49 results on '"Goveia J"'
Search Results
2. Umfassende Charakterisierung der Mikroumgebung des Prostatatumors identifiziert CXCR4/CXCL12-Crosstalk als neue Therapiestrategie beim Prostatakarzinom
- Author
-
Heidegger, I, Fotakis, G, Offermann, A, Goveia, J, Noureen, A, Timmer-Bosscha, H, Schäfer, G, Daum, S, Salcher, S, Walenkamp, A, Perner, S, Beatovic, A, Plattner, C, Krogsdam, A, Haybaeck, J, Sopper, S, Thaler, S, Klocker, H, Trajanoski, Z, Wolf, D, and Pircher, A
- Subjects
ddc: 610 ,Medicine and health - Abstract
Einleitung: Crosstalk zwischen neoplastischen und stromalen Zellen fördert das Fortschreiten des Prostatakarzinoms (PCa). Einblicke in Zell-Zell-Kommunikationsnetzwerke bieten neue therapeutische Wege, um Prozesse zu formen, die zu Veränderungen der PCa-Tumormikroumgebung (TME) beitragen. [zum vollständigen Text gelangen Sie über die oben angegebene URL]
- Published
- 2022
- Full Text
- View/download PDF
3. First identification and validation of prostate cancer specific tumor endothelial cell markers by multi-omic profiling
- Author
-
Heidegger, I., primary, Offermanns, A., additional, Timmer-Bosscha, H., additional, Schäfer, G., additional, Goveia, J., additional, Daum, S., additional, Walenkamp, A., additional, Perner, S., additional, Trajanoski, Z., additional, Krogsdam, A., additional, Haybaeck, J., additional, Plattner, C., additional, Salcher, S., additional, Thaler, S., additional, Keller, M., additional, Klocker, H., additional, Wolf, D., additional, and Pircher, A., additional
- Published
- 2021
- Full Text
- View/download PDF
4. Predicted genetic gains weighted by selection pressures for grain quality in irrigated rice
- Author
-
FACCHINELLO, P. H. K., CARVALHO, I. R., STRECK, E. A., AGUIAR, G. A., GOVEIA, J., FEIJÓ, M., PEREIRA, R. R., OLIVEIRA, V. F. de, FAGUNDES, P. R. R., MAIA, L. C. da, LAUTENCHLEGER, F., MAGALHAES JUNIOR, A. M. de, PAULO HENRIQUE KARLING FACCHINELLO, IVAN RICARDO CARVALHO, EDUARDO ANIBELE STRECK, GABRIEL ALMEIDA AGUIAR, JANAÍNA GOVEIA, MICHELE FEIJÓ, ROBERTO RAMOS PEREIRA, VICTORIA FREITAS DE OLIVEIRA, PAULO RICARDO REIS FAGUNDES, CPACT, LUCIANO CARLOS DA MAIA, FRANCINE LAUTENCHLEGER, and ARIANO MARTINS DE MAGALHAES JUNIOR, CPACT.
- Subjects
Arroz ,Oriza - Abstract
O objetivo deste trabalho foi analisar componentes de variâncias e herdabilidades, estimar o ganho de seleção e avaliar as diferentes pressões de seleção para atributos físicos de qualidade de grãos de famílias de gerações segregantes de arroz (Oryza sativa) irrigado, por meio da metodologia REML/BLUP. O experimento foi conduzido no Rio Grande do Sul, Brasil, onde foram realizadas seleções nas gerações segregantes F3, F4 e F5, nas safras 2015/2016, 2016/2017 e 2017/2018, em oito populações segregantes (famílias) oriundas de diferentes cruzamentos. Foram avaliados atributos físicos de qualidade intrínsecos, parâmetros genéticos e resposta à seleção, tendo-se utilizado diferentes pressões de seleção. Foram obtidas estimativas que apresentaram ganhos genéticos para os caracteres de qualidade dos grãos, principalmente para área calcárea total, alvura vítrea, alvura total, relação entre alvura vítrea e alvura total, e percentual de grãos inteiros e quebrados, em seleções nas primeiras gerações. As famílias utilizadas apresentam resultados satisfatórios, sendo superiores às cultivares-controle. Para a maioria dos caracteres, as estimativasde herdabilidade em sentido amplo são consideradas intermediárias, junto com os demais parâmetros, o que mostra a possibilidade de seleção genética para os atributos de qualidade do grão. A resposta à seleção com pressão de 10% é muito promissora para os atributos de qualidade de grãos de arroz. Made available in DSpace on 2021-11-12T02:10:53Z (GMT). No. of bitstreams: 1 PAB-2497-artigo-PH-2021.pdf: 606200 bytes, checksum: 92bb66245b8819d915f2810988087ed5 (MD5) Previous issue date: 2021 Título em português: Ganhos genéticos previstos ponderados por pressões de seleção para qualidade de grãos em arroz irrigado.
- Published
- 2021
5. Severe soft tissue infections of the extremities in patients admitted to an intensive care unit
- Author
-
Zahar, J.-R., Goveia, J., Lesprit, P., and Brun-Buisson, C.
- Published
- 2005
- Full Text
- View/download PDF
6. Endothelial cell heterogeneity in pathological angiogenesis characterized by metabolic transcriptome diversity : Heterogeniteit van endotheelcellen in pathologische angiogenese
- Author
-
Goveia, J, Eelen, G, and Carmeliet, P
- Abstract
Vascular endothelial cells (ECs) pervasively line the lumen of all blood vessels in the body. ECs are quiescent under physiological homeostasis but adopt one of three states in disease. EC activation includes the induction of cell adhesion molecules involved in immune cell recruitment and is associated with inflammatory diseases. EC dysfunction constitutes a state where ECs are no longer able to perform their homeostatic physiological functions and is a characteristic of prevalent diseases such as diabetes and atherosclerosis. Finally, ECs frequently adopt a blood vessel forming angiogenic state in diseases such as cancer and obesity. Therapeutic targeting of angiogenic ECs using agents that inhibit pro-angiogenic growth factor signaling is clinically approved to treat neovascularization in age-related macular degeneration (AMD) and cancer. Unfortunately, intrinsic and acquired resistance limit efficacy in both indications. Functional genetics in normal ECs showed that growth factor signaling converges on central carbon metabolism, and that EC metabolism can overrule pro-angiogenic growth factor signaling. However, whether metabolic reprogramming also occurs in diseased ECs, and whether this includes pathways beyond central carbon metabolism is almost entirely unknown. The goal of this work was to provide an atlas of metabolic heterogeneity in single diseased ECs in AMD and cancer. Since metabolomics is insufficiently sensitive to measure metabolite levels and fluxes in single ECs, and we previously documented that changes in metabolic gene expression are predictive of changes in metabolism in ECs, we used single cell RNA sequencing (scRNA-seq) to characterize the metabolic transcriptome of ECs at the single cell level. In the first study, we single-cell RNA-sequenced ECs from human and mouse lung tumors and detected 17 known and discovered 16 novel phenotypes. Integrated analysis of scRNA-seq data with orthogonal multi-omics and meta-analysis data across different human tumors, validated by functional analysis, identified collagen metabolism and modification as a putative angiogenic candidate pathway. In a second study, we used scRNA-sequencing to profile choroidal ECs isolated from mouse models of AMD. Trajectory inference suggested that ECs plastically upregulate genes in central carbon metabolism and collagen biosynthesis during angiogenic differentiation. Choroidal EC-tailored genome scale metabolic modeling integrated with scRNA-seq and gene expression meta-analysis identified rate limiting enzymes in cholesterol and proline biosynthesis as top-ranking candidates in proliferation and collagen production, respectively. In line with the stated ambition of this project, we developed user-friendly browseable databases to accompany each study in order to maximize resource value and facilitate the design of follow-up studies on the metabolic targets that can be derived from this work. status: published
- Published
- 2019
7. Increase in tumor-associated macrophages after antiangiogenic therapy is associated with poor survival among patients with recurrent glioblastoma
- Author
-
Lu-Emerson, C., primary, Snuderl, M., additional, Kirkpatrick, N. D., additional, Goveia, J., additional, Davidson, C., additional, Huang, Y., additional, Riedemann, L., additional, Taylor, J., additional, Ivy, P., additional, Duda, D. G., additional, Ancukiewicz, M., additional, Plotkin, S. R., additional, Chi, A. S., additional, Gerstner, E. R., additional, Eichler, A. F., additional, Dietrich, J., additional, Stemmer-Rachamimov, A. O., additional, Batchelor, T. T., additional, and Jain, R. K., additional
- Published
- 2013
- Full Text
- View/download PDF
8. Hypoxia determines survival outcomes of bacterial infection through HIF-1alpha dependent re-programming of leukocyte metabolism
- Author
-
Thompson, AAR, Dickinson, RS, Murphy, F, Thomson, JP, Marriott, HM, Tavares, A, Willson, J, Williams, L, Lewis, A, Mirchandani, A, Dos Santos Coelho, P, Doherty, C, Ryan, E, Watts, E, Morton, NM, Forbes, S, Stimson, RH, Hameed, AG, Arnold, N, Preston, JA, Lawrie, A, Finisguerra, V, Mazzone, M, Sadiku, P, Goveia, J, Taverna, F, Carmeliet, P, Foster, SJ, Chilvers, ER, Cowburn, AS, Dockrell, DH, Johnson, RS, Meehan, RR, Whyte, MKB, and Walmsley
- Subjects
Basic Science ,1108 Medical Microbiology ,2.1 Biological and endogenous factors ,Infection ,3. Good health - Abstract
Hypoxia and bacterial infection frequently co-exist, in both acute and chronic clinical settings, and typically result in adverse clinical outcomes. To ameliorate this morbidity, we investigated the interaction between hypoxia and the host response. In the context of acute hypoxia, both S. aureus and S. pneumoniae infections rapidly induced progressive neutrophil mediated morbidity and mortality, with associated hypothermia and cardiovascular compromise. Preconditioning animals through longer exposures to hypoxia, prior to infection, prevented these pathophysiological responses and profoundly dampened the transcriptome of circulating leukocytes. Specifically, perturbation of HIF pathway and glycolysis genes by hypoxic preconditioning was associated with reduced leukocyte glucose utilisation, resulting in systemic rescue from a global negative energy state and myocardial protection. Thus we demonstrate that hypoxia preconditions the innate immune response and determines survival outcomes following bacterial infection through suppression of HIF-1α and neutrophil metabolism. The therapeutic implications of this work are that in the context of systemic or tissue hypoxia therapies that target the host response could improve infection associated morbidity and mortality.
9. Partial and transient reduction of glycolysis by PFKFB3 blockade reduces pathological angiogenesis
- Author
-
Schoors S, De Bock K, Ar, Cantelmo, Georgiadou M, Ghesquière B, Cauwenberghs S, Kuchnio A, Bw, Wong, Quaegebeur A, Goveia J, Bifari F, Wang X, Blanco R, Tembuyser B, Cornelissen I, Bouché A, Vinckier S, Diaz-Moralli S, Gerhardt H, and Telang S
10. Integrated single-cell RNA-seq analysis reveals mitochondrial calcium signaling as a modulator of endothelial-to-mesenchymal transition.
- Author
-
Lebas M, Chinigò G, Courmont E, Bettaieb L, Machmouchi A, Goveia J, Beatovic A, Van Kerckhove J, Robil C, Angulo FS, Vedelago M, Errerd A, Treps L, Gao V, Delgado De la Herrán HC, Mayeuf-Louchart A, L'homme L, Chamlali M, Dejos C, Gouyer V, Garikipati VNS, Tomar D, Yin H, Fukui H, Vinckier S, Stolte A, Conradi LC, Infanti F, Lemonnier L, Zeisberg E, Luo Y, Lin L, Desseyn JL, Pickering J, Kishore R, Madesh M, Dombrowicz D, Perocchi F, Staels B, Pla AF, Gkika D, and Cantelmo AR
- Subjects
- Animals, Humans, Mice, Calcium Channels metabolism, Calcium Channels genetics, Ischemia metabolism, Ischemia pathology, Calcium metabolism, Single-Cell Gene Expression Analysis, Single-Cell Analysis, Calcium Signaling, Mitochondria metabolism, RNA-Seq methods, Endothelial Cells metabolism, Epithelial-Mesenchymal Transition genetics
- Abstract
Endothelial cells (ECs) are highly plastic, capable of differentiating into various cell types. Endothelial-to-mesenchymal transition (EndMT) is crucial during embryonic development and contributes substantially to vascular dysfunction in many cardiovascular diseases (CVDs). While targeting EndMT holds therapeutic promise, understanding its mechanisms and modulating its pathways remain challenging. Using single-cell RNA sequencing on three in vitro EndMT models, we identified conserved gene signatures. We validated original regulators in vitro and in vivo during embryonic heart development and peripheral artery disease. EndMT induction led to global expression changes in all EC subtypes rather than in mesenchymal clusters. We identified mitochondrial calcium uptake as a key driver of EndMT; inhibiting mitochondrial calcium uniporter (MCU) prevented EndMT in vitro, and conditional Mcu deletion in ECs blocked mesenchymal activation in a hind limb ischemia model. Tissues from patients with critical limb ischemia with EndMT features exhibited significantly elevated endothelial MCU. These findings highlight MCU as a regulator of EndMT and a potential therapeutic target.
- Published
- 2024
- Full Text
- View/download PDF
11. Nucleotide metabolism in cancer cells fuels a UDP-driven macrophage cross-talk, promoting immunosuppression and immunotherapy resistance.
- Author
-
Scolaro T, Manco M, Pecqueux M, Amorim R, Trotta R, Van Acker HH, Van Haele M, Shirgaonkar N, Naulaerts S, Daniluk J, Prenen F, Varamo C, Ponti D, Doglioni G, Ferreira Campos AM, Fernandez Garcia J, Radenkovic S, Rouhi P, Beatovic A, Wang L, Wang Y, Tzoumpa A, Antoranz A, Sargsian A, Di Matteo M, Berardi E, Goveia J, Ghesquière B, Roskams T, Soenen S, Voets T, Manshian B, Fendt SM, Carmeliet P, Garg AD, DasGupta R, Topal B, and Mazzone M
- Subjects
- Humans, Animals, Mice, Carcinoma, Pancreatic Ductal immunology, Carcinoma, Pancreatic Ductal therapy, Carcinoma, Pancreatic Ductal drug therapy, Cytidine Deaminase metabolism, Cytidine Deaminase genetics, Tumor-Associated Macrophages immunology, Tumor-Associated Macrophages metabolism, Cell Line, Tumor, Receptors, Purinergic P2 metabolism, Macrophages immunology, Macrophages metabolism, T-Lymphocytes, Cytotoxic immunology, T-Lymphocytes, Cytotoxic drug effects, Tumor Microenvironment immunology, Pancreatic Neoplasms immunology, Pancreatic Neoplasms therapy, Pancreatic Neoplasms drug therapy, Nucleotides metabolism, Immune Tolerance, Programmed Cell Death 1 Receptor, Uridine Diphosphate metabolism, Immunotherapy methods, Drug Resistance, Neoplasm immunology
- Abstract
Many individuals with cancer are resistant to immunotherapies. Here, we identify the gene encoding the pyrimidine salvage pathway enzyme cytidine deaminase (CDA) among the top upregulated metabolic genes in several immunotherapy-resistant tumors. We show that CDA in cancer cells contributes to the uridine diphosphate (UDP) pool. Extracellular UDP hijacks immunosuppressive tumor-associated macrophages (TAMs) through its receptor P2Y
6 . Pharmacologic or genetic inhibition of CDA in cancer cells (or P2Y6 in TAMs) disrupts TAM-mediated immunosuppression, promoting cytotoxic T cell entry and susceptibility to anti-programmed cell death protein 1 (anti-PD-1) treatment in resistant pancreatic ductal adenocarcinoma (PDAC) and melanoma models. Conversely, CDA overexpression in CDA-depleted PDACs or anti-PD-1-responsive colorectal tumors or systemic UDP administration (re)establishes resistance. In individuals with PDAC, high CDA levels in cancer cells correlate with increased TAMs, lower cytotoxic T cells and possibly anti-PD-1 resistance. In a pan-cancer single-cell atlas, CDAhigh cancer cells match with T cell cytotoxicity dysfunction and P2RY6high TAMs. Overall, we suggest CDA and P2Y6 as potential targets for cancer immunotherapy., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
12. FOXF1 promotes tumor vessel normalization and prevents lung cancer progression through FZD4.
- Author
-
Bian F, Goda C, Wang G, Lan YW, Deng Z, Gao W, Acharya A, Reza AA, Gomez-Arroyo J, Merjaneh N, Ren X, Goveia J, Carmeliet P, Kalinichenko VV, and Kalin TV
- Subjects
- Animals, Humans, Mice, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung blood supply, Disease Progression, Neovascularization, Pathologic genetics, Wnt Signaling Pathway, Endothelial Cells metabolism, Endothelial Cells pathology, Forkhead Transcription Factors metabolism, Forkhead Transcription Factors genetics, Frizzled Receptors metabolism, Frizzled Receptors genetics, Lung Neoplasms pathology, Lung Neoplasms genetics, Lung Neoplasms blood supply, Lung Neoplasms metabolism
- Abstract
Cancer cells re-program normal lung endothelial cells (EC) into tumor-associated endothelial cells (TEC) that form leaky vessels supporting carcinogenesis. Transcriptional regulators that control the reprogramming of EC into TEC are poorly understood. We identified Forkhead box F1 (FOXF1) as a critical regulator of EC-to-TEC transition. FOXF1 was highly expressed in normal lung vasculature but was decreased in TEC within non-small cell lung cancers (NSCLC). Low FOXF1 correlated with poor overall survival of NSCLC patients. In mice, endothelial-specific deletion of FOXF1 decreased pericyte coverage, increased vessel permeability and hypoxia, and promoted lung tumor growth and metastasis. Endothelial-specific overexpression of FOXF1 normalized tumor vessels and inhibited the progression of lung cancer. FOXF1 deficiency decreased Wnt/β-catenin signaling in TECs through direct transcriptional activation of Fzd4. Restoring FZD4 expression in FOXF1-deficient TECs through endothelial-specific nanoparticle delivery of Fzd4 cDNA rescued Wnt/β-catenin signaling in TECs, normalized tumor vessels and inhibited the progression of lung cancer. Altogether, FOXF1 increases tumor vessel stability, and inhibits lung cancer progression by stimulating FZD4/Wnt/β-catenin signaling in TECs. Nanoparticle delivery of FZD4 cDNA has promise for future therapies in NSCLC., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
13. Mitochondrial respiration supports autophagy to provide stress resistance during quiescence.
- Author
-
Magalhaes-Novais S, Blecha J, Naraine R, Mikesova J, Abaffy P, Pecinova A, Milosevic M, Bohuslavova R, Prochazka J, Khan S, Novotna E, Sindelka R, Machan R, Dewerchin M, Vlcak E, Kalucka J, Stemberkova Hubackova S, Benda A, Goveia J, Mracek T, Barinka C, Carmeliet P, Neuzil J, Rohlenova K, and Rohlena J
- Subjects
- AMP-Activated Protein Kinases metabolism, Adenosine Triphosphate metabolism, Animals, Cysteine metabolism, DNA, Mitochondrial metabolism, Dextrans metabolism, Endothelial Cells metabolism, Fibroblasts metabolism, Formaldehyde metabolism, Humans, Isothiocyanates, Lipopolysaccharides metabolism, Mechanistic Target of Rapamycin Complex 1 metabolism, Mice, Microtubule-Associated Proteins metabolism, Mitochondria metabolism, Phosphatidylethanolamines metabolism, Reactive Oxygen Species metabolism, Respiration, Sirolimus, Autophagy, Inflammatory Bowel Diseases metabolism
- Abstract
Mitochondrial oxidative phosphorylation (OXPHOS) generates ATP, but OXPHOS also supports biosynthesis during proliferation. In contrast, the role of OXPHOS during quiescence, beyond ATP production, is not well understood. Using mouse models of inducible OXPHOS deficiency in all cell types or specifically in the vascular endothelium that negligibly relies on OXPHOS-derived ATP, we show that selectively during quiescence OXPHOS provides oxidative stress resistance by supporting macroautophagy/autophagy. Mechanistically, OXPHOS constitutively generates low levels of endogenous ROS that induce autophagy via attenuation of ATG4B activity, which provides protection from ROS insult. Physiologically, the OXPHOS-autophagy system (i) protects healthy tissue from toxicity of ROS-based anticancer therapy, and (ii) provides ROS resistance in the endothelium, ameliorating systemic LPS-induced inflammation as well as inflammatory bowel disease. Hence, cells acquired mitochondria during evolution to profit from oxidative metabolism, but also built in an autophagy-based ROS-induced protective mechanism to guard against oxidative stress associated with OXPHOS function during quiescence. Abbreviations : AMPK: AMP-activated protein kinase; AOX: alternative oxidase; Baf A: bafilomycin A
1 ; CI, respiratory complexes I; DCF-DA: 2',7'-dichlordihydrofluorescein diacetate; DHE: dihydroethidium; DSS: dextran sodium sulfate; ΔΨmi: mitochondrial inner membrane potential; EdU: 5-ethynyl-2'-deoxyuridine; ETC: electron transport chain; FA: formaldehyde; HUVEC; human umbilical cord endothelial cells; IBD: inflammatory bowel disease; LC3B: microtubule associated protein 1 light chain 3 beta; LPS: lipopolysaccharide; MEFs: mouse embryonic fibroblasts; MTORC1: mechanistic target of rapamycin kinase complex 1; mtDNA: mitochondrial DNA; NAC: N-acetyl cysteine; OXPHOS: oxidative phosphorylation; PCs: proliferating cells; PE: phosphatidylethanolamine; PEITC: phenethyl isothiocyanate; QCs: quiescent cells; ROS: reactive oxygen species; PLA2: phospholipase A2, WB: western blot.- Published
- 2022
- Full Text
- View/download PDF
14. Identification of vascular cues contributing to cancer cell stemness and function.
- Author
-
Kumar S, Bar-Lev L, Sharife H, Grunewald M, Mogilevsky M, Licht T, Goveia J, Taverna F, Paldor I, Carmeliet P, and Keshet E
- Subjects
- Cell Line, Tumor, Cues, Humans, Neoplastic Stem Cells pathology, Brain Neoplasms pathology, Glioblastoma pathology, Glioma blood supply, Glioma genetics
- Abstract
Glioblastoma stem cells (GSCs) reside close to blood vessels (BVs) but vascular cues contributing to GSC stemness and the nature of GSC-BVs cross talk are not fully understood. Here, we dissected vascular cues influencing GSC gene expression and function to perfusion-based vascular cues, as well as to those requiring direct GSC-endothelial cell (EC) contacts. In light of our previous finding that perivascular tumor cells are metabolically different from tumor cells residing further downstream, cancer cells residing within a narrow, < 60 µm wide perivascular niche were isolated and confirmed to possess a superior tumor-initiation potential compared with those residing further downstream. To circumvent reliance on marker expression, perivascular GSCs were isolated from the respective locales based on their relative state of quiescence. Combined use of these procedures uncovered a large number of previously unrecognized differentially expressed GSC genes. We show that the unique metabolic milieu of the perivascular niche dominated by the highly restricted zone of mTOR activity is conducive for acquisition of GSC properties, primarily in the regulation of genes implicated in cell cycle control. A complementary role of vascular cues including those requiring direct glioma/EC contacts was revealed using glioma/EC co-cultures. Outstanding in the group of glioma cells impacted by nearby ECs were multiple genes responsible for maintaining GSCs in an undifferentiated state, a large fraction of which also relied on Notch-mediated signaling. Glioma-EC communication was found to be bidirectional, evidenced by extensive Notch-mediated EC reprogramming by contacting tumor cells, primarily metabolic EC reprogramming., (© 2022. The Author(s), under exclusive licence to Springer Nature B.V.)
- Published
- 2022
- Full Text
- View/download PDF
15. Comprehensive characterization of the prostate tumor microenvironment identifies CXCR4/CXCL12 crosstalk as a novel antiangiogenic therapeutic target in prostate cancer.
- Author
-
Heidegger I, Fotakis G, Offermann A, Goveia J, Daum S, Salcher S, Noureen A, Timmer-Bosscha H, Schäfer G, Walenkamp A, Perner S, Beatovic A, Moisse M, Plattner C, Krogsdam A, Haybaeck J, Sopper S, Thaler S, Keller MA, Klocker H, Trajanoski Z, Wolf D, and Pircher A
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation, Chemokine CXCL12 genetics, Chemokine CXCL12 metabolism, Endothelial Cells metabolism, Humans, Male, Mice, Receptors, CXCR4 genetics, Receptors, CXCR4 metabolism, Receptors, Epoprostenol, Tumor Microenvironment, Prostate metabolism, Prostatic Neoplasms drug therapy, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism
- Abstract
Background: Crosstalk between neoplastic and stromal cells fosters prostate cancer (PCa) progression and dissemination. Insight in cell-to-cell communication networks provides new therapeutic avenues to mold processes that contribute to PCa tumor microenvironment (TME) alterations. Here we performed a detailed characterization of PCa tumor endothelial cells (TEC) to delineate intercellular crosstalk between TEC and the PCa TME., Methods: TEC isolated from 67 fresh radical prostatectomy (RP) specimens underwent multi-omic ex vivo characterization as well as orthogonal validation of both TEC functions and key markers by immunohistochemistry (IHC) and immunofluorescence (IF). To identify cell-cell interaction targets in TEC, we performed single-cell RNA sequencing (scRNA-seq) in four PCa patients who underwent a RP to catalogue cellular TME composition. Targets were cross-validated using IHC, publicly available datasets, cell culture expriments as well as a PCa xenograft mouse model., Results: Compared to adjacent normal endothelial cells (NEC) bulk RNA-seq analysis revealed upregulation of genes associated with tumor vasculature, collagen modification and extracellular matrix remodeling in TEC. PTGIR, PLAC9, CXCL12 and VDR were identified as TEC markers and confirmed by IF and IHC in an independent patient cohort. By scRNA-seq we identified 27 cell (sub)types, including endothelial cells (EC) with arterial, venous and immature signatures, as well as angiogenic tip EC. A focused molecular analysis revealed that arterial TEC displayed highest CXCL12 mRNA expression levels when compared to all other TME cell (sub)populations and showed a negative prognostic role. Receptor-ligand interaction analysis predicted interactions between arterial TEC derived CXCL12 and its cognate receptor CXCR4 on angiogenic tip EC. CXCL12 was in vitro and in vivo validated as actionable TEC target by highlighting the vessel number- and density- reducing activity of the CXCR4-inhibitor AMD3100 in murine PCa as well as by inhibition of TEC proliferation and migration in vitro., Conclusions: Overall, our comprehensive analysis identified novel PCa TEC targets and highlights CXCR4/CXCL12 interaction as a potential novel target to interfere with tumor angiogenesis in PCa., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
16. Protocols for endothelial cell isolation from mouse tissues: brain, choroid, lung, and muscle.
- Author
-
Conchinha NV, Sokol L, Teuwen LA, Veys K, Dumas SJ, Meta E, García-Caballero M, Geldhof V, Chen R, Treps L, Borri M, de Zeeuw P, Falkenberg KD, Dubois C, Parys M, de Rooij LPMH, Rohlenova K, Goveia J, Schoonjans L, Dewerchin M, Eelen G, Li X, Kalucka J, and Carmeliet P
- Subjects
- Animals, Flow Cytometry methods, Male, Mice, Mice, Inbred C57BL, Brain cytology, Choroid cytology, Endothelial Cells cytology, Lung cytology, Muscles cytology
- Abstract
Endothelial cells (ECs) harbor distinct phenotypical and functional characteristics depending on their tissue localization and contribute to brain, eye, lung, and muscle diseases such as dementia, macular degeneration, pulmonary hypertension, and sarcopenia. To study their function, isolation of pure ECs in high quantities is crucial. Here, we describe protocols for rapid and reproducible blood vessel EC purification established for scRNA sequencing from murine tissues using mechanical and enzymatic digestion followed by magnetic and fluorescence-activated cell sorting. For complete details on the use and execution of these protocol, please refer to Kalucka et al. (2020), Rohlenova et al. (2020), and Goveia et al. (2020)., Competing Interests: The authors declare no competing interests., (© 2021 The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
17. Protocols for endothelial cell isolation from mouse tissues: kidney, spleen, and testis.
- Author
-
Dumas SJ, Meta E, Conchinha NV, Sokol L, Chen R, Borri M, Teuwen LA, Veys K, García-Caballero M, Geldhof V, Treps L, de Zeeuw P, Falkenberg KD, Dubois C, Parys M, de Rooij LPMH, Rohlenova K, Goveia J, Schoonjans L, Dewerchin M, Eelen G, Li X, Kalucka J, and Carmeliet P
- Subjects
- Animals, Flow Cytometry, Male, Mice, Endothelial Cells cytology, Kidney cytology, Spleen cytology, Testis cytology
- Abstract
Endothelial cells (ECs) exhibit phenotypic and functional tissue specificities, critical for studies in the vascular field and beyond. Thus, tissue-specific methods for isolation of highly purified ECs are necessary. Kidney, spleen, and testis ECs are relevant players in health and diseases such as chronic kidney disease, acute kidney injury, myelofibrosis, and cancer. Here, we provide tailored protocols for rapid and reproducible EC purification established for scRNA sequencing from these adult murine tissues using the combination of magnetic- and fluorescence-activated cell sorting. For complete details on the use and execution of these protocols, please refer to Kalucka et al. (2020) and Dumas et al. (2020)., Competing Interests: The authors declare no competing interests., (© 2021 The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
18. Protocols for endothelial cell isolation from mouse tissues: small intestine, colon, heart, and liver.
- Author
-
Sokol L, Geldhof V, García-Caballero M, Conchinha NV, Dumas SJ, Meta E, Teuwen LA, Veys K, Chen R, Treps L, Borri M, de Zeeuw P, Falkenberg KD, Dubois C, Parys M, de Rooij LPMH, Goveia J, Rohlenova K, Schoonjans L, Dewerchin M, Eelen G, Li X, Kalucka J, and Carmeliet P
- Subjects
- Animals, Cells, Cultured, Male, Mice, Mice, Inbred C57BL, Endothelial Cells cytology, Flow Cytometry methods, Intestines cytology, Liver cytology, Myocardium cytology
- Abstract
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)., Competing Interests: The authors declare no competing interests., (© 2021 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
19. Laboratory information system requirements to manage the COVID-19 pandemic: A report from the Belgian national reference testing center.
- Author
-
Weemaes M, Martens S, Cuypers L, Van Elslande J, Hoet K, Welkenhuysen J, Goossens R, Wouters S, Houben E, Jeuris K, Laenen L, Bruyninckx K, Beuselinck K, André E, Depypere M, Desmet S, Lagrou K, Van Ranst M, Verdonck AKLC, and Goveia J
- Subjects
- Academic Medical Centers, Belgium, Betacoronavirus, COVID-19, COVID-19 Testing, Change Management, Evidence-Based Medicine, Humans, Meaningful Use, Pandemics, SARS-CoV-2, Clinical Laboratory Information Systems, Clinical Laboratory Techniques, Coronavirus Infections diagnosis, Laboratories, Hospital organization & administration, Medical Order Entry Systems, Pneumonia, Viral diagnosis
- Abstract
Objective: The study sought to describe the development, implementation, and requirements of laboratory information system (LIS) functionality to manage test ordering, registration, sample flow, and result reporting during the coronavirus disease 2019 (COVID-19) pandemic., Materials and Methods: Our large (>12 000 000 tests/y) academic hospital laboratory is the Belgian National Reference Center for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) testing. We have performed a moving total of >25 000 SARS-CoV-2 polymerase chain reaction tests in parallel to standard routine testing since the start of the outbreak. A LIS implementation team dedicated to develop tools to remove the bottlenecks, primarily situated in the pre- and postanalytical phases, was established early in the crisis., Results: We outline the design, implementation, and requirements of LIS functionality related to managing increased test demand during the COVID-19 crisis, including tools for test ordering, standardized order sets integrated into a computerized provider order entry module, notifications on shipping requirements, automated triaging based on digital metadata forms, and the establishment of databases with contact details of other laboratories and primary care physicians to enable automated reporting. We also describe our approach to data mining and reporting of actionable daily summary statistics to governing bodies and other policymakers., Conclusions: Rapidly developed, agile extendable LIS functionality and its meaningful use alleviates the administrative burden on laboratory personnel and improves turnaround time of SARS-CoV-2 testing. It will be important to maintain an environment that is conducive for the rapid adoption of meaningful LIS tools after the COVID-19 crisis., (© The Author(s) 2020. Published by Oxford University Press on behalf of the American Medical Informatics Association.)
- Published
- 2020
- Full Text
- View/download PDF
20. Heterogeneous Effects of Calorie Content and Nutritional Components Underlie Dietary Influence on Pancreatic Cancer Susceptibility.
- Author
-
Dooley J, Lagou V, Goveia J, Ulrich A, Rohlenova K, Heirman N, Karakach T, Lampi Y, Khan S, Wang J, Dresselaers T, Himmelreich U, Gunter MJ, Prokopenko I, Carmeliet P, and Liston A
- Subjects
- Aged, Animals, Cell Cycle, Dietary Carbohydrates, Dietary Fats, Dietary Proteins, Female, Gene-Environment Interaction, Humans, Male, Mice, Inbred C57BL, Middle Aged, Obesity, Diet, Disease Susceptibility, Energy Intake, Nutritional Physiological Phenomena, Pancreatic Neoplasms pathology
- Abstract
Pancreatic cancer is a rare but fatal form of cancer, the fourth highest in absolute mortality. Known risk factors include obesity, diet, and type 2 diabetes; however, the low incidence rate and interconnection of these factors confound the isolation of individual effects. Here, we use epidemiological analysis of prospective human cohorts and parallel tracking of pancreatic cancer in mice to dissect the effects of obesity, diet, and diabetes on pancreatic cancer. Through longitudinal monitoring and multi-omics analysis in mice, we found distinct effects of protein, sugar, and fat dietary components, with dietary sugars increasing Mad2l1 expression and tumor proliferation. Using epidemiological approaches in humans, we find that dietary sugars give a MAD2L1 genotype-dependent increased susceptibility to pancreatic cancer. The translation of these results to a clinical setting could aid in the identification of the at-risk population for screening and potentially harness dietary modification as a therapeutic measure., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
21. BIOMEX: an interactive workflow for (single cell) omics data interpretation and visualization.
- Author
-
Taverna F, Goveia J, Karakach TK, Khan S, Rohlenova K, Treps L, Subramanian A, Schoonjans L, Dewerchin M, Eelen G, and Carmeliet P
- Subjects
- Algorithms, Bile Duct Neoplasms genetics, Cholangiocarcinoma genetics, Computer Graphics, Endothelial Cells metabolism, Humans, Metabolomics methods, Neoplasms mortality, Proteomics methods, Survival Analysis, Workflow, Gene Expression Profiling methods, Single-Cell Analysis methods, Software
- Abstract
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., (© The Author(s) 2020. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2020
- Full Text
- View/download PDF
22. Single-Cell RNA Sequencing Maps Endothelial Metabolic Plasticity in Pathological Angiogenesis.
- Author
-
Rohlenova K, Goveia J, García-Caballero M, Subramanian A, Kalucka J, Treps L, Falkenberg KD, de Rooij LPMH, Zheng Y, Lin L, Sokol L, Teuwen LA, Geldhof V, Taverna F, Pircher A, Conradi LC, Khan S, Stegen S, Panovska D, De Smet F, Staal FJT, Mclaughlin RJ, Vinckier S, Van Bergen T, Ectors N, De Haes P, Wang J, Bolund L, Schoonjans L, Karakach TK, Yang H, Carmeliet G, Liu Y, Thienpont B, Dewerchin M, Eelen G, Li X, Luo Y, and Carmeliet P
- Subjects
- Animals, Endothelial Cells cytology, Endothelial Cells pathology, HEK293 Cells, Human Umbilical Vein Endothelial Cells, Humans, Male, Mice, Mice, Inbred C57BL, Sequence Analysis, RNA, Single-Cell Analysis, Endothelial Cells metabolism, Lung Neoplasms metabolism, Macular Degeneration metabolism, Neovascularization, Pathologic metabolism, Transcriptome
- Abstract
Endothelial cell (EC) metabolism is an emerging target for anti-angiogenic therapy in tumor angiogenesis and choroidal neovascularization (CNV), but little is known about individual EC metabolic transcriptomes. By single-cell RNA sequencing 28,337 murine choroidal ECs (CECs) and sprouting CNV-ECs, we constructed a taxonomy to characterize their heterogeneity. Comparison with murine lung tumor ECs (TECs) revealed congruent marker gene expression by distinct EC phenotypes across tissues and diseases, suggesting similar angiogenic mechanisms. Trajectory inference predicted that differentiation of venous to angiogenic ECs was accompanied by metabolic transcriptome plasticity. ECs displayed metabolic transcriptome heterogeneity during cell-cycle progression and in quiescence. Hypothesizing that conserved genes are important, we used an integrated analysis, based on congruent transcriptome analysis, CEC-tailored genome-scale metabolic modeling, and gene expression meta-analysis in cross-species datasets, followed by in vitro and in vivo validation, to identify SQLE and ALDH18A1 as previously unknown metabolic angiogenic targets., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
23. An Integrated Gene Expression Landscape Profiling Approach to Identify Lung Tumor Endothelial Cell Heterogeneity and Angiogenic Candidates.
- Author
-
Goveia J, Rohlenova K, Taverna F, Treps L, Conradi LC, Pircher A, Geldhof V, de Rooij LPMH, Kalucka J, Sokol L, García-Caballero M, Zheng Y, Qian J, Teuwen LA, Khan S, Boeckx B, Wauters E, Decaluwé H, De Leyn P, Vansteenkiste J, Weynand B, Sagaert X, Verbeken E, Wolthuis A, Topal B, Everaerts W, Bohnenberger H, Emmert A, Panovska D, De Smet F, Staal FJT, Mclaughlin RJ, Impens F, Lagani V, Vinckier S, Mazzone M, Schoonjans L, Dewerchin M, Eelen G, Karakach TK, Yang H, Wang J, Bolund L, Lin L, Thienpont B, Li X, Lambrechts D, Luo Y, and Carmeliet P
- Published
- 2020
- Full Text
- View/download PDF
24. Single-Cell Transcriptome Atlas of Murine Endothelial Cells.
- Author
-
Kalucka J, de Rooij LPMH, Goveia J, Rohlenova K, Dumas SJ, Meta E, Conchinha NV, Taverna F, Teuwen LA, Veys K, García-Caballero M, Khan S, Geldhof V, Sokol L, Chen R, Treps L, Borri M, de Zeeuw P, Dubois C, Karakach TK, Falkenberg KD, Parys M, Yin X, Vinckier S, Du Y, Fenton RA, Schoonjans L, Dewerchin M, Eelen G, Thienpont B, Lin L, Bolund L, Li X, Luo Y, and Carmeliet P
- Subjects
- Animals, Brain cytology, Cardiovascular System cytology, Endothelial Cells classification, Endothelial Cells cytology, Gastrointestinal Tract cytology, Male, Mice, Mice, Inbred C57BL, Muscles cytology, Organ Specificity, RNA-Seq, Testis cytology, Endothelial Cells metabolism, Single-Cell Analysis, Transcriptome
- Abstract
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., Competing Interests: Declaration of Interests None of the authors have competing financial interests to declare., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
25. Single-Cell RNA Sequencing Reveals Renal Endothelium Heterogeneity and Metabolic Adaptation to Water Deprivation.
- Author
-
Dumas SJ, Meta E, Borri M, Goveia J, Rohlenova K, Conchinha NV, Falkenberg K, Teuwen LA, de Rooij L, Kalucka J, Chen R, Khan S, Taverna F, Lu W, Parys M, De Legher C, Vinckier S, Karakach TK, Schoonjans L, Lin L, Bolund L, Dewerchin M, Eelen G, Rabelink TJ, Li X, Luo Y, and Carmeliet P
- Subjects
- Animals, Endothelial Cells physiology, Male, Mice, Mice, Inbred C57BL, Phenotype, Adaptation, Physiological genetics, Endothelial Cells metabolism, Kidney cytology, Sequence Analysis, RNA, Water Deprivation physiology
- Abstract
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., (Copyright © 2020 by the American Society of Nephrology.)
- Published
- 2020
- Full Text
- View/download PDF
26. EndoDB: a database of endothelial cell transcriptomics data.
- Author
-
Khan S, Taverna F, Rohlenova K, Treps L, Geldhof V, de Rooij L, Sokol L, Pircher A, Conradi LC, Kalucka J, Schoonjans L, Eelen G, Dewerchin M, Karakach T, Li X, Goveia J, and Carmeliet P
- Subjects
- Animals, Endothelial Cells metabolism, Gene Expression Regulation genetics, Humans, Principal Component Analysis, Computational Biology, Databases, Genetic, Transcriptome genetics
- Abstract
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.
- Published
- 2019
- Full Text
- View/download PDF
27. Impairment of Angiogenesis by Fatty Acid Synthase Inhibition Involves mTOR Malonylation.
- Author
-
Bruning U, Morales-Rodriguez F, Kalucka J, Goveia J, Taverna F, Queiroz KCS, Dubois C, Cantelmo AR, Chen R, Loroch S, Timmerman E, Caixeta V, Bloch K, Conradi LC, Treps L, Staes A, Gevaert K, Tee A, Dewerchin M, Semenkovich CF, Impens F, Schilling B, Verdin E, Swinnen JV, Meier JL, Kulkarni RA, Sickmann A, Ghesquière B, Schoonjans L, Li X, Mazzone M, and Carmeliet P
- Subjects
- Acetyl-CoA Carboxylase antagonists & inhibitors, Animals, Cell Line, Tumor, Cell Proliferation, Fatty Acid Synthase, Type I antagonists & inhibitors, Fatty Acid Synthase, Type I genetics, Human Umbilical Vein Endothelial Cells cytology, Humans, Mechanistic Target of Rapamycin Complex 1 metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Orlistat therapeutic use, Protein Processing, Post-Translational, Retinal Neovascularization drug therapy, Fatty Acid Synthase, Type I physiology, Human Umbilical Vein Endothelial Cells metabolism, Malonyl Coenzyme A metabolism, Retinal Neovascularization pathology, TOR Serine-Threonine Kinases metabolism
- Abstract
The role of fatty acid synthesis in endothelial cells (ECs) remains incompletely characterized. We report that fatty acid synthase knockdown (FASN
KD ) in ECs impedes vessel sprouting by reducing proliferation. Endothelial loss of FASN impaired angiogenesis in vivo, while FASN blockade reduced pathological ocular neovascularization, at >10-fold lower doses than used for anti-cancer treatment. Impaired angiogenesis was not due to energy stress, redox imbalance, or palmitate depletion. Rather, FASNKD elevated malonyl-CoA levels, causing malonylation (a post-translational modification) of mTOR at lysine 1218 (K1218). mTOR K-1218 malonylation impaired mTOR complex 1 (mTORC1) kinase activity, thereby reducing phosphorylation of downstream targets (p70S6K/4EBP1). Silencing acetyl-CoA carboxylase 1 (an enzyme producing malonyl-CoA) normalized malonyl-CoA levels and reactivated mTOR in FASNKD ECs. Mutagenesis unveiled the importance of mTOR K1218 malonylation for angiogenesis. This study unveils a novel role of FASN in metabolite signaling that contributes to explaining the anti-angiogenic effect of FASN blockade., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
28. Quiescent Endothelial Cells Upregulate Fatty Acid β-Oxidation for Vasculoprotection via Redox Homeostasis.
- Author
-
Kalucka J, Bierhansl L, Conchinha NV, Missiaen R, Elia I, Brüning U, Scheinok S, Treps L, Cantelmo AR, Dubois C, de Zeeuw P, Goveia J, Zecchin A, Taverna F, Morales-Rodriguez F, Brajic A, Conradi LC, Schoors S, Harjes U, Vriens K, Pilz GA, Chen R, Cubbon R, Thienpont B, Cruys B, Wong BW, Ghesquière B, Dewerchin M, De Bock K, Sagaert X, Jessberger S, Jones EAV, Gallez B, Lambrechts D, Mazzone M, Eelen G, Li X, Fendt SM, and Carmeliet P
- Subjects
- Animals, Cell Proliferation, HEK293 Cells, Homeostasis, Humans, Mice, Mice, Inbred C57BL, Oxidation-Reduction, Oxidative Stress, Carnitine O-Palmitoyltransferase metabolism, Energy Metabolism, Fatty Acids metabolism, Human Umbilical Vein Endothelial Cells metabolism, NADP metabolism, Receptor, Notch1 metabolism
- Abstract
Little is known about the metabolism of quiescent endothelial cells (QECs). Nonetheless, when dysfunctional, QECs contribute to multiple diseases. Previously, we demonstrated that proliferating endothelial cells (PECs) use fatty acid β-oxidation (FAO) for de novo dNTP synthesis. We report now that QECs are not hypometabolic, but upregulate FAO >3-fold higher than PECs, not to support biomass or energy production but to sustain the tricarboxylic acid cycle for redox homeostasis through NADPH regeneration. Hence, endothelial loss of FAO-controlling CPT1A in CPT1A
ΔEC mice promotes EC dysfunction (leukocyte infiltration, barrier disruption) by increasing endothelial oxidative stress, rendering CPT1AΔEC mice more susceptible to LPS and inflammatory bowel disease. Mechanistically, Notch1 orchestrates the use of FAO for redox balance in QECs. Supplementation of acetate (metabolized to acetyl-coenzyme A) restores endothelial quiescence and counters oxidative stress-mediated EC dysfunction in CPT1AΔEC mice, offering therapeutic opportunities. Thus, QECs use FAO for vasculoprotection against oxidative stress-prone exposure., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
29. Serine Synthesis via PHGDH Is Essential for Heme Production in Endothelial Cells.
- Author
-
Vandekeere S, Dubois C, Kalucka J, Sullivan MR, García-Caballero M, Goveia J, Chen R, Diehl FF, Bar-Lev L, Souffreau J, Pircher A, Kumar S, Vinckier S, Hirabayashi Y, Furuya S, Schoonjans L, Eelen G, Ghesquière B, Keshet E, Li X, Vander Heiden MG, Dewerchin M, and Carmeliet P
- Subjects
- Apoptosis, Carbohydrate Metabolism, Inborn Errors metabolism, Cell Line, Tumor, Cell Proliferation, Cell Survival, Dietary Supplements, Gene Knockdown Techniques, Hemin metabolism, Human Umbilical Vein Endothelial Cells, Humans, Microcephaly metabolism, Mitochondria metabolism, Mitophagy, Neovascularization, Physiologic, Oxidative Stress, Phosphoglycerate Dehydrogenase deficiency, Protein Biosynthesis, Psychomotor Disorders metabolism, Purines metabolism, Reactive Oxygen Species metabolism, Seizures metabolism, Endothelial Cells metabolism, Heme metabolism, Phosphoglycerate Dehydrogenase genetics, Phosphoglycerate Dehydrogenase metabolism, Serine metabolism
- Abstract
The role of phosphoglycerate dehydrogenase (PHGDH), a key enzyme of the serine synthesis pathway (SSP), in endothelial cells (ECs) remains poorly characterized. We report that mouse neonates with EC-specific PHGDH deficiency suffer lethal vascular defects within days of gene inactivation, due to reduced EC proliferation and survival. In addition to nucleotide synthesis impairment, PHGDH knockdown (PHGDH
KD ) caused oxidative stress, due not only to decreased glutathione and NADPH synthesis but also to mitochondrial dysfunction. Electron transport chain (ETC) enzyme activities were compromised upon PHGDHKD because of insufficient heme production due to cellular serine depletion, not observed in other cell types. As a result of heme depletion, elevated reactive oxygen species levels caused EC demise. Supplementation of hemin in PHGDHKD ECs restored ETC function and rescued the apoptosis and angiogenesis defects. These data argue that ECs die upon PHGDH inhibition, even without external serine deprivation, illustrating an unusual importance of serine synthesis for ECs., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
30. Role of glutamine synthetase in angiogenesis beyond glutamine synthesis.
- Author
-
Eelen G, Dubois C, Cantelmo AR, Goveia J, Brüning U, DeRan M, Jarugumilli G, van Rijssel J, Saladino G, Comitani F, Zecchin A, Rocha S, Chen R, Huang H, Vandekeere S, Kalucka J, Lange C, Morales-Rodriguez F, Cruys B, Treps L, Ramer L, Vinckier S, Brepoels K, Wyns S, Souffreau J, Schoonjans L, Lamers WH, Wu Y, Haustraete J, Hofkens J, Liekens S, Cubbon R, Ghesquière B, Dewerchin M, Gervasio FL, Li X, van Buul JD, Wu X, and Carmeliet P
- Subjects
- Actins metabolism, Animals, Cell Movement, Endothelial Cells metabolism, Female, Glutamate-Ammonia Ligase deficiency, Glutamate-Ammonia Ligase genetics, Glutamate-Ammonia Ligase physiology, HEK293 Cells, Human Umbilical Vein Endothelial Cells cytology, Human Umbilical Vein Endothelial Cells enzymology, Human Umbilical Vein Endothelial Cells metabolism, Humans, Lipoylation, Mice, Palmitic Acid metabolism, Protein Processing, Post-Translational, Stress Fibers metabolism, rho GTP-Binding Proteins chemistry, rho GTP-Binding Proteins metabolism, rho-Associated Kinases metabolism, Endothelial Cells enzymology, Endothelial Cells pathology, Glutamate-Ammonia Ligase metabolism, Glutamine biosynthesis, Neovascularization, Pathologic
- Abstract
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.
- Published
- 2018
- Full Text
- View/download PDF
31. Role of glutamine and interlinked asparagine metabolism in vessel formation.
- Author
-
Huang H, Vandekeere S, Kalucka J, Bierhansl L, Zecchin A, Brüning U, Visnagri A, Yuldasheva N, Goveia J, Cruys B, Brepoels K, Wyns S, Rayport S, Ghesquière B, Vinckier S, Schoonjans L, Cubbon R, Dewerchin M, Eelen G, and Carmeliet P
- Subjects
- Culture Media chemistry, Endothelial Cells metabolism, Glutaminase metabolism, Human Umbilical Vein Endothelial Cells, Humans, Metabolic Networks and Pathways, Neovascularization, Pathologic, Asparagine metabolism, Cell Movement drug effects, Cell Proliferation drug effects, Endothelial Cells drug effects, Endothelial Cells physiology, Glutamine metabolism, Neovascularization, Physiologic drug effects
- Abstract
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., (© 2017 The Authors.)
- Published
- 2017
- Full Text
- View/download PDF
32. Hypoxia determines survival outcomes of bacterial infection through HIF-1alpha dependent re-programming of leukocyte metabolism.
- Author
-
Thompson AA, Dickinson RS, Murphy F, Thomson JP, Marriott HM, Tavares A, Willson J, Williams L, Lewis A, Mirchandani A, Dos Santos Coelho P, Doherty C, Ryan E, Watts E, Morton NM, Forbes S, Stimson RH, Hameed AG, Arnold N, Preston JA, Lawrie A, Finisguerra V, Mazzone M, Sadiku P, Goveia J, Taverna F, Carmeliet P, Foster SJ, Chilvers ER, Cowburn AS, Dockrell DH, Johnson RS, Meehan RR, Whyte MK, and Walmsley SR
- Abstract
Hypoxia and bacterial infection frequently co-exist, in both acute and chronic clinical settings, and typically result in adverse clinical outcomes. To ameliorate this morbidity, we investigated the interaction between hypoxia and the host response. In the context of acute hypoxia, both S. aureus and S. pneumoniae infections rapidly induced progressive neutrophil mediated morbidity and mortality, with associated hypothermia and cardiovascular compromise. Preconditioning animals through longer exposures to hypoxia, prior to infection, prevented these pathophysiological responses and profoundly dampened the transcriptome of circulating leukocytes. Specifically, perturbation of HIF pathway and glycolysis genes by hypoxic preconditioning was associated with reduced leukocyte glucose utilisation, resulting in systemic rescue from a global negative energy state and myocardial protection. Thus we demonstrate that hypoxia preconditions the innate immune response and determines survival outcomes following bacterial infection through suppression of HIF-1α and neutrophil metabolism. The therapeutic implications of this work are that in the context of systemic or tissue hypoxia therapies that target the host response could improve infection associated morbidity and mortality., Competing Interests: Competing Financial Interests The authors declare no competing financial interests.
- Published
- 2017
- Full Text
- View/download PDF
33. The role of fatty acid β-oxidation in lymphangiogenesis.
- Author
-
Wong BW, Wang X, Zecchin A, Thienpont B, Cornelissen I, Kalucka J, García-Caballero M, Missiaen R, Huang H, Brüning U, Blacher S, Vinckier S, Goveia J, Knobloch M, Zhao H, Dierkes C, Shi C, Hägerling R, Moral-Dardé V, Wyns S, Lippens M, Jessberger S, Fendt SM, Luttun A, Noel A, Kiefer F, Ghesquière B, Moons L, Schoonjans L, Dewerchin M, Eelen G, Lambrechts D, and Carmeliet P
- Subjects
- Acetates pharmacology, Acetyl Coenzyme A metabolism, Acetylation drug effects, Animals, Carnitine O-Palmitoyltransferase antagonists & inhibitors, Carnitine O-Palmitoyltransferase genetics, Carnitine O-Palmitoyltransferase metabolism, Cell Differentiation drug effects, Cell Differentiation genetics, Endothelial Cells cytology, Endothelial Cells drug effects, Endothelial Cells metabolism, Epigenesis, Genetic, Female, Histones metabolism, Homeodomain Proteins metabolism, Human Umbilical Vein Endothelial Cells, Humans, Lymphatic Vessels drug effects, Mice, Mice, Inbred C57BL, Oxidation-Reduction drug effects, Protein Biosynthesis, Transcription, Genetic, Tumor Suppressor Proteins metabolism, Umbilical Arteries cytology, Up-Regulation, Fatty Acids chemistry, Fatty Acids metabolism, Lymphangiogenesis drug effects, Lymphangiogenesis genetics, Lymphatic Vessels cytology, Lymphatic Vessels metabolism
- Abstract
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 β-oxidation, impairs lymphatic development. LECs use fatty acid β-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 β-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.
- Published
- 2017
- Full Text
- View/download PDF
34. Inhibition of the Glycolytic Activator PFKFB3 in Endothelium Induces Tumor Vessel Normalization, Impairs Metastasis, and Improves Chemotherapy.
- Author
-
Cantelmo AR, Conradi LC, Brajic A, Goveia J, Kalucka J, Pircher A, Chaturvedi P, Hol J, Thienpont B, Teuwen LA, Schoors S, Boeckx B, Vriens J, Kuchnio A, Veys K, Cruys B, Finotto L, Treps L, Stav-Noraas TE, Bifari F, Stapor P, Decimo I, Kampen K, De Bock K, Haraldsen G, Schoonjans L, Rabelink T, Eelen G, Ghesquière B, Rehman J, Lambrechts D, Malik AB, Dewerchin M, and Carmeliet P
- Subjects
- Animals, Cadherins genetics, Cell Line, Tumor, Cell Movement drug effects, Cisplatin pharmacology, Drug Synergism, Drug Therapy, Epithelial Cells pathology, Gene Expression Regulation, Neoplastic drug effects, Glycolysis drug effects, Human Umbilical Vein Endothelial Cells, Humans, Mice, Neoplasm Invasiveness, Neoplasm Metastasis, Neoplasm Transplantation, Neoplasms blood supply, Neoplasms drug therapy, Tamoxifen pharmacology, Cisplatin administration & dosage, Epithelial Cells metabolism, Neoplasms metabolism, Phosphofructokinase-2 antagonists & inhibitors, Tamoxifen administration & dosage
- Abstract
Abnormal tumor vessels promote metastasis and impair chemotherapy. Hence, tumor vessel normalization (TVN) is emerging as an anti-cancer treatment. Here, we show that tumor endothelial cells (ECs) have a hyper-glycolytic metabolism, shunting intermediates to nucleotide synthesis. EC haplo-deficiency or blockade of the glycolytic activator PFKFB3 did not affect tumor growth, but reduced cancer cell invasion, intravasation, and metastasis by normalizing tumor vessels, which improved vessel maturation and perfusion. Mechanistically, PFKFB3 inhibition tightened the vascular barrier by reducing VE-cadherin endocytosis in ECs, and rendering pericytes more quiescent and adhesive (via upregulation of N-cadherin) through glycolysis reduction; it also lowered the expression of cancer cell adhesion molecules in ECs by decreasing NF-κB signaling. PFKFB3-blockade treatment also improved chemotherapy of primary and metastatic tumors., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
35. Macrophage Metabolism Controls Tumor Blood Vessel Morphogenesis and Metastasis.
- Author
-
Wenes M, Shang M, Di Matteo M, Goveia J, Martín-Pérez R, Serneels J, Prenen H, Ghesquière B, Carmeliet P, and Mazzone M
- Subjects
- Animals, Blood Vessels metabolism, Cell Hypoxia, Disease Models, Animal, Endothelial Cells metabolism, Gene Deletion, Glucose metabolism, Glycolysis, Mice, Inbred C57BL, Mice, Knockout, Neoplasm Metastasis, Neoplasms pathology, Signal Transduction, TOR Serine-Threonine Kinases metabolism, Transcription Factors metabolism, Up-Regulation, Blood Vessels growth & development, Macrophages metabolism, Morphogenesis, Neoplasms blood supply, Neoplasms metabolism, Neovascularization, Pathologic metabolism
- Abstract
Hypoxic tumor-associated macrophages (TAMs) acquire angiogenic and immunosuppressive properties. Yet it remains unknown if metabolic changes influence these functions. Here, we argue that hypoxic TAMs strongly upregulate the expression of REDD1, a negative regulator of mTOR. REDD1-mediated mTOR inhibition hinders glycolysis in TAMs and curtails their excessive angiogenic response, with consequent formation of abnormal blood vessels. Accordingly, REDD1 deficiency in TAMs leads to the formation of smoothly aligned, pericyte-covered, functional vessels, which prevents vessel leakiness, hypoxia, and metastases. Mechanistically, highly glycolytic REDD1-deficient TAMs outcompete endothelial cells for glucose usage that thwarts vascular hyperactivation and promotes the formation of quiescent vascular junctions. Tuning down glycolysis in REDD1 knockout TAMs re-establishes abnormal angiogenesis and metastases. On this basis, we prove that the anti-tumor effect of mTOR inhibitors is partly countered by the deleterious outcome of these drugs on TAMs. Our data provide a functional link between TAM metabolism and tumor angiogenesis., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
36. Meta-analysis of clinical metabolic profiling studies in cancer: challenges and opportunities.
- Author
-
Goveia J, Pircher A, Conradi LC, Kalucka J, Lagani V, Dewerchin M, Eelen G, DeBerardinis RJ, Wilson ID, and Carmeliet P
- Subjects
- Humans, Metabolomics standards, Retrospective Studies, Metabolome, Metabolomics methods, Neoplasms pathology
- Abstract
Cancer cell metabolism has received increasing attention. Despite a boost in the application of clinical metabolic profiling (CMP) in cancer patients, a meta-analysis has not been performed. The primary goal of this study was to assess whether public accessibility of metabolomics data and identification and reporting of metabolites were sufficient to assess which metabolites were consistently altered in cancer patients. We therefore retrospectively curated data from CMP studies in cancer patients published during 5 recent years and used an established vote-counting method to perform a semiquantitative meta-analysis of metabolites in tumor tissue and blood. This analysis confirmed well-known increases in glycolytic metabolites, but also unveiled unprecedented changes in other metabolites such as ketone bodies and amino acids (histidine, tryptophan). However, this study also highlighted that insufficient public accessibility of metabolomics data, and inadequate metabolite identification and reporting hamper the discovery potential of meta-analyses of CMP studies, calling for improved standardization of metabolomics studies., (© 2016 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2016
- Full Text
- View/download PDF
37. Adequate hypoxia inducible factor 1α signaling is indispensable for bone regeneration.
- Author
-
Stegen S, Deprez S, Eelen G, Torrekens S, Van Looveren R, Goveia J, Ghesquière B, Carmeliet P, and Carmeliet G
- Subjects
- Animals, Cell Hypoxia, Cell Survival, Energy Metabolism, Gene Deletion, Gene Knockdown Techniques, Glycolysis, Human Umbilical Vein Endothelial Cells, Humans, Mice, Inbred C57BL, Neovascularization, Physiologic, Periosteum cytology, Reactive Oxygen Species metabolism, Bone Regeneration, Hypoxia-Inducible Factor 1, alpha Subunit metabolism
- Abstract
Engineered cell-based constructs are an appealing strategy to treat large skeletal defects. However, transplanted cells are often confronted with an environment that is deprived of oxygen and nutrients. Upon hypoxia, most cell types activate hypoxia-inducible factor 1α (HIF-1α) signaling, but its importance for implanted osteoprogenitor cells during bone regeneration is not elucidated. To this end, we specifically deleted the HIF--1α isoform in periosteal progenitor cells and show that activation of HIF-1α signaling in these cells is critical for bone repair by modulating angiogenic and metabolic processes. Activation of HIF-1α is not only crucial for blood vessel invasion, by enhancing angiogenic growth factor production, but also for periosteal cell survival early after implantation, when blood vessels have not yet invaded the construct. HIF-1α signaling limits oxygen consumption to avoid accumulation of harmful ROS and preserve redox balance, and additionally induces a switch to glycolysis to prevent energetic distress. Altogether, our results indicate that the proangiogenic capacity of implanted periosteal cells is HIF-1α regulated and that metabolic adaptations mediate post-implantation cell survival., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
38. Dual inhibition of Ang-2 and VEGF receptors normalizes tumor vasculature and prolongs survival in glioblastoma by altering macrophages.
- Author
-
Peterson TE, Kirkpatrick ND, Huang Y, Farrar CT, Marijt KA, Kloepper J, Datta M, Amoozgar Z, Seano G, Jung K, Kamoun WS, Vardam T, Snuderl M, Goveia J, Chatterjee S, Batista A, Muzikansky A, Leow CC, Xu L, Batchelor TT, Duda DG, Fukumura D, and Jain RK
- Subjects
- Animals, Cell Line, Tumor, Drug Screening Assays, Antitumor, Mice, Antibodies, Neoplasm pharmacology, Glioblastoma drug therapy, Glioblastoma metabolism, Glioblastoma pathology, Macrophages metabolism, Macrophages pathology, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins metabolism, Neoplasms, Experimental drug therapy, Neoplasms, Experimental metabolism, Neoplasms, Experimental pathology, Neovascularization, Pathologic drug therapy, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Quinazolines pharmacology, Receptors, Vascular Endothelial Growth Factor antagonists & inhibitors, Receptors, Vascular Endothelial Growth Factor metabolism, Ribonuclease, Pancreatic antagonists & inhibitors, Ribonuclease, Pancreatic metabolism
- Abstract
Glioblastomas (GBMs) rapidly become refractory to anti-VEGF therapies. We previously demonstrated that ectopic overexpression of angiopoietin-2 (Ang-2) compromises the benefits of anti-VEGF receptor (VEGFR) treatment in murine GBM models and that circulating Ang-2 levels in GBM patients rebound after an initial decrease following cediranib (a pan-VEGFR tyrosine kinase inhibitor) administration. Here we tested whether dual inhibition of VEGFR/Ang-2 could improve survival in two orthotopic models of GBM, Gl261 and U87. Dual therapy using cediranib and MEDI3617 (an anti-Ang-2-neutralizing antibody) improved survival over each therapy alone by delaying Gl261 growth and increasing U87 necrosis, effectively reducing viable tumor burden. Consistent with their vascular-modulating function, the dual therapies enhanced morphological normalization of vessels. Dual therapy also led to changes in tumor-associated macrophages (TAMs). Inhibition of TAM recruitment using an anti-colony-stimulating factor-1 antibody compromised the survival benefit of dual therapy. Thus, dual inhibition of VEGFR/Ang-2 prolongs survival in preclinical GBM models by reducing tumor burden, improving normalization, and altering TAMs. This approach may represent a potential therapeutic strategy to overcome the limitations of anti-VEGFR monotherapy in GBM patients by integrating the complementary effects of anti-Ang2 treatment on vessels and immune cells.
- Published
- 2016
- Full Text
- View/download PDF
39. HIF-1α Promotes Glutamine-Mediated Redox Homeostasis and Glycogen-Dependent Bioenergetics to Support Postimplantation Bone Cell Survival.
- Author
-
Stegen S, van Gastel N, Eelen G, Ghesquière B, D'Anna F, Thienpont B, Goveia J, Torrekens S, Van Looveren R, Luyten FP, Maxwell PH, Wielockx B, Lambrechts D, Fendt SM, Carmeliet P, and Carmeliet G
- Subjects
- Animals, Bone Regeneration, Cell Respiration, Cell Survival, Gene Deletion, Gene Knockdown Techniques, Gene Silencing, Glutaminase metabolism, Mice, Neovascularization, Physiologic, Osteocytes metabolism, Oxidation-Reduction, Oxidative Stress, Periosteum pathology, Reactive Oxygen Species metabolism, Energy Metabolism, Glutamine metabolism, Glycogen metabolism, Homeostasis, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Osteocytes transplantation
- Abstract
Cell-based therapy is a promising strategy in regenerative medicine, but the poor survival rate of the implanted cells remains a major challenge and limits clinical translation. We preconditioned periosteal cells to the hypoxic and ischemic environment of the bone defect site by deleting prolyl hydroxylase domain-containing protein 2 (PHD2), resulting in hypoxia-inducible factor 1 alpha (HIF-1α) stabilization. This strategy increased postimplantation cell survival and improved bone regeneration. The enhanced cell viability was angiogenesis independent but relied on combined changes in glutamine and glycogen metabolism. HIF-1α stabilization stimulated glutaminase-mediated glutathione synthesis, maintaining redox homeostasis at baseline and during oxidative or nutrient stress. Simultaneously, HIF-1α signaling increased glycogen storage, preventing an energy deficit during nutrient or oxygen deprivation. Pharmacological inhibition of PHD2 recapitulated the adaptations in glutamine and glycogen metabolism and, consequently, the beneficial effects on cell survival. Thus, targeting cellular metabolism is an appealing strategy for bone regeneration and cell-based therapy in general., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
40. Corrigendum: Fatty acid carbon is essential for dNTP synthesis in endothelial cells.
- Author
-
Schoors S, Bruning U, Missiaen R, Queiroz KC, Borgers G, Elia I, Zecchin A, Cantelmo AR, Christen S, Goveia J, Heggermont W, Goddë L, Vinckier S, Van Veldhoven PP, Eelen G, Schoonjans L, Gerhardt H, Dewerchin M, Baes M, De Bock K, Ghesquière B, Lunt SY, Fendt SM, and Carmeliet P
- Published
- 2015
- Full Text
- View/download PDF
41. Metabolic pathway compartmentalization: an underappreciated opportunity?
- Author
-
Zecchin A, Stapor PC, Goveia J, and Carmeliet P
- Subjects
- Adenosine Triphosphate biosynthesis, Animals, Cell Movement, Glycolysis, Humans, Mitochondria metabolism, Metabolic Networks and Pathways
- Abstract
For eukaryotic cells to function properly, they divide their intracellular space in subcellular compartments, each harboring specific metabolic activities. In recent years, it has become increasingly clear that compartmentalization of metabolic pathways is a prerequisite for certain cellular functions. This has for instance been documented for cellular migration, which relies on subcellular localization of glycolysis or mitochondrial respiration in a cell type-dependent manner. Although exciting, this field is still in its infancy, partly due to the limited availability of methods to study the directionality of metabolic pathways and to visualize metabolic processes in distinct cellular compartments. Nonetheless, advances in this field may offer opportunities for innovative strategies to target deregulated compartmentalized metabolism in disease., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
42. Fatty acid carbon is essential for dNTP synthesis in endothelial cells.
- Author
-
Schoors S, Bruning U, Missiaen R, Queiroz KC, Borgers G, Elia I, Zecchin A, Cantelmo AR, Christen S, Goveia J, Heggermont W, Goddé L, Vinckier S, Van Veldhoven PP, Eelen G, Schoonjans L, Gerhardt H, Dewerchin M, Baes M, De Bock K, Ghesquière B, Lunt SY, Fendt SM, and Carmeliet P
- Subjects
- Acetic Acid pharmacology, Adenosine Triphosphate metabolism, Animals, Blood Vessels cytology, Blood Vessels drug effects, Blood Vessels metabolism, Blood Vessels pathology, Carnitine O-Palmitoyltransferase antagonists & inhibitors, Carnitine O-Palmitoyltransferase deficiency, Carnitine O-Palmitoyltransferase genetics, Carnitine O-Palmitoyltransferase metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Citric Acid Cycle, DNA biosynthesis, Disease Models, Animal, Endothelial Cells cytology, Endothelial Cells drug effects, Endothelial Cells enzymology, Gene Silencing, Glucose metabolism, Human Umbilical Vein Endothelial Cells cytology, Human Umbilical Vein Endothelial Cells drug effects, Human Umbilical Vein Endothelial Cells metabolism, Human Umbilical Vein Endothelial Cells pathology, Humans, Mice, Neovascularization, Pathologic drug therapy, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Nucleotides chemistry, Nucleotides pharmacology, Oxidation-Reduction drug effects, Retinopathy of Prematurity drug therapy, Retinopathy of Prematurity metabolism, Retinopathy of Prematurity pathology, Carbon metabolism, Endothelial Cells metabolism, Fatty Acids chemistry, Fatty Acids metabolism, Nucleotides biosynthesis
- Abstract
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.
- Published
- 2015
- Full Text
- View/download PDF
43. Angiogenesis revisited - role and therapeutic potential of targeting endothelial metabolism.
- Author
-
Stapor P, Wang X, Goveia J, Moens S, and Carmeliet P
- Subjects
- Adenosine Triphosphate metabolism, Angiogenesis Inhibitors pharmacology, Animals, Endothelium, Vascular drug effects, Glycolysis drug effects, Humans, Molecular Targeted Therapy, Neovascularization, Pathologic metabolism, Phosphofructokinase-2 antagonists & inhibitors, Angiogenesis Inhibitors therapeutic use, Endothelium, Vascular metabolism, Neovascularization, Pathologic drug therapy, Neovascularization, Physiologic
- Abstract
Clinically approved therapies that target angiogenesis in tumors and ocular diseases focus on controlling pro-angiogenic growth factors in order to reduce aberrant microvascular growth. Although research on angiogenesis has revealed key mechanisms that regulate tissue vascularization, therapeutic success has been limited owing to insufficient efficacy, refractoriness and tumor resistance. Emerging concepts suggest that, in addition to growth factors, vascular metabolism also regulates angiogenesis and is a viable target for manipulating the microvasculature. Recent studies show that endothelial cells rely on glycolysis for ATP production, and that the key glycolytic regulator 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) regulates angiogenesis by controlling the balance of tip versus stalk cells. As endothelial cells acquire a tip cell phenotype, they increase glycolytic production of ATP for sprouting. Furthermore, pharmacological blockade of PFKFB3 causes a transient, partial reduction in glycolysis, and reduces pathological angiogenesis with minimal systemic harm. Although further assessment of endothelial cell metabolism is necessary, these results represent a paradigm shift in anti-angiogenic therapy from targeting angiogenic factors to focusing on vascular metabolism, warranting research on the metabolic pathways that govern angiogenesis., (© 2014. Published by The Company of Biologists Ltd.)
- Published
- 2014
- Full Text
- View/download PDF
44. Principles of targeting endothelial cell metabolism to treat angiogenesis and endothelial cell dysfunction in disease.
- Author
-
Goveia J, Stapor P, and Carmeliet P
- Subjects
- Apoptosis, Atherosclerosis metabolism, Atherosclerosis pathology, Cell Proliferation, Diabetes Mellitus metabolism, Diabetes Mellitus pathology, Endothelium, Vascular metabolism, Glycolysis, Humans, Metabolic Networks and Pathways, Models, Biological, Neovascularization, Pathologic metabolism, Signal Transduction, Endothelial Cells metabolism, Endothelium, Vascular pathology, Neovascularization, Pathologic pathology
- Abstract
The endothelium is the orchestral conductor of blood vessel function. Pathological blood vessel formation (a process termed pathological angiogenesis) or the inability of endothelial cells (ECs) to perform their physiological function (a condition known as EC dysfunction) are defining features of various diseases. Therapeutic intervention to inhibit aberrant angiogenesis or ameliorate EC dysfunction could be beneficial in diseases such as cancer and cardiovascular disease, respectively, but current strategies have limited efficacy. Based on recent findings that pathological angiogenesis and EC dysfunction are accompanied by EC-specific metabolic alterations, targeting EC metabolism is emerging as a novel therapeutic strategy. Here, we review recent progress in our understanding of how EC metabolism is altered in disease and discuss potential metabolic targets and strategies to reverse EC dysfunction and inhibit pathological angiogenesis., (© 2014 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2014
- Full Text
- View/download PDF
45. The multifaceted activity of VEGF in angiogenesis - Implications for therapy responses.
- Author
-
Moens S, Goveia J, Stapor PC, Cantelmo AR, and Carmeliet P
- Subjects
- Bone Marrow Cells metabolism, Endothelial Cells cytology, Fibroblasts metabolism, Humans, Myeloid Cells metabolism, Myocytes, Smooth Muscle metabolism, Signal Transduction, Tumor Microenvironment, Angiogenesis Inhibitors therapeutic use, Neoplasms blood supply, Neoplasms drug therapy, Neovascularization, Pathologic drug therapy, Vascular Endothelial Growth Factor A antagonists & inhibitors
- Abstract
Vascular endothelial growth factor (VEGF) is a key growth factor driving angiogenesis (i.e. the formation of new blood vessels) in health and disease. Pharmacological blockade of VEGF signaling to inhibit tumor angiogenesis is clinically approved but the survival benefit is limited as patients invariably acquire resistance. This is partially mediated by the intrinsic flexibility of tumor cells to adapt to VEGF-blockade. However, it has become clear that tumor stromal cells also contribute to the resistance. Originally, VEGF was thought to specifically target endothelial cells (ECs) but it is now clear that many stromal cells also respond to VEGF signaling, making anti-VEGF therapy more complex than initially anticipated. A more comprehensive understanding of the complex responses of stromal cells to VEGF-blockade might inform the design of improved anti-angiogenic agents., (Copyright © 2014. Published by Elsevier Ltd.)
- Published
- 2014
- Full Text
- View/download PDF
46. Endothelial cell differentiation by SOX17: promoting the tip cell or stalking its neighbor instead?
- Author
-
Goveia J, Zecchin A, Rodriguez FM, Moens S, Stapor P, and Carmeliet P
- Subjects
- Animals, Humans, Endothelial Cells metabolism, HMGB Proteins physiology, Neovascularization, Pathologic physiopathology, Neovascularization, Physiologic physiology, Receptors, Notch physiology, SOXF Transcription Factors physiology, Signal Transduction physiology
- Published
- 2014
- Full Text
- View/download PDF
47. Analytical characterization of an assay designed to detect and identify diverse agents of disseminated viral infection.
- Author
-
Metzgar D, Lovari R, Ray K, Baynes D, Drapp D, Frinder M, Vijesurier R, Stemler M, Ofsaiof R, Carolan H, Welk J, Toleno D, Ranken R, Hall TA, Massire C, Sampath R, Blyn LB, Goveia J, and Schneider G
- Subjects
- Humans, Limit of Detection, Polymerase Chain Reaction, Reproducibility of Results, Viruses classification, Viruses genetics, Viruses isolation & purification, Pathology, Molecular methods, Viremia diagnosis, Viremia virology, Virology methods
- Abstract
Background: Diverse viruses often reactivate in or infect cancer patients, patients with immunocompromising infections or genetic conditions, and transplant recipients undergoing immunosuppressive therapy. These infections can disseminate, leading to death, transplant rejection, and other severe outcomes., Objectives: To develop and characterize an assay capable of inclusive and accurate identification of diverse potentially disseminating viruses directly from plasma specimens., Study Design: We developed a PCR/electrospray ionization mass spectrometry (PCR/ESI-MS) assay designed to simultaneously detect and identify adenovirus, enterovirus, polyomaviruses JC and BK, parvovirus B19, HSV-1, HSV-2, VZV, EBV, CMV, and herpesviruses 6-8 in plasma specimens. The assay performance was characterized analytically, and the results from clinical plasma samples were compared to the results obtained from single-analyte real time PCR tests currently used in clinical practice., Results: The assay demonstrated sensitivity and specificity to diverse strains of the targeted viral families and robustness to interfering substances and potentially cross reacting organisms. The assay yielded 94% sensitivity when testing clinical plasma samples previously identified as positive using standard-of-care real-time PCR tests for a single target virus (available samples included positive samples for 11 viruses targeted by the assay)., Conclusions: The assay functioned as designed, providing simultaneous broad-spectrum detection and identification of diverse agents of disseminated viral infection. Among 156 clinical samples tested, 37 detections were made in addition to the detections matching the initial clinical positive results., (Copyright © 2013 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
48. Partial and transient reduction of glycolysis by PFKFB3 blockade reduces pathological angiogenesis.
- Author
-
Schoors S, De Bock K, Cantelmo AR, Georgiadou M, Ghesquière B, Cauwenberghs S, Kuchnio A, Wong BW, Quaegebeur A, Goveia J, Bifari F, Wang X, Blanco R, Tembuyser B, Cornelissen I, Bouché A, Vinckier S, Diaz-Moralli S, Gerhardt H, Telang S, Cascante M, Chesney J, Dewerchin M, and Carmeliet P
- Subjects
- Angiogenesis Inhibitors pharmacology, Animals, Cell Movement drug effects, Cell Proliferation drug effects, Disease Models, Animal, Gene Expression Regulation drug effects, Human Umbilical Vein Endothelial Cells drug effects, Human Umbilical Vein Endothelial Cells enzymology, Human Umbilical Vein Endothelial Cells pathology, Humans, Mice, Mice, Inbred C57BL, Neovascularization, Pathologic genetics, Neovascularization, Physiologic drug effects, Neovascularization, Physiologic genetics, Phosphofructokinase-2 metabolism, Pyridines pharmacology, Retinal Vessels drug effects, Retinal Vessels growth & development, Retinal Vessels pathology, Vascular Endothelial Growth Factor Receptor-1 antagonists & inhibitors, Vascular Endothelial Growth Factor Receptor-1 metabolism, Zebrafish, Glycolysis drug effects, Neovascularization, Pathologic enzymology, Phosphofructokinase-2 antagonists & inhibitors
- Abstract
Strategies targeting pathological angiogenesis have focused primarily on blocking vascular endothelial growth factor (VEGF), but resistance and insufficient efficacy limit their success, mandating alternative antiangiogenic strategies. We recently provided genetic evidence that the glycolytic activator phosphofructokinase-2/fructose-2,6-bisphosphatase 3 (PFKFB3) promotes vessel formation but did not explore the antiangiogenic therapeutic potential of PFKFB3 blockade. Here, we show that blockade of PFKFB3 by the small molecule 3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO) reduced vessel sprouting in endothelial cell (EC) spheroids, zebrafish embryos, and the postnatal mouse retina by inhibiting EC proliferation and migration. 3PO also suppressed vascular hyperbranching induced by inhibition of Notch or VEGF receptor 1 (VEGFR1) and amplified the antiangiogenic effect of VEGF blockade. Although 3PO reduced glycolysis only partially and transiently in vivo, this sufficed to decrease pathological neovascularization in ocular and inflammatory models. These insights may offer therapeutic antiangiogenic opportunities., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
49. Educational interventions to improve the meaningful use of Electronic Health Records: a review of the literature: BEME Guide No. 29.
- Author
-
Goveia J, Van Stiphout F, Cheung Z, Kamta B, Keijsers C, Valk G, and Ter Braak E
- Subjects
- Cost-Benefit Analysis, Counseling, Feedback, Humans, Policy, Professional Competence, Quality Improvement organization & administration, Electronic Health Records organization & administration, Health Personnel education, Meaningful Use organization & administration
- Abstract
Background: Electronic health records (EHRs) are increasingly available and this was expected to reduce healthcare costs and medical errors. This promise has not been realized because healthcare professionals are unable to use EHRs in a manner that contributes to significant improvements in care, i.e. meaningful. Policymakers now acknowledge that training healthcare professionals in meaningful use is essential for successful EHR implementation. To help educators and policymakers design evidence based educational interventions (i.e. interventions that involve educational activities but no practical lessons) and training (i.e. interventions that involve practical components), we summarized all evidence regarding the efficacy of different educational interventions to improve meaningful use of EHRs., Methods: We used a predefined search filter to search eight databases for studies that considered an educational intervention to promote meaningful use of EHRs by healthcare professionals., Results: Seven of the 4507 reviewed articles met the in- and exclusion criteria., Conclusions: These studies suggest that a combination of classroom training, computer-based training and feedback is most effective to improve meaningful use. In addition, the training should be tailored to the needs of the trainees and they should be able to practice in their own time. However, the evidence is very limited and we recommend that governments, hospitals and other policymakers invest more in the development of evidence based educational interventions to improve meaningful use of EHRs.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.