Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Mol Metab ; 47: 101168, 2021 05.
Article in English | MEDLINE | ID: mdl-33465519

ABSTRACT

OBJECTIVE: Storage of triglycerides in lipid droplets is governed by a set of lipid droplet-associated proteins. One of these lipid droplet-associated proteins, hypoxia-inducible lipid droplet-associated (HILPDA), was found to impair lipid droplet breakdown in macrophages and cancer cells by inhibiting adipose triglyceride lipase. Here, we aimed to better characterize the role and mechanism of action of HILPDA in hepatocytes. METHODS: We performed studies in HILPDA-deficient and HILPDA-overexpressing liver cells, liver slices, and mice. The functional role and physical interactions of HILPDA were investigated using a variety of biochemical and microscopic techniques, including real-time fluorescence live-cell imaging and Förster resonance energy transfer-fluorescence lifetime imaging microscopy (FRET-FLIM). RESULTS: Levels of HILPDA were markedly induced by fatty acids in several hepatoma cell lines. Hepatocyte-specific deficiency of HILPDA in mice modestly but significantly reduced hepatic triglycerides in mice with non-alcoholic steatohepatitis. Similarly, deficiency of HILPDA in mouse liver slices and primary hepatocytes reduced lipid storage and accumulation of fluorescently-labeled fatty acids in lipid droplets, respectively, which was independent of adipose triglyceride lipase. Fluorescence microscopy showed that HILPDA partly colocalizes with lipid droplets and with the endoplasmic reticulum, is especially abundant in perinuclear areas, and mainly associates with newly added fatty acids. Real-time fluorescence live-cell imaging further revealed that HILPDA preferentially localizes to lipid droplets that are being remodeled. Overexpression of HILPDA in liver cells increased the activity of diacylglycerol acyltransferases (DGAT) and DGAT1 protein levels, concurrent with increased lipid storage. Confocal microscopy coupled to FRET-FLIM analysis demonstrated that HILPDA physically interacts with DGAT1 in living liver cells. The stimulatory effect of HILPDA on lipid storage via DGAT1 was corroborated in adipocytes. CONCLUSIONS: Our data indicate that HILPDA physically interacts with DGAT1 and increases DGAT activity. Our findings suggest a novel regulatory mechanism by which fatty acids promote triglyceride synthesis and storage.


Subject(s)
Diacylglycerol O-Acyltransferase/metabolism , Hepatocytes/metabolism , Hypoxia/metabolism , Lipid Droplets/metabolism , Adipocytes/metabolism , Animals , Carcinoma, Hepatocellular , Diacylglycerol O-Acyltransferase/genetics , Fatty Acids/metabolism , Gene Expression , Hep G2 Cells , Humans , Lipid Metabolism , Lipogenesis , Liver/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Neoplasm Proteins/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Triglycerides/metabolism
2.
FASEB J ; 31(11): 4971-4984, 2017 11.
Article in English | MEDLINE | ID: mdl-28760743

ABSTRACT

Recently we identified hypoxia-inducible protein 2 (HIG2)/hypoxia-inducible lipid droplet-associated (HILPDA) as lipid droplet (LD) protein. Because HILPDA is highly expressed in atherosclerotic plaques, we examined its regulation and function in murine macrophages, compared it to the LD adipose differentiation-related protein (Adrp)/perilipin 2 (Plin2), and investigated its effects on atherogenesis in apolipoprotein E-deficient (ApoE-/-) mice. Tie2-Cre-driven Hilpda conditional knockout (cKO) did not affect viability, proliferation, and ATP levels in macrophages. Hilpda proved to be a target of hypoxia-inducible factor 1 (Hif-1) and peroxisome proliferator-activated receptors. In contrast, Adrp/Plin2 was not induced by Hif-1. Hilpda localized to the endoplasmic reticulum-LD interface, the site of LD formation. Hypoxic lipid accumulation and storage of oxidized LDL, cholesteryl esters and triglycerides were abolished in Hilpda cKO macrophages, independent of the glycolytic switch, fatty acid or lipoprotein uptake. Hilpda depletion reduced resistance against lipid overload and increased production of reactive oxygen species after reoxygenation. LPS-stimulated prostaglandin-E2 production was dysregulated in macrophages, demonstrating the substrate buffer and reservoir function of LDs for eicosanoid production. In ApoE-/- Hilpda cKO mice, total aortic plaque area, plaque macrophages and vascular Vegf expression were reduced. Thus, macrophage Hilpda is crucial to foam-cell formation and lipid deposition, and to controlled prostaglandin-E2 production. By these means Hilpda promotes lesion formation and progression of atherosclerosis.-Maier, A., Wu, H., Cordasic, N., Oefner, P., Dietel, B., Thiele, C., Weidemann, A., Eckardt, K.-U., Warnecke, C. Hypoxia-inducible protein 2 Hig2/Hilpda mediates neutral lipid accumulation in macrophages and contributes to atherosclerosis in apolipoprotein E-deficient mice.


