even in the presence of glutamine and asparagine. Asparagine further proved crucial in glutamine-deprived ECs to restore protein synthesis, suppress ER stress, and reactivate mTOR signaling. These findings reveal a novel link between endothelial glutamine and asparagine metabolism in vessel sprouting.


Asunto(s)
Asparagina/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Glutamina/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Medios de Cultivo/química , Células Endoteliales/metabolismo , Glutaminasa/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Redes y Vías Metabólicas , Neovascularización Patológica
8.
Nature ; 520(7546): 192-197, 2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25830893

RESUMEN

The metabolism of endothelial cells during vessel sprouting remains poorly studied. Here we report that endothelial loss of CPT1A, a rate-limiting enzyme of fatty acid oxidation (FAO), causes vascular sprouting defects due to impaired proliferation, not migration, of human and murine endothelial cells. Reduction of FAO in endothelial cells did not cause energy depletion or disturb redox homeostasis, but impaired de novo nucleotide synthesis for DNA replication. Isotope labelling studies in control endothelial cells showed that fatty acid carbons substantially replenished the Krebs cycle, and were incorporated into aspartate (a nucleotide precursor), uridine monophosphate (a precursor of pyrimidine nucleoside triphosphates) and DNA. CPT1A silencing reduced these processes and depleted endothelial cell stores of aspartate and deoxyribonucleoside triphosphates. Acetate (metabolized to acetyl-CoA, thereby substituting for the depleted FAO-derived acetyl-CoA) or a nucleoside mix rescued the phenotype of CPT1A-silenced endothelial cells. Finally, CPT1 blockade inhibited pathological ocular angiogenesis in mice, suggesting a novel strategy for blocking angiogenesis.


Asunto(s)
Carbono/metabolismo , Células Endoteliales/metabolismo , Ácidos Grasos/química , Ácidos Grasos/metabolismo , Nucleótidos/biosíntesis , Ácido Acético/farmacología , Adenosina Trifosfato/metabolismo , Animales , Vasos Sanguíneos/citología , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Carnitina O-Palmitoiltransferasa/antagonistas & inhibidores , Carnitina O-Palmitoiltransferasa/deficiencia , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclo del Ácido Cítrico , ADN/biosíntesis , Modelos Animales de Enfermedad , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Silenciador del Gen , Glucosa/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Nucleótidos/química , Nucleótidos/farmacología , Oxidación-Reducción/efectos de los fármacos , Retinopatía de la Prematuridad/tratamiento farmacológico , Retinopatía de la Prematuridad/metabolismo , Retinopatía de la Prematuridad/patología
9.
Nucleic Acids Res ; 47(D1): D736-D744, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30357379

RESUMEN

Endothelial cells (ECs) line blood vessels, regulate homeostatic processes (blood flow, immune cell trafficking), but are also involved in many prevalent diseases. The increasing use of high-throughput technologies such as gene expression microarrays and (single cell) RNA sequencing generated a wealth of data on the molecular basis of EC (dys-)function. Extracting biological insight from these datasets is challenging for scientists who are not proficient in bioinformatics. To facilitate the re-use of publicly available EC transcriptomics data, we developed the endothelial database EndoDB, a web-accessible collection of expert curated, quality assured and pre-analyzed data collected from 360 datasets comprising a total of 4741 bulk and 5847 single cell endothelial transcriptomes from six different organisms. Unlike other added-value databases, EndoDB allows to easily retrieve and explore data of specific studies, determine under which conditions genes and pathways of interest are deregulated and assess reprogramming of metabolism via principal component analysis, differential gene expression analysis, gene set enrichment analysis, heatmaps and metabolic and transcription factor analysis, while single cell data are visualized as gene expression color-coded t-SNE plots. Plots and tables in EndoDB are customizable, downloadable and interactive. EndoDB is freely available at https://vibcancer.be/software-tools/endodb, and will be updated to include new studies.


