Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Blood Adv ; 8(11): 2870-2879, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38588481

ABSTRACT

ABSTRACT: Hemojuvelin (HJV) is a glycosylphosphatidylinositol-anchored protein of the repulsive guidance molecule family acting as a bone morphogenetic protein (BMP) coreceptor to induce the hepatic iron regulatory protein hepcidin. Hepcidin causes ubiquitination and degradation of the sole known iron exporter ferroportin, thereby limiting iron availability. The detailed signaling mechanism of HJV in vivo has yet to be investigated. In the current manuscript, we used an established model of adeno-associated virus (AAV)-mediated liver-specific overexpression of HJV in murine models of hepatocyte-specific deficiency of the BMP type I receptors Alk2 or Alk3. In control mice, HJV overexpression increased hepatic Hamp messenger RNA (mRNA) levels, soluble HJV (sHJV), splenic iron content (SIC), as well as phosphorylated small mothers against decapentaplegic protein (pSMAD1/5/8) levels. In contrast, in Alk2fl/fl;Alb-Cre and Alk3fl/fl;Alb-Cre mice, which present with moderate and severe iron overload, respectively, the administration of AAV-HJV induced HJV and sHJV. However, it did not rescue the iron overload phenotypes of those mice. Serum iron levels were induced in Alk2fl/fl;Alb-Cre mice after HJV overexpression. In phosphate-buffered saline-injected Alk3fl/fl;Alb-Cre mice, serum iron levels and the expression of duodenal ferroportin remained high, whereas Hamp mRNA levels were decreased to 1% to 5% of the levels detected in controls. This was reduced even further by AAV-HJV overexpression. SIC remained low in mice with hepatocyte-specific Alk2 or Alk3 deficiency, reflecting disturbed iron homeostasis with high serum iron levels and transferrin saturation and an inability to induce hepcidin by HJV overexpression. The data indicate that ALK2 and ALK3 are both required in vivo for the HJV-mediated induction of hepcidin.


Subject(s)
GPI-Linked Proteins , Hemochromatosis Protein , Hepcidins , Animals , Mice , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/genetics , Hepcidins/metabolism , Hepcidins/genetics , Hemochromatosis Protein/metabolism , Hemochromatosis Protein/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type I/genetics , Liver/metabolism , Iron/metabolism , Iron Overload/metabolism , Iron Overload/genetics , Activin Receptors, Type I/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type II
2.
Cancer Res ; 82(7): 1380-1395, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35105690

ABSTRACT

The activation and differentiation of cancer-associated fibroblasts (CAF) are involved in tumor progression. Here, we show that the tumor-promoting lipid mediator prostaglandin E2 (PGE2) plays a paradoxical role in CAF activation and tumor progression. Restricting PGE2 signaling via knockout of microsomal prostaglandin E synthase-1 (mPGES-1) in PyMT mice or of the prostanoid E receptor 3 (EP3) in CAFs stunted mammary carcinoma growth associated with strong CAF proliferation. CAF proliferation upon EP3 inhibition required p38 MAPK signaling. Mechanistically, TGFß-activated kinase-like protein (TAK1L), which was identified as a negative regulator of p38 MAPK activation, was decreased following ablation of mPGES-1 or EP3. In contrast with its effects on primary tumor growth, disruption of PGE2 signaling in CAFs induced epithelial-to-mesenchymal transition in cancer organoids and promoted metastasis in mice. Moreover, TAK1L expression in CAFs was associated with decreased CAF activation, reduced metastasis, and prolonged survival in human breast cancer. These data characterize a new pathway of regulating inflammatory CAF activation, which affects breast cancer progression. SIGNIFICANCE: The inflammatory lipid prostaglandin E2 suppresses cancer-associated fibroblast expansion and activation to limit primary mammary tumor growth while promoting metastasis.


Subject(s)
Breast Neoplasms , Cancer-Associated Fibroblasts , Carcinoma , Animals , Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/metabolism , Carcinoma/pathology , Dinoprostone/metabolism , Female , Fibroblasts/metabolism , Humans , Mice , Prostaglandin-E Synthases/genetics , Prostaglandin-E Synthases/metabolism , Prostaglandin-E Synthases/pharmacology
3.
Int J Mol Sci ; 22(13)2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34281173

ABSTRACT

Epoxides and diols of polyunsaturated fatty acids (PUFAs) are bioactive and can influence processes such as tumor cell proliferation and angiogenesis. Studies with inhibitors of the soluble epoxide hydrolase (sEH) in animals overexpressing cytochrome P450 enzymes or following the systemic administration of specific epoxides revealed a markedly increased incidence of tumor metastases. To determine whether PUFA epoxides increased metastases in a model of spontaneous breast cancer, sEH-/- mice were crossed onto the polyoma middle T oncogene (PyMT) background. We found that the deletion of the sEH accelerated the growth of primary tumors and increased both the tumor macrophage count and angiogenesis. There were small differences in the epoxide/diol content of tumors, particularly in epoxyoctadecamonoenic acid versus dihydroxyoctadecenoic acid, and marked changes in the expression of proteins linked with cell proliferation and metabolism. However, there was no consequence of sEH inhibition on the formation of metastases in the lymph node or lung. Taken together, our results confirm previous reports of increased tumor growth in animals lacking sEH but fail to substantiate reports of enhanced lymph node or pulmonary metastases.