Subject(s)
Atherosclerosis/metabolism , Foam Cells/metabolism , Lipid Metabolism , Neoplasm Proteins/metabolism , Plaque, Atherosclerotic/metabolism , Animals , Apolipoproteins E/deficiency , Atherosclerosis/genetics , Atherosclerosis/pathology , Dinoprostone/genetics , Dinoprostone/metabolism , Disease Models, Animal , Female , Foam Cells/pathology , Humans , Male , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Perilipin-2/genetics , Perilipin-2/metabolism , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
3.
Oncoimmunology ; 6(1): e1256526, 2017.
Article in English | MEDLINE | ID: mdl-28197372

ABSTRACT

Genome-wide association studies (GWAS) associated Family with sequence similarity 13, member A (FAM13A) with non-small cell lung cancer (NSCLC) occurrence. Here, we found increased numbers of FAM13A protein expressing cells in the tumoral region of lung tissues from a cohort of patients with NSCLC. Moreover, FAM13A inversely correlated with CTLA4 but directly correlated with HIF1α levels in the control region of these patients. Consistently, FAM13A RhoGAP was found to be associated with T cell effector molecules like HIF1α and Tbet and was downregulated in immunosuppressive CD4+CD25+Foxp3+CTLA4+ T cells. TGFß, a tumor suppressor factor, as well as siRNA to FAM13A, suppressed both isoforms of FAM13A and inhibited tumor cell proliferation. RNA-Seq analysis confirmed this finding. Moreover, siRNA to FAM13A induced TGFß levels. Finally, in experimental tumor cell migration, FAM13A was induced and TGFß accelerated this process by inducing cell migration, HIF1α, and the FAM13A RhoGAP isoform. Furthermore, siRNA to FAM13A inhibited tumor cell proliferation and induced cell migration without affecting HIF1α. In conclusion, FAM13A is involved in tumor cell proliferation and downstream of TGFß and HIF1α, FAM13A RhoGAP is associated with Th1 gene expression and lung tumor cell migration. These findings identify FAM13A as key regulator of NSCLC growth and progression.

4.
Kidney Int ; 91(3): 616-627, 2017 03.
Article in English | MEDLINE | ID: mdl-27927598

ABSTRACT

Chronic kidney disease (CKD) is associated with increased risk and worse prognosis of cardiovascular disease, including peripheral artery disease. An impaired angiogenic response to ischemia may contribute to poor outcomes of peripheral artery disease in patients with CKD. Hypoxia inducible factors (HIF) are master regulators of angiogenesis and therefore represent a promising target for therapeutic intervention. To test this we induced hind-limb ischemia in rats with CKD caused by 5/6 nephrectomy and administered two different treatments known to stabilize HIF protein in vivo: carbon monoxide and a pharmacological inhibitor of prolyl hydroxylation 2-(1-chloro-4- hydroxyisoquinoline-3-carboxamido) acetate (ICA). Expression levels of pro-angiogenic HIF target genes (Vegf, Vegf-r1, Vegf-r2, Ho-1) were measured by qRT-PCR. Capillary density was measured by CD31 immunofluorescence staining and HIF expression was evaluated by immunohistochemistry. Capillary density in ischemic skeletal muscle was significantly lower in CKD animals compared to sham controls. Rats with CKD showed significantly lower expression of HIF and all measured pro-angiogenic HIF target genes, including VEGF. Both HIF stabilizing treatments rescued HIF target gene expression in animals with CKD and led to significantly higher ischemia-induced capillary sprouting compared to untreated controls. ICA was effective regardless of whether it was administered before or after induction of ischemia and led to a HIF expression in skeletal muscle. Thus, impaired ischemia-induced angiogenesis in rats with CKD can be improved by HIF stabilization, even if started after onset of ischemia.


Subject(s)
Capillaries/drug effects , Carbon Monoxide/pharmacology , Glycine/analogs & derivatives , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ischemia/drug therapy , Isoquinolines/pharmacology , Muscle, Skeletal/blood supply , Neovascularization, Physiologic/drug effects , Renal Insufficiency, Chronic/metabolism , Signal Transduction/drug effects , Animals , Capillaries/metabolism , Capillaries/physiopathology , Cell Line , Disease Models, Animal , Gene Expression Regulation , Glycine/pharmacology , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Hindlimb , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Male , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Protein Stability , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/physiopathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
5.
Circ Res ; 117(6): 513-24, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26208651

ABSTRACT

RATIONALE: Accelerated arterial stiffening is a major complication of diabetes mellitus with no specific therapy available to date. OBJECTIVE: The present study investigates the role of the osteogenic transcription factor runt-related transcription factor 2 (Runx2) as a potential mediator and therapeutic target of aortic fibrosis and aortic stiffening in diabetes mellitus. METHODS AND RESULTS: Using a murine model of type 2 diabetes mellitus (db/db mice), we identify progressive structural aortic stiffening that precedes the onset of arterial hypertension. At the same time, Runx2 is aberrantly upregulated in the medial layer of db/db aortae, as well as in thoracic aortic samples from patients with type 2 diabetes mellitus. Vascular smooth muscle cell-specific overexpression of Runx2 in transgenic mice increases expression of its target genes, Col1a1 and Col1a2, leading to medial fibrosis and aortic stiffening. Interestingly, increased Runx2 expression per se is not sufficient to induce aortic calcification. Using in vivo and in vitro approaches, we further demonstrate that expression of Runx2 in diabetes mellitus is regulated via a redox-sensitive pathway that involves a direct interaction of NF-κB with the Runx2 promoter. CONCLUSIONS: In conclusion, this study highlights Runx2 as a previously unrecognized inducer of vascular fibrosis in the setting of diabetes mellitus, promoting arterial stiffness irrespective of calcification.


Subject(s)
Aorta/metabolism , Aorta/pathology , Core Binding Factor Alpha 1 Subunit/biosynthesis , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Vascular Stiffness/physiology , Aged , Animals , Cells, Cultured , Female , Fibrosis , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Transcription Factors/biosynthesis
6.
Free Radic Biol Med ; 81: 58-68, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25557012

ABSTRACT

GPx8 is a mammalian Cys-glutathione peroxidase of the endoplasmic reticulum membrane, involved in protein folding. Its regulation is mostly unknown. We addressed both the functionality of two hypoxia-response elements (HREs) within the promoter, GPx8 HRE1 and GPx8 HRE2, and the GPx8 physiological role. In HeLa cells, treatment with HIFα stabilizers, such as diethyl succinate (DES) or 2-2'-bipyridyl (BP), induces GPx8 expression at both mRNA and protein level. Luciferase activity of pGL3(GPx8wt), containing a fragment of the GPx8 promoter including the two HREs, is also induced by DES/BP or by overexpressing either individual HIFα subunit. Mutating GPx8 HRE1 within pGL3(GPx8wt) resulted in a significantly higher inhibition of luciferase activity than mutating GPx8 HRE2. Electrophoretic mobility-shift assay showed that both HREs exhibit enhanced binding to a nuclear extract from DES/BP-treated cells, with stronger binding by GPx8 HRE1. In DES-treated cells transfected with pGL3(GPx8wt) or mutants thereof, silencing of HIF2α, but not HIF1α, abolishes luciferase activity. Thus GPx8 is a novel HIF target preferentially responding to HIF2α binding at its two novel functional GPx8 HREs, with GPx8 HRE1 playing the major role. Fibroblast growth factor (FGF) treatment increases GPx8 mRNA expression, and reporter gene experiments indicate that induction occurs via HIF. Comparing the effects of depleting GPx8 on the downstream effectors of FGF or insulin signaling revealed that absence of GPx8 results in a 16- or 12-fold increase in phosphorylated ERK1/2 by FGF or insulin treatment, respectively. Furthermore, in GPx8-depleted cells, phosphorylation of AKT by insulin treatment increases 2.5-fold. We suggest that induction of GPx8 expression by HIF slows down proliferative signaling during hypoxia and/or growth stimulation through receptor tyrosine kinases.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Endoplasmic Reticulum/drug effects , Fibroblast Growth Factors/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Peroxidases/genetics , 2,2'-Dipyridyl/pharmacology , Amino Acid Sequence , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Genes, Reporter , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin/pharmacology , Luciferases/genetics , Luciferases/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Sequence Data , Peroxidases/metabolism , Phosphorylation , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Response Elements , Sequence Alignment , Signal Transduction , Succinates/pharmacology , Transcription, Genetic
7.
Mol Carcinog ; 53(12): 970-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-23818324

ABSTRACT

Hypoxia leads to the upregulation of a variety of genes mediated largely via the hypoxia inducible transcription factor (HIF). Prominent HIF-regulated target genes such as the vascular endothelial growth factor (VEGF), the glucose transporter 1 (Glut-1), or erythropoietin (EPO) help to assure survival of cells and organisms in a low oxygenated environment. Here, we are the first to report the hypoxic regulation of the sperm associated antigen 4 (SPAG4). SPAG4 is a member of the cancer testis (CT) gene family and to date little is known about its physiological function or its involvement in tumor biology. A number of CT family candidate genes are therefore currently being investigated as potential cancer markers, due to their predominant testicular expression pattern. We analyzed RNA and protein expression by RNAse protection assay, immunofluorescent as well as immunohistological stainings. To evaluate the influence of SPAG4 on migration and invasion capabilities, siRNA knockdown as well as transient overexpression was performed prior to scratch or invasion assay analysis. The hypoxic regulation of SPAG4 is clearly mediated in a HIF-1 and VHL dependent manner. We furthermore show upregulation of SPAG4 expression in human renal clear cell carcinoma (RCC) and co-localization within the nucleolus in physiological human testis tissue. SPAG4 knockdown reduces the invasion capability of RCC cells in vitro and overexpression leads to enhancement of tumor cell migration. Together, SPAG4 could possibly play a role in the invasion capability and growth of renal tumors and could represent an interesting target for clinical intervention.


Subject(s)
Carcinoma, Renal Cell/genetics , Carrier Proteins/genetics , Cell Movement/genetics , Hypoxia-Inducible Factor 1/genetics , Hypoxia/genetics , Kidney Neoplasms/genetics , Neoplasm Invasiveness/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , HeLa Cells , Humans , Up-Regulation/genetics
8.
PLoS One ; 7(1): e31034, 2012.
Article in English | MEDLINE | ID: mdl-22299048

ABSTRACT

The Hypoxia-inducible transcription Factor (HIF) represents an important adaptive mechanism under hypoxia, whereas sustained activation may also have deleterious effects. HIF activity is determined by the oxygen regulated α-subunits HIF-1α or HIF-2α. Both are regulated by oxygen dependent degradation, which is controlled by the tumor suppressor "von Hippel-Lindau" (VHL), the gatekeeper of renal tubular growth control. HIF appears to play a particular role for the kidney, where renal EPO production, organ preservation from ischemia-reperfusion injury and renal tumorigenesis are prominent examples. Whereas HIF-1α is inducible in physiological renal mouse, rat and human tubular epithelia, HIF-2α is never detected in these cells, in any species. In contrast, distinct early lesions of biallelic VHL inactivation in kidneys of the hereditary VHL syndrome show strong HIF-2α expression. Furthermore, knockout of VHL in the mouse tubular apparatus enables HIF-2α expression. Continuous transgenic expression of HIF-2α by the Ksp-Cadherin promotor leads to renal fibrosis and insufficiency, next to multiple renal cysts. In conclusion, VHL appears to specifically repress HIF-2α in renal epithelia. Unphysiological expression of HIF-2α in tubular epithelia has deleterious effects. Our data are compatible with dedifferentiation of renal epithelial cells by sustained HIF-2α expression. However, HIF-2α overexpression alone is insufficient to induce tumors. Thus, our data bear implications for renal tumorigenesis, epithelial differentiation and renal repair mechanisms.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression , Kidney Diseases, Cystic/genetics , Kidney Tubules/metabolism , Kidney Tubules/pathology , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , COS Cells , Chlorocebus aethiops , Fibrosis/genetics , Gene Expression/physiology , Gene Silencing/physiology , HEK293 Cells , HeLa Cells , Humans , Kidney Diseases, Cystic/metabolism , Kidney Diseases, Cystic/pathology , Kidney Tubules/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Opossums , Rats , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
9.
Biochim Biophys Acta ; 1813(1): 1-13, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21056597

ABSTRACT

Hypoxia is a common pathogenic stress, which requires adaptive activation of the Hypoxia-inducible transcription factor (HIF). In concert transcriptional HIF targets enhance oxygen availability and simultaneously reduce oxygen demand, enabling survival in a hypoxic microenvironment. Here, we describe the characterization of a new HIF-1 target gene, Rab20, which is a member of the Rab family of small GTP-binding proteins, regulating intracellular trafficking and vesicle formation. Rab20 is directly regulated by HIF-1, resulting in rapid upregulation of Rab20 mRNA as well as protein under hypoxia. Furthermore, exogenous as well as endogenous Rab20 protein colocalizes with mitochondria. Knockdown studies reveal that Rab20 is involved in hypoxia induced apoptosis. Since mitochondria play a key role in the control of cell death, we suggest that regulating mitochondrial homeostasis in hypoxia is a key function of Rab20. Furthermore, our study implicates that cellular transport pathways play a role in oxygen homeostasis. Hypoxia-induced Rab20 may influence tissue homeostasis and repair during and after hypoxic stress.


Subject(s)
Apoptosis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/pathology , Mitochondria/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Blotting, Western , Caspases/metabolism , Cells, Cultured , Electrophoretic Mobility Shift Assay , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoenzyme Techniques , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , rab GTP-Binding Proteins/antagonists & inhibitors , rab GTP-Binding Proteins/genetics
10.
Transcription ; 2(6): 269-75, 2011.
Article in English | MEDLINE | ID: mdl-22223045

ABSTRACT

Hypoxia-inducible factors (HIF1α/HIF2α) are key transcription factors that promote angiogenesis. The overexpression of degradation-resistant HIF mutants is considered a promising pro-angiogenic therapeutic tool. We compared the transcriptional activity of HIF1α/HIF2α mutants that obtained their resistance to oxygen-dependent degradation either by deletion of their entire oxygen-dependent degradation (ODD) domain or by replacement of prolyl residues that are crucial for oxygen-dependent degradation. Although all HIF mutants translocated into the nucleus, HIF1α and HIF2α mutants inclosing the point mutations were significantly more effective in trans-activating the target gene VEGF and in inducing tube formation of endothelial cells than mutants lacking the complete ODD domain.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Deletion , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Point Mutation , Transcription, Genetic/genetics , Active Transport, Cell Nucleus , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites/genetics , Cell Line , Cell Nucleus/metabolism , Endothelial Cells/metabolism , Endothelial Cells/physiology , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoblotting , Luciferases/genetics , Luciferases/metabolism , Mice , Microscopy, Fluorescence , Neovascularization, Physiologic , Oxygen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcriptional Activation , Transfection , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
11.
Kidney Int ; 78(9): 857-67, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20720525

ABSTRACT

The two hypoxia-inducible factors (HIF-1α and HIF-2α) are transcription factors that regulate the response to hypoxia. Recently, the factor inhibiting HIF (FIH1) was identified as a molecular oxygen-dependent dioxygenase that blunts the transcriptional activity of HIF and has also been implicated in HIF-dependent and -independent hypoxia responses. Interestingly, HIF accumulation in the kidney has been shown to confer renal protection and to also cause glomerular injury or enhance renal fibrosis. In order to better understand the regulation of hypoxia-inducible genes, we determined the expression of FIH1 in the kidney and its functional role in isolated renal cells. FIH1 was expressed only in distal tubules and in podocytes, thus showing a very distinct expression pattern, partially overlapping with sites of HIF-1α expression. In tubular cells, RNA silencing of FIH1 caused transcriptional activation of HIF target genes during hypoxia. In contrast, FIH1 silencing in podocytes enhanced transcription of hypoxia-inducible genes in an HIF-independent manner. Using the anti-Thy.1 rat model of glomerulonephritis, we found a gradual decrease of glomerular FIH1 expression during disease progression paralleled by an increase in hypoxia-inducible genes including CXCR4, a mediator of glomerular inflammation. Thus, FIH1 appears to be a suppressor of oxygen-dependent genes in the kidney, operating through HIF-dependent and -independent mechanisms.


Subject(s)
Glomerulonephritis, Membranoproliferative/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Tubules, Distal/metabolism , Mixed Function Oxygenases/metabolism , Oxygen/metabolism , Podocytes/metabolism , Repressor Proteins/metabolism , Animals , Cell Hypoxia , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Glomerulonephritis, Membranoproliferative/genetics , Glomerulonephritis, Membranoproliferative/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Isoantibodies , Male , Mice , Mice, Inbred C57BL , Mixed Function Oxygenases/genetics , RNA Interference , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Repressor Proteins/genetics , Time Factors
12.
FASEB J ; 24(11): 4443-58, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20624928

ABSTRACT

Hypoxia-inducible protein 2 (HIG2) has been implicated in canonical Wnt signaling, both as target and activator. The potential link between hypoxia and an oncogenic signaling pathway might play a pivotal role in renal clear-cell carcinoma characterized by constitutive activation of hypoxia-inducible factors (HIFs), and hence prompted us to analyze HIG2 regulation and function in detail. HIG2 was up-regulated by hypoxia and HIF inducers in all cell types and mouse organs investigated and abundantly expressed in renal clear-cell carcinomas. Promoter analyses, gel shifts, and siRNA studies revealed that HIG2 is a direct and specific target of HIF-1, but not responsive to HIF-2. Surprisingly, HIG2 was not secreted, and HIG2 overexpression neither stimulated proliferation nor activated Wnt signaling. Instead, we show that HIG2 decorates the hemimembrane of lipid droplets, whose number and size increase on hypoxic inhibition of fatty acid ß-oxidation, and colocalizes with the lipid droplet proteins adipophilin and TIP47. Normoxic overexpression of HIG2 was sufficient to increase neutral lipid deposition in HeLa cells and stimulated cytokine expression. HIG2 could be detected in atherosclerotic arteries and fatty liver disease, suggesting that this ubiquitously inducible HIF-1 target gene may play an important functional role in diseases associated with pathological lipid accumulation.


Subject(s)
Carcinoma, Renal Cell/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/metabolism , Neoplasm Proteins/metabolism , Animals , Carcinoma, Renal Cell/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation , Cytokines/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Kidney Neoplasms/pathology , Lipid Metabolism , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasm Proteins/drug effects , Neoplasm Proteins/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Signal Transduction , Transcriptional Activation/genetics , Wnt1 Protein/metabolism
13.
Eur J Cancer ; 46(6): 1159-69, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20185296

ABSTRACT

The hypoxia-inducible factor-1 (HIF-1), which consists of the constitutive HIF-1beta and the oxygen-responsive HIF-1alpha subunit, is the master activator of the cellular transcriptional response to hypoxia coordinating gene expression during reduced oxygen tension. Overexpression of HIF-1 and increased transcriptional activity induced by hypoxia are linked to progression of many tumour types such as head and neck cancer, cervical carcinoma, leukaemia and renal cell carcinoma. In this study, we demonstrate that HIF activity is increased in malignant melanoma cells already under normoxic conditions in contrast to other tumour types. HIF-1alpha and -2alpha knockdown by siRNA transfection revealed that this effect is due to constitutive HIF-1alpha expression. Furthermore, the inhibition or activation of reactive oxygen species (ROS) decreased or activated, respectively, HIF-1 activity and HIF-1alpha protein expression. Interestingly, the inhibition of the NFkappaB pathway also reduced the accumulation of HIF-1alpha assuming a context between ROS and NFkappaB, and suggesting that ROS and NFkappaB activity contribute to HIF-1alpha accumulation. In summary, we identified an increased HIF-1alpha protein expression and activity in melanoma under normoxia mediated by ROS and the NFkappaB pathway.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Melanoma/metabolism , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Skin Neoplasms/metabolism , Animals , Cell Hypoxia/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1/genetics , Melanoma/genetics , Mice , NF-kappa B/genetics , Skin Neoplasms/genetics , Transcriptional Activation
14.
J Biol Chem ; 285(9): 6658-69, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20026874

ABSTRACT

Hypoxia has been shown to promote tumor metastasis and lead to therapy resistance. Recent work has demonstrated that hypoxia represses E-cadherin expression, a hallmark of epithelial to mesenchymal transition, which is believed to amplify tumor aggressiveness. The molecular mechanism of E-cadherin repression is unknown, yet lysyl oxidases have been implicated to be involved. Gene expression of lysyl oxidase (LOX) and the related LOX-like 2 (LOXL2) is strongly induced by hypoxia. In addition to the previously demonstrated LOX, we characterize LOXL2 as a direct transcriptional target of HIF-1. We demonstrate that activation of lysyl oxidases is required and sufficient for hypoxic repression of E-cadherin, which mediates cellular transformation and takes effect in cellular invasion assays. Our data support a molecular pathway from hypoxia to cellular transformation. It includes up-regulation of HIF and subsequent transcriptional induction of LOX and LOXL2, which repress E-cadherin and induce epithelial to mesenchymal transition. Lysyl oxidases could be an attractive molecular target for cancers of epithelial origin, in particular because they are partly extracellular.


Subject(s)
Amino Acid Oxidoreductases/physiology , Cadherins/antagonists & inhibitors , Cell Transformation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Hypoxia/metabolism , Protein-Lysine 6-Oxidase/physiology , Amino Acid Oxidoreductases/genetics , Cell Line , Epithelial Cells , Gene Expression Regulation, Enzymologic , Humans , Hypoxia/enzymology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mesoderm/cytology , Neoplasm Metastasis , Protein-Lysine 6-Oxidase/genetics , RNA, Messenger/analysis , Up-Regulation/genetics
15.
J Biol Chem ; 285(7): 4328-36, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20018872

ABSTRACT

Hypoxia, a driving force in neovascularization, promotes alterations in gene expression mediated by hypoxia-inducible factor (HIF)-1alpha. Connective tissue growth factor (CTGF, CCN2) is a modulator of endothelial cell growth and migration, but its regulation by hypoxia is poorly understood. Therefore, we analyzed signaling pathways involved in the regulation of CTGF by hypoxia in endothelial cells. Exposure to low oxygen tension or treatment with the hypoxia-mimetic dimethyloxalyl glycine (DMOG) stabilized HIF-1alpha and up-regulated CTGF in human umbilical vein endothelial cells and in a murine microvascular endothelial cell line. Induction of CTGF correlated with a HIF-dependent increase in protein and mRNA levels, and nuclear accumulation of the transcription factor FoxO3a. By contrast, gene expression and cellular localization of FoxO1 were not significantly altered by hypoxia. Expression of CTGF was strongly reduced by siRNA silencing of FoxO1 or FoxO3a. Furthermore, nuclear exclusion of FoxO1/3a transcription factors by inhibition of serine/threonine protein phosphatases by okadaic acid inhibited CTGF expression, providing evidence for both FoxO proteins as regulators of CTGF expression. The DMOG-stimulated induction of CTGF was further increased when endothelial cells were co-incubated with transforming growth factor-beta, an activator of Smad signaling. Activation of RhoA-Rho kinase signaling by the microtubule-disrupting drug combretastatin A4 also enhanced the DMOG-induced CTGF expression, thus placing CTGF induction by hypoxia in a network of interacting signaling pathways. Our findings provide evidence that FoxO1, hypoxia-stimulated expression of FoxO3a and its nuclear accumulation are required for the induction of CTGF by hypoxia in endothelial cells.


Subject(s)
Cell Hypoxia/physiology , Connective Tissue Growth Factor/metabolism , Endothelial Cells/metabolism , Forkhead Transcription Factors/metabolism , Amino Acids, Dicarboxylic/pharmacology , Animals , Blotting, Western , Cell Hypoxia/drug effects , Cell Line , Cells, Cultured , Connective Tissue Growth Factor/genetics , Endothelial Cells/drug effects , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Mice , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction
16.
PLoS One ; 4(11): e7875, 2009 Nov 18.
Article in English | MEDLINE | ID: mdl-19924283

ABSTRACT

BACKGROUND: Hepcidin is a major regulator of iron metabolism and plays a key role in anemia of chronic disease, reducing intestinal iron uptake and release from body iron stores. Hypoxia and chemical stabilizers of the hypoxia-inducible transcription factor (HIF) have been shown to suppress hepcidin expression. We therefore investigated the role of HIF in hepcidin regulation. METHODOLOGY/PRINCIPAL FINDINGS: Hepcidin mRNA was down-regulated in hepatoma cells by chemical HIF stabilizers and iron chelators, respectively. In contrast, the response to hypoxia was variable. The decrease in hepcidin mRNA was not reversed by HIF-1alpha or HIF-2alpha knock-down or by depletion of the HIF and iron regulatory protein (IRP) target transferrin receptor 1 (TfR1). However, the response of hepcidin to hypoxia and chemical HIF inducers paralleled the regulation of transferrin receptor 2 (TfR2), one of the genes critical to hepcidin expression. Hepcidin expression was also markedly and rapidly decreased by serum deprivation, independent of transferrin-bound iron, and by the phosphatidylinositol 3 (PI3) kinase inhibitor LY294002, indicating that growth factors are required for hepcidin expression in vitro. Hepcidin promoter constructs mirrored the response of mRNA levels to interleukin-6 and bone morphogenetic proteins, but not consistently to hypoxia or HIF stabilizers, and deletion of the putative HIF binding motifs did not alter the response to different hypoxic stimuli. In mice exposed to carbon monoxide, hypoxia or the chemical HIF inducer N-oxalylglycine, liver hepcidin 1 mRNA was elevated rather than decreased. CONCLUSIONS/SIGNIFICANCE: Taken together, these data indicate that hepcidin is neither a direct target of HIF, nor indirectly regulated by HIF through induction of TfR1 expression. Hepcidin mRNA expression in vitro is highly sensitive to the presence of serum factors and PI3 kinase inhibition and parallels TfR2 expression.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Hypoxia-Inducible Factor 1/metabolism , Iron-Regulatory Proteins/chemistry , Amino Acid Motifs , Animals , Antigens, CD/metabolism , Base Sequence , Chromones/pharmacology , Hepcidins , Humans , Interleukin-6/metabolism , Mice , Molecular Sequence Data , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic , Receptors, Transferrin/metabolism
17.
Biochem J ; 424(1): 143-51, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19694616

ABSTRACT

HIF (hypoxia-inducible factor)-3alpha is the third member of the HIF transcription factor family. Whereas HIF-1alpha and -2alpha play critical roles in the cellular and systemic adaptation to hypoxia, little is known about the regulation and function of HIF-3alpha. At least five different splice variants may be expressed from the human HIF-3alpha locus that are suggested to exert primarily negative regulatory effects on hypoxic gene induction. In the present paper, we report that hypoxia induces the human HIF-3alpha gene at the transcriptional level in a HIF-1-dependent manner. HIF-3alpha2 and HIF-3alpha4 transcripts, the HIF-3alpha splice variants expressed in Caki-1 renal carcinoma cells, rapidly increased after exposure to hypoxia or chemical hypoxia mimetics. siRNA (small interfering RNA)-mediated HIF-alpha knockdown demonstrated that HIF-3alpha is a specific target gene of HIF-1alpha, but is not affected by HIF-2alpha knockdown. In contrast with HIF-1alpha and HIF-2alpha, HIF-3alpha is not regulated at the level of protein stability. HIF-3alpha protein could be detected under normoxia in the cytoplasm and nuclei, but increased under hypoxic conditions. Promoter analyses and chromatin immunoprecipitation experiments localized a functional hypoxia-responsive element 5' to the transcriptional start of HIF-3alpha2. siRNA-mediated knockdown of HIF-3alpha increased transactivation of a HIF-driven reporter construct and mRNA expression of lysyl oxidase. Immunohistochemistry revealed an overlap of HIF-1alpha-positive and HIF-3alpha-positive areas in human renal cell carcinomas. These findings shed light on a novel aspect of HIF-3alpha as a HIF-1 target gene and point to a possible role as a modulator of hypoxic gene induction.


Subject(s)
Cell Hypoxia/physiology , Gene Expression Regulation , Hypoxia-Inducible Factor 1/metabolism , Transcription Factors/metabolism , Apoptosis Regulatory Proteins , Basic Helix-Loop-Helix Transcription Factors , Cell Hypoxia/genetics , Cell Line , Cell Line, Tumor , Chromatin Immunoprecipitation , Humans , Hypoxia-Inducible Factor 1/genetics , Immunoblotting , Immunohistochemistry , Promoter Regions, Genetic/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
18.
J Pathol ; 218(4): 520-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19431154

ABSTRACT

Striking similarities exist between molecular mechanisms driving embryonic liver development and progression of hepatocellular carcinoma (HCC). Bone morphogenetic proteins (BMPs), particularly BMP4, have been proposed to regulate embryonic hepatic development. BMP expression has been observed in neoplasia but the expression and biological role of BMP4 in human HCC are unknown. We found increased BMP4 mRNA and protein in HCC cell lines and tissue samples compared to primary human hepatocytes and corresponding non-tumourous tissue. Hypoxia further induced BMP4 expression in HCC cells, which was abolished by transfection of a dominant negative form of HIF-1 alpha (dnHIF-1 alpha). However, gel shift assays revealed only minor binding activity in nuclear extracts from (hypoxic) HCC cells to a putative hypoxia-response element in the BMP4 promoter. Sequence analysis of the BMP4 promoter revealed two Ets-1 binding sites, and Ets-1 activity was increased in HCC cells under hypoxic conditions. Transfection of dnHIF-1 alpha completely abrogated hypoxia-induced Ets-1 activity as well as BMP4 expression. Overexpression of Ets-1 markedly enhanced BMP4 promoter activity, while antisense Ets-1 almost completely abolished basal as well as hypoxia-induced BMP4 expression. These data demonstrate that Ets-1 activity contributes to baseline expression of the BMP4 gene and is the predominant mediator of the HIF-dependent BMP4 induction under hypoxic conditions. To determine the functional relevance of BMP4 expression, HCC cell lines were treated with antisense BMP4 constructs or siRNA against BMP4. BMP4 suppression resulted in a strong reduction of the migratory and invasive potential and anchorage-independent growth. Furthermore, tube formation assays indicated that BMP4 expressed by HCC cells promotes vasculogenesis. Our findings demonstrate that BMP4 is increased in HCC and promotes HCC progression. Therefore, BMP4 expression may have clinical relevance, and interfering with BMP4 signalling appears as an attractive therapeutic target for this highly aggressive tumour.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Bone Morphogenetic Protein 4/genetics , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/pathology , Cell Hypoxia , Cell Line, Tumor , Collagen , Drug Combinations , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Laminin , Liver Neoplasms/blood supply , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness , Neovascularization, Pathologic , Proteoglycans , Proto-Oncogene Protein c-ets-1/metabolism , RNA Interference , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transfection/methods
19.
Cell Cycle ; 8(9): 1386-95, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19342889

ABSTRACT

Hypoxia is a severe stress which induces physiological and molecular adaptations, where the latter is dominated by the Hypoxia-inducible transcription Factor (HIF). A well described response on cellular level upon exposure to hypoxia is a reversible cell cycle arrest, which probably renders the cells more resistant to the difficult environment. The individual roles of hypoxia itself and of the isoforms HIF-1alpha and HIF-2alpha in cell cycle regulation are poorly understood and discussed controversially. In order to characterize the isolated effect of both HIFalpha isoforms on the cell cycle we generated tetracycline inducible, HIF-1alpha and -2alpha expressing NIH3T3 cells. The cDNAs for HIFalpha were mutated to generate stable and active HIF under normoxia. Upon activation of both HIFalpha subunits, the total number of living cells was reduced and long-term stimulation of HIF led to complete loss of transgene expression, implicating a strong negative selection pressure. Equally, colony forming activity was reduced by activation of both HIFalpha subunits. Cell cycle analyses showed that HIF activation resulted in a prominent cell cycle arrest in G(1)-phase, similarly to the hypoxic effect. Both, HIF-1alpha and HIF-2alpha were able to induce the expression of the cyclin-dependent kinase inhibitor p27 on reporter gene and protein level. Our study shows that HIF-1 and HIF-2 can individually arrest the cell cycle independent from hypoxia. These findings have implications for the resistance of tumor cells to the environment and treatment, but also for physiological cells. Importantly, recent approaches to stabilize HIFalpha in normoxia could have deleterious effects on proliferating tissues.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle , Fibroblasts/cytology , Fibroblasts/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Animals , Apoptosis , Cell Hypoxia , Cell Proliferation , Clone Cells , Cyclin-Dependent Kinase Inhibitor p27/metabolism , G1 Phase , Genes, Reporter , Mice , NIH 3T3 Cells , Protein Subunits/metabolism , Transgenes
20.
Am J Pathol ; 174(5): 1663-74, 2009 May.
Article in English | MEDLINE | ID: mdl-19349364

ABSTRACT

Hypoxia-inducible transcription factors (HIFs) play important roles in the response of the kidney to systemic and regional hypoxia. Degradation of HIFs is mediated by three oxygen-dependent HIF-prolyl hydroxylases (PHDs), which have partially overlapping characteristics. Although PHD inhibitors, which can induce HIFs in the presence of oxygen, are already in clinical development, little is known about the expression and regulation of these enzymes in the kidney. Therefore, we investigated the expression levels of the three PHDs in both isolated tubular cells and rat kidneys. All three PHDs were present in the kidney and were expressed predominantly in three different cell populations: (a) in distal convoluted tubules and collecting ducts (PHD1,2,3), (b) in glomerular podocytes (PHD1,3), and (c) in interstitial fibroblasts (PHD1,3). Higher levels of PHDs were found in tubular segments of the inner medulla where oxygen tensions are known to be physiologically low. PHD expression levels were unchanged in HIF-positive tubular and interstitial cells after induction by systemic hypoxia. In rat models of acute renal injury, changes in PHD expression levels were variable; while cisplatin and ischemia/reperfusion led to significant decreases in PHD2 and 3 expression levels, no changes were seen in a model of contrast media-induced nephropathy. These results implicate the non-uniform expression of HIF-regulating enzymes that modify the hypoxic response in the kidney under both regional and temporal conditions.


Subject(s)
Acute Kidney Injury/enzymology , DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immediate-Early Proteins/metabolism , Kidney/enzymology , Procollagen-Proline Dioxygenase/metabolism , Animals , Antineoplastic Agents/toxicity , Blotting, Western , Cisplatin/toxicity , Contrast Media/pharmacology , DNA-Binding Proteins/genetics , Fibroblasts/drug effects , Fibroblasts/enzymology , Gene Expression Regulation, Enzymologic/physiology , Homeodomain Proteins/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases , Immediate-Early Proteins/genetics , Immunoenzyme Techniques , Ischemia/metabolism , Ischemia/pathology , Kidney/drug effects , Kidney/injuries , Kidney Medulla/drug effects , Kidney Medulla/enzymology , Kidney Tubules/drug effects , Kidney Tubules/enzymology , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/enzymology , Male , Mice , Mice, Inbred C57BL , Oxygen/metabolism , Podocytes/drug effects , Podocytes/enzymology , Procollagen-Proline Dioxygenase/genetics , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...