Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(21)2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34768857

ABSTRACT

The mammalian exclusive Orai3 channel participates in the generation and/or modulation of two independent Ca2+ currents, the store-operated current, Icrac, involving functional interactions between the stromal interaction molecules (STIM), STIM1/STIM2, and Orai1/Orai2/Orai3, as well as the store-independent arachidonic acid (AA) (or leukotriene C4)-regulated current Iarc, which involves Orai1, Orai3 and STIM1. Overexpression of functional Orai3 has been described in different neoplastic cells and cancer tissue samples as compared to non-tumor cells or normal adjacent tissue. In these cells, Orai3 exhibits a cell-specific relevance in Ca2+ influx. In estrogen receptor-positive breast cancer cells and non-small cell lung cancer (NSCLC) cells store-operated Ca2+ entry (SOCE) is strongly dependent on Orai3 expression while in colorectal cancer and pancreatic adenocarcinoma cells Orai3 predominantly modulates SOCE. On the other hand, in prostate cancer cells Orai3 expression has been associated with the formation of Orai1/Orai3 heteromeric channels regulated by AA and reduction in SOCE, thus leading to enhanced proliferation. Orai3 overexpression is associated with supporting several cancer hallmarks, including cell cycle progression, proliferation, migration, and apoptosis resistance. This review summarizes the current knowledge concerning the functional role of Orai3 in the pathogenesis of cancer.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling/physiology , Neoplasms/pathology , Animals , Apoptosis/physiology , Calcium/metabolism , Calcium Channels/genetics , Cell Cycle/physiology , Cell Movement/physiology , Cell Proliferation/physiology , Humans
2.
STAR Protoc ; 2(3): 100508, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34585146

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).


Subject(s)
Brain/cytology , Choroid/cytology , Endothelial Cells/cytology , Lung/cytology , Muscles/cytology , Animals , Flow Cytometry/methods , Male , Mice , Mice, Inbred C57BL
3.
STAR Protoc ; 2(3): 100523, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34382011

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).


Subject(s)
Endothelial Cells/cytology , Kidney/cytology , Spleen/cytology , Testis/cytology , Animals , Flow Cytometry , Male , Mice
4.
STAR Protoc ; 2(2): 100489, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34007969

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).


Subject(s)
Endothelial Cells/cytology , Flow Cytometry/methods , Intestines/cytology , Liver/cytology , Myocardium/cytology , Animals , Cells, Cultured , Male , Mice , Mice, Inbred C57BL
5.
Biochim Biophys Acta Mol Cell Res ; 1868(7): 119023, 2021 06.
Article in English | MEDLINE | ID: mdl-33798603

ABSTRACT

Changes in cytosolic free Ca2+ concentration play a central role in many fundamental cellular processes including muscle contraction, neurotransmission, cell proliferation, differentiation, gene transcription and cell death. Many of these processes are known to be regulated by store-operated calcium channels (SOCs), among which ORAI1 is the most studied in cancer cells, leaving the role of other ORAI channels yet inadequately addressed. Here we demonstrate that ORAI3 channels are expressed in both normal (HPDE) and pancreatic ductal adenocarcinoma (PDAC) cell lines, where they form functional channels, their knockdown affecting store operated calcium entry (SOCE). More specifically, ORAI3 silencing increased SOCE in PDAC cell lines, while decreasing SOCE in normal pancreatic cell line. We also show the role of ORAI3 in proliferation, cell cycle, viability, mitotic catastrophe and cell death. Finally, we demonstrate that ORAI3 silencing impairs pancreatic tumor growth and induces cell death in vivo, suggesting that ORAI3 could represent a potential therapeutic target in PDAC treatment.


Subject(s)
Calcium Channels/metabolism , Pancreatic Neoplasms/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Apoptosis/genetics , Calcium/metabolism , Calcium Channels/genetics , Calcium Signaling/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing/physiology , Humans , Mitosis/genetics , ORAI1 Protein/metabolism , Pancreatic Neoplasms/metabolism , RNA, Small Interfering/metabolism , Pancreatic Neoplasms
6.
J Autism Dev Disord ; 51(12): 4587-4604, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33586083

ABSTRACT

Episodic future thinking (EFT) has been suggested to underlie anticipatory pleasure (AP), itself known to play a crucial role in social functioning (SF). Both AP and SF are impaired in various clinical populations, including autism spectrum disorders (ASD) and 22q11.2 deletion syndrome (22q11DS). Therefore, the relationship between EFT, AP and SF was investigated, as well as the potential role of projecting oneself in a social vs. non-social context. Seventy-seven participants [24 with 22q11DS, 20 with ASD, 33 typically developing controls (TDs)] (aged 12-25) were included. They were assessed with a future thinking task in which they were asked to recall a memory and produce a likely event. Narratives were rated based of specificity, richness and imaginability. Participants completed questionnaires assessing AP and SF. Narratives from ASD and 22q11DS participants were rated as less vivid compared to TDs. However, the characteristics of the narratives differed between ASD and 22q11DS participants in terms of specificity and level of details, as well as in reaction to social condition. Moreover, correlations were found between AP and EFT in both ASD and 22q11DS participants, and between SF and EFT in ASD participants. These results point towards impairments in EFT in both ASD and 22q11DS participants but with a specific profile in each condition. The observed associations between EFT and AP suggest that decreased autonoetic consciousness might underlie AP impairments. In ASD individuals, the association between SF and EFT highlights the need to better characterize EFT since EFT could be another mechanism contributing to social difficulties.


Subject(s)
Autism Spectrum Disorder , DiGeorge Syndrome , Memory, Episodic , Humans , Mental Recall , Pleasure , Social Interaction
7.
iScience ; 23(7): 101263, 2020 Jul 24.
Article in English | MEDLINE | ID: mdl-32585596

ABSTRACT

Mitochondria are important cell death checkpoints, and mitochondrial Ca2+ overload is considered as a potent apoptotic intrinsic pathway inducer. Here, we report that this Ca2+ apoptosis link is largely ineffective in inducing cell-death just by itself and required a concomitant inhibition of autophagy to counteract its pro-survival action. In such condition, an acute mitochondrial stress revealed by a DRP1-mediated mitochondrial dynamic remodeling is observed concomitantly with mitochondrial depolarization, release of cytochrome c, and efficient apoptosis induction. We also uncover that mitochondrial Ca2+ status modulates the function of autophagy as a sensitizer for chemotherapies. This priming mediated by mitochondrial Ca2+ overload and inhibition of autophagy sensitizes many cancer cells types to different chemotherapies with independent mechanisms of action. Collectively, our results redefine an important cell signaling pathway, uncovering new combined therapies for the treatment of diseases associated with mitochondrial Ca2+ homeostasis disorders such as cancer.

8.
Cell ; 180(4): 764-779.e20, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32059779

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.


Subject(s)
Endothelial Cells/metabolism , Single-Cell Analysis , Transcriptome , 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
10.
Genet Med ; 21(2): 441-450, 2019 02.
Article in English | MEDLINE | ID: mdl-29930394

ABSTRACT

PURPOSE: Malignant hyperthermia (MH) is a pharmacogenetic disorder arising from uncontrolled muscle calcium release due to an abnormality in the sarcoplasmic reticulum (SR) calcium-release mechanism triggered by halogenated inhalational anesthetics. However, the molecular mechanisms involved are still incomplete. METHODS: We aimed to identify transient receptor potential vanilloid 1 (TRPV1) variants within the entire coding sequence in patients who developed sensitivity to MH of unknown etiology. In vitro and in vivo functional studies were performed in heterologous expression system, trpv1-/- mice, and a murine model of human MH. RESULTS: We identified TRPV1 variants in two patients and their heterologous expression in muscles of trpv1-/- mice strongly enhanced calcium release from SR upon halogenated anesthetic stimulation, suggesting they could be responsible for the MH phenotype. We confirmed the in vivo significance by using mice with a knock-in mutation (Y524S) in the type I ryanodine receptor (Ryr1), a mutation analogous to the Y522S mutation associated with MH in humans. We showed that the TRPV1 antagonist capsazepine slows the heat-induced hypermetabolic response in this model. CONCLUSION: We propose that TRPV1 contributes to MH and could represent an actionable therapeutic target for prevention of the pathology and also be responsible for MH sensitivity when mutated.


Subject(s)
Calcium Signaling , Genetic Predisposition to Disease , Malignant Hyperthermia/genetics , TRPV Cation Channels/genetics , Anesthetics/pharmacology , Animals , Calcium , Disease Models, Animal , Female , Gene Expression/drug effects , Gene Knock-In Techniques , HEK293 Cells , Homeostasis , Humans , Male , Malignant Hyperthermia/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , TRPV Cation Channels/metabolism
11.
Methods Mol Biol ; 1843: 95-106, 2018.
Article in English | MEDLINE | ID: mdl-30203280

ABSTRACT

Since last decade real-time qPCR has become a routine and robust approach for measuring the expression of genes of interest. Indeed, using qPCR, expression profile analyses are now possible and participate to the understanding of physiological or pathological role of channels such as calcium release-activated channels (CRAC). Initially discovered in lymphocyte T and immunity perturbations, recent studies have highlighted the role of CRAC channels in other pathologies such as cancer. Here we describe a protocol to quantify CRAC components expression, in tissue sample and cell lines, to validate knockdown strategies or identify their roles in physiological and pathological conditions (Hoth and Penner, J Physiol 465:359-386, 1993; Hoth and Penner, Nature 355:353-356, 1992; and Zweifach and Lewis, Proc Natl Acad Sci U S A 90:6295-6299, 1993).


Subject(s)
Calcium Release Activated Calcium Channels/genetics , Calcium Release Activated Calcium Channels/metabolism , Gene Expression , Ion Channel Gating , Real-Time Polymerase Chain Reaction , Calcium/metabolism , Calcium Signaling , Cell Line , Humans , Real-Time Polymerase Chain Reaction/methods
12.
Methods Mol Biol ; 1843: 107-113, 2018.
Article in English | MEDLINE | ID: mdl-30203281

ABSTRACT

The characterization of protein-protein interactions through methods such as co-immunoprecipitation, followed by Western blot analysis, is a crucial step in the understanding of protein functions and the biology of the cell. Since the discovery of ORAI and STIM proteins as component of store-operated channel (SOC), overexpressing systems have been used to demonstrate how ORAI and STIM can associate with physiological and pathological conditions. Here we describe a protocol allowing endogenous studies.


Subject(s)
Blotting, Western , Carrier Proteins/metabolism , Immunoprecipitation , Protein Interaction Mapping , Stromal Interaction Molecules/metabolism , Blotting, Western/methods , Calcium/metabolism , Calcium Channels/metabolism , Cell Line, Tumor , Humans , Immunoprecipitation/methods , Protein Binding , Protein Interaction Mapping/methods
13.
Nature ; 561(7721): 63-69, 2018 09.
Article in English | MEDLINE | ID: mdl-30158707

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.


Subject(s)
Endothelial Cells/enzymology , Endothelial Cells/pathology , Glutamate-Ammonia Ligase/metabolism , Glutamine/biosynthesis , Neovascularization, Pathologic , 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
14.
Cell Metab ; 28(6): 866-880.e15, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30146486

ABSTRACT

The role of fatty acid synthesis in endothelial cells (ECs) remains incompletely characterized. We report that fatty acid synthase knockdown (FASNKD) 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.


Subject(s)
Fatty Acid Synthase, Type I/physiology , Human Umbilical Vein Endothelial Cells/metabolism , Malonyl Coenzyme A/metabolism , Retinal Neovascularization/pathology , TOR Serine-Threonine Kinases/metabolism , 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
15.
Cell Metab ; 28(6): 881-894.e13, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30146488

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 ß-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 CPT1AΔEC 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.


Subject(s)
Carnitine O-Palmitoyltransferase/metabolism , Energy Metabolism , Fatty Acids/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , NADP/metabolism , Receptor, Notch1/metabolism , Animals , Cell Proliferation , HEK293 Cells , Homeostasis , Humans , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Oxidative Stress
16.
Cell Metab ; 28(4): 573-587.e13, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30017355

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 (PHGDHKD) 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.


Subject(s)
Endothelial Cells/metabolism , Heme/metabolism , Phosphoglycerate Dehydrogenase/genetics , Phosphoglycerate Dehydrogenase/metabolism , Serine/metabolism , 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
17.
Sci Rep ; 7(1): 15896, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29162859

ABSTRACT

Despite the tremendous progress in medicine, cancer remains one of the most serious global health problems awaiting new effective therapies. Here we present ferroquine (FQ), the next generation antimalarial drug, as a promising candidate for repositioning as cancer therapeutics. We report that FQ potently inhibits autophagy, perturbs lysosomal function and impairs prostate tumor growth in vivo. We demonstrate that FQ negatively regulates Akt kinase and hypoxia-inducible factor-1α (HIF-1α) and is particularly effective in starved and hypoxic conditions frequently observed in advanced solid cancers. FQ enhances the anticancer activity of several chemotherapeutics suggesting its potential application as an adjuvant to existing anticancer therapy. Alike its parent compound chloroquine (CQ), FQ accumulates within and deacidifies lysosomes. Further, FQ induces lysosomal membrane permeabilization, mitochondrial depolarization and caspase-independent cancer cell death. Overall, our work identifies ferroquine as a promising new drug with a potent anticancer activity.


Subject(s)
Aminoquinolines/pharmacology , Antimalarials/pharmacology , Antineoplastic Agents/pharmacology , Ferrous Compounds/pharmacology , Aminoquinolines/chemistry , Animals , Antimalarials/chemistry , Autophagy/drug effects , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chloroquine/chemistry , Chloroquine/pharmacology , Female , Ferrous Compounds/chemistry , Hydrogen-Ion Concentration , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Metallocenes , Mice, Nude , Neoplasms/pathology , Permeability , Stress, Physiological , Xenograft Model Antitumor Assays
18.
Curr Opin Hematol ; 24(3): 240-247, 2017 May.
Article in English | MEDLINE | ID: mdl-28212191

ABSTRACT

PURPOSE OF REVIEW: Endothelial cell metabolism has recently emerged as an important coregulator of angiogenesis and is therefore a promising new target in various angiogenesis-associated illnesses, like cancer. In this review, we discuss recent insights in endothelial cell metabolism in both physiological and pathological conditions and discuss possible translational implications. RECENT FINDINGS: Two metabolic pathways that determine the performance of endothelial cells are glycolysis and fatty acid oxidation (FAO). Glycolysis is essential as endothelial cells primarily rely on this pathway for ATP production. 6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) is a key regulator of glycolysis in endothelial cells. As endothelial cells increase glycolysis even further during angiogenesis, PFKFB3 also controls vessel sprouting and promotes endothelial cell migration. Moreover, in tumors, additional PFKFB3 upregulation leads to a more immature and dysfunctional vasculature. PFKFB3 blockade therefore results in tumor vessel normalization, with beneficial therapeutic effects on reduced metastasis and improved chemotherapy. Also, FAO stimulates endothelial cell proliferation through affecting DNA synthesis, and is critical for lymphangiogenesis, in part through epigenetic changes in histone acetylation. As FAO is controlled by carnitine palmitoyltransferase 1a, inhibition of this key enzyme decreases pathological angiogenesis. SUMMARY: Both PFKFB3 and carnitine palmitoyltransferase 1a are key metabolic regulators of vessel sprouting and promising new therapeutic targets in diseases associated with pathological angiogenesis.


Subject(s)
Endothelial Cells/metabolism , Animals , Biomarkers , Blood Vessels/metabolism , Cell Communication , Energy Metabolism , Gene Expression Regulation , Homeostasis , Humans , Metabolic Networks and Pathways , Neoplasms/etiology , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic , Signal Transduction
19.
Front Immunol ; 8: 1750, 2017.
Article in English | MEDLINE | ID: mdl-29321777

ABSTRACT

Endothelial cells (ECs) line blood vessels, i.e., vital conduits for oxygen and nutrient delivery to distant tissues. While mostly present as quiescent "phalanx" cells throughout adult life, ECs can rapidly switch to a migratory "tip" cell and a proliferative "stalk" cell, and sprout into avascular tissue to form new blood vessels. The angiogenic switch has long been considered to be primarily orchestrated by the activity of angiogenic molecules. However, recent evidence illustrates an instrumental role of cellular metabolism in vessel sprouting, whereby ECs require specific metabolic adaptations to grow. Here, we overview the emerging picture that tip, stalk, and phalanx cells have distinct metabolic signatures and discuss how these signatures can become deregulated in pathological conditions, such as in cancer.

20.
Oncotarget ; 7(20): 29063-80, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27074561

ABSTRACT

Since its cloning a decade ago, TRPM8 channel has emerged as a promising prognostic marker and a putative therapeutic target in prostate cancer (PCa). However, recent studies have brought to light the complexity of TRPM8 isoforms in PCa. Consequently, the respective role of each TRPM8 isoform needs to be deciphered prior to considering TRPM8 as an attractive therapeutic target. Full-length (6 transmembrane (TM)-domain) TRPM8 channel is overexpressed in early PCa and repressed in advanced prostate tumors whereas the localization of the truncated, 4TM-TRPM8 channel (4 transmembrane (TM)-domain), in the membranes of endoplasmic reticulum (ER) is independent of the pathogenic status of epithelial cells. In the same line, expression of non-channel cytoplasmic small TRPM8 isoforms (namely sM8) is conserved in cancer cells. In this study, we identify sM8s as putative regulator of PCa cell death. Indeed, suppression of sM8 isoforms was found to induce concomitantly ER stress, oxidative stress, p21 expression and apoptosis in human epithelial prostate cancer cells. We furthermore demonstrate that induction of such mechanisms required the activity of 4TM-TRPM8 channels at the ER-mitochondria junction. Our study thus suggests that targeting sM8 could be an appropriate strategy to fight prostate cancer.


Subject(s)
Prostatic Neoplasms/pathology , TRPM Cation Channels/metabolism , Animals , Apoptosis/physiology , Cell Line, Tumor , Female , Heterografts , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/metabolism , Protein Isoforms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...