Subject(s)
Breast Neoplasms/metabolism , Epoxide Hydrolases/metabolism , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogenesis , Cell Proliferation/physiology , Cell Transformation, Neoplastic , Cytochrome P-450 Enzyme System/metabolism , Disease Models, Animal , Epoxide Hydrolases/genetics , Epoxy Compounds/metabolism , Fatty Acids, Unsaturated/metabolism , Female , Gene Deletion , Mice , Mice, Knockout , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism
4.
EMBO Rep ; 22(2): e48961, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33512764

ABSTRACT

Endothelial tip cells are essential for VEGF-induced angiogenesis, but underlying mechanisms are elusive. The Ena/VASP protein family, consisting of EVL, VASP, and Mena, plays a pivotal role in axon guidance. Given that axonal growth cones and endothelial tip cells share many common features, from the morphological to the molecular level, we investigated the role of Ena/VASP proteins in angiogenesis. EVL and VASP, but not Mena, are expressed in endothelial cells of the postnatal mouse retina. Global deletion of EVL (but not VASP) compromises the radial sprouting of the vascular plexus in mice. Similarly, endothelial-specific EVL deletion compromises the radial sprouting of the vascular plexus and reduces the endothelial tip cell density and filopodia formation. Gene sets involved in blood vessel development and angiogenesis are down-regulated in EVL-deficient P5-retinal endothelial cells. Consistently, EVL deletion impairs VEGF-induced endothelial cell proliferation and sprouting, and reduces the internalization and phosphorylation of VEGF receptor 2 and its downstream signaling via the MAPK/ERK pathway. Together, we show that endothelial EVL regulates sprouting angiogenesis via VEGF receptor-2 internalization and signaling.


Subject(s)
Cell Adhesion Molecules/physiology , Endothelial Cells , Neovascularization, Physiologic , Vascular Endothelial Growth Factor Receptor-2 , Animals , Endothelial Cells/metabolism , Mice , Morphogenesis , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
5.
Theranostics ; 10(17): 7857-7871, 2020.
Article in English | MEDLINE | ID: mdl-32685025

ABSTRACT

Rationale: Vasoregression secondary to glial activation develops in various retinal diseases, including retinal degeneration and diabetic retinopathy. Photoreceptor degeneration and subsequent retinal vasoregression, characterized by pericyte loss and acellular capillary formation in the absence diabetes, are also seen in transgenic rats expressing the polycystic kidney disease (PKD) gene. Activated Müller glia contributes to retinal vasodegeneration, at least in part via the expression of the soluble epoxide hydrolase (sEH). Given that an increase in sEH expression triggered vascular destabilization in diabetes, and that vasoregression is similar in diabetic mice and PKD rats, the aim of the present study was to determine whether sEH inhibition could prevent retinal vasoregression in the PKD rat. Methods: One-month old male homozygous transgenic PKD rats were randomly allocated to receive vehicle or a sEH inhibitor (sEH-I; Sar5399, 30 mg/kg) for four weeks. Wild-type Sprague-Dawley (SD) littermates received vehicle as controls. Retinal sEH expression and activity were measured by Western blotting and LC-MS, and vasoregression was quantified in retinal digestion preparations. Microglial activation and immune response cytokines were assessed by immunofluorescence and quantitative PCR, respectively. 19,20-dihydroxydocosapentaenoic acid (19,20-DHDP) mediated Notch signaling, microglial activation and migration were assessed in vivo and in vitro. Results: This study demonstrates that sEH expression and activity were increased in PKD retinae, which led to elevated production of 19,20-DHDP and the depression of Notch signaling. The latter changes elicited pericyte loss and the recruitment of CD11b+/CD74+ microglia to the perivascular region. Microglial activation increased the expression of immune-response cytokines, and reduced levels of Notch3 and delta-like ligand 4 (Dll4). Treatment with Sar5399 decreased 19,20-DHDP generation and increased Notch3 expression. Sar5399 also prevented vasoregression by reducing pericyte loss and suppressed microglial activation as well as the expression of immune-response cytokines. Mechanistically, the activation of Notch signaling by Dll4 maintained a quiescent microglial cell phenotype, i.e. reduced both the surface presentation of CD74 and microglial migration. In contrast, in retinal explants, 19,20-DHDP and Notch inhibition both promoted CD74 expression and reversed the Dll4-induced decrease in migration. Conclusions: Our data indicate that 19,20-DHDP-induced alterations in Notch-signaling result in microglia activation and pericyte loss and contribute to retinal vasoregression in polycystic kidney disease. Moreover, sEH inhibition can ameliorate vasoregression through reduced activity of inflammatory microglia. sEH inhibition is thus an attractive new therapeutic approach to prevent retinal vasoregression.