Asunto(s)
Biología Computacional , Bases de Datos Genéticas , Transcriptoma/genética , Animales , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Análisis de Componente Principal
10.
J Am Soc Nephrol ; 31(1): 118-138, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31818909

RESUMEN

BACKGROUND: Renal endothelial cells from glomerular, cortical, and medullary kidney compartments are exposed to different microenvironmental conditions and support specific kidney processes. However, the heterogeneous phenotypes of these cells remain incompletely inventoried. Osmotic homeostasis is vitally important for regulating cell volume and function, and in mammals, osmotic equilibrium is regulated through the countercurrent system in the renal medulla, where water exchange through endothelium occurs against an osmotic pressure gradient. Dehydration exposes medullary renal endothelial cells to extreme hyperosmolarity, and how these cells adapt to and survive in this hypertonic milieu is unknown. METHODS: We inventoried renal endothelial cell heterogeneity by single-cell RNA sequencing >40,000 mouse renal endothelial cells, and studied transcriptome changes during osmotic adaptation upon water deprivation. We validated our findings by immunostaining and functionally by targeting oxidative phosphorylation in a hyperosmolarity model in vitro and in dehydrated mice in vivo. RESULTS: We identified 24 renal endothelial cell phenotypes (of which eight were novel), highlighting extensive heterogeneity of these cells between and within the cortex, glomeruli, and medulla. In response to dehydration and hypertonicity, medullary renal endothelial cells upregulated the expression of genes involved in the hypoxia response, glycolysis, and-surprisingly-oxidative phosphorylation. Endothelial cells increased oxygen consumption when exposed to hyperosmolarity, whereas blocking oxidative phosphorylation compromised endothelial cell viability during hyperosmotic stress and impaired urine concentration during dehydration. CONCLUSIONS: This study provides a high-resolution atlas of the renal endothelium and highlights extensive renal endothelial cell phenotypic heterogeneity, as well as a previously unrecognized role of oxidative phosphorylation in the metabolic adaptation of medullary renal endothelial cells to water deprivation.


Asunto(s)
Adaptación Fisiológica/genética , Células Endoteliales/metabolismo , Riñón/citología , Análisis de Secuencia de ARN , Privación de Agua/fisiología , Animales , Células Endoteliales/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo
12.
Nat Cell Biol ; 9(6): 666-74, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17515931

RESUMEN

Caspase-14 belongs to a conserved family of aspartate-specific proteinases. Its expression is restricted almost exclusively to the suprabasal layers of the epidermis and the hair follicles. Moreover, the proteolytic activation of caspase-14 is associated with stratum corneum formation, implicating caspase-14 in terminal keratinocyte differentiation and cornification. Here, we show that the skin of caspase-14-deficient mice was shiny and lichenified, indicating an altered stratum-corneum composition. Caspase-14-deficient epidermis contained significantly more alveolar keratohyalin F-granules, the profilaggrin stores. Accordingly, caspase-14-deficient epidermis is characterized by an altered profilaggrin processing pattern and we show that recombinant caspase-14 can directly cleave profilaggrin in vitro. Caspase-14-deficient epidermis is characterized by reduced skin-hydration levels and increased water loss. In view of the important role of filaggrin in the structure and moisturization of the skin, the knockout phenotype could be explained by an aberrant processing of filaggrin. Importantly, the skin of caspase-14-deficient mice was highly sensitive to the formation of cyclobutane pyrimidine dimers after UVB irradiation, leading to increased levels of UVB-induced apoptosis. Removal of the stratum corneum indicate that caspase-14 controls the UVB scavenging capacity of the stratum corneum.


Asunto(s)
Envejecimiento/fisiología , Caspasas/genética , Deshidratación/enzimología , Epidermis/enzimología , Queratinocitos/enzimología , Rayos Ultravioleta/efectos adversos , Envejecimiento/efectos de la radiación , Animales , Animales Recién Nacidos , Diferenciación Celular/genética , Diferenciación Celular/efectos de la radiación , Células Cultivadas , Deshidratación/fisiopatología , Epidermis/fisiopatología , Epidermis/efectos de la radiación , Proteínas Filagrina , Proteínas de Filamentos Intermediarios/metabolismo , Queratinocitos/patología , Queratinocitos/efectos de la radiación , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Trastornos por Fotosensibilidad/enzimología , Trastornos por Fotosensibilidad/genética , Trastornos por Fotosensibilidad/fisiopatología , Dímeros de Pirimidina/metabolismo , Equilibrio Hidroelectrolítico/genética , Equilibrio Hidroelectrolítico/efectos de la radiación
13.
Nucleic Acids Res ; 40(22): 11477-89, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23066103

RESUMEN

Telomere position effect (TPE) is the influence of telomeres on subtelomeric epigenetic marks and gene expression. Previous studies suggested that TPE depends on genetic background. As these analyses were performed on different chromosomes, cell types and species, it remains unclear whether TPE represents a chromosome-rather than genetic background-specific regulation. We describe the development of a Linear Human Artificial Chromosome (L-HAC) as a new tool for telomere studies. The L-HAC was generated through the Cre-loxP-mediated addition of telomere ends to an existing circular HAC (C-HAC). As it can be transferred to genetically distinct cell lines and animal models the L-HAC enables the study of TPE in an unprecedented manner. The HAC was relocated to four telomerase-positive cell lines via microcell-mediated chromosome transfer and subsequently to mice via blastocyst injection of L-HAC(+)-ES-cells. We could show consistent genetic background-dependent adaptation of telomere length and telomere-associated de novo subtelomeric DNA methylation in mouse ES-R1 cells as well as in mice. Expression of the subtelomeric neomycin gene was inversely correlated with telomere length and subtelomeric methylation. We thus provide a new tool for functional telomere studies and provide strong evidence that telomere length, subtelomeric chromatin marks and expression of subtelomeric genes are genetic background dependent.


Asunto(s)
Efectos de la Posición Cromosómica , Cromosomas Artificiales Humanos , Homeostasis del Telómero , Telómero/fisiología , Animales , Células Cultivadas , Cromatina/metabolismo , Cricetinae , Metilación de ADN , Humanos , Ratones , Ratones Endogámicos BALB C
14.
Commun Biol ; 7(1): 618, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38783087

RESUMEN

Endothelial cells (ECs) are highly glycolytic, but whether they generate glycolytic intermediates via gluconeogenesis (GNG) in glucose-deprived conditions remains unknown. Here, we report that glucose-deprived ECs upregulate the GNG enzyme PCK2 and rely on a PCK2-dependent truncated GNG, whereby lactate and glutamine are used for the synthesis of lower glycolytic intermediates that enter the serine and glycerophospholipid biosynthesis pathways, which can play key roles in redox homeostasis and phospholipid synthesis, respectively. Unexpectedly, however, even in normal glucose conditions, and independent of its enzymatic activity, PCK2 silencing perturbs proteostasis, beyond its traditional GNG role. Indeed, PCK2-silenced ECs have an impaired unfolded protein response, leading to accumulation of misfolded proteins, which due to defective proteasomes and impaired autophagy, results in the accumulation of protein aggregates in lysosomes and EC demise. Ultimately, loss of PCK2 in ECs impaired vessel sprouting. This study identifies a role for PCK2 in proteostasis beyond GNG.


Asunto(s)
Células Endoteliales , Gluconeogénesis , Fosfoenolpiruvato Carboxiquinasa (GTP) , Proteostasis , Gluconeogénesis/genética , Humanos , Células Endoteliales/metabolismo , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Glucosa/metabolismo , Autofagia , Respuesta de Proteína Desplegada , Fosfoenolpiruvato Carboxiquinasa (ATP)
15.
Commun Biol ; 6(1): 648, 2023 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-37330599

RESUMEN

Translation of academic results into clinical practice is a formidable unmet medical need. Single-cell RNA-sequencing (scRNA-seq) studies generate long descriptive ranks of markers with predicted biological function, but without functional validation, it remains challenging to know which markers truly exert the putative function. Given the lengthy/costly nature of validation studies, gene prioritization is required to select candidates. We address these issues by studying tip endothelial cell (EC) marker genes because of their importance for angiogenesis. Here, by tailoring Guidelines On Target Assessment for Innovative Therapeutics, we in silico prioritize previously unreported/poorly described, high-ranking tip EC markers. Notably, functional validation reveals that four of six candidates behave as tip EC genes. We even discover a tip EC function for a gene lacking in-depth functional annotation. Thus, validating prioritized genes from scRNA-seq studies offers opportunities for identifying targets to be considered for possible translation, but not all top-ranked scRNA-seq markers exert the predicted function.


Asunto(s)
Perfilación de la Expresión Génica , Transcriptoma , Perfilación de la Expresión Génica/métodos
16.
Signal Transduct Target Ther ; 8(1): 305, 2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37591843

RESUMEN

Although VEGF-B was discovered as a VEGF-A homolog a long time ago, the angiogenic effect of VEGF-B remains poorly understood with limited and diverse findings from different groups. Notwithstanding, drugs that inhibit VEGF-B together with other VEGF family members are being used to treat patients with various neovascular diseases. It is therefore critical to have a better understanding of the angiogenic effect of VEGF-B and the underlying mechanisms. Using comprehensive in vitro and in vivo methods and models, we reveal here for the first time an unexpected and surprising function of VEGF-B as an endogenous inhibitor of angiogenesis by inhibiting the FGF2/FGFR1 pathway when the latter is abundantly expressed. Mechanistically, we unveil that VEGF-B binds to FGFR1, induces FGFR1/VEGFR1 complex formation, and suppresses FGF2-induced Erk activation, and inhibits FGF2-driven angiogenesis and tumor growth. Our work uncovers a previously unrecognized novel function of VEGF-B in tethering the FGF2/FGFR1 pathway. Given the anti-angiogenic nature of VEGF-B under conditions of high FGF2/FGFR1 levels, caution is warranted when modulating VEGF-B activity to treat neovascular diseases.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos , Factor B de Crecimiento Endotelial Vascular , Humanos , Factor 2 de Crecimiento de Fibroblastos/genética , Inmunoterapia , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética
17.
Cardiovasc Res ; 119(2): 520-535, 2023 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35998078

RESUMEN

AIMS: Severe acute respiratory syndrome coronavirus-2 infection causes COVID-19, which in severe cases evokes life-threatening acute respiratory distress syndrome (ARDS). Transcriptome signatures and the functional relevance of non-vascular cell types (e.g. immune and epithelial cells) in COVID-19 are becoming increasingly evident. However, despite its known contribution to vascular inflammation, recruitment/invasion of immune cells, vascular leakage, and perturbed haemostasis in the lungs of severe COVID-19 patients, an in-depth interrogation of the endothelial cell (EC) compartment in lethal COVID-19 is lacking. Moreover, progressive fibrotic lung disease represents one of the complications of COVID-19 pneumonia and ARDS. Analogous features between idiopathic pulmonary fibrosis (IPF) and COVID-19 suggest partial similarities in their pathophysiology, yet, a head-to-head comparison of pulmonary cell transcriptomes between both conditions has not been implemented to date. METHODS AND RESULTS: We performed single-nucleus RNA-sequencing on frozen lungs from 7 deceased COVID-19 patients, 6 IPF explant lungs, and 12 controls. The vascular fraction, comprising 38 794 nuclei, could be subclustered into 14 distinct EC subtypes. Non-vascular cell types, comprising 137 746 nuclei, were subclustered and used for EC-interactome analyses. Pulmonary ECs of deceased COVID-19 patients showed an enrichment of genes involved in cellular stress, as well as signatures suggestive of dampened immunomodulation and impaired vessel wall integrity. In addition, increased abundance of a population of systemic capillary and venous ECs was identified in COVID-19 and IPF. COVID-19 systemic ECs closely resembled their IPF counterparts, and a set of 30 genes was found congruently enriched in systemic ECs across studies. Receptor-ligand interaction analysis of ECs with non-vascular cell types in the pulmonary micro-environment revealed numerous previously unknown interactions specifically enriched/depleted in COVID-19 and/or IPF. CONCLUSIONS: This study uncovered novel insights into the abundance, expression patterns, and interactomes of EC subtypes in COVID-19 and IPF, relevant for future investigations into the progression and treatment of both lethal conditions.


Asunto(s)
COVID-19 , Fibrosis Pulmonar Idiopática , Síndrome de Dificultad Respiratoria , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Transcriptoma
18.
Nat Commun ; 13(1): 5511, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36127427

RESUMEN

Since a detailed inventory of endothelial cell (EC) heterogeneity in breast cancer (BC) is lacking, here we perform single cell RNA-sequencing of 26,515 cells (including 8433 ECs) from 9 BC patients and compare them to published EC taxonomies from lung tumors. Angiogenic ECs are phenotypically similar, while other EC subtypes are different. Predictive interactome analysis reveals known but also previously unreported receptor-ligand interactions between ECs and immune cells, suggesting an involvement of breast EC subtypes in immune responses. We also identify a capillary EC subtype (LIPEC (Lipid Processing EC)), which expresses genes involved in lipid processing that are regulated by PPAR-γ and is more abundant in peri-tumoral breast tissue. Retrospective analysis of 4648 BC patients reveals that treatment with metformin (an indirect PPAR-γ signaling activator) provides long-lasting clinical benefit and is positively associated with LIPEC abundance. Our findings warrant further exploration of this LIPEC/PPAR-γ link for BC treatment.


Asunto(s)
Neoplasias de la Mama , Metformina , Neoplasias de la Mama/patología , Células Endoteliales/patología , Femenino , Humanos , Inmunidad , Ligandos , Lípidos , Metformina/farmacología , PPAR gamma/genética , ARN , Estudios Retrospectivos
19.
Stem Cells ; 28(3): 399-406, 2010 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-20049903

RESUMEN

Alzheimer's disease amyloid precursor protein (APP) has been implicated in many neurobiologic processes, but supporting evidence remains indirect. Studies are confounded by the existence of two partially redundant APP homologues, APLP1 and APLP2. APP/APLP1/APLP2 triple knockout (APP tKO) mice display cobblestone lissencephaly and are perinatally lethal. To circumvent this problem, we generated APP triple knockout embryonic stem (ES) cells and differentiated these to APP triple knockout neurons in vitro and in vivo. In comparison with wild-type (WT) ES cell-derived neurons, APP tKO neurons formed equally pure neuronal cultures, had unaltered in vitro migratory capacities, had a similar acquisition of polarity, and were capable of extending long neurites and forming active excitatory synapses. These data were confirmed in vivo in chimeric mice with APP tKO neurons expressing the enhanced green fluorescent protein (eGFP) present in a WT background brain. The results suggest that the loss of the APP family of proteins has no major effect on these critical neuronal processes and that the apparent multitude of functions in which APP has been implicated might be characterized by molecular redundancy. Our stem cell culture provides an excellent tool to circumvent the problem of lack of viability of APP/APLP triple knockout mice and will help to explore the function of this intriguing protein further in vitro and in vivo.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Encéfalo/embriología , Encéfalo/metabolismo , Diferenciación Celular/fisiología , Células Madre Embrionarias/metabolismo , Neurogénesis/genética , Neuronas/metabolismo , Animales , Encéfalo/citología , Técnicas de Cultivo de Célula , Movimiento Celular/genética , Polaridad Celular/genética , Células Cultivadas , Quimera , Lisencefalia de Cobblestone/genética , Lisencefalia de Cobblestone/metabolismo , Lisencefalia de Cobblestone/fisiopatología , Células Madre Embrionarias/citología , Femenino , Proteínas Fluorescentes Verdes/genética , Masculino , Ratones , Ratones Noqueados , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/metabolismo , Malformaciones del Sistema Nervioso/fisiopatología , Vías Nerviosas/citología , Vías Nerviosas/embriología , Vías Nerviosas/metabolismo , Neuritas/metabolismo , Neuritas/ultraestructura , Neuronas/citología
20.
STAR Protoc ; 2(2): 100489, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-34007969

RESUMEN

Endothelial cells (ECs) from the small intestine, colon, liver, and heart have distinct phenotypes and functional adaptations that are dependent on their physiological environment. Gut ECs adapt to low oxygen, heart ECs to contractile forces, and liver ECs to low flow rates. Isolating high-purity ECs in sufficient quantities is crucial to study their functions. Here, we describe protocols combining magnetic and fluorescent activated cell sorting for rapid and reproducible EC purification from four adult murine tissues. For complete details on the use and execution of these protocols, please refer to Kalucka et al. (2020).


Asunto(s)
Células Endoteliales/citología , Citometría de Flujo/métodos , Intestinos/citología , Hígado/citología , Miocardio/citología , Animales , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda