98 results on '"CARMELIET, PETER"'
Search Results
2. Metabolic and Innate Immune Cues Merge into a Specific Inflammatory Response via the UPR
- Author
-
Mogilenko, Denis A., Haas, Joel T., Lhomme, Laurent, Fleury, Sebastien, Quemener, Sandrine, Levavasseur, Matthieu, Becquart, Coralie, Wartelle, Julien, Bogomolova, Alexandra, Pineau, Laurent, Molendi-Coste, Olivier, Lancel, Steve, Dehondt, Helene, Gheeraert, Celine, Melchior, Aurelie, Dewas, Cedric, Nikitin, Artemii, Pic, Samuel, Rabhi, Nabil, Annicotte, Jean-Sebastien, Oyadomari, Seiichi, Velasco-Hernandez, Talia, Cammenga, Jörg, Foretz, Marc, Viollet, Benoit, Vukovic, Milica, Villacreces, Arnaud, Kranc, Kamil, Carmeliet, Peter, Marot, Guillemette, Boulter, Alexis, Tavernier, Simon, Berod, Luciana, Longhi, Maria P., Paget, Christophe, Janssens, Sophie, Staumont-Salle, Delphine, Aksoy, Ezra, Staels, Bart, Dombrowicz, David, Mogilenko, Denis A., Haas, Joel T., Lhomme, Laurent, Fleury, Sebastien, Quemener, Sandrine, Levavasseur, Matthieu, Becquart, Coralie, Wartelle, Julien, Bogomolova, Alexandra, Pineau, Laurent, Molendi-Coste, Olivier, Lancel, Steve, Dehondt, Helene, Gheeraert, Celine, Melchior, Aurelie, Dewas, Cedric, Nikitin, Artemii, Pic, Samuel, Rabhi, Nabil, Annicotte, Jean-Sebastien, Oyadomari, Seiichi, Velasco-Hernandez, Talia, Cammenga, Jörg, Foretz, Marc, Viollet, Benoit, Vukovic, Milica, Villacreces, Arnaud, Kranc, Kamil, Carmeliet, Peter, Marot, Guillemette, Boulter, Alexis, Tavernier, Simon, Berod, Luciana, Longhi, Maria P., Paget, Christophe, Janssens, Sophie, Staumont-Salle, Delphine, Aksoy, Ezra, Staels, Bart, and Dombrowicz, David
- Abstract
Innate immune responses are intricately linked with intracellular metabolism of myeloid cells. Toll-like receptor (TLR) stimulation shifts intracellular metabolism toward glycolysis, while anti-inflammatory signals depend on enhanced mitochondrial respiration. How exogenous metabolic signals affect the immune response is unknown. We demonstrate that TLR-dependent responses of dendritic cells (DCs) are exacerbated by a high-fatty-acid (FA) metabolic environment. FAs suppress the TLR-induced hexokinase activity and perturb tricarboxylic acid cycle metabolism. These metabolic changes enhance mitochondria! reactive oxygen species (mtROS) production and, in turn, the unfolded protein response (UPR), leading to a distinct transcriptomic signature with IL-23 as hallmark. Interestingly, chemical or genetic suppression of glycolysis was sufficient to induce this specific immune response. Conversely, reducing mtROS production or DC-specific deficiency in XBP1 attenuated IL-23 expression and skin inflammation in an IL-23-dependent model of psoriasis. Thus, fine-tuning of innate immunity depends on optimization of metabolic demands and minimization of mtROS-induced UPR., Funding Agencies|ANR; European Union [EGID ANR-10-LABX-46, ANR-17-CE15-0030-02]; National Psoriasis Foundation (USA) Early Career Research Grant; EMBO Long-Term Fellowship; MRC [MR/M023230/1]; CRUK [C29967/A14633, C29967/A26787]; ERC advanced grant (ERC-2016-AdG) [694717]
- Published
- 2019
- Full Text
- View/download PDF
3. Role of PFKFB3-driven glycolysis in vessel sprouting.
- Author
-
UCL - SSS/DDUV - Institut de Duve, De Bock, Katrien, Georgiadou, Maria, Schoors, Sandra, Kuchnio, Anna, Wong, Brian W, Cantelmo, Anna Rita, Quaegebeur, Annelies, Ghesquière, Bart, Cauwenberghs, Sandra, Eelen, Guy, Phng, Li-Kun, Betz, Inge, Tembuyser, Bieke, Brepoels, Katleen, Welti, Jonathan, Geudens, Ilse, Segura, Inmaculada, Cruys, Bert, Bifari, Franscesco, Decimo, Ilaria, Blanco, Raquel, Wyns, Sabine, Vangindertael, Jeroen, Rocha, Susana, Collins, Russel T, Munck, Sebastian, Daelemans, Dirk, Imamura, Hiromi, Devlieger, Roland, Rider, Mark, Van Veldhoven, Paul P, Schuit, Frans, Bartrons, Ramon, Hofkens, Johan, Fraisl, Peter, Telang, Sucheta, Deberardinis, Ralph J, Schoonjans, Luc, Vinckier, Stefan, Chesney, Jason, Gerhardt, Holger, Dewerchin, Mieke, Carmeliet, Peter, UCL - SSS/DDUV - Institut de Duve, De Bock, Katrien, Georgiadou, Maria, Schoors, Sandra, Kuchnio, Anna, Wong, Brian W, Cantelmo, Anna Rita, Quaegebeur, Annelies, Ghesquière, Bart, Cauwenberghs, Sandra, Eelen, Guy, Phng, Li-Kun, Betz, Inge, Tembuyser, Bieke, Brepoels, Katleen, Welti, Jonathan, Geudens, Ilse, Segura, Inmaculada, Cruys, Bert, Bifari, Franscesco, Decimo, Ilaria, Blanco, Raquel, Wyns, Sabine, Vangindertael, Jeroen, Rocha, Susana, Collins, Russel T, Munck, Sebastian, Daelemans, Dirk, Imamura, Hiromi, Devlieger, Roland, Rider, Mark, Van Veldhoven, Paul P, Schuit, Frans, Bartrons, Ramon, Hofkens, Johan, Fraisl, Peter, Telang, Sucheta, Deberardinis, Ralph J, Schoonjans, Luc, Vinckier, Stefan, Chesney, Jason, Gerhardt, Holger, Dewerchin, Mieke, and Carmeliet, Peter
- Abstract
Vessel sprouting by migrating tip and proliferating stalk endothelial cells (ECs) is controlled by genetic signals (such as Notch), but it is unknown whether metabolism also regulates this process. Here, we show that ECs relied on glycolysis rather than on oxidative phosphorylation for ATP production and that loss of the glycolytic activator PFKFB3 in ECs impaired vessel formation. Mechanistically, PFKFB3 not only regulated EC proliferation but also controlled the formation of filopodia/lamellipodia and directional migration, in part by compartmentalizing with F-actin in motile protrusions. Mosaic in vitro and in vivo sprouting assays further revealed that PFKFB3 overexpression overruled the pro-stalk activity of Notch, whereas PFKFB3 deficiency impaired tip cell formation upon Notch blockade, implying that glycolysis regulates vessel branching.
- Published
- 2013
4. FAF1, a gene that is disrupted in cleft palate and has conserved function in zebrafish.
- Author
-
UCL - SSS/DDUV - Institut de Duve, UCL - (SLuc) Centre de génétique médicale UCL, UCL - (SLuc) Service de chirurgie plastique, Ghassibé, Michella, Desmyter, Laurence, Langenberg, Tobias, Claes, Filip, Boute, Odile, Bayet, Bénédicte, Pellerin, Philippe, Hermans, Karlien, Backx, Liesbeth, Mansilla, Maria Adela, Imoehl, Sandra, Nowak, Stefanie, Ludwig, Kerstin U, Baluardo, Carlotta, Ferrian, Melissa, Mossey, Peter A, Noethen, Markus, Dewerchin, Mieke, François, Geneviève, Revencu, Nicole, Vanwijck, Romain, Hecht, Jacqueline, Mangold, Elisabeth, Murray, Jeffrey, Rubini, Michele, Vermeesch, Joris R, Poirel, Hélène, Carmeliet, Peter, Vikkula, Miikka, UCL - SSS/DDUV - Institut de Duve, UCL - (SLuc) Centre de génétique médicale UCL, UCL - (SLuc) Service de chirurgie plastique, Ghassibé, Michella, Desmyter, Laurence, Langenberg, Tobias, Claes, Filip, Boute, Odile, Bayet, Bénédicte, Pellerin, Philippe, Hermans, Karlien, Backx, Liesbeth, Mansilla, Maria Adela, Imoehl, Sandra, Nowak, Stefanie, Ludwig, Kerstin U, Baluardo, Carlotta, Ferrian, Melissa, Mossey, Peter A, Noethen, Markus, Dewerchin, Mieke, François, Geneviève, Revencu, Nicole, Vanwijck, Romain, Hecht, Jacqueline, Mangold, Elisabeth, Murray, Jeffrey, Rubini, Michele, Vermeesch, Joris R, Poirel, Hélène, Carmeliet, Peter, and Vikkula, Miikka
- Abstract
Cranial neural crest (CNC) is a multipotent migratory cell population that gives rise to most of the craniofacial bones. An intricate network mediates CNC formation, epithelial-mesenchymal transition, migration along distinct paths, and differentiation. Errors in these processes lead to craniofacial abnormalities, including cleft lip and palate. Clefts are the most common congenital craniofacial defects. Patients have complications with feeding, speech, hearing, and dental and psychological development. Affected by both genetic predisposition and environmental factors, the complex etiology of clefts remains largely unknown. Here we show that Fas-associated factor-1 (FAF1) is disrupted and that its expression is decreased in a Pierre Robin family with an inherited translocation. Furthermore, the locus is strongly associated with cleft palate and shows an increased relative risk. Expression studies show that faf1 is highly expressed in zebrafish cartilages during embryogenesis. Knockdown of zebrafish faf1 leads to pharyngeal cartilage defects and jaw abnormality as a result of a failure of CNC to differentiate into and express cartilage-specific markers, such as sox9a and col2a1. Administration of faf1 mRNA rescues this phenotype. Our findings therefore identify FAF1 as a regulator of CNC differentiation and show that it predisposes humans to cleft palate and is necessary for lower jaw development in zebrafish.
- Published
- 2011
5. Heterozygous deficiency of PHD2 restores tumor oxygenation and inhibits metastasis via endothelial normalization
- Author
-
UCL - MD/FARM - Ecole de pharmacie, UCL - (SLuc) Service de médecine nucléaire, Mazzone, Massimiliano, Dettori, Daniela, Leite de Oliveira, Rodrigo, Loges, Sonja, Schmidt, Thomas, Jonckx, Bart, Tian, Ya-Min, Lanahan, Anthony A., Pollard, Patrick, Ruiz de Almodovar, Carmen, De Smet, Frederik, Vinckier, Stefan, Aragonés, Julián, Debackere, Koen, Luttun, Aernout, Wyns, Sabine, Jordan, Bénédicte, Pisacane, Alberto, Gallez, Bernard, Lampugnani, Maria Grazia, Dejana, Elisabetta, Simons, Michael, Ratcliffe, Peter, Maxwell, Patrick, Carmeliet, Peter, UCL - MD/FARM - Ecole de pharmacie, UCL - (SLuc) Service de médecine nucléaire, Mazzone, Massimiliano, Dettori, Daniela, Leite de Oliveira, Rodrigo, Loges, Sonja, Schmidt, Thomas, Jonckx, Bart, Tian, Ya-Min, Lanahan, Anthony A., Pollard, Patrick, Ruiz de Almodovar, Carmen, De Smet, Frederik, Vinckier, Stefan, Aragonés, Julián, Debackere, Koen, Luttun, Aernout, Wyns, Sabine, Jordan, Bénédicte, Pisacane, Alberto, Gallez, Bernard, Lampugnani, Maria Grazia, Dejana, Elisabetta, Simons, Michael, Ratcliffe, Peter, Maxwell, Patrick, and Carmeliet, Peter
- Abstract
A key function of blood vessels, to supply oxygen, is impaired in tumors because of abnormalities in their endothelial lining. PHD proteins serve as oxygen sensors and may regulate oxygen delivery. We therefore studied the role of endothelial PHD2 in vessel shaping by implanting tumors in PHD2(+/-) mice. Haplodeficiency of PHD2 did not affect tumor vessel density or lumen size, but normalized the endothelial lining and vessel maturation. This resulted in improved tumor perfusion and oxygenation and inhibited tumor cell invasion, intravasation, and metastasis. Haplodeficiency of PHD2 redirected the specification of endothelial tip cells to a more quiescent cell type, lacking filopodia and arrayed in a phalanx formation. This transition relied on HIF-driven upregulation of (soluble) VEGFR-1 and VE-cadherin. Thus, decreased activity of an oxygen sensor in hypoxic conditions prompts endothelial cells to readjust their shape and phenotype to restore oxygen supply. Inhibition of PHD2 may offer alternative therapeutic opportunities for anticancer therapy.
- Published
- 2009
6. Sculpting Heart Valves with NFATc and VEGF
- Author
-
Lambrechts, Diether and Carmeliet, Peter
- Subjects
- *
HEART valves , *VASCULAR endothelial growth factors , *HEART physiology , *MEDICAL sciences , *MEDICAL care , *LIFE sciences - Abstract
Heart valves are of vital importance for our moment-to-moment existence, but how they form remains a mystery. In this issue of Cell, Chang et al. reveal a novel role for calcineurin, NFATs, and VEGF in valve formation . Dynamic changes in NFAT/VEGF expression in regional myocardial and endocardial fields and developmental windows orchestrate this complex process. [Copyright &y& Elsevier]
- Published
- 2004
- Full Text
- View/download PDF
7. SnapShot: Tumor Angiogenesis
- Author
-
Jain, Rakesh K. and Carmeliet, Peter
- Published
- 2012
- Full Text
- View/download PDF
8. Neutrophil trapping and nexocytosis, mast cell-mediated processes for inflammatory signal relay.
- Author
-
Mihlan, Michael, Wissmann, Stefanie, Gavrilov, Alina, Kaltenbach, Lukas, Britz, Marie, Franke, Kristin, Hummel, Barbara, Imle, Andrea, Suzuki, Ryo, Stecher, Manuel, Glaser, Katharina M., Lorentz, Axel, Carmeliet, Peter, Yokomizo, Takehiko, Hilgendorf, Ingo, Sawarkar, Ritwick, Diz-Muñoz, Alba, Buescher, Joerg M., Mittler, Gerhard, and Maurer, Marcus
- Subjects
- *
CELL communication , *TYPE I interferons , *MAST cells , *ALLERGIES , *IMMUNE response , *NEUTROPHILS - Abstract
Neutrophils are sentinel immune cells with essential roles for antimicrobial defense. Most of our knowledge on neutrophil tissue navigation derived from wounding and infection models, whereas allergic conditions remained largely neglected. Here, we analyzed allergen-challenged mouse tissues and discovered that degranulating mast cells (MCs) trap living neutrophils inside them. MCs release the attractant leukotriene B4 to re-route neutrophils toward them, thus exploiting a chemotactic system that neutrophils normally use for intercellular communication. After MC intracellular trap (MIT) formation, neutrophils die, but their undigested material remains inside MC vacuoles over days. MCs benefit from MIT formation, increasing their functional and metabolic fitness. Additionally, they are more pro-inflammatory and can exocytose active neutrophilic compounds with a time delay (nexocytosis), eliciting a type 1 interferon response in surrounding macrophages. Together, our study highlights neutrophil trapping and nexocytosis as MC-mediated processes, which may relay neutrophilic features over the course of chronic allergic inflammation. [Display omitted] • MCs induce neutrophil swarms upon IgE-mediated degranulation in tissues • Living neutrophils are trapped by MCs and form a cell-in-cell structure • MITs show increased functional and metabolic fitness • MITs are more pro-inflammatory and can exocytose active neutrophilic compounds During allergen challenge, degranulating mast cells re-route and trap living neutrophils in vivo , resulting in functional and metabolic alteration of the mast cell and the acquisition of neutrophilic features. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Basic and Therapeutic Aspects of Angiogenesis
- Author
-
Potente, Michael, Gerhardt, Holger, and Carmeliet, Peter
- Subjects
- *
NEOVASCULARIZATION , *BLOOD-vessel abnormalities , *TISSUES , *INFLAMMATION , *CANCER invasiveness , *ENDOTHELIUM , *TREATMENT effectiveness , *BLOOD-vessel development - Abstract
Blood vessels form extensive networks that nurture all tissues in the body. Abnormal vessel growth and function are hallmarks of cancer and ischemic and inflammatory diseases, and they contribute to disease progression. Therapeutic approaches to block vascular supply have reached the clinic, but limited efficacy and resistance pose unresolved challenges. Recent insights establish how endothelial cells communicate with each other and with their environment to form a branched vascular network. The emerging principles of vascular growth provide exciting new perspectives, the translation of which might overcome the current limitations of pro- and antiangiogenic medicine. [Copyright &y& Elsevier]
- Published
- 2011
- Full Text
- View/download PDF
10. The Neurovascular Link in Health and Disease: Molecular Mechanisms and Therapeutic Implications
- Author
-
Quaegebeur, Annelies, Lange, Christian, and Carmeliet, Peter
- Subjects
- *
NERVOUS system blood-vessels , *MOLECULAR dynamics , *NEUROLOGICAL disorders , *BLOOD cells , *NEUROLOGY , *NEUROSCIENCES - Abstract
At first sight, the nervous and vascular systems seem to share little in common. However, neural and vascular cells not only are anatomically closely tied to each other, but they also utilize and respond to similar classes of signals to establish correct connectivity and wiring of their networks. Recent studies further provide evidence that this neurovascular crosstalk is more important for understanding the molecular basis of neurological disease than originally anticipated. Moreover, neurovascular strategies offer novel therapeutic opportunities for neurodegenerative disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
11. An SDF-1 Trap for Myeloid Cells Stimulates Angiogenesis
- Author
-
Ruiz de Almodovar, Carmen, Luttun, Aernout, and Carmeliet, Peter
- Subjects
- *
HEMATOPOIETIC stem cells , *MYELOID leukemia , *NEOVASCULARIZATION , *GENE expression , *VASCULAR endothelial growth factors , *BONE marrow , *CELLS - Abstract
In this issue of Cell, examine the role of hematopoietic cells in the formation of new blood vessels. They show that organ-specific expression of vascular endothelial growth factor (VEGF) is sufficient to mobilize and recruit hematopoietic cells from the bone marrow to the blood, but retention of the proangiogenic subpopulation of hematopoietic cells in peripheral organs requires an additional factor, stromal-derived factor 1 (SDF-1). [Copyright &y& Elsevier]
- Published
- 2006
- Full Text
- View/download PDF
12. Pericytes: Blood-Brain Barrier Safeguards against Neurodegeneration?
- Author
-
Quaegebeur, Annelies, Segura, Inmaculada, and Carmeliet, Peter
- Subjects
- *
NEURONS , *BLOOD-brain barrier , *NEURODEGENERATION , *BIOLOGICAL periodicals , *COGNITION disorders , *DISEASES in older people - Abstract
The role of pericytes in the control of blood-brain barrier (BBB) integrity has remained enigmatic. In this issue, Bell et al. and two concurrent studies highlight that pericyte loss causes BBB breakdown and hypoperfusion. Remarkably, these vascular changes precede neurodegeneration and cognitive defects in old age. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
13. Anti-PlGF Inhibits Growth of VEGF(R)-Inhibitor-Resistant Tumors without Affecting Healthy Vessels
- Author
-
Fischer, Christian, Jonckx, Bart, Mazzone, Massimiliano, Zacchigna, Serena, Loges, Sonja, Pattarini, Lucia, Chorianopoulos, Emmanuel, Liesenborghs, Laurens, Koch, Marta, De Mol, Maria, Autiero, Monica, Wyns, Sabine, Plaisance, Stephane, Moons, Lieve, van Rooijen, Nico, Giacca, Mauro, Stassen, Jean-Marie, Dewerchin, Mieke, Collen, Desire, and Carmeliet, Peter
- Subjects
- *
VASCULAR endothelial growth factors , *TUMORS , *GROWTH factors , *NEOVASCULARIZATION - Abstract
Summary: Novel antiangiogenic strategies with complementary mechanisms are needed to maximize efficacy and minimize resistance to current angiogenesis inhibitors. We explored the therapeutic potential and mechanisms of αPlGF, an antibody against placental growth factor (PlGF), a VEGF homolog, which regulates the angiogenic switch in disease, but not in health. αPlGF inhibited growth and metastasis of various tumors, including those resistant to VEGF(R) inhibitors (VEGF R Is), and enhanced the efficacy of chemotherapy and VEGF R Is. αPlGF inhibited angiogenesis, lymphangiogenesis, and tumor cell motility. Distinct from VEGF R Is, αPlGF prevented infiltration of angiogenic macrophages and severe tumor hypoxia, and thus, did not switch on the angiogenic rescue program responsible for resistance to VEGF R Is. Moreover, it did not cause or enhance VEGF R I-related side effects. The efficacy and safety of αPlGF, its pleiotropic and complementary mechanism to VEGF R Is, and the negligible induction of an angiogenic rescue program suggest that αPlGF may constitute a novel approach for cancer treatment. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
14. The ER Stress Sensor PERK Coordinates ER-Plasma Membrane Contact Site Formation through Interaction with Filamin-A and F-Actin Remodeling.
- Author
-
van Vliet, Alexander R., Giordano, Francesca, Gerlo, Sarah, Segura, Inmaculada, Van Eygen, Sofie, Molenberghs, Geert, Rocha, Susana, Houcine, Audrey, Derua, Rita, Verfaillie, Tom, Vangindertael, Jeroen, De Keersmaecker, Herlinde, Waelkens, Etienne, Tavernier, Jan, Hofkens, Johan, Annaert, Wim, Carmeliet, Peter, Samali, Afshin, Mizuno, Hideaki, and Agostinis, Patrizia
- Subjects
- *
ENDOPLASMIC reticulum , *F-actin , *SYNAPTOTAGMINS , *MICROFILAMENT proteins , *CELLULAR signal transduction - Abstract
Summary Loss of ER Ca 2+ homeostasis triggers endoplasmic reticulum (ER) stress and drives ER-PM contact sites formation in order to refill ER-luminal Ca 2+ . Recent studies suggest that the ER stress sensor and mediator of the unfolded protein response (UPR) PERK regulates intracellular Ca 2+ fluxes, but the mechanisms remain elusive. Here, using proximity-dependent biotin identification (BioID), we identified the actin-binding protein Filamin A (FLNA) as a key PERK interactor. Cells lacking PERK accumulate F-actin at the cell edges and display reduced ER-PM contacts. Following ER-Ca 2+ store depletion, the PERK-FLNA interaction drives the expansion of ER-PM juxtapositions by regulating F-actin-assisted relocation of the ER-associated tethering proteins Stromal Interaction Molecule 1 (STIM1) and Extended Synaptotagmin-1 (E-Syt1) to the PM. Cytosolic Ca 2+ elevation elicits rapid and UPR-independent PERK dimerization, which enforces PERK-FLNA-mediated ER-PM juxtapositions. Collectively, our data unravel an unprecedented role of PERK in the regulation of ER-PM appositions through the modulation of the actin cytoskeleton. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
15. Immunomodulation by endothelial cells: prospects for cancer therapy.
- Author
-
Alnaqbi H, Becker LM, Mousa M, Alshamsi F, Azzam SK, Emini Veseli B, Hymel LA, Alhosani K, Alhusain M, Mazzone M, Alsafar H, and Carmeliet P
- Subjects
- Humans, Animals, Neoplasms immunology, Neoplasms therapy, Neoplasms pathology, Tumor Microenvironment immunology, Endothelial Cells immunology, Endothelial Cells metabolism, Immunomodulation, Immunotherapy methods
- Abstract
Growing evidence highlights the importance of tumor endothelial cells (TECs) in the tumor microenvironment (TME) for promoting tumor growth and evading immune responses. Immunomodulatory endothelial cells (IMECs) represent a distinct plastic phenotype of ECs that exerts the ability to modulate immunity in health and disease. This review discusses our current understanding of IMECs in cancer biology, scrutinizing insights from single-cell reports to compare their characteristics and function dynamics across diverse tumor types, conditions, and species. We investigate possible implications of exploiting IMECs in the context of cancer treatment, particularly examining their influence on the efficacy of existing therapies and the potential to leverage them as targets in optimizing immunotherapeutic strategies., Competing Interests: Declaration of interests None are declared by the authors., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
16. Targeting EGLN2/PHD1 protects motor neurons and normalizes the astrocytic interferon response.
- Author
-
Germeys C, Vandoorne T, Davie K, Poovathingal S, Heeren K, Vermeire W, Nami F, Moisse M, Quaegebeur A, Sierksma A, Rué L, Sicart A, Eykens C, De Cock L, De Strooper B, Carmeliet P, Van Damme P, De Bock K, and Van Den Bosch L
- Subjects
- Animals, Humans, Mice, Disease Models, Animal, Hypoxia-Inducible Factor-Proline Dioxygenases antagonists & inhibitors, Hypoxia-Inducible Factor-Proline Dioxygenases genetics, Hypoxia-Inducible Factor-Proline Dioxygenases metabolism, Induced Pluripotent Stem Cells metabolism, Interferons metabolism, Amyotrophic Lateral Sclerosis drug therapy, Amyotrophic Lateral Sclerosis genetics, Amyotrophic Lateral Sclerosis metabolism, Amyotrophic Lateral Sclerosis pathology, Astrocytes metabolism, Motor Neurons metabolism, Zebrafish metabolism
- Abstract
Neuroinflammation and dysregulated energy metabolism are linked to motor neuron degeneration in amyotrophic lateral sclerosis (ALS). The egl-9 family hypoxia-inducible factor (EGLN) enzymes, also known as prolyl hydroxylase domain (PHD) enzymes, are metabolic sensors regulating cellular inflammation and metabolism. Using an oligonucleotide-based and a genetic approach, we showed that the downregulation of Egln2 protected motor neurons and mitigated the ALS phenotype in two zebrafish models and a mouse model of ALS. Single-nucleus RNA sequencing of the murine spinal cord revealed that the loss of EGLN2 induced an astrocyte-specific downregulation of interferon-stimulated genes, mediated via the stimulator of interferon genes (STING) protein. In addition, we found that the genetic deletion of EGLN2 restored this interferon response in patient induced pluripotent stem cell (iPSC)-derived astrocytes, confirming the link between EGLN2 and astrocytic interferon signaling. In conclusion, we identified EGLN2 as a motor neuron protective target normalizing the astrocytic interferon-dependent inflammatory axis in vivo, as well as in patient-derived cells., Competing Interests: Declaration of interests T.V. is an employee of Bristol-Myers Squibb (Princeton, USA). B.D.S. has been a consultant for Eli Lilly and Company (Indianapolis, USA), Biogen (Cambridge, USA), Janssen Pharmaceutica (Beerse, Belgium), AbbVie, Inc. (North Chicago, USA), and others and is now a consultant for Eisai (Nutley, USA), Remynd (Leuven, Belgium), and Muna Therapeutics (Copenhagen, Denmark). B.D.S. is a scientific founder and stockholder of Muna Therapeutics. P.V.D. has served on advisory boards for Biogen, CSL Behring (King of Prussia, USA), Alexion Pharmaceuticals (Boston, USA), Ferrer (Barcelona, Spain), QurAlis (Cambridge, UK), Cytokinetics (South San Francisco, USA), Argenx (Boston, USA), UCB (Brussels, Belgium), Muna Therapeutics, Alector (South San Francisco, USA), Augustine Therapeutics (Leuven, Belgium), VectorY (Amsterdam, the Netherlands), Zambon (Bresso, Italy), and Amylyx (Cambridge, UK) (paid to institution). P.V.D. has received speaker fees from Biogen and Amylyx (paid to institution). P.V.D. has participated as an investigator in clinical trials on ALS sponsored by Biogen, Cytokinetics, Ferrer, Amylyx, Wave Life Sciences (Cambridge, UK), Corcept Therapeutics (Menlo Park, USA), Transposon Therapeutics (Westport, USA), Sanofi (Paris, France), AB Science (Paris, France), IONIS Pharmaceuticals (Carlsbad, USA), Apellis Pharmaceuticals (Waltham, USA), Alexion Pharmaceuticals, Orphazyme (Copenhagen, Denmark), Orion Pharma (Espoo, Finland), and AL-S Pharma (Schlieren, Switzerland). P.V.D. is supported by the E. von Behring Chair for Neuromuscular and Neurodegenerative Disorders (from CSL Behring, paid to institution). L.V.D.B. is head of the scientific advisory board of Augustine Therapeutics and is part of the investment advisory board of Droia Ventures (Meise, Belgium). L.V.D.B. and B.D.S. are scientific founders of Augustine Therapeutics., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
17. The Receptor Tyrosine Kinase AXL Is Required at Multiple Steps of the Metastatic Cascade during HER2-Positive Breast Cancer Progression.
- Author
-
Goyette MA, Duhamel S, Aubert L, Pelletier A, Savage P, Thibault MP, Johnson RM, Carmeliet P, Basik M, Gaboury L, Muller WJ, Park M, Roux PP, Gratton JP, and Côté JF
- Published
- 2023
- Full Text
- View/download PDF
18. VEGF Mediates Commissural Axon Chemoattraction through Its Receptor Flk1.
- Author
-
Ruiz de Almodovar C, Fabre PJ, Knevels E, Coulon C, Segura I, Haddick PCG, Aerts L, Delattin N, Strasser G, Oh WJ, Lange C, Vinckier S, Haigh J, Fouquet C, Gu C, Alitalo K, Castellani V, Tessier-Lavigne M, Chedotal A, Charron F, and Carmeliet P
- Published
- 2023
- Full Text
- View/download PDF
19. Generation of vessel co-option lung metastases mouse models for single-cell isolation of metastases-derived cells and endothelial cells.
- Author
-
Cuypers A, Teuwen LA, Bridgeman VL, de Rooij LPMH, Eelen G, Dewerchin M, Cantelmo AR, Kalucka J, Bouché A, Vinckier S, Carton A, Manderveld A, Vermeulen PB, Reynolds AR, and Carmeliet P
- Subjects
- Mice, Animals, Endothelial Cells, Disease Models, Animal, Neovascularization, Pathologic pathology, Lung Neoplasms pathology
- Abstract
Tumor vessel co-option, a process in which cancer cells "hijack" pre-existing blood vessels to grow and invade healthy tissue, is poorly understood but is a proposed resistance mechanism against anti-angiogenic therapy (AAT). Here, we describe protocols for establishing murine renal (RENCA) and breast (4T1) cancer lung vessel co-option metastases models. Moreover, we outline a reproducible protocol for single-cell isolation from murine lung metastases using magnetic-activated cell sorting as well as immunohistochemical stainings to distinguish vessel co-option from angiogenesis. For complete details on the use and execution of this protocol, please refer to Teuwen et al. (2021)., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
20. CD8 + T cell metabolic rewiring defined by scRNA-seq identifies a critical role of ASNS expression dynamics in T cell differentiation.
- Author
-
Fernández-García J, Franco F, Parik S, Altea-Manzano P, Pane AA, Broekaert D, van Elsen J, Di Conza G, Vermeire I, Schalley T, Planque M, van Brussel T, Schepers R, Modave E, Karakach TK, Carmeliet P, Lambrechts D, Ho PC, and Fendt SM
- Subjects
- Mice, Animals, Single-Cell Analysis, Lymphocyte Activation, Cell Differentiation, Disease Models, Animal, CD8-Positive T-Lymphocytes, Melanoma metabolism
- Abstract
T cells dynamically rewire their metabolism during an immune response. We applied single-cell RNA sequencing to CD8
+ T cells activated and differentiated in vitro in physiological medium to resolve these metabolic dynamics. We identify a differential time-dependent reliance of activating T cells on the synthesis versus uptake of various non-essential amino acids, which we corroborate with functional assays. We also identify metabolic genes that potentially dictate the outcome of T cell differentiation, by ranking them based on their expression dynamics. Among them, we find asparagine synthetase (Asns), whose expression peaks for effector T cells and decays toward memory formation. Disrupting these expression dynamics by ASNS overexpression promotes an effector phenotype, enhancing the anti-tumor response of adoptively transferred CD8+ T cells in a mouse melanoma model. We thus provide a resource of dynamic expression changes during CD8+ T cell activation and differentiation, and identify ASNS expression dynamics as a modulator of CD8+ T cell differentiation., Competing Interests: Declaration of interests P.-C.H. is on the scientific advisory board for Elixiron Immunotherapeutics, Acepodia, and Novartis; has received funding from Elixiron Immunotherapeutics; and is the founder of Pilatus Biosciences. S.-M.F. has received funding from Bayer, Merck, Black Belt Therapeutics, and Alesta Therapeutics; has consulted for Fund+; and is on the scientific advisory board for Alesta Therapeutics and the editorial board of Cell Reports., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
21. Skeletal progenitors preserve proliferation and self-renewal upon inhibition of mitochondrial respiration by rerouting the TCA cycle.
- Author
-
Tournaire G, Loopmans S, Stegen S, Rinaldi G, Eelen G, Torrekens S, Moermans K, Carmeliet P, Ghesquière B, Thienpont B, Fendt SM, van Gastel N, and Carmeliet G
- Subjects
- Cell Proliferation, Energy Metabolism physiology, Respiration, Citric Acid Cycle, Mitochondria metabolism
- Abstract
A functional electron transport chain (ETC) is crucial for supporting bioenergetics and biosynthesis. Accordingly, ETC inhibition decreases proliferation in cancer cells but does not seem to impair stem cell proliferation. However, it remains unclear how stem cells metabolically adapt. In this study, we show that pharmacological inhibition of complex III of the ETC in skeletal stem and progenitor cells induces glycolysis side pathways and reroutes the tricarboxylic acid (TCA) cycle to regenerate NAD
+ and preserve cell proliferation. These metabolic changes also culminate in increased succinate and 2-hydroxyglutarate levels that inhibit Ten-eleven translocation (TET) DNA demethylase activity, thereby preserving self-renewal and multilineage potential. Mechanistically, mitochondrial malate dehydrogenase and reverse succinate dehydrogenase activity proved to be essential for the metabolic rewiring in response to ETC inhibition. Together, these data show that the metabolic plasticity of skeletal stem and progenitor cells allows them to bypass ETC blockade and preserve their self-renewal., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
22. 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
23. Combined glucocorticoid resistance and hyperlactatemia contributes to lethal shock in sepsis.
- Author
-
Vandewalle J, Timmermans S, Paakinaho V, Vancraeynest L, Dewyse L, Vanderhaeghen T, Wallaeys C, Van Wyngene L, Van Looveren K, Nuyttens L, Eggermont M, Dewaele S, Velho TR, Moita LF, Weis S, Sponholz C, van Grunsven LA, Dewerchin M, Carmeliet P, De Bosscher K, Van de Voorde J, Palvimo JJ, and Libert C
- Subjects
- Animals, Glucocorticoids, Lactic Acid, Mice, Receptors, Glucocorticoid metabolism, Vascular Endothelial Growth Factor A, Hyperlactatemia, Sepsis complications, Sepsis metabolism
- Abstract
Sepsis is a potentially lethal syndrome resulting from a maladaptive response to infection. Upon infection, glucocorticoids are produced as a part of the compensatory response to tolerate sepsis. This tolerance is, however, mitigated in sepsis due to a quickly induced glucocorticoid resistance at the level of the glucocorticoid receptor. Here, we show that defects in the glucocorticoid receptor signaling pathway aggravate sepsis pathophysiology by lowering lactate clearance and sensitizing mice to lactate-induced toxicity. The latter is exerted via an uncontrolled production of vascular endothelial growth factor, resulting in vascular leakage and collapse with severe hypotension, organ damage, and death, all being typical features of a lethal form of sepsis. In conclusion, sepsis leads to glucocorticoid receptor failure and hyperlactatemia, which collectively leads to a lethal vascular collapse., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
24. 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
25. Tumor vessel co-option probed by single-cell analysis.
- Author
-
Teuwen LA, De Rooij LPMH, Cuypers A, Rohlenova K, Dumas SJ, García-Caballero M, Meta E, Amersfoort J, Taverna F, Becker LM, Veiga N, Cantelmo AR, Geldhof V, Conchinha NV, Kalucka J, Treps L, Conradi LC, Khan S, Karakach TK, Soenen S, Vinckier S, Schoonjans L, Eelen G, Van Laere S, Dewerchin M, Dirix L, Mazzone M, Luo Y, Vermeulen P, and Carmeliet P
- Subjects
- Animals, Cell Line, Tumor, Endothelial Cells pathology, Female, Kidney Neoplasms pathology, Lung Neoplasms secondary, Macrophages pathology, Mice, Inbred BALB C, Myeloid Cells pathology, Pericytes pathology, Mice, Neoplasms blood supply, Neoplasms pathology, Single-Cell Analysis
- Abstract
Tumor vessel co-option is poorly understood, yet it is a resistance mechanism against anti-angiogenic therapy (AAT). The heterogeneity of co-opted endothelial cells (ECs) and pericytes, co-opting cancer and myeloid cells in tumors growing via vessel co-option, has not been investigated at the single-cell level. Here, we use a murine AAT-resistant lung tumor model, in which VEGF-targeting induces vessel co-option for continued growth. Single-cell RNA sequencing (scRNA-seq) of 31,964 cells reveals, unexpectedly, a largely similar transcriptome of co-opted tumor ECs (TECs) and pericytes as their healthy counterparts. Notably, we identify cell types that might contribute to vessel co-option, i.e., an invasive cancer-cell subtype, possibly assisted by a matrix-remodeling macrophage population, and another M1-like macrophage subtype, possibly involved in keeping or rendering vascular cells quiescent., Competing Interests: Declaration of interest The authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
26. High-fat diet-activated fatty acid oxidation mediates intestinal stemness and tumorigenicity.
- Author
-
Mana MD, Hussey AM, Tzouanas CN, Imada S, Barrera Millan Y, Bahceci D, Saiz DR, Webb AT, Lewis CA, Carmeliet P, Mihaylova MM, Shalek AK, and Yilmaz ÖH
- Subjects
- Animals, Humans, Mice, Oxidation-Reduction, Carcinogenesis pathology, Diet, High-Fat adverse effects, Fatty Acids metabolism, Intestines pathology, Obesity physiopathology, PPAR alpha metabolism, Stem Cells metabolism
- Abstract
Obesity is an established risk factor for cancer in many tissues. In the mammalian intestine, a pro-obesity high-fat diet (HFD) promotes regeneration and tumorigenesis by enhancing intestinal stem cell (ISC) numbers, proliferation, and function. Although PPAR (peroxisome proliferator-activated receptor) nuclear receptor activity has been proposed to facilitate these effects, their exact role is unclear. Here we find that, in loss-of-function in vivo models, PPARα and PPARδ contribute to the HFD response in ISCs. Mechanistically, both PPARs do so by robustly inducing a downstream fatty acid oxidation (FAO) metabolic program. Pharmacologic and genetic disruption of CPT1A (the rate-controlling enzyme of mitochondrial FAO) blunts the HFD phenotype in ISCs. Furthermore, inhibition of CPT1A dampens the pro-tumorigenic consequences of a HFD on early tumor incidence and progression. These findings demonstrate that inhibition of a HFD-activated FAO program creates a therapeutic opportunity to counter the effects of a HFD on ISCs and intestinal tumorigenesis., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
27. Neutrophils fuel effective immune responses through gluconeogenesis and glycogenesis.
- Author
-
Sadiku P, Willson JA, Ryan EM, Sammut D, Coelho P, Watts ER, Grecian R, Young JM, Bewley M, Arienti S, Mirchandani AS, Sanchez Garcia MA, Morrison T, Zhang A, Reyes L, Griessler T, Jheeta P, Paterson GG, Graham CJ, Thomson JP, Baillie K, Thompson AAR, Morgan JM, Acosta-Sanchez A, Dardé VM, Duran J, Guinovart JJ, Rodriguez-Blanco G, Von Kriegsheim A, Meehan RR, Mazzone M, Dockrell DH, Ghesquiere B, Carmeliet P, Whyte MKB, and Walmsley SR
- Published
- 2021
- Full Text
- View/download PDF
28. 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
29. 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
30. Glutamine Metabolism Controls Chondrocyte Identity and Function.
- Author
-
Stegen S, Rinaldi G, Loopmans S, Stockmans I, Moermans K, Thienpont B, Fendt SM, Carmeliet P, and Carmeliet G
- Subjects
- Animals, Cell Differentiation, Cell Proliferation, Cells, Cultured, Chondrocytes cytology, Chondrocytes physiology, Female, Glutaminase metabolism, Male, Mice, SOX9 Transcription Factor metabolism, Chondrocytes metabolism, Glutamine metabolism
- Abstract
Correct functioning of chondrocytes is crucial for long bone growth and fracture repair. These cells are highly anabolic but survive and function in an avascular environment, implying specific metabolic requirements that are, however, poorly characterized. Here, we show that chondrocyte identity and function are closely linked with glutamine metabolism in a feedforward process. The master chondrogenic transcription factor SOX9 stimulates glutamine metabolism by increasing glutamine consumption and levels of glutaminase 1 (GLS1), a rate-controlling enzyme in this pathway. Consecutively, GLS1 action is critical for chondrocyte properties and function via a tripartite mechanism. First, glutamine controls chondrogenic gene expression epigenetically through glutamate dehydrogenase-dependent acetyl-CoA synthesis, necessary for histone acetylation. Second, transaminase-mediated aspartate synthesis supports chondrocyte proliferation and matrix synthesis. Third, glutamine-derived glutathione synthesis avoids harmful reactive oxygen species accumulation and allows chondrocyte survival in the avascular growth plate. Collectively, our study identifies glutamine as a metabolic regulator of cartilage fitness during bone development., 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
31. Endothelial Lactate Controls Muscle Regeneration from Ischemia by Inducing M2-like Macrophage Polarization.
- Author
-
Zhang J, Muri J, Fitzgerald G, Gorski T, Gianni-Barrera R, Masschelein E, D'Hulst G, Gilardoni P, Turiel G, Fan Z, Wang T, Planque M, Carmeliet P, Pellerin L, Wolfrum C, Fendt SM, Banfi A, Stockmann C, Soro-Arnáiz I, Kopf M, and De Bock K
- Subjects
- Animals, Cells, Cultured, Ischemia pathology, Macrophage Activation drug effects, Macrophages metabolism, Mice, Mice, Knockout, Mice, Transgenic, Muscle, Skeletal metabolism, Endothelial Cells chemistry, Ischemia metabolism, Lactates pharmacology, Macrophages drug effects, Muscle, Skeletal drug effects
- Abstract
Endothelial cell (EC)-derived signals contribute to organ regeneration, but angiocrine metabolic communication is not described. We found that EC-specific loss of the glycolytic regulator pfkfb3 reduced ischemic hindlimb revascularization and impaired muscle regeneration. This was caused by the reduced ability of macrophages to adopt a proangiogenic and proregenerative M2-like phenotype. Mechanistically, loss of pfkfb3 reduced lactate secretion by ECs and lowered lactate levels in the ischemic muscle. Addition of lactate to pfkfb3-deficient ECs restored M2-like polarization in an MCT1-dependent fashion. Lactate shuttling by ECs enabled macrophages to promote proliferation and fusion of muscle progenitors. Moreover, VEGF production by lactate-polarized macrophages was increased, resulting in a positive feedback loop that further stimulated angiogenesis. Finally, increasing lactate levels during ischemia rescued macrophage polarization and improved muscle reperfusion and regeneration, whereas macrophage-specific mct1 deletion prevented M2-like polarization. In summary, ECs exploit glycolysis for angiocrine lactate shuttling to steer muscle regeneration from ischemia., Competing Interests: Declaration of interests Authors declare no competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
32. 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
33. 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
34. 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
35. Loss or Inhibition of Stromal-Derived PlGF Prolongs Survival of Mice with Imatinib-Resistant Bcr-Abl1 + Leukemia.
- Author
-
Schmidt T, Kharabi Masouleh B, Loges S, Cauwenberghs S, Fraisl P, Maes C, Jonckx B, De Keersmaecker K, Kleppe M, Tjwa M, Schenk T, Vinckier S, Fragoso R, De Mol M, Beel K, Dias S, Verfaillie C, Clark RE, Brümmendorf TH, Vandenberghe P, Rafii S, Holyoake T, Hochhaus A, Cools J, Karin M, Carmeliet G, Dewerchin M, and Carmeliet P
- Published
- 2020
- Full Text
- View/download PDF
36. Closing the Mitochondrial Permeability Transition Pore in hiPSC-Derived Endothelial Cells Induces Glycocalyx Formation and Functional Maturation.
- Author
-
Tiemeier GL, Wang G, Dumas SJ, Sol WMPJ, Avramut MC, Karakach T, Orlova VV, van den Berg CW, Mummery CL, Carmeliet P, van den Berg BM, and Rabelink TJ
- Subjects
- Cell Differentiation, Cell Line, Endothelial Cells metabolism, Humans, Induced Pluripotent Stem Cells metabolism, Mitochondria ultrastructure, Mitochondrial Permeability Transition Pore, Reactive Oxygen Species metabolism, Endothelial Cells cytology, Glycocalyx metabolism, Induced Pluripotent Stem Cells cytology, Mitochondria metabolism, Mitochondrial Membrane Transport Proteins metabolism
- Abstract
Human induced pluripotent stem cells (hiPSCs) are used to study organogenesis and model disease as well as being developed for regenerative medicine. Endothelial cells are among the many cell types differentiated from hiPSCs, but their maturation and stabilization fall short of that in adult endothelium. We examined whether shear stress alone or in combination with pericyte co-culture would induce flow alignment and maturation of hiPSC-derived endothelial cells (hiPSC-ECs) but found no effects comparable with those in primary microvascular ECs. In addition, hiPSC-ECs lacked a luminal glycocalyx, critical for vasculature homeostasis, shear stress sensing, and signaling. We noted, however, that hiPSC-ECs have dysfunctional mitochondrial permeability transition pores, resulting in reduced mitochondrial function and increased reactive oxygen species. Closure of these pores by cyclosporine A improved EC mitochondrial function but also restored the glycocalyx such that alignment to flow took place. These results indicated that mitochondrial maturation is required for proper hiPSC-EC functionality., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
37. Leigh Syndrome Mouse Model Can Be Rescued by Interventions that Normalize Brain Hyperoxia, but Not HIF Activation.
- Author
-
Jain IH, Zazzeron L, Goldberger O, Marutani E, Wojtkiewicz GR, Ast T, Wang H, Schleifer G, Stepanova A, Brepoels K, Schoonjans L, Carmeliet P, Galkin A, Ichinose F, Zapol WM, and Mootha VK
- Subjects
- Anemia metabolism, Animals, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Brain pathology, Disease Models, Animal, Hyperoxia metabolism, Hypoxia metabolism, Hypoxia-Inducible Factor 1 genetics, Hypoxia-Inducible Factor 1 metabolism, Leigh Disease metabolism, Mice, Brain metabolism, Carbon Monoxide therapeutic use, Hyperoxia therapy, Leigh Disease therapy, Oxygen metabolism
- Abstract
Leigh syndrome is a devastating mitochondrial disease for which there are no proven therapies. We previously showed that breathing chronic, continuous hypoxia can prevent and even reverse neurological disease in the Ndufs4 knockout (KO) mouse model of complex I (CI) deficiency and Leigh syndrome. Here, we show that genetic activation of the hypoxia-inducible factor transcriptional program via any of four different strategies is insufficient to rescue disease. Rather, we observe an age-dependent decline in whole-body oxygen consumption. These mice exhibit brain tissue hyperoxia, which is normalized by hypoxic breathing. Alternative experimental strategies to reduce oxygen delivery, including breathing carbon monoxide (600 ppm in air) or severe anemia, can reverse neurological disease. Therefore, unused oxygen is the most likely culprit in the pathology of this disease. While pharmacologic activation of the hypoxia response is unlikely to alleviate disease in vivo, interventions that safely normalize brain tissue hyperoxia may hold therapeutic potential., (Copyright © 2019. Published by Elsevier Inc.)
- Published
- 2019
- Full Text
- View/download PDF
38. Hallmarks of Endothelial Cell Metabolism in Health and Disease.
- Author
-
Li X, Sun X, and Carmeliet P
- Subjects
- Animals, Fibroblast Growth Factors metabolism, Glycolysis, Humans, Mice, Neoplasms metabolism, Retinal Diseases metabolism, Vascular Diseases metabolism, Vascular Endothelial Growth Factors antagonists & inhibitors, Vascular Endothelial Growth Factors metabolism, Endothelial Cells metabolism, Neovascularization, Pathologic metabolism, Neovascularization, Physiologic physiology
- Abstract
In 2009, it was postulated that endothelial cells (ECs) would only be able to execute the orders of growth factors if these cells would accordingly adapt their metabolism. Ten years later, it has become clear that ECs, often differently from other cell types, rely on distinct metabolic pathways to survive and form new blood vessels; that manipulation of EC metabolic pathways alone (even without changing angiogenic signaling) suffices to alter vessel sprouting; and that perturbations of these metabolic pathways can underlie excess formation of new blood vessels (angiogenesis) in cancer and ocular diseases. Initial proof of evidence has been provided that targeting (normalizing) these metabolic perturbations in diseased ECs and delivery of metabolites deserve increasing attention as novel therapeutic approaches for inhibiting or stimulating vessel growth in multiple disorders., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
39. Metabolic and Innate Immune Cues Merge into a Specific Inflammatory Response via the UPR.
- Author
-
Mogilenko DA, Haas JT, L'homme L, Fleury S, Quemener S, Levavasseur M, Becquart C, Wartelle J, Bogomolova A, Pineau L, Molendi-Coste O, Lancel S, Dehondt H, Gheeraert C, Melchior A, Dewas C, Nikitin A, Pic S, Rabhi N, Annicotte JS, Oyadomari S, Velasco-Hernandez T, Cammenga J, Foretz M, Viollet B, Vukovic M, Villacreces A, Kranc K, Carmeliet P, Marot G, Boulter A, Tavernier S, Berod L, Longhi MP, Paget C, Janssens S, Staumont-Sallé D, Aksoy E, Staels B, and Dombrowicz D
- Published
- 2019
- Full Text
- View/download PDF
40. 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
41. 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
42. 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
43. CPT1a-Dependent Long-Chain Fatty Acid Oxidation Contributes to Maintaining Glucagon Secretion from Pancreatic Islets.
- Author
-
Briant LJB, Dodd MS, Chibalina MV, Rorsman NJG, Johnson PRV, Carmeliet P, Rorsman P, and Knudsen JG
- Subjects
- Adenosine Triphosphate metabolism, Animals, Blood Glucose analysis, Carnitine O-Palmitoyltransferase antagonists & inhibitors, Carnitine O-Palmitoyltransferase genetics, Fatty Acids chemistry, Glucose metabolism, Glucose pharmacology, Humans, KATP Channels metabolism, Membrane Potentials drug effects, Metabolic Networks and Pathways, Mice, Mice, Inbred C57BL, Mice, Knockout, Protein Isoforms genetics, Protein Isoforms metabolism, RNA Interference, RNA, Small Interfering metabolism, Sodium-Potassium-Exchanging ATPase metabolism, Carnitine O-Palmitoyltransferase metabolism, Fatty Acids metabolism, Glucagon metabolism, Islets of Langerhans metabolism
- Abstract
Glucagon, the principal hyperglycemic hormone, is secreted from pancreatic islet α cells as part of the counter-regulatory response to hypoglycemia. Hence, secretory output from α cells is under high demand in conditions of low glucose supply. Many tissues oxidize fat as an alternate energy substrate. Here, we show that glucagon secretion in low glucose conditions is maintained by fatty acid metabolism in both mouse and human islets, and that inhibiting this metabolic pathway profoundly decreases glucagon output by depolarizing α cell membrane potential and decreasing action potential amplitude. We demonstrate, by using experimental and computational approaches, that this is not mediated by the K
ATP channel, but instead due to reduced operation of the Na+ -K+ pump. These data suggest that counter-regulatory secretion of glucagon is driven by fatty acid metabolism, and that the Na+ -K+ pump is an important ATP-dependent regulator of α cell function., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
44. Emerging Concepts in Organ-Specific Lymphatic Vessels and Metabolic Regulation of Lymphatic Development.
- Author
-
Wong BW, Zecchin A, García-Caballero M, and Carmeliet P
- Subjects
- Animals, Humans, Lymphangiogenesis physiology, Lymphatic System physiology, Lymphatic Vessels physiology, Metabolic Networks and Pathways, Metabolic Syndrome physiopathology
- Abstract
The lymphatic system has been less well characterized than the blood vascular system; however, work in recent years has uncovered novel regulators and non-venous lineages that contribute to lymphatic formation in various organs. Further, the identification of organ-specific lymphatic beds underscores their potential interaction with organ development and function, and highlights the possibility of targeting these organ-specific lymphatics beds in disease. This review focuses on newly described metabolic and epigenetic regulators of lymphangiogenesis and the interplay between lymphatic development and function in a number of major organ systems., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
45. Fasting Activates Fatty Acid Oxidation to Enhance Intestinal Stem Cell Function during Homeostasis and Aging.
- Author
-
Mihaylova MM, Cheng CW, Cao AQ, Tripathi S, Mana MD, Bauer-Rowe KE, Abu-Remaileh M, Clavain L, Erdemir A, Lewis CA, Freinkman E, Dickey AS, La Spada AR, Huang Y, Bell GW, Deshpande V, Carmeliet P, Katajisto P, Sabatini DM, and Yilmaz ÖH
- Subjects
- Animals, Cells, Cultured, Mice, Mice, Inbred Strains, Oxidation-Reduction, Aging, Fasting metabolism, Fatty Acids metabolism, Homeostasis, Intestines cytology, Stem Cells cytology, Stem Cells metabolism
- Abstract
Diet has a profound effect on tissue regeneration in diverse organisms, and low caloric states such as intermittent fasting have beneficial effects on organismal health and age-associated loss of tissue function. The role of adult stem and progenitor cells in responding to short-term fasting and whether such responses improve regeneration are not well studied. Here we show that a 24 hr fast augments intestinal stem cell (ISC) function in young and aged mice by inducing a fatty acid oxidation (FAO) program and that pharmacological activation of this program mimics many effects of fasting. Acute genetic disruption of Cpt1a, the rate-limiting enzyme in FAO, abrogates ISC-enhancing effects of fasting, but long-term Cpt1a deletion decreases ISC numbers and function, implicating a role for FAO in ISC maintenance. These findings highlight a role for FAO in mediating pro-regenerative effects of fasting in intestinal biology, and they may represent a viable strategy for enhancing intestinal regeneration., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
46. The Receptor Tyrosine Kinase AXL Is Required at Multiple Steps of the Metastatic Cascade during HER2-Positive Breast Cancer Progression.
- Author
-
Goyette MA, Duhamel S, Aubert L, Pelletier A, Savage P, Thibault MP, Johnson RM, Carmeliet P, Basik M, Gaboury L, Muller WJ, Park M, Roux PP, Gratton JP, and Côté JF
- Subjects
- Animals, Breast Neoplasms genetics, Breast Neoplasms pathology, Female, Heterografts, Humans, Mice, Neoplasm Invasiveness, Neoplasm Metastasis, Neoplasm Transplantation, Proto-Oncogene Proteins antagonists & inhibitors, Proto-Oncogene Proteins genetics, Receptor Protein-Tyrosine Kinases antagonists & inhibitors, Receptor Protein-Tyrosine Kinases genetics, Receptor, ErbB-2 genetics, Axl Receptor Tyrosine Kinase, Breast Neoplasms mortality, Epithelial-Mesenchymal Transition, Proto-Oncogene Proteins metabolism, Receptor Protein-Tyrosine Kinases metabolism, Receptor, ErbB-2 metabolism
- Abstract
AXL is activated by its ligand GAS6 and is expressed in triple-negative breast cancer cells. In the current study, we report AXL expression in HER2-positive (HER2
+ ) breast cancers where it correlates with poor patient survival. Using murine models of HER2+ breast cancer, Axl, but not its ligand Gas6, was found to be essential for metastasis. We determined that AXL is required for intravasation, extravasation, and growth at the metastatic site. We found that AXL is expressed in HER2+ cancers displaying epithelial-to-mesenchymal transition (EMT) signatures where it contributes to sustain EMT. Interfering with AXL in a patient-derived xenograft (PDX) impaired transforming growth factor β (TGF-β)-induced cell invasion. Last, pharmacological inhibition of AXL specifically decreased the metastatic burden of mice developing HER2+ breast cancer. Our data identify AXL as a potential anti-metastatic co-therapeutic target for the treatment of HER2+ breast cancers., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
47. A Fatty Acid Oxidation-Dependent Metabolic Shift Regulates Adult Neural Stem Cell Activity.
- Author
-
Knobloch M, Pilz GA, Ghesquière B, Kovacs WJ, Wegleiter T, Moore DL, Hruzova M, Zamboni N, Carmeliet P, and Jessberger S
- Subjects
- Animals, Carnitine O-Palmitoyltransferase deficiency, Carnitine O-Palmitoyltransferase metabolism, Cell Cycle, Cell Proliferation, Hippocampus enzymology, Malonyl Coenzyme A metabolism, Mice, Knockout, Neural Stem Cells cytology, Neural Stem Cells enzymology, Neurogenesis, Oxidation-Reduction, Fatty Acids metabolism, Neural Stem Cells metabolism
- Abstract
Hippocampal neurogenesis is important for certain forms of cognition, and failing neurogenesis has been implicated in neuropsychiatric diseases. The neurogenic capacity of hippocampal neural stem/progenitor cells (NSPCs) depends on a balance between quiescent and proliferative states. Here, we show that the rate of fatty acid oxidation (FAO) regulates the activity of NSPCs. Quiescent NSPCs show high levels of carnitine palmitoyltransferase 1a (Cpt1a)-dependent FAO, which is downregulated in proliferating NSPCs. Pharmacological inhibition and conditional deletion of Cpt1a in vitro and in vivo leads to altered NSPC behavior, showing that Cpt1a-dependent FAO is required for stem cell maintenance and proper neurogenesis. Strikingly, manipulation of malonyl-CoA, the metabolite that regulates levels of FAO, is sufficient to induce exit from quiescence and to enhance NSPC proliferation. Thus, the data presented here identify a shift in FAO metabolism that governs NSPC behavior and suggest an instructive role for fatty acid metabolism in regulating NSPC activity., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
48. Myocardial Infarction Primes Autoreactive T Cells through Activation of Dendritic Cells.
- Author
-
Van der Borght K, Scott CL, Nindl V, Bouché A, Martens L, Sichien D, Van Moorleghem J, Vanheerswynghels M, De Prijck S, Saeys Y, Ludewig B, Gillebert T, Guilliams M, Carmeliet P, and Lambrecht BN
- Subjects
- Animals, CD11c Antigen metabolism, CD4-Positive T-Lymphocytes immunology, Cell Movement, Interferon Regulatory Factors metabolism, Lymph Nodes metabolism, Mice, Inbred BALB C, Mice, Inbred C57BL, Monocytes pathology, Myocardium pathology, Myosins metabolism, Phenotype, Transcription Factors metabolism, Dendritic Cells immunology, Myocardial Infarction immunology, Myocardial Infarction pathology, T-Lymphocytes immunology
- Abstract
Peripheral tolerance is crucial for avoiding activation of self-reactive T cells to tissue-restricted antigens. Sterile tissue injury can break peripheral tolerance, but it is unclear how autoreactive T cells get activated in response to self. An example of a sterile injury is myocardial infarction (MI). We hypothesized that tissue necrosis is an activator of dendritic cells (DCs), which control tolerance to self-antigens. DC subsets of a murine healthy heart consisted of IRF8-dependent conventional (c)DC1, IRF4-dependent cDC2, and monocyte-derived DCs. In steady state, cardiac self-antigen α-myosin was presented in the heart-draining mediastinal lymph node (mLN) by cDC1s, driving the proliferation of antigen-specific CD4
+ TCR-M T cells and their differentiation into regulatory cells (Tregs). Following MI, all DC subsets infiltrated the heart, whereas only cDCs migrated to the mLN. Here, cDC2s induced TCR-M proliferation and differentiation into interleukin-(IL)-17/interferon-(IFN)γ-producing effector cells. Thus, cardiac-specific autoreactive T cells get activated by mature DCs following myocardial infarction., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
49. Neurogenic Radial Glia-like Cells in Meninges Migrate and Differentiate into Functionally Integrated Neurons in the Neonatal Cortex.
- Author
-
Bifari F, Decimo I, Pino A, Llorens-Bobadilla E, Zhao S, Lange C, Panuccio G, Boeckx B, Thienpont B, Vinckier S, Wyns S, Bouché A, Lambrechts D, Giugliano M, Dewerchin M, Martin-Villalba A, and Carmeliet P
- Subjects
- Animals, Animals, Newborn, Cell Lineage, Embryo, Mammalian cytology, Excitatory Amino Acid Transporter 1 metabolism, Gene Expression Profiling, HEK293 Cells, Humans, Mice, Inbred C57BL, Nestin metabolism, Receptor, Platelet-Derived Growth Factor beta metabolism, Reproducibility of Results, Single-Cell Analysis, Spheroids, Cellular cytology, Staining and Labeling, Transcriptome genetics, Cell Differentiation, Cell Movement, Cerebral Cortex cytology, Meninges cytology, Neurogenesis, Neuroglia cytology, Neurons cytology
- Abstract
Whether new neurons are added in the postnatal cerebral cortex is still debated. Here, we report that the meninges of perinatal mice contain a population of neurogenic progenitors formed during embryonic development that migrate to the caudal cortex and differentiate into Satb2
+ neurons in cortical layers II-IV. The resulting neurons are electrically functional and integrated into local microcircuits. Single-cell RNA sequencing identified meningeal cells with distinct transcriptome signatures characteristic of (1) neurogenic radial glia-like cells (resembling neural stem cells in the SVZ), (2) neuronal cells, and (3) a cell type with an intermediate phenotype, possibly representing radial glia-like meningeal cells differentiating to neuronal cells. Thus, we have identified a pool of embryonically derived radial glia-like cells present in the meninges that migrate and differentiate into functional neurons in the neonatal cerebral cortex., (Copyright © 2016 Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
50. 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
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.