Subject(s)
Epoxide Hydrolases/antagonists & inhibitors , Polycystic Kidney Diseases/complications , Retinal Degeneration/drug therapy , Retinal Vessels/drug effects , Animals , Disease Models, Animal , Epoxide Hydrolases/metabolism , Fatty Acids, Unsaturated/metabolism , Humans , Male , Microglia/drug effects , Microglia/immunology , Polycystic Kidney Diseases/genetics , Rats , Rats, Transgenic , Retina/cytology , Retina/drug effects , Retina/immunology , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/immunology , Retinal Degeneration/pathology , Retinal Vessels/pathology , TRPP Cation Channels/genetics
6.
Proc Natl Acad Sci U S A ; 117(11): 5923-5930, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32123095

ABSTRACT

Arachidonic acid epoxides generated by cytochrome P450 (CYP) enzymes have been linked to increased tumor growth and metastasis, largely on the basis of overexpression studies and the application of exogenous epoxides. Here we studied tumor growth and metastasis in Cyp2c44-/- mice crossed onto the polyoma middle T oncogene (PyMT) background. The resulting PyMT2c44 mice developed more primary tumors earlier than PyMT mice, with increased lymph and lung metastasis. Primary tumors from Cyp2c44-deficient mice contained higher numbers of tumor-associated macrophages, as well as more lymphatic endothelial cells than tumors from PyMT mice. While epoxide and diol levels were comparable in tumors from both genotypes, prostaglandin (PG) levels were higher in the PyMTΔ2c44 tumors. This could be accounted for by the finding that Cyp2c44 metabolized the PG precursor, PGH2 to 12(S)-hydroxyheptadeca-5Z,8E,10E-trienoic acid (12-HHT), thus effectively reducing levels of effector PGs (including PGE2). Next, proteomic analyses revealed an up-regulation of WD repeating domain FYVE1 (WDFY1) in tumors from PyMTΔ2c44 mice, a phenomenon that was reproduced in Cyp2c44-deficient macrophages as well as by PGE2 Mechanistically, WDFY1 was involved in Toll-like receptor signaling, and its down-regulation in human monocytes attenuated the LPS-induced phosphorylation of IFN regulatory factor 3 and nuclear factor-κB. Taken together, our results indicate that Cyp2c44 protects against tumor growth and metastasis by preventing the synthesis of PGE2 The latter eicosanoid influenced macrophages at least in part by enhancing Toll-like receptor signaling via the up-regulation of WDFY1.


Subject(s)
Breast Neoplasms/metabolism , Cytochrome P450 Family 2/metabolism , Lymphangiogenesis/physiology , Prostaglandins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cytochrome P450 Family 2/genetics , Disease Models, Animal , Endothelial Cells/pathology , Fatty Acids, Unsaturated/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lymphangiogenesis/genetics , Macrophages , Mice , Mice, Knockout , Monocytes , Neoplastic Processes , Proteomics , Signal Transduction , Toll-Like Receptors , Up-Regulation
7.
Acta Physiol (Oxf) ; 228(1): e13325, 2020 01.
Article in English | MEDLINE | ID: mdl-31162891

ABSTRACT

AIM: Protein kinase (PK) A anchoring protein (AKAP) 12 is a scaffolding protein that anchors PKA to compartmentalize cyclic AMP signalling. This study assessed the consequences of the downregulation or deletion of AKAP12 on endothelial cell migration and angiogenesis. METHODS: The consequences of siRNA-mediated downregulation AKAP12 were studied in primary cultures of human endothelial cells as well as in endothelial cells and retinas from wild-type versus AKAP12-/- mice. Molecular interactions were investigated using a combination of immunoprecipitation and mass spectrometry. RESULTS: AKAP12 was expressed at low levels in confluent endothelial cells but its expression was increased in actively migrating cells, where it localized to lamellipodia. In the postnatal retina, AKAP12 was expressed by actively migrating tip cells at the angiogenic front, and its deletion resulted in defective extension of the vascular plexus. In migrating endothelial cells, AKAP12 was co-localized with the PKA type II-α regulatory subunit as well as multiple key regulators of actin dynamics and actin filament-based movement; including components of the Arp2/3 complex and the vasodilator-stimulated phosphoprotein (VASP). Fitting with the evidence of a physical VASP/AKAP12/PKA complex, it was possible to demonstrate that the VEGF-stimulated and PKA-dependent phosphorylation of VASP was dependent on AKAP12. Indeed, AKAP12 colocalized with phospho-Ser157 VASP at the leading edge of migrating endothelial cells. CONCLUSION: The results suggest that compartmentalized AKAP12/PKA signalling mediates VASP phosphorylation at the leading edge of migrating endothelial cells to translate angiogenic stimuli into altered actin dynamics and cell movement.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cell Cycle Proteins/metabolism , Endothelial Cells/drug effects , Vascular Endothelial Growth Factor A/pharmacology , A Kinase Anchor Proteins/genetics , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Cycle Proteins/genetics , Cell Movement/drug effects , Cell Movement/physiology , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Endothelial Cells/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Mice , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Vascular Endothelial Growth Factor A/administration & dosage
8.
Front Oncol ; 9: 1022, 2019.
Article in English | MEDLINE | ID: mdl-31637217

ABSTRACT

IL-27 regulates inflammatory diseases by exerting a pleiotropic impact on immune cells. In cancer, IL-27 restricts tumor growth by acting on tumor cells directly, while its role in the tumor microenvironment is still controversially discussed. To explore IL-27 signaling in the tumor stroma, we used a mammary carcinoma syngraft approach in IL27Rα-deficient mice. Tumor growth in animals lacking IL27Rα was markedly reduced. We noticed a decrease in immune cell infiltrates, enhanced tumor cell death, and fibroblast accumulation. However, most striking changes pertain the tumor vasculature. Tumors in IL27Rα-deficient mice were unable to form functional vessels. Blocking IL-27-STAT1 signaling in endothelial cells in vitro provoked an overshooting migration/sprouting of endothelial cells. Apparently, the lack of the IL-27 receptor caused endothelial cell hyper-activation via STAT1 that limited vessel maturation. Our data reveal a so far unappreciated role of IL-27 in endothelial cells with importance in pathological vessel formation.

9.
J Clin Invest ; 129(12): 5204-5218, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31479425

ABSTRACT

Polyunsaturated fatty acids such as docosahexaenoic acid (DHA) positively affect the outcome of retinopathy of prematurity (ROP). Given that DHA metabolism by cytochrome P450 and soluble epoxide hydrolase (sEH) enzymes affects retinal angiogenesis and vascular stability, we investigated the role of sEH in a mouse model of ROP. In WT mice, hyperoxia elicited tyrosine nitration and inhibition of sEH and decreased generation of the DHA-derived diol 19,20-dihydroxydocosapentaenoic acid (19,20-DHDP). Correspondingly, in a murine model of ROP, sEH-/- mice developed a larger central avascular zone and peripheral pathological vascular tuft formation than did their WT littermates. Astrocytes were the cells most affected by sEH deletion, and hyperoxia increased astrocyte apoptosis. In rescue experiments, 19,20-DHDP prevented astrocyte loss by targeting the mitochondrial membrane to prevent the hyperoxia-induced dissociation of presenilin-1 and presenilin-1-associated protein to attenuate poly ADP-ribose polymerase activation and mitochondrial DNA damage. Therapeutic intravitreal administration of 19,20-DHDP not only suppressed astrocyte loss, but also reduced pathological vascular tuft formation in sEH-/- mice. Our data indicate that sEH activity is required for mitochondrial integrity and retinal astrocyte survival in ROP. Moreover, 19,20-DHDP may be more effective than DHA as a nutritional supplement for preventing retinopathy in preterm infants.


Subject(s)
Astrocytes/cytology , DNA Damage , DNA, Mitochondrial/metabolism , Epoxide Hydrolases/metabolism , Retina/enzymology , Retinopathy of Prematurity/enzymology , Animals , Animals, Newborn , Apoptosis , Astrocytes/enzymology , Cell Survival , Fatty Acids, Unsaturated/metabolism , HEK293 Cells , Humans , Hyperoxia/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Neovascularization, Physiologic , Oxygen/metabolism , Phenotype , Tyrosine/metabolism
10.
Invest Ophthalmol Vis Sci ; 59(2): 653-661, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29392309

ABSTRACT

Purpose: Neovascularization is a major cause of blindness in various ocular diseases. Bioactive sphingosine 1-phosphate (S1P), synthesized by two sphingosine kinases (Sphk1, Sphk2), emerged as a key player in a multitude of cellular processes, including cell survival, proliferation, inflammation, migration, and angiogenesis. We investigated the role of Sphk2, S1P, and S1P receptors (S1PR) during retinal neovascularization using the oxygen-induced retinopathy mouse model (OIR). Methods: Sphk2 overexpressing (tgSphk2) and Sphk2 knockout (Sphk2-/-) mice were used in the OIR model, exposed to 75% O2 over 5 days from postnatal day (P)7 to 12 to initiate vessel regression. After returning to room air, these mice developed a marked neovascularization. Retinae recovered from untreated and treated eyes at P7, P12, P14, and P17 were used for lectin-stained retinal whole mounts, mass spectrometry, and quantitative real-time PCR. Results: tgSphk2 mice showed higher retinal S1P concentrations, accelerated retinal angiogenesis, and increased neovascularization. Expression of S1PR, vascular endothelial growth factor α (VEGFα), and angiopoietin 1 and 2 was differentially regulated during the course of OIR in the different genotypes. Sphk2-/- displayed a markedly reduced retinal angiogenesis and neovascularization as well as decreased VEGFα and angiopoietin expression. Conclusions: Using genetic models of Sphk2 overexpression or deletion we demonstrate a strong impact of Sphk2/S1P on retinal vasculopathy and expression of vascular growth factors like VEGF and angiopoietin in the retina. Consequently, Sphk2, S1P, and S1PR may offer attractive novel therapeutic targets for ischemic retinopathies.


Subject(s)
Disease Models, Animal , Phosphotransferases (Alcohol Group Acceptor)/physiology , Retinal Neovascularization/enzymology , Retinopathy of Prematurity/enzymology , Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Animals , Chromatography, Liquid , Lysophospholipids/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oxygen/toxicity , Real-Time Polymerase Chain Reaction , Receptors, Lysosphingolipid/metabolism , Retina/metabolism , Retinal Neovascularization/pathology , Retinopathy of Prematurity/chemically induced , Retinopathy of Prematurity/pathology , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Tandem Mass Spectrometry , Vascular Endothelial Growth Factor A/metabolism
11.
Nature ; 552(7684): 248-252, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29211719

ABSTRACT

Diabetic retinopathy is an important cause of blindness in adults, and is characterized by progressive loss of vascular cells and slow dissolution of inter-vascular junctions, which result in vascular leakage and retinal oedema. Later stages of the disease are characterized by inflammatory cell infiltration, tissue destruction and neovascularization. Here we identify soluble epoxide hydrolase (sEH) as a key enzyme that initiates pericyte loss and breakdown of endothelial barrier function by generating the diol 19,20-dihydroxydocosapentaenoic acid, derived from docosahexaenoic acid. The expression of sEH and the accumulation of 19,20-dihydroxydocosapentaenoic acid were increased in diabetic mouse retinas and in the retinas and vitreous humour of patients with diabetes. Mechanistically, the diol targeted the cell membrane to alter the localization of cholesterol-binding proteins, and prevented the association of presenilin 1 with N-cadherin and VE-cadherin, thereby compromising pericyte-endothelial cell interactions and inter-endothelial cell junctions. Treating diabetic mice with a specific sEH inhibitor prevented the pericyte loss and vascular permeability that are characteristic of non-proliferative diabetic retinopathy. Conversely, overexpression of sEH in the retinal Müller glial cells of non-diabetic mice resulted in similar vessel abnormalities to those seen in diabetic mice with retinopathy. Thus, increased expression of sEH is a key determinant in the pathogenesis of diabetic retinopathy, and inhibition of sEH can prevent progression of the disease.


Subject(s)
Diabetic Retinopathy/enzymology , Diabetic Retinopathy/prevention & control , Epoxide Hydrolases/antagonists & inhibitors , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Capillary Permeability/drug effects , Carrier Proteins/metabolism , Cell Membrane/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Disease Models, Animal , Disease Progression , Docosahexaenoic Acids/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Ependymoglial Cells , Fatty Acids, Unsaturated/metabolism , Female , Humans , Intercellular Junctions/drug effects , Intercellular Junctions/pathology , Male , Mice , Mice, Inbred C57BL , Pancreatic Elastase/metabolism , Pericytes/drug effects , Pericytes/pathology , Presenilin-1/metabolism , Retina/drug effects , Retina/enzymology , Retina/metabolism , Retina/pathology , Solubility , Vitreous Body/metabolism
12.
J Exp Med ; 214(9): 2695-2713, 2017 Sep 04.
Article in English | MEDLINE | ID: mdl-28739604

ABSTRACT

Metastasis is the primary cause of cancer death. The inflammatory tumor microenvironment contributes to metastasis, for instance, by recruiting blood and lymph vessels. Among tumor-infiltrating immune cells, tumor-associated macrophages (TAMs) take a center stage in promoting both tumor angiogenesis and metastatic spread. We found that genetic deletion of the S1P receptor 1 (S1pr1) alone in CD11bhi CD206+ TAMs infiltrating mouse breast tumors prevents pulmonary metastasis and tumor lymphangiogenesis. Reduced lymphangiogenesis was also observed in the nonrelated methylcholanthrene-induced fibrosarcoma model. Transcriptome analysis of isolated TAMs from both entities revealed reduced expression of the inflammasome component Nlrp3 in S1PR1-deficient TAMs. Macrophage-dependent lymphangiogenesis in vitro was triggered upon inflammasome activation and required both S1PR1 signaling and IL-1ß production. Finally, NLRP3 expression in tumor-infiltrating macrophages correlated with survival, lymph node invasion, and metastasis of mammary carcinoma patients. Conceptually, our study indicates an unappreciated role of the NLRP3 inflammasome in promoting metastasis via the lymphatics downstream of S1PR1 signaling in macrophages.


Subject(s)
Interleukin-1beta/physiology , Lymphangiogenesis/physiology , Macrophages/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Neoplasm Metastasis/physiopathology , Receptors, Lysosphingolipid/physiology , Animals , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Female , Fibrosarcoma/physiopathology , Humans , Lymphatic Metastasis , Mammary Neoplasms, Experimental/physiopathology , Mice , Mice, Inbred C57BL , Sphingosine-1-Phosphate Receptors
13.
Prostaglandins Other Lipid Mediat ; 133: 93-102, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28442442

ABSTRACT

Polyunsaturated fatty acids (PUFA) and their cytochrome P450 (CYP450) metabolites have been linked to angiogenesis and vessel homeostasis. However, the role of individual CYP isoforms and their endogenous metabolites in those processes are not clear. Here, we focused on the role of Cyp2c44 in postnatal retinal angiogenesis and report that Cyp2c44 is highly expressed in Müller glial cells in the retina. The constitutive as well as inducible postnatal genetic deletion of Cyp2c44 resulted in an increased vessel network density without affecting vessel radial expansion during the first postnatal week. This phenotype was associated with an increased endothelial cell proliferation and attenuated Notch signaling. LC-MS/MS analyses revealed that levels of hydroxydocosahexaenoic acids (HDHA), i.e., 10-, 17- and 20-HDHA were significantly elevated in retinas from 5day old Cyp2c44-/- mice compared to their wild-type littermates. Enzymatic activity assays revealed that HDHAs were potential substrates for Cyp2c44 which could account for the increased levels of HDHAs in retinas from Cyp2c44-/- mice. These data indicate that Cyp2c44 is expressed in the murine retina and, like the soluble epoxide hydrolase, is expressed in Müller glia cells. The enhanced endothelial cell proliferation and Notch inhibition seen in retinas from Cyp2c44-deficient mice indicate a role for Cyp2c44-derived lipid mediators in physiological angiogenesis.


Subject(s)
Cytochrome P450 Family 2/metabolism , Ependymoglial Cells/enzymology , Neovascularization, Physiologic , Retina/physiology , Animals , Cell Proliferation , Cytochrome P450 Family 2/deficiency , Cytochrome P450 Family 2/genetics , Docosahexaenoic Acids/metabolism , Gene Deletion , Gene Expression Regulation, Enzymologic , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Retina/cytology
14.
Sci Signal ; 9(434): ra64, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27353364

ABSTRACT

Tumor cell-derived factors skew macrophages toward a tumor-supporting phenotype associated with the secretion of protumorigenic mediators. Apoptosing tumor cells release sphingosine 1-phosphate (S1P), which stimulates the production of lipocalin 2 (LCN2) in tumor-associated macrophages and is associated with tumor metastasis. We explored the mechanism by which S1P induces LCN2 in macrophages and investigated how this contributed to tumor growth and metastasis. Knockdown of S1P receptor 1 (S1PR1) in primary human macrophages and experiments with bone marrow-derived macrophages from S1PR1-deficient mice showed that S1P signaled through S1PR1 to induce LCN2 expression. The LCN2 promoter contains a consensus sequence for signal transducer and activator of transcription 3 (STAT3), and deletion of the STAT3 recognition sequence reduced expression of an LCN2-controlled reporter gene. Conditioned medium from coculture experiments indicated that the release of LCN2 from macrophages induced tube formation and proliferation in cultures of primary human lymphatic endothelial cells in a manner dependent on the kinase PI3K and subsequent induction of the growth factor VEGFC, which functioned as an autocrine signal stimulating the receptor VEGFR3. Knockout of Lcn2 attenuated tumor-associated lymphangiogenesis and breast tumor metastasis both in the breast cancer model MMTV-PyMT mice and in mice bearing orthotopic wild-type tumors. Our findings indicate that macrophages respond to dying tumor cells by producing signals that promote lymphangiogenesis, which enables metastasis.


Subject(s)
Breast Neoplasms/metabolism , Lipocalin-2/metabolism , Lymphangiogenesis , Macrophages/metabolism , Mammary Neoplasms, Experimental/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Humans , Lipocalin-2/genetics , Lysophospholipids , MCF-7 Cells , Macrophages/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Sphingosine/analogs & derivatives
15.
Cardiovasc Res ; 111(3): 184-93, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27013635

ABSTRACT

AIMS: Pulmonary hypertension is a progressive disease with poor prognosis, characterized by pathological inward remodelling and loss of patency of the lung vasculature. The right ventricle is co-affected by pulmonary hypertension, which triggers events such as hypoxia and/or increased mechanical load. Initially the right ventricle responds with 'adaptive' hypertrophy, which is often rapidly followed by 'maladaptive' changes leading to right heart decompensation and failure, which is the ultimate cause of death. METHODS AND RESULTS: We report here that miR-223 is expressed in the murine lung and right ventricle at higher levels than in the left ventricle. Moreover, lung and right-ventricular miR-223 levels were markedly down-regulated by hypoxia. Correspondingly, increasing right-ventricular load by pulmonary artery banding, induced right-ventricular ischaemia, and the down-regulation of miR-223. Lung and right ventricle miR-223 down-regulation were linked with increased expression of the miR-223 target; insulin-like growth factor-I receptor (IGF-IR) and IGF-I downstream signalling. Similarly, miR-223 was decreased and IGF-IR increased in human pulmonary hypertension. Notably in young mice, miR-223 overexpression, the genetic inactivation or pharmacological inhibition of IGF-IR, all attenuated right-ventricular hypertrophy and improved right heart function under conditions of hypoxia or increased afterload. CONCLUSION: These findings highlight the early role of pulmonary and right-ventricular miR-223 and the IGF-IR in the right heart failure programme initiated by pulmonary hypoxia and increased mechanical load and may lead to the development of novel therapeutic strategies that target the development of PH and right heart failure.


Subject(s)
Heart Failure/metabolism , Heart Ventricles/metabolism , Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Lung/metabolism , MicroRNAs/metabolism , Receptor, IGF Type 1/metabolism , Receptors, Somatomedin/metabolism , Ventricular Dysfunction, Right/metabolism , Ventricular Function, Right , Animals , Gene Expression Regulation , Genetic Predisposition to Disease , Heart Failure/genetics , Heart Failure/physiopathology , Heart Failure/prevention & control , Heart Ventricles/physiopathology , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/prevention & control , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/complications , Hypoxia/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Imidazoles/pharmacology , Male , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Phenotype , Pyridines/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/deficiency , Receptor, IGF Type 1/genetics , Signal Transduction , Ventricular Dysfunction, Right/genetics , Ventricular Dysfunction, Right/physiopathology , Ventricular Dysfunction, Right/prevention & control
16.
Thromb Haemost ; 114(3): 569-78, 2015 Aug 31.
Article in English | MEDLINE | ID: mdl-25993908

ABSTRACT

Polycystic ovary syndrome (PCOS) is associated with decreased fertility, insulin resistance and an increased risk of developing cardiovascular disease. Treating PCOS patients with metformin improves fertility and decreases cardiovascular complications. Given that platelet activation contributes to both infertility and cardiovascular disease development, we assessed platelet reactivity in PCOS patients and the consequences of metformin treatment. Compared to washed platelets from healthy donors, platelets from PCOS patients demonstrated enhanced reactivity and impaired activation of the AMP-activated kinase (AMPK). PCOS platelets also demonstrated enhanced expression of mitochondrial proteins such as the cytochrome c reductase, ATP synthase and the voltage-dependent anion channel-1. However, mitochondrial function was impaired as demonstrated by a decreased respiration rate. In parallel, the phosphorylation of dynamin-related protein-1 (Drp-1) on Ser616 was increased while that on Ser637 decreased. The latter changes were accompanied by decreased mitochondrial size. In insulin-resistant PCOS patients (HOMA-IR> 2) metformin treatment (1.7 g per day for 4 weeks to 6 months) improved insulin sensitivity, restored mitochondrial integrity and function and normalised platelet aggregation. Treatment was without effect in PCOS patients with HOMA-IR< 2. Moreover, treatment of megakaryocytes with metformin enhanced mitochondrial content and in the same cells metformin enhanced the phosphorylation of the Drp-1 on Ser637 via an AMPKα1-dependent mechanism. In conclusion, the improvement of mitochondrial integrity and platelet reactivity may contribute to the beneficial effects of metformin on cardiovascular disease.


Subject(s)
Blood Platelets/drug effects , Metformin/therapeutic use , Mitochondria/drug effects , Platelet Activation/drug effects , Polycystic Ovary Syndrome/drug therapy , AMP-Activated Protein Kinases/blood , AMP-Activated Protein Kinases/genetics , Adult , Blood Platelets/enzymology , Blood Platelets/ultrastructure , Case-Control Studies , Cell Line , Dose-Response Relationship, Drug , Dynamins , Enzyme Activation , Female , GTP Phosphohydrolases/blood , Humans , Insulin Resistance , Microtubule-Associated Proteins/blood , Mitochondria/enzymology , Mitochondria/ultrastructure , Mitochondrial Dynamics/drug effects , Mitochondrial Proteins/blood , Mitochondrial Size/drug effects , Phosphorylation , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/diagnosis , Polycystic Ovary Syndrome/enzymology , Polycystic Ovary Syndrome/genetics , RNA Interference , Signal Transduction , Time Factors , Transfection , Treatment Outcome
17.
Cardiovasc Res ; 106(2): 284-94, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25750188

ABSTRACT

AIMS: Secreted modular calcium-binding protein 1 (SMOC1) is a matricellular protein that potentially interferes with growth factor receptor signalling. The aim of this study was to determine how its expression is regulated in endothelial cells and its role in the regulation of endothelial cell function. METHODS AND RESULTS: SMOC1 was expressed by native murine endothelial cells as well as by cultured human, porcine, and murine endothelial cells. SMOC1 expression in cultured cells was increased by hypoxia via the down-regulation of miR-223, and SMOC1 expression was increased in lungs from miR-223-deficient mice. Silencing SMOC1 (small interfering RNA) attenuated endothelial cell proliferation, migration, and sprouting in in vitro angiogenesis assays. Similarly endothelial cell sprouting from aortic rings ex vivo as well as postnatal retinal angiogenesis in vivo was attenuated in SMOC1(+/-) mice. In endothelial cells, transforming growth factor (TGF)-ß signalling via activin-like kinase (ALK) 5 leads to quiescence, whereas TGF-ß signalling via ALK1 results in endothelial cell activation. SMOC1 acted as a negative regulator of ALK5/SMAD2 signalling, resulting in altered α2 integrin levels. Mechanistically, SMOC1 associated (immunohistochemistry, proximity ligation assay, and co-immunoprecipitation) with endoglin; an endothelium-specific type III auxiliary receptor for the TGF-ß super family and the effects of SMOC1 down-regulation on SMAD2 phosphorylation were abolished by the down-regulation of endoglin. CONCLUSION: These results indicate that SMOC1 is an ALK5 antagonist produced by endothelial cells that tips TGF-ß signalling towards ALK1 activation, thus promoting endothelial cell proliferation and angiogenesis.


Subject(s)
Endothelial Cells/metabolism , Osteonectin/metabolism , Transforming Growth Factor beta/metabolism , Activin Receptors, Type I/metabolism , Activin Receptors, Type II , Animals , Cell Proliferation , Endothelium, Vascular/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Physiologic/genetics , Osteonectin/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Swine
18.
Biol Cell ; 107(6): 175-88, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25757011

ABSTRACT

BACKGROUND INFORMATION: Tumour-associated lymphangiogenesis was identified as an important clinical determinant for the prognosis of hepatocellular carcinoma (HCC) and significantly influences patient survival. However, in this context, little is known about regulation of lymphangiogenesis by hypoxia-inducible factors (HIF). In HCC, mainly HIF-1α was positively correlated with lymphatic invasion and metastasis, whereas a defined role of HIF-2α is missing. RESULTS: We created a stable knockdown (k/d) of HIF-1α and HIF-2α in HepG2 cells and generated co-cultures of HepG2 spheroids with embryonic bodies. This constitutes an in vitro tumour model mimicking the cancer microenvironment and allows addressing the role of distinct HIF isoforms in regulating HCC lymphangiogenesis. In co-cultures with a HIF-2α k/d, lymphangiogenesis was significantly increased, whereas the k/d of HIF-1α showed no effect. The HIF-2α-dependent lymphangiogenic phenotype was confirmed in vivo using matrigel plug assays with supernatants of HIF-2α k/d HepG2 cells. We identified and verified insulin-like growth factor binding protein 1 (IGFBP1) as a HIF-2α target gene. The potential of HepG2 cells to induce lymphangiogenesis in two independent functional assays was significantly enhanced either by a k/d of HIF-2α or by silencing IGFBP1. Moreover, we confirmed IGF as a potent pro-lymphatic growth factor with IGFBP1 being its negative modulator. CONCLUSIONS: We propose that HIF-2α acts as an important negative regulator of hepatic lymphangiogenesis in vitro and in vivo by inducing IGFBP1 and thus, interfering with IGF signalling. Therefore, HIF-2α may constitute a critical target in HCC therapy.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Hepatocellular/genetics , Insulin-Like Growth Factor Binding Protein 1/genetics , Liver Neoplasms/genetics , Lymphangiogenesis/genetics , Up-Regulation/genetics , Animals , Cell Line , Cell Line, Tumor , Coculture Techniques , Female , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lymphatic Metastasis/genetics , Mice , Mice, Inbred C57BL
19.
Exp Cell Res ; 331(1): 46-57, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25489981

ABSTRACT

Hypoxia promotes progression of hepatocellular carcinoma (HCC), not only affecting tumor cell proliferation and invasion, but also angiogenesis and thus, increasing the risk of metastasis. Hypoxia inducible factors (HIF)-1α and -2α cause adaptation of tumors to hypoxia, still with uncertainties towards the angiogenic switch. We created a stable knockdown of HIF-1α and HIF-2α in HepG2 cells and generated cocultures of HepG2 spheroids with embryonic bodies as an in vitro tumor model mimicking the cancer microenvironment. The naturally occuring oxygen and nutrient gradients within the cocultures allow us to question the role of distinct HIF isoforms in regulating HCC angiogenesis. In cocultures with a HIF-2α knockdown, angiogenesis was attenuated, while the knockdown of HIF-1α was without effect. Microarray analysis identified plasminogen activator inhibitor 1 (PAI-1) as a HIF-2α target gene in HepG2 cells. The knockdown of PAI-1 in HepG2 cells also lowered angiogenesis. Blocking plasmin, the downstream target of PAI-1, with aprotinin in HIF-2α knockdown (k/d) cells proved a cause-effect relation and restored angiogenesis, with no effect on control cocultures. Suggestively, HIF-2α increases PAI-1 to lower concentrations of active plasmin, thereby supporting angiogenesis. We conclude that the HIF-2α target gene PAI-1 favors the angiogenic switch in HCC.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Hepatocellular/blood supply , Gene Expression Regulation, Neoplastic , Liver Neoplasms/blood supply , Neovascularization, Pathologic , Plasminogen Activator Inhibitor 1/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Oligonucleotide Array Sequence Analysis , Plasminogen Activator Inhibitor 1/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
20.
J Pharmacol Exp Ther ; 350(1): 14-21, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24763066

ABSTRACT

Cytochrome P450-derived epoxides of arachidonic acid [i.e., the epoxyeicosatrienoic acids (EETs)] are important lipid signaling molecules involved in the regulation of vascular tone and angiogenesis. Because many actions of 11,12-cis-epoxyeicosatrienoic acid (EET) are dependent on the activation of protein kinase A (PKA), the existence of a cell-surface G(s)-coupled receptor has been postulated. To assess whether the responses of endothelial cells to 11,12-EET are enantiomer specific and linked to a potential G protein-coupled receptor, we assessed 11,12-EET-induced, PKA-dependent translocation of transient receptor potential (TRP) C6 channels, as well as angiogenesis. In primary cultures of human endothelial cells, (±)-11,12-EET led to the rapid (30 seconds) translocation a TRPC6-V5 fusion protein, an effect reproduced by 11(R),12(S)-EET, but not by 11(S),12(R)-EET or (±)-14,15-EET. Similarly, endothelial cell migration and tube formation were stimulated by (±)-11,12-EET and 11(R),12(S)-EET, whereas 11(S),12(R)-EET and 11,12-dihydroxyeicosatrienoic acid were without effect. The effects of (±)-11,12-EET on TRP channel translocation and angiogenesis were sensitive to EET antagonists, and TRP channel trafficking was also prevented by a PKA inhibitor. The small interfering RNA-mediated downregulation of G(s) in endothelial cells had no significant effect on responses stimulated by vascular endothelial growth or a PKA activator but abolished responses to (±)-11,12-EET. The downregulation of G(q)/11 failed to prevent 11,12-EET-induced TRPC6 channel translocation or the formation of capillary-like structures. Taken together, our results suggest that a G(s)-coupled receptor in the endothelial cell membrane responds to 11(R),12(S)-EET and mediates the PKA-dependent translocation and activation of TRPC6 channels, as well as angiogenesis.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , GTP-Binding Protein alpha Subunits, Gs/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , TRPC Cation Channels/metabolism , 8,11,14-Eicosatrienoic Acid/antagonists & inhibitors , 8,11,14-Eicosatrienoic Acid/pharmacology , Angiogenesis Inducing Agents/pharmacology , Cell Movement/drug effects , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Down-Regulation , Humans , Primary Cell Culture , RNA, Small Interfering/pharmacology , Stereoisomerism , TRPC6 Cation Channel , Vascular Endothelial Growth Factor A/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL